1
|
Watkins OC, Yong HEJ, Sharma N, Chan SY. A review of the role of inositols in conditions of insulin dysregulation and in uncomplicated and pathological pregnancy. Crit Rev Food Sci Nutr 2020; 62:1626-1673. [PMID: 33280430 DOI: 10.1080/10408398.2020.1845604] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inositols, a group of 6-carbon polyols, are highly bioactive molecules derived from diet and endogenous synthesis. Inositols and their derivatives are involved in glucose and lipid metabolism and participate in insulin-signaling, with perturbations in inositol processing being associated with conditions involving insulin resistance, dysglycemia and dyslipidemia such as polycystic ovary syndrome and diabetes. Pregnancy is similarly characterized by substantial and complex changes in glycemic and lipidomic regulation as part of maternal adaptation and is also associated with physiological alterations in inositol processing. Disruptions in maternal adaptation are postulated to have a critical pathophysiological role in pregnancy complications such as gestational diabetes and pre-eclampsia. Inositol supplementation has shown promise as an intervention for the alleviation of symptoms in conditions of insulin resistance and for gestational diabetes prevention. However, the mechanisms behind these affects are not fully understood. In this review, we explore the role of inositols in conditions of insulin dysregulation and in pregnancy, and identify priority areas for research. We particularly examine the role and function of inositols within the maternal-placental-fetal axis in both uncomplicated and pathological pregnancies. We also discuss how inositols may mediate maternal-placental-fetal cross-talk, and regulate fetal growth and development, and suggest that inositols play a vital role in promoting healthy pregnancy.
Collapse
Affiliation(s)
- Oliver C Watkins
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hannah E J Yong
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| | - Neha Sharma
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shiao-Yng Chan
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research, Singapore, Singapore
| |
Collapse
|
2
|
Abbott GW. KCNQs: Ligand- and Voltage-Gated Potassium Channels. Front Physiol 2020; 11:583. [PMID: 32655402 PMCID: PMC7324551 DOI: 10.3389/fphys.2020.00583] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022] Open
Abstract
Voltage-gated potassium (Kv) channels in the KCNQ (Kv7) family are essential features of a broad range of excitable and non-excitable cell types and are found in organisms ranging from Hydra vulgaris to Homo sapiens. Although they are firmly in the superfamily of S4 domain-bearing voltage-sensing ion channels, KCNQ channels are highly sensitive to a range of endogenous and exogenous small molecules that act directly on the pore, the voltage-sensing domain, or the interface between the two. The focus of this review is regulation of KCNQs by direct binding of neurotransmitters and metabolites from both animals and plants and the role of the latter in the effects of plants consumed for food and as traditional folk medicines. The conceptual question arises: Are KCNQs voltage-gated channels that are also sensitive to ligands or ligand-gated channels that are also sensitive to voltage?
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
3
|
|
4
|
Yang XL, Wang X, Peng BW. NFAT5 Has a Job in the Brain. Dev Neurosci 2018; 40:289-300. [PMID: 30391952 DOI: 10.1159/000493789] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/14/2018] [Indexed: 11/19/2022] Open
Abstract
Nuclear factor of activated T cells 5 (NFAT5) has recently been classified as a new member of the Rel family. In addition, there are 5 more well-defined members (NF-κB and NFAT1-4) in the Rel family, which participate in regulating the expression of immune and inflammatory response-related genes. NFAT5 was initially identified in renal medullary cells where it regulated the expression of osmoprotective-related genes during the osmotic response. Many studies have demonstrated that NFAT5 is highly expressed in the nuclei of neurons in fetal and adult brains. Additionally, its expression is approximately 10-fold higher in fetal brains. With the development of detection technologies (laser scanning confocal microscopy, transgene technology, etc.), recent studies suggest that NFAT5 is also expressed in glial cells and plays a more diverse functional role. This article aims to summarize the current knowledge regarding the expression of NFAT5, its regulation of activation, and varied biological functions in the brain.
Collapse
Affiliation(s)
- Xing-Liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xin Wang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China,
| |
Collapse
|
5
|
Saiardi A, Mudge AW. Lithium and fluoxetine regulate the rate of phosphoinositide synthesis in neurons: a new view of their mechanisms of action in bipolar disorder. Transl Psychiatry 2018; 8:175. [PMID: 30171184 PMCID: PMC6119186 DOI: 10.1038/s41398-018-0235-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022] Open
Abstract
Lithium is widely used to treat bipolar disorder, but its primary mechanism of action is uncertain. One proposal has been that lithium's ability to inhibit the enzyme inositol monophosphatase (IMPase) reduces the supply of recycled inositol used for membrane phosphoinositide (PIns) synthesis. This 28-year-old hypothesis is still widely debated, however, largely because total levels of PIns in brain or in cultured neurons do not decrease after lithium treatment. Here we use mature cultured cortical neurons to show that, although lithium has little effect on steady-state levels of either inositol or PIns, it markedly inhibits the rate of PIns synthesis. Moreover, we show that rapid synthesis of membrane PIns preferentially uses inositol newly imported from the extracellular space. Unexpectedly, we also find that the antidepressant drug fluoxetine (FLUO: Prozac) stimulates the rate of PIns synthesis. The convergence of both lithium and FLUO in regulating the rate of synthesis of PIns in opposite ways highlights PIns turnover in neurons as a potential new drug target, as well as for understanding mood control in BD. Our results also indicate new avenues for investigation of how neurons regulate their supply of inositol.
