1
|
Normoyle KP, Lillis KP, Egawa K, McNally MA, Paulchakrabarti M, Coudhury BP, Lau L, Shiu FH, Staley KJ. Displacement of extracellular chloride by immobile anionic constituents of the brain's extracellular matrix. J Physiol 2025; 603:353-378. [PMID: 39621449 PMCID: PMC11747837 DOI: 10.1113/jp285463] [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: 08/14/2023] [Accepted: 11/01/2024] [Indexed: 01/19/2025] Open
Abstract
GABA is the primary inhibitory neurotransmitter. Membrane currents evoked by GABAA receptor activation have uniquely small driving forces: their reversal potential (EGABA) is very close to the resting membrane potential. As a consequence, GABAA currents can flow in either direction, depending on both the membrane potential and the local intra and extracellular concentrations of the primary permeant ion, chloride (Cl). Local cytoplasmic Cl concentrations vary widely because of displacement of mobile Cl ions by relatively immobile anions. Here, we use new reporters of extracellular chloride (Cl- o) to demonstrate that Cl is displaced in the extracellular space by high and spatially heterogenous concentrations of immobile anions including sulfated glycosaminoglycans (sGAGs). Cl- o varies widely, and the mean Cl- o is only half the canonical concentration (i.e. the Cl concentration in the cerebrospinal fluid). These unexpectedly low and heterogenous Cl- o domains provide a mechanism to link the varied but highly stable distribution of sGAGs and other immobile anions in the brain's extracellular space to neuronal signal processing via the effects on the amplitude and direction of GABAA transmembrane Cl currents. KEY POINTS: Extracellular chloride concentrations in the brain were measured using a new chloride-sensitive organic fluorophore and two-photon fluorescence lifetime imaging. In vivo, the extracellular chloride concentration was spatially heterogenous and only half of the cerebrospinal fluid chloride concentration Stable displacement of extracellular chloride by immobile extracellular anions was responsible for the low extracellular chloride concentration The changes in extracellular chloride were of sufficient magnitude to alter the conductance and reversal potential of GABAA chloride currents The stability of the extracellular matrix, the impact of the component immobile anions, including sulfated glycosaminoglycans on extracellular chloride concentrations, and the consequent effect on GABAA signalling suggests a previously unappreciated mechanism for modulating GABAA signalling.
Collapse
Affiliation(s)
- Kieran P Normoyle
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kyle P Lillis
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kiyoshi Egawa
- Department of Medicine, Hokaiddo University, Sapporo, Hokaiddo, Japan
| | - Melanie A McNally
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | | | - Biswa P Coudhury
- GlycoAnalytics Core, University of California San Diego, La Jolla, CA, USA
| | - Lauren Lau
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Fu Hung Shiu
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Kevin J Staley
- Department of Neurology, Division of Child Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Bangash MA, Cubuk C, Iseppon F, Haroun R, Garcia C, Luiz AP, Arcangeletti M, Gossage SJ, Santana-Varela S, Cox JJ, Lewis MJ, Wood JN, Zhao J. Analgesic targets identified in mouse sensory neuron somata and terminal pain translatomes. Cell Rep 2024; 43:114614. [PMID: 39163201 DOI: 10.1016/j.celrep.2024.114614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 06/07/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024] Open
Abstract
The relationship between transcription and protein expression is complex. We identified polysome-associated RNA transcripts in the somata and central terminals of mouse sensory neurons in control, painful (plus nerve growth factor), and pain-free conditions (Nav1.7-null mice). The majority (98%) of translated transcripts are shared between male and female mice in both the somata and terminals. Some transcripts are highly enriched in the somata or terminals. Changes in the translatome in painful and pain-free conditions include novel and known regulators of pain pathways. Antisense knockdown of selected somatic and terminal polysome-associated transcripts that correlate with pain states diminished pain behavior. Terminal-enriched transcripts included those encoding synaptic proteins (e.g., synaptotagmin), non-coding RNAs, transcription factors (e.g., Znf431), proteins associated with transsynaptic trafficking (HoxC9), GABA-generating enzymes (Gad1 and Gad2), and neuropeptides (Penk). Thus, central terminal translation may well be a significant regulatory locus for peripheral input from sensory neurons.
Collapse
Affiliation(s)
- M Ali Bangash
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Cankut Cubuk
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Rayan Haroun
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Chloe Garcia
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Ana P Luiz
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Manuel Arcangeletti
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Samuel J Gossage
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Sonia Santana-Varela
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - James J Cox
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK
| | - Myles J Lewis
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - John N Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| | - Jing Zhao
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London WC1E 6BT, UK.
| |
Collapse
|
3
|
Channer B, Matt SM, Nickoloff-Bybel EA, Pappa V, Agarwal Y, Wickman J, Gaskill PJ. Dopamine, Immunity, and Disease. Pharmacol Rev 2023; 75:62-158. [PMID: 36757901 PMCID: PMC9832385 DOI: 10.1124/pharmrev.122.000618] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
The neurotransmitter dopamine is a key factor in central nervous system (CNS) function, regulating many processes including reward, movement, and cognition. Dopamine also regulates critical functions in peripheral organs, such as blood pressure, renal activity, and intestinal motility. Beyond these functions, a growing body of evidence indicates that dopamine is an important immunoregulatory factor. Most types of immune cells express dopamine receptors and other dopaminergic proteins, and many immune cells take up, produce, store, and/or release dopamine, suggesting that dopaminergic immunomodulation is important for immune function. Targeting these pathways could be a promising avenue for the treatment of inflammation and disease, but despite increasing research in this area, data on the specific effects of dopamine on many immune cells and disease processes remain inconsistent and poorly understood. Therefore, this review integrates the current knowledge of the role of dopamine in immune cell function and inflammatory signaling across systems. We also discuss the current understanding of dopaminergic regulation of immune signaling in the CNS and peripheral tissues, highlighting the role of dopaminergic immunomodulation in diseases such as Parkinson's disease, several neuropsychiatric conditions, neurologic human immunodeficiency virus, inflammatory bowel disease, rheumatoid arthritis, and others. Careful consideration is given to the influence of experimental design on results, and we note a number of areas in need of further research. Overall, this review integrates our knowledge of dopaminergic immunology at the cellular, tissue, and disease level and prompts the development of therapeutics and strategies targeted toward ameliorating disease through dopaminergic regulation of immunity. SIGNIFICANCE STATEMENT: Canonically, dopamine is recognized as a neurotransmitter involved in the regulation of movement, cognition, and reward. However, dopamine also acts as an immune modulator in the central nervous system and periphery. This review comprehensively assesses the current knowledge of dopaminergic immunomodulation and the role of dopamine in disease pathogenesis at the cellular and tissue level. This will provide broad access to this information across fields, identify areas in need of further investigation, and drive the development of dopaminergic therapeutic strategies.