Collapse
Affiliation(s)
- Adolfo Saiardi
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Anne W. Mudge
- 0000000121901201grid.83440.3bMedical Research Council Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT UK
| |
Collapse
|
6
|
Maeba R, Araki A, Fujiwara Y. Serum Ethanolamine Plasmalogen and Urine Myo-Inositol as Cognitive Decline Markers. Adv Clin Chem 2018; 87:69-111. [PMID: 30342713 DOI: 10.1016/bs.acc.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent studies have suggested that metabolic disorders, particularly type 2 diabetes mellitus (T2DM), and dementia, including Alzheimer's disease (AD), were linked at the clinical and molecular levels. Brain insulin deficiency and resistance may be key events in AD pathology mechanistically linking AD to T2DM. Ethanolamine plasmalogens (PlsEtns) are abundant in the brain and play essential roles in neuronal function and myelin formation. As such, PlsEtn deficiency may be pathologically relevant in some neurodegenerative disorders such as AD. Decreased brain PlsEtn associated with dementia may reflect serum PlsEtn deficiency. We hypothesized that myo-inositol plays a role in myelin formation through its facilitation of PlsEtn biosynthesis. Excessive urinary myo-inositol (UMI) loss would likely result in PlsEtn deficiency potentially leading to demyelinating diseases such as dementia. Accordingly, measurement of both serum PlsEtn and baseline UMI excretion could improve the detection of cognitive impairment (CI) in a more specific and reliable manner. To verify our hypothesis, we conducted a clinical observational study of memory clinic outpatients (MCO) and cognitively normal elderly (NE) for nearly 4.5years. We demonstrated that serum PlsEtn concentration associated with UMI excretion was useful for predicting advancing dementia in patients with mild CI. Because hyperglycemia and associated insulin resistance might be a leading cause of increased baseline UMI excretion, serum PlsEtn quantitation would be useful in detecting CI among the elderly with hyperglycemia. Our findings suggest that myo-inositol is a novel candidate molecule linking T2DM to AD.
Collapse
Affiliation(s)
- Ryouta Maeba
- Department of Biochemistry, Teikyo University School of Medicine, Tokyo, Japan
| | - Atsushi Araki
- Department of Diabetes, Metabolism and Endocrinology, Tokyo Metropolitan Geriatric Hospital, Tokyo, Japan
| | - Yoshinori Fujiwara
- Research Team for Social Participation and Community Health, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
7
|
The KCNE2 K⁺ channel regulatory subunit: Ubiquitous influence, complex pathobiology. Gene 2015; 569:162-72. [PMID: 26123744 DOI: 10.1016/j.gene.2015.06.061] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/15/2015] [Accepted: 06/23/2015] [Indexed: 02/05/2023]
Abstract
The KCNE single-span transmembrane subunits are encoded by five-member gene families in the human and mouse genomes. Primarily recognized for co-assembling with and functionally regulating the voltage-gated potassium channels, the broad influence of KCNE subunits in mammalian physiology belies their small size. KCNE2 has been widely studied since we first discovered one of its roles in the heart and its association with inherited and acquired human Long QT syndrome. Since then, physiological analyses together with human and mouse genetics studies have uncovered a startling array of functions for KCNE2, in the heart, stomach, thyroid and choroid plexus. The other side of this coin is the variety of interconnected disease manifestations caused by KCNE2 disruption, involving both excitable cells such as cardiomyocytes, and non-excitable, polarized epithelia. Kcne2 deletion in mice has been particularly instrumental in illustrating the potential ramifications within a monogenic arrhythmia syndrome, with removal of one piece revealing the unexpected complexity of the puzzle. Here, we review current knowledge of the function and pathobiology of KCNE2.
Collapse
|
8
|
Abbott GW, Tai KK, Neverisky DL, Hansler A, Hu Z, Roepke TK, Lerner DJ, Chen Q, Liu L, Zupan B, Toth M, Haynes R, Huang X, Demirbas D, Buccafusca R, Gross SS, Kanda VA, Berry GT. KCNQ1, KCNE2, and Na+-coupled solute transporters form reciprocally regulating complexes that affect neuronal excitability. Sci Signal 2014; 7:ra22. [PMID: 24595108 DOI: 10.1126/scisignal.2005025] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Na(+)-coupled solute transport is crucial for the uptake of nutrients and metabolic precursors, such as myo-inositol, an important osmolyte and precursor for various cell signaling molecules. We found that various solute transporters and potassium channel subunits formed complexes and reciprocally regulated each other in vitro and in vivo. Global metabolite profiling revealed that mice lacking KCNE2, a K(+) channel β subunit, showed a reduction in myo-inositol concentration in cerebrospinal fluid (CSF) but not in serum. Increased behavioral responsiveness to stress and seizure susceptibility in Kcne2(-/-) mice were alleviated by injections of myo-inositol. Suspecting a defect in myo-inositol transport, we found that KCNE2 and KCNQ1, a voltage-gated potassium channel α subunit, colocalized and coimmunoprecipitated with SMIT1, a Na(+)-coupled myo-inositol transporter, in the choroid plexus epithelium. Heterologous coexpression demonstrated that myo-inositol transport by SMIT1 was augmented by coexpression of KCNQ1 but was inhibited by coexpression of both KCNQ1 and KCNE2, which form a constitutively active, heteromeric K(+) channel. SMIT1 and the related transporter SMIT2 were also inhibited by a constitutively active mutant form of KCNQ1. The activities of KCNQ1 and KCNQ1-KCNE2 were augmented by SMIT1 and the glucose transporter SGLT1 but were suppressed by SMIT2. Channel-transporter signaling complexes may be a widespread mechanism to facilitate solute transport and electrochemical crosstalk.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- 1Bioelectricity Laboratory, Department of Pharmacology, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Rae CD. A Guide to the Metabolic Pathways and Function of Metabolites Observed in Human Brain 1H Magnetic Resonance Spectra. Neurochem Res 2013; 39:1-36. [PMID: 24258018 DOI: 10.1007/s11064-013-1199-5] [Citation(s) in RCA: 338] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Revised: 11/08/2013] [Accepted: 11/11/2013] [Indexed: 12/20/2022]
|
10
|
Shin JA, Kwon HM, Han KH, Lee HY. TonEBP and SMIT expression in human placenta. Anat Cell Biol 2012; 45:155-9. [PMID: 23094203 PMCID: PMC3472141 DOI: 10.5115/acb.2012.45.3.155] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 07/13/2012] [Accepted: 08/09/2012] [Indexed: 01/30/2023] Open
Abstract
Tonicity-responsive enhancer binding protein (TonEBP) is a signal transcription factor of transporters such as sodium-myo-inositol cotransporter (SMIT), aldose reductase. TonEBP has a variety of functions such as control of intracellular osmolytes and immunomodulating. It is known that TonEBP is abundant in the placenta, but location and function aren't known. The aim of this study is to describe the localization of TonEBP in the placenta. We assayed the immunohistochemistry of TonEBP and performed in situ hybridization of SMIT in normal human full term placenta. In normal human full term placenta, TonEBP was in villous trophoblasts, extravillous trophoblasts and some endothelial cells. The result of the in situ hybridization of SMIT was similar to that of immunohistochemistry of TonEBP. Neither TonEBP nor SMIT was present in TonEBP knockout mouse placenta. This shows TonEBP is a key factor in SMIT transcription. TonEBP may play an important role in transporting of inositol to fetus in placenta.
Collapse
Affiliation(s)
- Jung-A Shin
- Department of Anatomy, Ewha Womans University School of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
11
|
Ho HTB, Dahlin A, Wang J. Expression Profiling of Solute Carrier Gene Families at the Blood-CSF Barrier. Front Pharmacol 2012; 3:154. [PMID: 22936914 PMCID: PMC3426838 DOI: 10.3389/fphar.2012.00154] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 08/01/2012] [Indexed: 12/12/2022] Open
Abstract
The choroid plexus (CP) is a highly vascularized tissue in the brain ventricles and acts as the blood-cerebrospinal fluid (CSF) barrier (BCSFB). A main function of the CP is to secrete CSF, which is accomplished by active transport of small ions and water from the blood side to the CSF side. The CP also supplies the brain with certain nutrients, hormones, and metal ions, while removing metabolites and xenobiotics from the CSF. Numerous membrane transporters are expressed in the CP in order to facilitate the solute exchange between the blood and the CSF. The solute carrier (SLC) superfamily represents a major class of transporters in the CP that constitutes the molecular mechanisms for CP function. Recently, we systematically and quantitatively examined Slc gene expression in 20 anatomically comprehensive brain areas in the adult mouse brain using high-quality in situ hybridization data generated by the Allen Brain Atlas. Here we focus our analysis on Slc gene expression at the BCSFB using previously obtained data. Of the 252 Slc genes present in the mouse brain, 202 Slc genes were found at detectable levels in the CP. Unsupervised hierarchical cluster analysis showed that the CP Slc gene expression pattern is substantially different from the other 19 analyzed brain regions. The majority of the Slc genes in the CP are expressed at low to moderate levels, whereas 28 Slc genes are present in the CP at the highest levels. These highly expressed Slc genes encode transporters involved in CSF secretion, energy production, and transport of nutrients, hormones, neurotransmitters, sulfate, and metal ions. In this review, the functional characteristics and potential importance of these Slc transporters in the CP are discussed, with particular emphasis on their localization and physiological functions at the BCSFB.
Collapse
Affiliation(s)
- Horace T B Ho
- Department of Pharmaceutics, University of Washington Seattle, WA, USA
| | | | | |
Collapse
|
12
|
Investigation of the H+–myo-inositol transporter (HMIT) as a neuronal regulator of phosphoinositide signalling. Biochem Soc Trans 2009; 37:1139-43. [DOI: 10.1042/bst0371139] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Phosphoinositide signalling regulates a series of important neuronal processes that are thought to be altered in mood disorders. Furthermore, mood-stabilizing drugs inhibit key enzymes that regulate phosphoinositide production and alter neuronal growth cone morphology in an inositol-reversible manner. Inositol is taken up by neurons from the extracellular fluid, presumably via membrane transporters; it can also be synthesized by the enzyme MIP-synthase (myo-inositol-1-phosphate synthase) and, in addition, it is generated by inositol phospholipid hydrolysis. The neuronal-specific HMIT (H+–myo-inositol transporter) represents a potential regulator of inositol signalling in neurons that warrants further investigation.