Collapse
Affiliation(s)
- Breana Channer
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Stephanie M Matt
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Emily A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Vasiliki Pappa
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Yash Agarwal
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Jason Wickman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| | - Peter J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania (B.C., S.M.M., E.A.N-B., Y.A., J.W., P.J.G.); and The Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania (V.P.)
| |
Collapse
|
4
|
Taghizadeh A, Taghizadeh M, Yazdi MK, Zarrintaj P, Ramsey JD, Seidi F, Stadler FJ, Lee H, Saeb MR, Mozafari M. Mussel-inspired biomaterials: From chemistry to clinic. Bioeng Transl Med 2022; 7:e10385. [PMID: 36176595 PMCID: PMC9472010 DOI: 10.1002/btm2.10385] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/25/2022] [Accepted: 07/16/2022] [Indexed: 11/18/2022] Open
Abstract
After several billions of years, nature still makes decisions on its own to identify, develop, and direct the most effective material for phenomena/challenges faced. Likewise, and inspired by the nature, we learned how to take steps in developing new technologies and materials innovations. Wet and strong adhesion by Mytilidae mussels (among which Mytilus edulis-blue mussel and Mytilus californianus-California mussel are the most well-known species) has been an inspiration in developing advanced adhesives for the moist condition. The wet adhesion phenomenon is significant in designing tissue adhesives and surgical sealants. However, a deep understanding of engaged chemical moieties, microenvironmental conditions of secreted proteins, and other contributing mechanisms for outstanding wet adhesion mussels are essential for the optimal design of wet glues. In this review, all aspects of wet adhesion of Mytilidae mussels, as well as different strategies needed for designing and fabricating wet adhesives are discussed from a chemistry point of view. Developed muscle-inspired chemistry is a versatile technique when designing not only wet adhesive, but also, in several more applications, especially in the bioengineering area. The applications of muscle-inspired biomaterials in various medical applications are summarized for future developments in the field.
Collapse
Affiliation(s)
- Ali Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook UniversityCheonanRepublic of Korea
| | - Mohsen Taghizadeh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook UniversityCheonanRepublic of Korea
| | - Mohsen Khodadadi Yazdi
- Center of Excellence in ElectrochemistrySchool of Chemistry, College of Science, University of TehranTehranIran
| | - Payam Zarrintaj
- School of Chemical Engineering, Oklahoma State UniversityStillwaterOklahomaUSA
| | - Joshua D. Ramsey
- School of Chemical Engineering, Oklahoma State UniversityStillwaterOklahomaUSA
| | - Farzad Seidi
- Jiangsu Co‐Innovation Center of Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and MaterialsNanjing Forestry UniversityNanjingChina
| | - Florian J. Stadler
- College of Materials Science and EngineeringShenzhen Key Laboratory of Polymer Science and TechnologyGuangdongChina
| | - Haeshin Lee
- Department of ChemistryKorea Advanced Institute of Science and Technology (KAIST)DaejeonRepublic of Korea
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative MedicineIran University of Medical SciencesTehranIran
- Present address:
Lunenfeld‐Tanenbaum Research InstituteMount Sinai Hospital, University of TorontoToronto, ONCanada
| |
Collapse
|
5
|
Nenadov DS, Pogrmic-Majkic K, Tesic B, Kokai D, Fa Nedeljkovic S, Stanic B, Andric N. Impact of In Vitro Long-Term Low-Level DEHP Exposure on Gene Expression Profile in Human Granulosa Cells. Cells 2022; 11:cells11152304. [PMID: 35892601 PMCID: PMC9332775 DOI: 10.3390/cells11152304] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/08/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022] Open
Abstract
Here, we applied a model of long-term exposure of human granulosa cells to low environmentally relevant levels of di(2-ethylhexyl) phthalate (DEHP). This approach provides more relevant data regarding the impact of DEHP on the function of human granulosa cells. The immortalized human granulosa cells HGrC1 were exposed to 50 nM and 250 nM DEHP for four weeks. The cells were collected every week to analyze the basal granulosa cells’ functions. A portion of the DEHP-exposed cells was stimulated with forskolin (FOR) for 48 h. Steroidogenesis was investigated using ELISA, whereas DNBQ sequencing and RT-qPCR were used to analyze gene expression. The results show that steroidogenesis was not affected by DEHP exposure. RNAsequencing shows that DEHP caused week- and concentration-specific changes in various genes and functions in HGrC1. Sulfotransferase family 1A member 3 (SULT1A3) and 4 (SULT1A4), which are involved in catecholamine metabolism, were the most prominent genes affected by DEHP under both the basal and FOR-stimulated conditions in all four weeks of exposure. This study showed, for the first time, that SULT1A3 and SULT1A4 are expressed in human granulosa cells, are regulated by FOR, and are affected by low-level DEHP exposure. These data provide new insight into the relationship between DEHP, SULT1A3, and SULT1A4 in human granulosa cells.