Collapse
|
13
|
Brighina E, Bresolin N, Pardi G, Rango M. Human fetal brain chemistry as detected by proton magnetic resonance spectroscopy. Pediatr Neurol 2009; 40:327-42. [PMID: 19380068 DOI: 10.1016/j.pediatrneurol.2008.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2008] [Revised: 11/03/2008] [Accepted: 11/19/2008] [Indexed: 11/25/2022]
Abstract
Magnetic resonance spectroscopy represents an invaluable tool for the in vivo study of brain development at the chemistry level. Whereas magnetic resonance spectroscopy has received wide attention in pediatric and adult settings, only a few studies were performed on the human fetal brain. They revealed changes occurring throughout gestation in the levels of the main metabolites detected by proton magnetic resonance spectroscopy (N-acetylaspartate, choline, myo-inositol, creatine, and glutamate), providing a reference for the normal metabolic brain development. Throughout the third trimester of gestation, N-acetylaspartate gradually increases, whereas choline undergoes a slow reduction during the process of myelination. Less clear are the modifications in creatine, myo-inositol, and glutamate levels. Under conditions of fetal distress, the meaning of lactate detection is unclear, and further studies are needed. Another field for investigation involves the possibility of early detection of glutamate levels in fetuses at risk for hypoxic-ischemic encephalopathy, because the role of glutamate excitotoxicity in this context is well-established. Because metabolic modifications may precede functional or morphologic changes in the central nervous system, magnetic resonance spectroscopy may likely serve as a powerful, noninvasive tool for the early diagnosis and prognosis of different pathologic conditions.
Collapse
Affiliation(s)
- Erika Brighina
- Foundation Instituto di Ricerca e Cura a Carattere Scientifico Policlinico, Mangiagalli and Regina Elena, University of Milan, Milan, Italy
| | | | | | | |
Collapse
|
14
|
Buccafusca R, Venditti CP, Kenyon LC, Johanson RA, Van Bockstaele E, Ren J, Pagliardini S, Minarcik J, Golden JA, Coady MJ, Greer JJ, Berry GT. Characterization of the null murine sodium/myo-inositol cotransporter 1 (Smit1 or Slc5a3) phenotype: myo-inositol rescue is independent of expression of its cognate mitochondrial ribosomal protein subunit 6 (Mrps6) gene and of phosphatidylinositol levels in neonatal brain. Mol Genet Metab 2008; 95:81-95. [PMID: 18675571 DOI: 10.1016/j.ymgme.2008.05.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Revised: 05/30/2008] [Accepted: 05/30/2008] [Indexed: 12/13/2022]
Abstract
Ablation of the murine Slc5a3 gene results in severe myo-inositol (Ins) deficiency and congenital central apnea due to abnormal respiratory rhythmogenesis. The lethal knockout phenotype may be rescued by supplementing the maternal drinking water with 1% Ins. In order to test the hypothesis that Ins deficiency leads to inositide deficiencies, which are corrected by prenatal treatment, we measured the effects of Ins rescue on Ins, phosphatidylinositol (PtdIns) and myo-inositol polyphosphate levels in brains of E18.5 knockout fetuses. As the Slc5a3 gene structure is unique in the sodium/solute cotransporter (SLC5) family, and exon 1 is shared with the mitochondrial ribosomal protein subunit 6 (Mrps6) gene, we also sought to determine whether expression of its cognate Mrps6 gene is abnormal in knockout fetuses. The mean level of Ins was increased by 92% in brains of rescued Slc5a3 knockout fetuses (0.48 versus 0.25 nmol/mg), but was still greatly reduced in comparison to wildtype (6.97 nmol/mg). The PtdIns, InsP(5) and InsP(6) levels were normal without treatment. Mrps6 gene expression was unaffected in the E18.5 knockout fetuses. This enigmatic model is not associated with neonatal PtdIns deficiency and rescue of the phenotype may be accomplished without restoration of Ins. The biochemical mechanism that both uniformly leads to death and allows for Ins rescue remains unknown. In conclusion, in neonatal brain tissue, Mrps6 gene expression may not be contingent on function of its embedded Slc5a3 gene, while inositide deficiency may not be the mechanism of lethal apnea in null Slc5a3 mice.
Collapse
Affiliation(s)
- Roberto Buccafusca
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
McGrath BM, McKay R, Dave S, Seres P, Weljie AM, Slupsky CM, Hanstock CC, Greenshaw AJ, Silverstone PH. Acute dextro-amphetamine administration does not alter brain myo-inositol levels in humans and animals: MRS investigations at 3 and 18.8 T. Neurosci Res 2008; 61:351-9. [PMID: 18508145 DOI: 10.1016/j.neures.2008.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 04/05/2008] [Accepted: 04/09/2008] [Indexed: 11/24/2022]
Abstract
The pathophysiological underpinnings of bipolar disorder are not fully understood. However, they may be due in part to changes in the phosphatidylinositol second messenger system (PI-cycle) generally, or changes in myo-inositol concentrations more specifically. Dextro-amphetamine has been used as a model for mania in several human studies as it causes similar subjective and physiological symptoms. We wanted to determine if dextro-amphetamine altered myo-inositol concentrations in vivo as it would clearly define a mechanism linking putative changes in the PI-cycle to the subjective psychological changes seen with dextro-amphetamine administration. Fifteen healthy human volunteers received a baseline scan, followed by second scan 75 min after receiving a 25 mg oral dose of dextro-amphetamine. Stimulated echo proton magnetic resonance spectroscopy (MRS) scans were preformed at 3.0 Tesla (T) in the dorsal medial prefrontal cortex (DMPFC). Metabolite data were adjusted for tissue composition and analyzed using LCModel. Twelve adult male rats were treated acutely with a 5-mg/kg intraperitoneal dose of dextro-amphetamine. After 1 h rats were decapitated and the brains were rapidly removed and frozen until dissection. Rat brains were dissected into frontal, temporal, and occipital cortical areas, as well as hippocampus. Tissue was analyzed using a Varian 18.8 T spectrometer. Metabolites were identified and quantified using Chenomx Profiler software. The main finding in the present study was that myo-inositol concentrations in the DMPFC of human volunteers and in the four rat brain regions were not altered by acute dextro-amphetamine. While it remains possible that the PI-cycle may be involved in the pathophysiology of bipolar disorder, it is not likely that the subjective and physiological of dextro-amphetamine are mediated, directly or indirectly, via alternations in myo-inositol concentrations.
Collapse
Affiliation(s)
- Brent M McGrath
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada T6G 2B7.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Di Daniel E, Cheng L, Maycox PR, Mudge AW. The common inositol-reversible effect of mood stabilizers on neurons does not involve GSK3 inhibition, myo-inositol-1-phosphate synthase or the sodium-dependent myo-inositol transporters. Mol Cell Neurosci 2006; 32:27-36. [PMID: 16531065 DOI: 10.1016/j.mcn.2006.01.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2005] [Revised: 01/13/2006] [Accepted: 01/30/2006] [Indexed: 01/07/2023] Open
Abstract
We previously showed that the mood stabilizers lithium, valproate (VPA), and carbamazepine (CBZ) have a common, inositol-reversible effect on the dynamic behavior of sensory neurons, suggesting that they all inhibit phosphoinositide (PIns) synthesis. We now report similar effects of the drugs in cortical neurons and show by mRNA analysis that these neurons do not express myo-inositol-1-phosphate synthase (MIP-synthase) or the sodium-dependent myo-inositol transporters (SMIT1 and SMIT2), but they do express the H+/myo-inositol transporter (HMIT) mRNA and protein. We used glycogen synthase kinase-3 (GSK3) inhibitors and Western blotting of GSK3 targets to confirm that the common effects of the drugs on both sensory and cortical neuron growth cones are inositol-dependent and GSK3-independent. Moreover, the anti-convulsant drugs gabapentin and phenytoin do not mimic the mood stabilizers. These results confirm that the common inositol-reversible effect of mood stabilizers on neurons does not involve GSK3 and further show that the effects are independent of MIP-synthase and SMIT transporters.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antimanic Agents/pharmacology
- Carbamazepine/pharmacology
- Cells, Cultured
- Cerebral Cortex/cytology
- Cerebral Cortex/drug effects
- Cerebral Cortex/growth & development
- Enzyme Inhibitors/pharmacology
- Ganglia, Spinal/cytology
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Glucose Transport Proteins, Facilitative/genetics
- Glucose Transport Proteins, Facilitative/metabolism
- Glycogen Synthase Kinase 3/drug effects
- Glycogen Synthase Kinase 3/metabolism
- Growth Cones/drug effects
- Growth Cones/metabolism
- Growth Cones/ultrastructure
- Lithium/pharmacology
- Myo-Inositol-1-Phosphate Synthase/genetics
- Neurons/drug effects
- Neurons/metabolism
- Neurons, Afferent/cytology
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Symporters/genetics
- Valproic Acid/pharmacology
Collapse
Affiliation(s)
- Elena Di Daniel
- Schizophrenia and Bipolar Neurophysiology and Pharmacology Research Department, Psychiatry Centre of Excellence for Drug Discovery, GlaxoSmithKline Pharmaceuticals, Third Avenue, Harlow, Essex, CM19 5AW, UK.
| | | | | | | |
Collapse
|
17
|
Chau JFL, Lee MK, Law JWS, Chung SK, Chung SSM. Sodium/myo-inositol cotransporter-1 is essential for the development and function of the peripheral nerves. FASEB J 2005; 19:1887-9. [PMID: 16174787 DOI: 10.1096/fj.05-4192fje] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Sodium/myo-inositol cotransporter-1 (SMIT-1) is one of the transporters responsible for importing myo-inositol (MI) into the cells. MI is a precursor for a family of signal transduction molecules, phosphatidylinositol, and its derivatives that regulates many cellular functions. SMIT-1 null mice died soon after birth due to respiratory failure, but neonatal lethality was prevented by prenatal maternal MI supplement. Although the lung air sacs were closed, lung development was not significantly affected in the SMIT-1 null mice. The development of the peripheral nerves, including the brachial plexus, facial, vagus, and intercostal nerves, and the phrenic nerve that innervates the diaphragm was severely affected. All of these peripheral nerve abnormalities were corrected by prenatal MI supplement, indicating that MI is essential for the development of peripheral nerve and that neonatal lethality of the SMIT-1 knockout mice is most likely due to abnormal development of the nerves that control breathing. In the adult SMIT-1 deficient mice rescued by MI supplement, MI content in their brain, kidney, skeletal muscle, liver, and sciatic nerve was greatly reduced. The sciatic nerve, in particular, was most dependent on SMIT-1 for the accumulation of MI, and nerve conduction velocity and protein kinase C activity in this tissue were significantly reduced by SMIT-1 deficiency.