Collapse
|
6
|
Cook I, Leyh TS. The N-Terminus of Human Sulfotransferase 2B1b─a Sterol-Sensing Allosteric Site. Biochemistry 2022; 61:843-855. [PMID: 35523209 DOI: 10.1021/acs.biochem.1c00740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Among human cytosolic sulfotransferases, SULT2B1b is highly specific for oxysterols─oxidized cholesterol derivatives, including nuclear-receptor ligands causally linked to skin and neurodegerative diseases, cancer and atherosclerosis. Sulfonation of signaling oxysterols redirects their receptor-binding functions, and controlling these functions is expected to prove valuable in disease prevention and treatment. SULT2B1b is distinct among the human SULT2 isoforms by virtue of its atypically long N-terminus, which extends 15 residues beyond the next longest N-terminus in the family. Here, in silico studies are used to predict that the N-terminal extension forms an allosteric pocket and to identify potential allosteres. One such allostere, quercetin, is used to confirm the existence of the pocket and to demonstrate that allostere binding inhibits turnover. The structure of the pocket is obtained by positioning quercetin on the enzyme, using spin-label-triangulation NMR, followed by NMR distance-constrained molecular dynamics docking. The model is confirmed using a combination of site-directed mutagenesis and initial-rate studies. Stopped-flow ligand-binding studies demonstrate that inhibition is achieved by stabilizing the closed form of the enzyme active-site cap, which encapsulates the nucleotide, slowing its release. Finally, endogenous oxysterols are shown to bind to the site in a highly selective fashion─one of the two immediate biosynthetic precursors of cholesterol (7-dehydrocholesterol) is an inhibitor, while the other (24-dehydrocholesterol) is not. These findings provide insights into the allosteric dialogue in which SULT2B1b participates in in vivo and establishes a template against which to develop isoform-specific inhibitors to control SULT2B1b biology.
Collapse
Affiliation(s)
- Ian Cook
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461-1926, United States
| | - Thomas S Leyh
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, New York 10461-1926, United States
| |
Collapse
|
7
|
Mlynarska-Cieslak A, Chrominski M, Spiewla T, Baranowski MR, Bednarczyk M, Jemielity J, Kowalska J. Fluorinated Phosphoadenosine 5'-Phosphosulfate Analogues for Continuous Sulfotransferase Activity Monitoring and Inhibitor Screening by 19F NMR Spectroscopy. ACS Chem Biol 2022; 17:661-669. [PMID: 35196009 PMCID: PMC8938925 DOI: 10.1021/acschembio.1c00978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
Sulfotransferases
(STs) are ubiquitous enzymes that participate
in a vast number of biological processes involving sulfuryl group
(SO3) transfer. 3′-phosphoadenosine 5′-phosphosulfate
(PAPS) is the universal ST cofactor, serving as the “active
sulfate” source in cells. Herein, we report the synthesis of
three fluorinated PAPS analogues that bear fluorine or trifluoromethyl
substituents at the C2 or C8 positions of adenine and their evaluation
as substitute cofactors that enable ST activity to be quantified and
real-time-monitored by fluorine-19 nuclear magnetic resonance (19F NMR) spectroscopy. Using plant AtSOT18 and human SULT1A3
as two model enzymes, we reveal that the fluorinated PAPS analogues
show complementary properties with regard to recognition by enzymes
and the working 19F NMR pH range and are attractive versatile
tools for studying STs. Finally, we developed an 19F NMR
assay for screening potential inhibitors against SULT1A3, thereby
highlighting the possible use of fluorinated PAPS analogues for the
discovery of drugs for ST-related diseases.
Collapse
Affiliation(s)
- Agnieszka Mlynarska-Cieslak
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Mikolaj Chrominski
- Centre of New Technologies University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland
| | - Tomasz Spiewla
- Centre of New Technologies University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland
| | - Marek R. Baranowski
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| | - Marcelina Bednarczyk
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
- Centre of New Technologies University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland
| | - Jacek Jemielity
- Centre of New Technologies University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland
| | - Joanna Kowalska
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, Pasteura 5, 02-093 Warsaw, Poland
| |
Collapse
|
8
|
Vuralli D, Arslan B, Topa E, de Morais AL, Gulbahar O, Ayata C, Bolay H. Migraine susceptibility is modulated by food triggers and analgesic overuse via sulfotransferase inhibition. J Headache Pain 2022; 23:36. [PMID: 35282834 PMCID: PMC8919627 DOI: 10.1186/s10194-022-01405-z] [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: 01/27/2022] [Accepted: 02/27/2022] [Indexed: 11/11/2022] Open
Abstract
Background/aim Certain constituents in migraine food triggers and non-steroidal anti-inflammatory drugs (NSAIDs) inhibit sulfotransferases (SULTs) that detoxify drugs/chemicals and play role in the metabolism of neurotransmitters. We aimed to dissect SULT1A1 modulation of CSD susceptibility and behavior in an in vivo experimental model using hesperidin, a SULT1A1 inhibitor found in citrus fruits (known migraine triggers) and mefenamic acid (SULT1A1 inhibitor), an NSAID to simulate medication overuse. Methods Hesperidin was used as SULT1A1 inhibitor found in citrus fruits, known migraine triggers and mefenamic acid (NSAID), another SULT1A1 inhibitor, was used to induce MO in rats. The groups were; 1) Hesperidin (ip) or its vehicle-DMSO (ip) 2) Chronic (4 weeks) mefenamic acid (ip) or its vehicle (ip) 3) Chronic mefenamic acid+hesperidin (ip) or DMSO (ip). CSD susceptibility was evaluated and behavioral testing was performed. SULT1A1 enzyme activity was measured in brain samples. Results Single-dose of hesperidin neither changed CSD susceptibility nor resulted in any behavioral change. Chronic mefenamic acid exposure resulted in increased CSD susceptibility, mechanical-thermal hypersensitivity, increased head shake, grooming and freezing and decreased locomotion. Single dose hesperidin administration after chronic mefenamic acid exposure resulted in increased CSD susceptibility and mechanical-thermal hypersensitivity, increased freezing and decreased locomotion. SULT1A1 enzyme activity was lower in mefenamic acid and mefenamic acid+hesperidin groups compared to their vehicles. Conclusion Mefenamic acid and hesperidin have synergistic effect in modulating CSD susceptibility and pain behavior. Sulfotransferase inhibition may be the common mechanism by which food triggers and NSAIDs modulate migraine susceptibility. Further investigations regarding human provocation studies using hesperidin in migraine patients with medication overuse are needed.