Collapse
Affiliation(s)
- Jenny F L Chau
- Institute of Molecular Biology, The University of Hong Kong, Hong Kong, SAR, China
| | | | | | | | | |
Collapse
|
18
|
Berry GT, Wu S, Buccafusca R, Ren J, Gonzales LW, Ballard PL, Golden JA, Stevens MJ, Greer JJ. Loss of murine Na+/myo-inositol cotransporter leads to brain myo-inositol depletion and central apnea. J Biol Chem 2003; 278:18297-302. [PMID: 12582158 DOI: 10.1074/jbc.m213176200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
myo-Inositol (Ins) and its polyphosphoinositide derivatives that are important in membrane signaling have long been held to play a special role in brain metabolism. As polyphosphoinositides turn over rapidly and are exceptionally abundant in nervous tissue, high Ins levels in the range of 2-15 mm that have been observed in brain may be necessary to maintain the rates of phosphoinositide synthesis in diverse membrane locations within neurons. Cellular concentration gradients of this magnitude indicate a dependence on active Ins transport, especially at the time of growth and differentiation. The Na(+)/myo-inositol cotransporter (SMIT1 or SLC5A3) gene is highly expressed prenatally in the central nervous system and placenta. To gain more insight into brain Ins metabolism, while ascertaining the importance of SMIT1 as a transporter, we generated mice with a homozygous targeted deletion of this gene. Newborn SMIT1(-/-) animals have no evidence of SMIT1 mRNA, a 92% reduction in the level of brain Ins, an 84% reduction in whole body Ins, and expire shortly after birth due to hypoventilation. Gross pathologic and light microscopic examinations of each organ, as well as the placenta, of embryonic day 18.5 fetuses at near term gestation were normal. Based on [(3)H]acetate incorporation into phospholipids of lung tissue explants, immunostaining of lung tissue for surfactant protein A, B, and C, and electron microscopic examination of alveolar cells, there was no evidence of abnormal pulmonary surfactant production by type 2 pneumocytes in lung. Although no histologic lesions were detected in the nervous system, electrophysiological studies of the brainstem pre-Bötzinger respiratory control center demonstrated an abnormal rhythm discharge with periods of central apnea. The cause of death can be explained by the regulatory defect in brainstem control of ventilation. This model demonstrates the critical importance of SMIT1 in the developing nervous system. The high affinity SMIT1 transporter is responsible for the Ins concentration gradient in the murine fetal-placental unit.
Collapse
Affiliation(s)
- Gerard T Berry
- Department of Pediatrics, University of Pennsylvania School of Medicine, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kimura T, Yamada K, Masago A, Shimada S. Subarachnoid hemorrhage induces Na+/myo-inositol cotransporter in the rat brain. Neurol Med Chir (Tokyo) 2003; 43:74-8; discussion 79. [PMID: 12627883 DOI: 10.2176/nmc.43.74] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Neurons and glial cells respond to extracellular hyperosmolarity by accumulating small organic solutes, called "osmolytes." Na+/myo-inositol is one of the major organic osmolytes in the brain and Na+/myo-inositol cotransporter (SMIT) regulates extracellular Na+/myo-inositol content. Subarachnoid hemorrhage (SAH) is an osmotic stress-inducing event of the brain. The expression of SMIT messenger ribonucleic acid (mRNA) and protein was investigated with in situ hybridization and immunohistochemistry in rat brains with SAH induced by endovascular perforation. SMIT riboprobe was raised from a 490-bp rat SMIT complementary deoxyribonucleic acid. Anti-SMIT antibody was raised in rabbits. SMIT mRNA was expressed strongly in the cortex, hippocampus, and hypothalamus of the perforated side at 6 to 24 hours after SAH. Mild upregulation was noted in the contralateral cortex, hippocampus, and hypothalamus. The ventral aspect of the pons showed mild upregulation. Microautoradiography and immunostaining showed SMIT expression mainly in the neurons, but also in some non-neural cells in the hippocampus. The present results indicate that diffuse osmotic stress occurs in the host brain after SAH.
Collapse
Affiliation(s)
- Tomohiro Kimura
- Departments of Neurosurgery, Nagoya City University Medical School, Nagoya, Japan
| | | | | | | |
Collapse
|
20
|
Fisher SK, Novak JE, Agranoff BW. Inositol and higher inositol phosphates in neural tissues: homeostasis, metabolism and functional significance. J Neurochem 2002; 82:736-54. [PMID: 12358779 DOI: 10.1046/j.1471-4159.2002.01041.x] [Citation(s) in RCA: 478] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Inositol phospholipids and inositol phosphates mediate well-established functions in signal transduction and in Ca2+ homeostasis in the CNS and non-neural tissues. More recently, there has been renewed interest in other roles that both myo-inositol and its highly phosphorylated forms may play in neural function. We review evidence that myo-inositol serves as a clinically relevant osmolyte in the CNS, and that its hexakisphosphate and pyrophosphorylated derivatives may play roles in such diverse cellular functions as DNA repair, nuclear RNA export and synaptic membrane trafficking.
Collapse
Affiliation(s)
- Stephen K Fisher
- Mental Health Research Institute, and Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | |
Collapse
|
21
|
Maouyo D, Kim JY, Lee SD, Wu Y, Woo SK, Kwon HM. Mouse TonEBP-NFAT5: expression in early development and alternative splicing. Am J Physiol Renal Physiol 2002; 282:F802-9. [PMID: 11934689 DOI: 10.1152/ajprenal.00123.2001] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tonicity-responsive enhancer binding protein (TonEBP)- nuclear factor of activated T cell family 5 is a DNA binding protein that plays a key role in the response of cells to hypertonicity. However, TonEBP is expressed and active in tissues that are in an isotonic milieu. To explore the biological role of TonEBP, we cloned mouse TonEBP that shares 92% of amino acids with the human counterpart. TonEBP is expressed in embryonic stem cells and throughout the stages of fetal development. Immunohistochemical analysis shows expression of TonEBP in most, if not all, developing tissues, including the brain, colon, heart, muscle, and eyes. Widespread alternative splicing in exons 2-4 was detected throughout development and in different adult tissues. As a result, four different polypeptides are produced with different lengths at the NH(2) terminus. Two of the isoforms differ in their ability to stimulate transcription. In conclusion, the presence of TonEBP mRNA during mouse embryogenesis suggests that TonEBP functions at all stages of mouse development, as well as in isotonic adult tissues.
Collapse
Affiliation(s)
- Djikolngar Maouyo
- Division of Nephrology, School of Medicine, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
22
|
Robertson NJ, Lewis RH, Cowan FM, Allsop JM, Counsell SJ, Edwards AD, Cox IJ. Early increases in brain myo-inositol measured by proton magnetic resonance spectroscopy in term infants with neonatal encephalopathy. Pediatr Res 2001; 50:692-700. [PMID: 11726726 DOI: 10.1203/00006450-200112000-00011] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Our aim was to assess brain myo-inositol/creatine plus phosphocreatine (Cr) in the first week in term infants with neonatal encephalopathy using localized short echo time proton magnetic resonance spectroscopy and to relate this to measures of brain injury, specifically lactate/Cr in the first week, basal ganglia changes on magnetic resonance imaging (MRI), and neurodevelopmental outcome at 1 y. Fourteen term infants with neonatal encephalopathy of gestational age (mean +/- SD) 39.6 +/- 1.6 wk, birth weight 3270 +/- 490 g, underwent MRI and magnetic resonance spectroscopy at 3.5 +/- 2.1 d. Five infants were entered in a pilot study of treatment with moderate whole-body hypothermia for neonatal encephalopathy; two were being cooled at the time of the scan. T(1)- and T(2)-weighted transverse magnetic resonance images were graded as normal or abnormal according to the presence or absence of the normal signal intensity of the posterior limb of the internal capsule and signal intensity changes in the basal ganglia. Localized proton magnetic resonance spectroscopy data were obtained from an 8-cm(3) voxel in the basal ganglia using echo times of 40 and 270 ms, and the peak area ratios of myo-inositol/Cr and lactate/Cr were measured. Outcome was scored using Griffith's development scales and neurodevelopmental examination at 1 y. MRI and outcome were normal in six infants and abnormal in eight. myo-Inositol/Cr and lactate/Cr were higher in infants with abnormal MRI and outcome (p < 0.01, p < 0.01, respectively). myo-Inositol/Cr and lactate/Cr were correlated (p < 0.01) and were both correlated to the Griffith's developmental scales (p < 0.01, p < 0.01, respectively). In conclusion, these preliminary data suggest that early increases in brain basal ganglia myo-inositol/Cr in infants with neonatal encephalopathy are associated with increased lactate/Cr, MRI changes of severe injury, and a poor neurodevelopmental outcome at 1 y.
Collapse
Affiliation(s)
- N J Robertson
- Department of Paediatrics, Faculty of Medicine, Imperial College of Science, Technology and Medicine, Hammersmith Hospital Campus, London W12 0HS, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
23
|
Nishiwaki A, Asai K, Tada T, Ueda T, Shimada S, Ogura Y, Kato T. Expression of glia maturation factor during retinal development in the rat. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 95:103-9. [PMID: 11687281 DOI: 10.1016/s0169-328x(01)00252-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glia maturation factor plays important roles in the development and growth of glia and neurons. We investigated the expression and localization of Glia maturation factor-beta (GMFB) and Glia maturation factor-gamma (GMFG) in the rat retina. By northern blot analysis, both GMFB and GMFG mRNAs were detected in retina as early as embryonic day (E) 18 and persisted until adult. The expression of GMFB mRNA was always much greater than that of GMFG mRNA. In situ hybridization showed that the GMFB mRNA signal was positive in the retina from E14 till adult. Immunostaining revealed that GMFB protein was present in the inner layer of retina at E14 and P1, and in Müller cells in adult. GMFG immunoreactivity was observed only in the inner limiting membrane from E14 to P1 rat retina, and was not detected in the adult retina. These results show that GMFs are synthesized and localized mainly in Müller cells in the rat retina, and suggest that they may contribute to the development and growth of glia and neurons.
Collapse
Affiliation(s)
- A Nishiwaki
- Department of Ophthalmology, Nagoya City University Medical School, 1 Kawasumi, Mizuho-cho, Mizuho-ku, 467-8601, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
24
|
Ueda T, Ugawa S, Saishin Y, Shimada S. Expression of receptor-activity modifying protein (RAMP) mRNAs in the mouse brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 93:36-45. [PMID: 11532336 DOI: 10.1016/s0169-328x(01)00179-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Receptor activity modifying proteins (RAMPs) comprise a family of accessory proteins for G protein-coupled receptors (GPCRs). They function as receptor modulators that determine the ligand specificity of receptors for calcitonin gene-related peptide (CGRP), amylin and adrenomedullin (ADM). Here we demonstrate for the first time the characteristic distributions of the RAMP family mRNAs in the brain. Northern blot analysis revealed that mRAMP 1 and 3 mRNAs were intensely expressed in the brain, but mRAMP2 mRNA less abundantly. In situ hybridization studies showed the heterogenous and unique distributions of mRAMP mRNAs; RAMP1 mRNA was widely expressed throughout the brain including the cerebral cortex, caudate putamen, amygdaloid complex, hippocampus, cerebellum and ependyma, mRAMP2 was most abundant in the hippocampus, cerebellum, pia mater and blood vessels, while mRAMP3 was specifically distributed in a variety of thalamic nuclei and the cerebellum. In addition, RAMP1 and -3 genes were also detected in the subfornical organ and area postrema, which are members of circumventricular organs lacking blood-brain barrier. The present results help in understanding the diversification and regulation of receptor functions for calcitonin family peptides, and potentially other GPCRs in the brain.