Collapse
Affiliation(s)
- Doga Vuralli
- Department of Neurology and Algology, Neuropsychiatry Center, Neuroscience and Neurotechnology Center (NÖROM), Gazi University Faculty of Medicine, Besevler, Ankara, Turkey.,Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Charlestown, USA
| | - Burak Arslan
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Besevler, Ankara, Turkey
| | - Elif Topa
- Neuropsychiatry Center, Gazi University, Besevler, Ankara, Turkey
| | - Andreia Lopes de Morais
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Charlestown, USA
| | - Ozlem Gulbahar
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, Besevler, Ankara, Turkey
| | - Cenk Ayata
- Neurovascular Research Lab, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Charlestown, USA.,Stroke Service and Neuroscience Intensive Care Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MA, Charlestown, USA
| | - Hayrunnisa Bolay
- Department of Neurology and Algology, Neuropsychiatry Center, Neuroscience and Neurotechnology Center (NÖROM), Gazi University Faculty of Medicine, Besevler, Ankara, Turkey.
| |
Collapse
|
9
|
Yang X, Sun W, Wu Q, Lin H, Lu Z, Shen X, Chen Y, Zhou Y, Huang L, Wu F, Liu F, Chu D. Excess Folic Acid Supplementation before and during Pregnancy and Lactation Alters Behaviors and Brain Gene Expression in Female Mouse Offspring. Nutrients 2021; 14:nu14010066. [PMID: 35010941 PMCID: PMC8746785 DOI: 10.3390/nu14010066] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Use of folic acid (FA) during early pregnancy protects against birth defects. However, excess FA has shown gender-specific neurodevelopmental toxicity. Previously, we fed the mice with 2.5 times the recommended amount of FA one week prior to mating and during the pregnancy and lactation periods, and detected the activated expression of Fos and related genes in the brains of weaning male offspring, as well as behavioral abnormalities in the adults. Here, we studied whether female offspring were affected by the same dosage of FA. An open field test, three-chamber social approach and social novelty test, an elevated plus-maze, rotarod test and the Morris water maze task were used to evaluate their behaviors. RNA sequencing was performed to identify differentially expressed genes in the brains. Quantitative real time-PCR (qRT-PCR) and Western blots were applied to verify the changes in gene expression. We found increased anxiety and impaired exploratory behavior, motor coordination and spatial memory in FA-exposed females. The brain transcriptome revealed 36 up-regulated and 79 down-regulated genes in their brains at weaning. The increase of Tlr1; Sult1a1; Tph2; Acacb; Etnppl; Angptl4 and Apold1, as well as a decrease of Ppara mRNA were confirmed by qRT-PCR. Among these genes; the mRNA levels of Etnppl; Angptl4andApold1 were increased in the both FA-exposed female and male brains. The elevation of Sult1a1 protein was confirmed by Western blots. Our data suggest that excess FA alteres brain gene expression and behaviors in female offspring, of which certain genes show apparent gender specificity.
Collapse
Affiliation(s)
- Xingyue Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Wenyan Sun
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Qian Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Hongyan Lin
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Zhixing Lu
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Xin Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Yongqi Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Yan Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine, Nantong University, Nantong 226001, China; (W.S.); (Z.L.); (Y.C.); (Y.Z.)
| | - Li Huang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong 226001, China; (H.L.); (F.W.)
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY 10314, USA
- Correspondence: (F.L.); (D.C.)
| | - Dandan Chu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; (X.Y.); (Q.W.); (X.S.); (L.H.)
- Correspondence: (F.L.); (D.C.)
| |
Collapse
|
10
|
Chen B, Tong X, Zhang X, Gui W, Ai G, Huang L, Ding D, Zhang J, Kang L. Sulfation modification of dopamine in brain regulates aggregative behavior of animals. Natl Sci Rev 2021; 9:nwab163. [PMID: 35530433 PMCID: PMC9072122 DOI: 10.1093/nsr/nwab163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 11/17/2022] Open
Abstract
Behavioral plasticity and the underlying neuronal plasticity represent a fundamental capacity of animals to cope with environmental stimuli. Behavioral plasticity is controlled by complex molecular networks that act under different layers of regulation. While various molecules have been found to be involved in the regulation of plastic behaviors across species, less is known about how organisms orchestrate the activity of these molecules as part of a coherent behavioral response to varying environments. Here we discover a mechanism for the regulation of animal behavioral plasticity involving molecular sulfation in the brain, a modification of substrate molecules by sulfotransferase (ST)-catalyzed addition of a sulfonate group (SO3) from an obligate donor, 3′-phosphoadenosine 5′-phosphosulfate (PAPS) to the substrates. We investigated aggregation behaviors of migratory locusts, which are well-known for extreme phase change plasticity triggered by population density. The processes of PAPS biosynthesis acted efficiently on induction of locust behavioral transition: Inhibition of PAPS synthesis solicited a behavioral shift from gregarious to solitarious states; external PAPS dosage, by contrast, promoted aggregation in solitarious locusts. Genetic or pharmacological intervention in the sulfation catalyzation resulted into pronounced solitarizing effects. Analysis of substrate-specific STs suggests a widespread involvement of sulfated neurotransmitters in the behavioral response. Dopamine in the brain was finally identified to be actively sulfate conjugated, and the sulfate conjugation enhanced the free DA-mediated behavioral aggregation. Similar results in Caenorhabditis elegans and mice indicate that sulfation may be involved more broadly in the modulation of animal aggregation. These findings reveal a general mechanism that effectively regulates animal social-like behavioral plasticity, possibly through sulfation-mediated modification of neural networks.
Collapse
Affiliation(s)
- Bing Chen
- School of Life Science, Institutes of Life Science and Green Development, Hebei University, Baoding 071002, China
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiwen Tong
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Sciences, South China Normal University, Guangzhou, 510631 China
| | - Xia Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wanying Gui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Guoming Ai
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lihua Huang
- School of Life Sciences, South China Normal University, Guangzhou, 510631 China
| | - Ding Ding
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiangxu Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Le Kang
- School of Life Science, Institutes of Life Science and Green Development, Hebei University, Baoding 071002, China
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
11
|
Silva-Adaya D, Garza-Lombó C, Gonsebatt ME. Xenobiotic transport and metabolism in the human brain. Neurotoxicology 2021; 86:125-138. [PMID: 34371026 DOI: 10.1016/j.neuro.2021.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 02/06/2023]
Abstract
Organisms have metabolic pathways responsible for eliminating endogenous and exogenous toxicants. Generally, we associate the liver par excellence as the organ in charge of detoxifying the body; however, this process occurs in all tissues, including the brain. Due to the presence of the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB), the Central Nervous System (CNS) is considered a partially isolated organ, but similar to other organs, the CNS possess xenobiotic transporters and metabolic pathways associated with the elimination of xenobiotic agents. In this review, we describe the different systems related to the detoxification of xenobiotics in the CNS, providing examples in which their association with neurodegenerative processes is suspected. The CNS detoxifying systems include carrier-mediated, active efflux and receptor-mediated transport, and detoxifying systems that include phase I and phase II enzymes, as well as those enzymes in charge of neutralizing compounds such as electrophilic agents, reactive oxygen species (ROS), and free radicals, which are products of the bioactivation of xenobiotics. Moreover, we discuss the differential expression of these systems in different regions of the CNS, showing the different detoxifying needs and the composition of each region in terms of the cell type, neurotransmitter content, and the accumulation of xenobiotics and/or reactive compounds.
Collapse
Affiliation(s)
- Daniela Silva-Adaya
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico; Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía, Mexico, 14269, Mexico
| | - Carla Garza-Lombó
- Department of Pharmacology and Toxicology, The Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 West 15th Street, NB, Indianapolis, IN, 46202, USA
| | - María E Gonsebatt
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
12
|
Baglia RA, Mills KR, Mitra K, Tutol JN, Ball D, Page KM, Kallu J, Gottipolu S, D'Arcy S, Nielsen SO, Dodani SC. An activity-based fluorescent sensor for the detection of the phenol sulfotransferase SULT1A1 in living cells. RSC Chem Biol 2021; 2:830-834. [PMID: 34212150 PMCID: PMC8190907 DOI: 10.1039/d0cb00231c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/04/2021] [Indexed: 11/21/2022] Open
Abstract
Human phenol sulfotransferases mediate the transfer of a sulfuryl moiety from the activated sulfate donor PAPS to hydroxy-containing substrates, altering substrate solubility and charge to affect phase II metabolism and cell signaling. Here, we present the development, computational modeling, in vitro enzymology, and biological application of STS-3, an activity-based fluorescent sensor for the SULT1A1 isoform.
Collapse
Affiliation(s)
- Regina A Baglia
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Kira R Mills
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Koushambi Mitra
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Jasmine N Tutol
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Darby Ball
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Kierstin M Page
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Jyothi Kallu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sriharika Gottipolu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sheena D'Arcy
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Steven O Nielsen
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| | - Sheel C Dodani
- Department of Chemistry and Biochemistry, The University of Texas at Dallas Richardson TX 75080 USA
| |
Collapse
|
13
|
Xie Y, Xie W. The Role of Sulfotransferases in Liver Diseases. Drug Metab Dispos 2020; 48:742-749. [PMID: 32587100 PMCID: PMC7469250 DOI: 10.1124/dmd.120.000074] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022] Open
Abstract
The cytosolic sulfotransferases (SULTs) are phase II conjugating enzymes that catalyze the transfer of a sulfonate group from the universal sulfate donor 3'-phosphoadenosine-5'-phosphosulfate to a nucleophilic group of their substrates to generate hydrophilic products. Sulfation has a major effect on the chemical and functional homeostasis of substrate chemicals. SULTs are widely expressed in metabolically active or hormonally responsive tissues, including the liver and many extrahepatic tissues. The expression of SULTs exhibits isoform-, tissue-, sex-, and development-specific regulations. SULTs display a broad range of substrates including xenobiotics and endobiotics. The expression of SULTs has been shown to be transcriptionally regulated by members of the nuclear receptor superfamily, such as the peroxisome proliferator-activated receptors, pregnane X receptor, constitutive androstane receptor, vitamin D receptor, liver X receptors, farnesoid X receptor, retinoid-related orphan receptors, estrogen-related receptors, and hepatocyte nuclear factor 4α These nuclear receptors can be activated by numerous xenobiotics and endobiotics, such as fatty acids, bile acids, and oxysterols, many of which are substrates of SULTs. Due to their metabolism of xenobiotics and endobiotics, SULTs and their regulations are implicated in the pathogenesis of many diseases. This review is aimed to summarize the central role of major SULTs, including the SULT1 and SULT2 subfamilies, in the pathophysiology of liver and liver-related diseases. SIGNIFICANCE STATEMENT: Sulfotransferases (SULTs) are indispensable in the homeostasis of xenobiotics and endobiotics. Knowing SULTs and their regulations are implicated in human diseases, it is hoped that genetic or pharmacological manipulations of the expression and/or activity of SULTs can be used to affect the clinical outcome of diseases.
Collapse
Affiliation(s)
- Yang Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (Y.X., W.X.) and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (W.X.)
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania (Y.X., W.X.) and Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania (W.X.)
| |
Collapse
|
14
|
Idris M, Mitchell DJ, Gordon R, Sidharthan NP, Butcher NJ, Minchin RF. Interaction of the Brain-Selective Sulfotransferase SULT4A1 with Other Cytosolic Sulfotransferases: Effects on Protein Expression and Function. Drug Metab Dispos 2020; 48:337-344. [PMID: 32152050 DOI: 10.1124/dmd.119.089714] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Sulfotransferase (SULT) 4A1 is a brain-selective sulfotransferase-like protein that has recently been shown to be essential for normal neuronal development in mice. In the present study, SULT4A1 was found to colocalize with SULT1A1/3 in human brain neurons. Using immunoprecipitation, SULT4A1 was shown to interact with both SULT1A1 and SULT1A3 when expressed in human cells. Mutation of the conserved dimerization motif located in the C terminus of the sulfotransferases prevented this interaction. Both ectopically expressed and endogenous SULT4A1 decreased SULT1A1/3 protein levels in neuronal cells, and this was also prevented by mutation of the dimerization motif. During differentiation of neuronal SH-SY5Y cells, there was a loss in SULT1A1/3 protein but an increase in SULT4A1 protein. This resulted in an increase in the toxicity of dopamine, a substrate for SULT1A3. Inhibition of SULT4A1 using small interference RNA abrogated the loss in SULT1A1/3 and reversed dopamine toxicity. These results show a reciprocal relationship between SULT4A1 and the other sulfotransferases, suggesting that it may act as a chaperone to control the expression of SULT1A1/3 in neuronal cells. SIGNIFICANCE STATEMENT: The catalytically inactive sulfotransferase (SULT) 4A1 may regulate the function of other SULTs by interacting with them via a conserved dimerization motif. In neuron-like cells, SULT4A1 is able to modulate dopamine toxicity by interacting with SULT1A3, potentially decreasing the metabolism of dopamine.
Collapse
Affiliation(s)
- Misgana Idris
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Deanne J Mitchell
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Richard Gordon
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Neelima P Sidharthan
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Neville J Butcher
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| | - Rodney F Minchin
- School of Biomedical Sciences, University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
15
|
Chtioui H, Sadowski SM, Winzeler B, Tschopp O, Grouzmann E, Abid K. High concentration of plasma methoxytyramine: dopamine-producing tumour or Parkinson's disease therapy? Ann Clin Biochem 2019; 56:466-471. [PMID: 30974961 DOI: 10.1177/0004563219835263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background Levodopa (L-DOPA) provided to patients with Parkinson’s disease causes an increase in dopamine and methoxytyramine blood concentration which may lead to erroneous diagnosis of dopamine-producing tumours based on a plasma fractionated metanephrines and methoxytyramine assay. Considering that oral L-DOPA is mainly transformed in the gut wall into dopamine and methoxytyramine, we hypothesize that patients treated with L-DOPA produce predominantly sulphated methoxytyramine, whereas dopamine-producing tumours, devoid of sulfotransferase, will secrete free methoxytyramine. These metabolic differences may allow for discrimination between the two groups of patients through methoxytyramine plasma concentration. Methods We retrospectively investigated a cohort of 16 patients with a dopamine-secreting pheochromocytoma or paraganglioma and 22 patients treated for Parkinson’s disease to see whether the metabolic ratio of free and sulphated methoxytyramine differs. Results Receiver operating characteristic curve analysis indicates an absolute separation between the two groups when using a cut-off of free/total methoxytyramine (sum of free and sulphated methoxytyramine) ratio of 0.0059, corresponding to a free methoxytyramine fraction of 0.59% ( P < 0.0001, AUC 1.0 indicating 100% sensitivity and specificity). Conclusion Dopamine secreted by tumours and exogenous dopamine (from Parkinson’s disease treatment) follow different metabolic pathways. We observed that free/total methoxytyramine ratio may be a useful tool in distinguishing between patients with a dopamine-secreting tumour from patients treated with L-DOPA when clinical information is incomplete or lacking.
Collapse
Affiliation(s)
- Haithem Chtioui
- 1 Service of Clinical Pharmacology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Samira M Sadowski
- 2 Service of Thoracic and Endocrine Surgery, University Hospital of Geneva, Geneva, Switzerland
| | - Bettina Winzeler
- 3 Department of Endocrinology, University Hospital of Basel, Basel, Switzerland
| | - Oliver Tschopp
- 4 Department of Endocrinology, University Hospital of Zurich, Zurich, Switzerland
| | - Eric Grouzmann
- 1 Service of Clinical Pharmacology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Karim Abid
- 1 Service of Clinical Pharmacology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
16
|
Bairam AF, Rasool MI, Alherz FA, Abunnaja MS, El Daibani AA, Gohal SA, Kurogi K, Sakakibara Y, Suiko M, Liu MC. Sulfation of catecholamines and serotonin by SULT1A3 allozymes. Biochem Pharmacol 2018. [PMID: 29524394 DOI: 10.1016/j.bcp.2018.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Previous studies have demonstrated the involvement of sulfoconjugation in the metabolism of catecholamines and serotonin. The current study aimed to clarify the effects of single nucleotide polymorphisms (SNPs) of human SULT1A3 and SULT1A4 genes on the enzymatic characteristics of the sulfation of dopamine, epinephrine, norepinephrine and serotonin by SULT1A3 allozymes. Following a comprehensive search of different SULT1A3 and SULT1A4 genotypes, twelve non-synonymous (missense) coding SNPs (cSNPs) of SULT1A3/SULT1A4 were identified. cDNAs encoding the corresponding SULT1A3 allozymes, packaged in pGEX-2T vector were generated by site-directed mutagenesis. SULT1A3 allozymes were expressed, and purified. Purified SULT1A3 allozymes exhibited differential sulfating activity toward catecholamines and serotonin. Kinetic analyses demonstrated differences in both substrate affinity and catalytic efficiency of the SULT1A3 allozymes. Collectively, these findings provide useful information relevant to the differential metabolism of dopamine, epinephrine, norepinephrine and serotonin through sulfoconjugation in individuals having different SULT1A3/SULT1A4 genotypes.
Collapse
Affiliation(s)
- Ahsan F Bairam
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Department of Pharmacology, College of Pharmacy, University of Kufa, Najaf, Iraq
| | - Mohammed I Rasool
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Department of Pharmacology, College of Pharmacy, University of Karbala, Karbala, Iraq
| | - Fatemah A Alherz
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Maryam S Abunnaja
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Amal A El Daibani
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Saud A Gohal
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA
| | - Katsuhisa Kurogi
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA; Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Yoichi Sakakibara
- Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Masahito Suiko
- Biochemistry and Applied Biosciences, University of Miyazaki, Miyazaki 889-2192, Japan
| | - Ming-Cheh Liu
- Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH 43614, USA.
| |
Collapse
|
17
|
Walton E, Cecil CA, Suderman M, Liu J, Turner JA, Calhoun V, Ehrlich S, Relton CL, Barker ED. Longitudinal epigenetic predictors of amygdala:hippocampus volume ratio. J Child Psychol Psychiatry 2017; 58:1341-1350. [PMID: 28480579 PMCID: PMC5677591 DOI: 10.1111/jcpp.12740] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND The ratio between amygdala:hippocampal (AH) volume has been associated with multiple psychiatric problems, including anxiety and aggression. Yet, little is known about its biological underpinnings. Here, we used a methylome-wide approach to test (a) whether DNA methylation in early life (birth, age 7) prospectively associates with total AH volume ratio in early adulthood, and (b) whether significant DNA methylation markers are influenced by prenatal risk factors. METHODS Analyses were based on a subsample (n = 109 males) from the Avon Longitudinal Study of Parents and Children, which included measures of prenatal risk, DNA methylation (Infinium Illumina 450k), T1-weighted brain scans and psychopathology in early adulthood (age 18-21). Amygdala and hippocampus measures were derived using Freesurfer 5.3.0. Methylation markers related to AH volume ratio across time were identified using longitudinal multilevel modeling. RESULTS Amygdala:hippocampal volume ratio correlated positively with age 18 psychosis-like symptoms (p = .007). Methylation of a probe in the gene SP6 associated longitudinally with (a) higher AH volume ratio (FDR q-value = .01) and (b) higher stressful life events during pregnancy (p = .046). SP6 is expressed in the hippocampus and amygdala and has been implicated in cognitive decline in Alzheimer's disease. The association between SP6 DNA methylation, AH volume ratio and psychopathology was replicated in an independent dataset of 101 patients with schizophrenia and 111 healthy controls. CONCLUSIONS Our findings suggest that epigenetic alterations in genes implicated in neurodevelopment may contribute to a brain-based biomarker of psychopathology.
Collapse
Affiliation(s)
- Esther Walton
- Department of PsychologyInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- Department of PsychologyGeorgia State UniversityAtlantaGAUSA
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
| | - Charlotte A.M. Cecil
- Department of PsychologyInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Matthew Suderman
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
| | - Jingyu Liu
- The Mind Research NetworkAlbuquerqueNMUSA
- Department of Electrical EngineeringUniversity of New MexicoAlbuquerqueNMUSA
| | | | - Vince Calhoun
- The Mind Research NetworkAlbuquerqueNMUSA
- Department of Electrical EngineeringUniversity of New MexicoAlbuquerqueNMUSA
| | - Stefan Ehrlich
- Division of Psychological and Social Medicine and Developmental NeurosciencesFaculty of MedicineTU DresdenDresdenGermany
| | - Caroline L. Relton
- Medical Research Council Integrative Epidemiology UnitUniversity of BristolBristolUK
| | - Edward D. Barker
- Department of PsychologyInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
18
|
Cook I, Wang T, Leyh TS. Tetrahydrobiopterin regulates monoamine neurotransmitter sulfonation. Proc Natl Acad Sci U S A 2017; 114:E5317-E5324. [PMID: 28630292 PMCID: PMC5502633 DOI: 10.1073/pnas.1704500114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Monoamine neurotransmitters are among the hundreds of signaling small molecules whose target interactions are switched "on" and "off" via transfer of the sulfuryl-moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfate) to the hydroxyls and amines of their scaffolds. These transfer reactions are catalyzed by a small family of broad-specificity enzymes-the human cytosolic sulfotransferases (SULTs). The first structure of a SULT allosteric-binding site (that of SULT1A1) has recently come to light. The site is conserved among SULT1 family members and is promiscuous-it binds catechins, a naturally occurring family of flavanols. Here, the catechin-binding site of SULT1A3, which sulfonates monoamine neurotransmitters, is modeled on that of 1A1 and used to screen in silico for endogenous metabolite 1A3 allosteres. Screening predicted a single high-affinity allostere, tetrahydrobiopterin (THB), an essential cofactor in monoamine neurotransmitter biosynthesis. THB is shown to bind and inhibit SULT1A3 with high affinity, 23 (±2) nM, and to bind weakly, if at all, to the four other major SULTs found in brain and liver. The structure of the THB-bound binding site is determined and confirms that THB binds the catechin site. A structural comparison of SULT1A3 with SULT1A1 (its immediate evolutionary progenitor) reveals how SULT1A3 acquired high affinity for THB and that the majority of residue changes needed to transform 1A1 into 1A3 are clustered at the allosteric and active sites. Finally, sequence records reveal that the coevolution of these sites played an essential role in the evolution of simian neurotransmitter metabolism.
Collapse
Affiliation(s)
- Ian Cook
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461-1926
| | - Ting Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461-1926
| | - Thomas S Leyh
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461-1926
| |
Collapse
|
19
|
Butcher NJ, Horne MK, Mellick GD, Fowler CJ, Masters CL, Minchin RF. Sulfotransferase 1A3/4 copy number variation is associated with neurodegenerative disease. THE PHARMACOGENOMICS JOURNAL 2017; 18:209-214. [DOI: 10.1038/tpj.2017.4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 12/12/2016] [Accepted: 01/17/2017] [Indexed: 12/14/2022]
|
20
|
Advances in drug metabolism and pharmacogenetics research in Australia. Pharmacol Res 2017; 116:7-19. [DOI: 10.1016/j.phrs.2016.12.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 12/07/2016] [Accepted: 12/08/2016] [Indexed: 01/04/2023]
|
21
|
Rote JC, Malkowski SN, Cochrane CS, Bailey GE, Brown NS, Cafiero M, Peterson LW. Catechol reactivity: Synthesis of dopamine derivatives substituted at the 6-position. SYNTHETIC COMMUN 2017. [DOI: 10.1080/00397911.2016.1269350] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
| | | | | | | | - Noah S. Brown
- Department of Chemistry, Rhodes College, Memphis, TN, USA
| | | | | |
Collapse
|
22
|
Suiko M, Kurogi K, Hashiguchi T, Sakakibara Y, Liu MC. Updated perspectives on the cytosolic sulfotransferases (SULTs) and SULT-mediated sulfation. Biosci Biotechnol Biochem 2016; 81:63-72. [PMID: 27649811 DOI: 10.1080/09168451.2016.1222266] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The cytosolic sulfotransferases (SULTs) are Phase II detoxifying enzymes that mediate the sulfate conjugation of numerous xenobiotic molecules. While the research on the SULTs has lagged behind the research on Phase I cytochrome P-450 enzymes and other Phase II conjugating enzymes, it has gained more momentum in recent years. This review aims to summarize information obtained in several fronts of the research on the SULTs, including the range of the SULTs in different life forms, concerted actions of the SULTs and other Phase II enzymes, insights into the structure-function relationships of the SULTs, regulation of SULT expression and activity, developmental expression of SULTs, as well as the use of a zebrafish model for studying the developmental pharmacology/toxicology.
Collapse
Affiliation(s)
- Masahito Suiko
- a Faculty of Agriculture, Department of Biochemistry and Applied Biosciences , University of Miyazaki , Miyazaki , Japan
| | - Katsuhisa Kurogi
- a Faculty of Agriculture, Department of Biochemistry and Applied Biosciences , University of Miyazaki , Miyazaki , Japan.,b Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences , University of Toledo Health Science Campus , Toledo , OH , USA
| | - Takuyu Hashiguchi
- a Faculty of Agriculture, Department of Biochemistry and Applied Biosciences , University of Miyazaki , Miyazaki , Japan
| | - Yoichi Sakakibara
- a Faculty of Agriculture, Department of Biochemistry and Applied Biosciences , University of Miyazaki , Miyazaki , Japan
| | - Ming-Cheh Liu
- b Department of Pharmacology, College of Pharmacy and Pharmaceutical Sciences , University of Toledo Health Science Campus , Toledo , OH , USA
| |
Collapse
|
23
|
Meisel JD, Kim DH. Inhibition of Lithium-Sensitive Phosphatase BPNT-1 Causes Selective Neuronal Dysfunction in C. elegans. Curr Biol 2016; 26:1922-8. [PMID: 27397889 DOI: 10.1016/j.cub.2016.05.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/09/2016] [Accepted: 05/19/2016] [Indexed: 11/26/2022]
Abstract
Lithium has been a mainstay for the treatment of bipolar disorder, yet the molecular mechanisms underlying its action remain enigmatic. Bisphosphate 3'-nucleotidase (BPNT-1) is a lithium-sensitive phosphatase that catalyzes the breakdown of cytosolic 3'-phosphoadenosine 5'-phosphate (PAP), a byproduct of sulfation reactions utilizing the universal sulfate group donor 3'-phosphoadenosine 5'-phosphosulfate (PAPS) [1-3]. Loss of BPNT-1 leads to the toxic accumulation of PAP in yeast and non-neuronal cell types in mice [4, 5]. Intriguingly, BPNT-1 is expressed throughout the mammalian brain [4], and it has been hypothesized that inhibition of BPNT-1 could contribute to the effects of lithium on behavior [5]. Here, we show that loss of BPNT-1 in Caenorhabditis elegans results in the selective dysfunction of two neurons, the bilaterally symmetric pair of ASJ chemosensory neurons. As a result, BPNT-1 mutants are defective in behaviors dependent on the ASJ neurons, such as dauer exit and pathogen avoidance. Acute treatment with lithium also causes dysfunction of the ASJ neurons, and we show that this effect is reversible and mediated specifically through inhibition of BPNT-1. Finally, we show that the selective effect of lithium on the nervous system is due in part to the limited expression of the cytosolic sulfotransferase SSU-1 in the ASJ neuron pair. Our data suggest that lithium, through inhibition of BPNT-1 in the nervous system, can cause selective toxicity to specific neurons, resulting in corresponding effects on behavior of C. elegans.
Collapse
Affiliation(s)
- Joshua D Meisel
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Dennis H Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA.
| |
Collapse
|
24
|
Heteroreceptor Complexes Formed by Dopamine D 1, Histamine H 3, and N-Methyl-D-Aspartate Glutamate Receptors as Targets to Prevent Neuronal Death in Alzheimer's Disease. Mol Neurobiol 2016; 54:4537-4550. [PMID: 27370794 DOI: 10.1007/s12035-016-9995-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder causing progressive memory loss and cognitive dysfunction. Anti-AD strategies targeting cell receptors consider them as isolated units. However, many cell surface receptors cooperate and physically contact each other forming complexes having different biochemical properties than individual receptors. We here report the discovery of dopamine D1, histamine H3, and N-methyl-D-aspartate (NMDA) glutamate receptor heteromers in heterologous systems and in rodent brain cortex. Heteromers were detected by co-immunoprecipitation and in situ proximity ligation assays (PLA) in the rat cortex where H3 receptor agonists, via negative cross-talk, and H3 receptor antagonists, via cross-antagonism, decreased D1 receptor agonist signaling determined by ERK1/2 or Akt phosphorylation, and counteracted D1 receptor-mediated excitotoxic cell death. Both D1 and H3 receptor antagonists also counteracted NMDA toxicity suggesting a complex interaction between NMDA receptors and D1-H3 receptor heteromer function. Likely due to heteromerization, H3 receptors act as allosteric regulator for D1 and NMDA receptors. By bioluminescence resonance energy transfer (BRET), we demonstrated that D1 or H3 receptors form heteromers with NR1A/NR2B NMDA receptor subunits. D1-H3-NMDA receptor complexes were confirmed by BRET combined with fluorescence complementation. The endogenous expression of complexes in mouse cortex was determined by PLA and similar expression was observed in wild-type and APP/PS1 mice. Consistent with allosteric receptor-receptor interactions within the complex, H3 receptor antagonists reduced NMDA or D1 receptor-mediated excitotoxic cell death in cortical organotypic cultures. Moreover, H3 receptor antagonists reverted the toxicity induced by ß1-42-amyloid peptide. Thus, histamine H3 receptors in D1-H3-NMDA heteroreceptor complexes arise as promising targets to prevent neurodegeneration.
Collapse
|
25
|
Shao X, Li J, Wang S, Chen G, Xu J, Ji X, Li L, Lu W, Zhou T. Exogenous dopamine induces dehydroepiandrosterone sulfotransferase (rSULT2A1) in rat liver and changes the pharmacokinetic profile of moxifloxacin in rats. Drug Metab Pharmacokinet 2015; 30:97-104. [DOI: 10.1016/j.dmpk.2014.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/17/2014] [Accepted: 10/06/2014] [Indexed: 01/11/2023]
|
26
|
Bigler DJ, Peterson LW, Cafiero M. Effects of implicit solvent and relaxed amino acid side chains on the MP2 and DFT calculations of ligand–protein structure and electronic interaction energies of dopaminergic ligands in the SULT1A3 enzyme active site. COMPUT THEOR CHEM 2015. [DOI: 10.1016/j.comptc.2014.10.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
27
|
El-Gowelli HM, El-Mas MM. Central modulation of cyclosporine-induced hypertension. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:351-61. [DOI: 10.1007/s00210-014-1074-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 11/18/2014] [Indexed: 12/25/2022]
|