Collapse
Affiliation(s)
- T Ueda
- Department of Anatomy and Neuroscience, Nagoya City University Medical School, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan.
| | | | | | | |
Collapse
|
25
|
Ueda T, Iwata A, Komatsu H, Aihara N, Yamada K, Ugawa S, Shimada S. Diffuse brain injury induces local expression of Na+/myo-inositol cotransporter in the rat brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 86:63-9. [PMID: 11165372 DOI: 10.1016/s0169-328x(00)00261-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We studied expression of an osmoprotective gene, sodium/myo-inositol cotransporter (SMIT) in Marmarou's animal model for human diffuse brain injury by in situ hybridization and immunohistochemistry. In rats with diffuse brain injury, transient upregulation of SMIT mRNA was exclusively observed in the lateral area of pyramidal tract in lower brainstem. The expression was induced at 1 h after injury, peaked at 24 h, and returned to almost control levels at 48 h. Upregulated expression was found mainly in small glia-like cells. By immunohistochemistry using antibodies to phosphorylated mitogen-activated protein (MAP) kinases, inductions of phosphorylated p44/42 MAP kinase were also observed after diffuse brain injury. Interestingly, the distribution patterns of induced phosphorylated p44/42 MAP kinase were completely coincident with those of upregulated SMIT mRNA after diffuse brain injury. These results suggest that diffuse brain injury induces local expression of SMIT by activation of p44/42 MAP kinase cascade. The confined SMIT induction may reflect regional differences of damage and/or cellular differences in sensitivity to neuropathological stresses caused by this injury.
Collapse
Affiliation(s)
- T Ueda
- Department of Anatomy II, Nagoya City University Medical School, 1 Kawasumi, Mizuho-cho, Mizuho-ku, 467-8601, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
26
|
Shetty HU, Siarey RJ, Galdzicki Z, Stoll J, Rapoport SI. Ts65Dn mouse, a Down syndrome model, exhibits elevated myo-inositol in selected brain regions and peripheral tissues. Neurochem Res 2000; 25:431-5. [PMID: 10823574 DOI: 10.1023/a:1007592006005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
myo-Inositol is elevated in the Down syndrome (DS; trisomy 21) brain and may play a role in mental retardation. In the present study, we examined brain regions and peripheral tissues of Ts65Dn mouse, a recently characterized genetic model of DS, for abnormal myo-inositol accumulation. A GC/MS technique was used to quantitate myo-inositol and other polyol species (ribitol, arabitol, xylitol, and 1,5-anhydrosorbitol) in tissues from the Ts65Dn mice and control diploid mice. myo-Inositol was found to be elevated in frontal cortex, hippocampus, and brain stem but not in cerebellum of the Ts65Dn mouse. Among peripheral organs examined, liver and skeletal muscle were found to excessively accumulate myo-inositol. In all tissues, concentrations of polyol internal controls were normal. The Ts65Dn mouse is useful to study the possible effect of elevated myo-inositol on cellular processes.
Collapse
Affiliation(s)
- H U Shetty
- Section on Brain Physiology and Metabolism, National Institute on Aging, National Institutes of Health, Bethesda, Maryland 20892-1582, USA.
| | | | | | | | | |
Collapse
|
27
|
Pavlova A, Sakurai H, Leclercq B, Beier DR, Yu AS, Nigam SK. Developmentally regulated expression of organic ion transporters NKT (OAT1), OCT1, NLT (OAT2), and Roct. Am J Physiol Renal Physiol 2000; 278:F635-43. [PMID: 10751225 DOI: 10.1152/ajprenal.2000.278.4.f635] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Several xenobiotic (organic cation and anion) transporters have recently been identified, although their endogenous substrates, if such exist, remain unknown. When we initially identified NKT, also known as OAT1, the first member of the organic anion transporter (OAT) family (Lopez-Nieto CE, You G, Bush KT, Barros EJ, Beier DR, and Nigam SK. J Biol Chem 272: 6471-6478, 1997), we noted its expression in the embryonic kidney. We have now demonstrated its transporter function and more fully examined the spatiotemporal expression patterns of representative organic ion transporters, [NKT (OAT1), Roct, OCT1, and NLT, also known as OAT2] during murine development. In the kidney, NKT (OAT1), OCT1, and Roct transcripts appeared at midgestation, coinciding with proximal tubule differentiation, and gradually increased during nephron maturation. A similar pattern was observed for NLT (OAT2) in the liver and kidney, although, in the kidney, NLT (OAT2) transcription did not increase as dramatically. The roughly cotemporal expression of these related transporters in the developing proximal tubule may indicate common transcriptional regulation. Expression during embryogenesis in extrarenal sites could suggest a role in the formation and maintenance of nonrenal tissues. Importantly, all four genes were expressed in unexpected places during nonrenal organogenesis: Roct in the fetal liver (temporally coinciding with the onset of hematopoiesis) and neural tissue; NKT (OAT1) in the fetal brain; OCT1 in the ascending aorta and atrium; and NLT (OAT2) in the fetal lung, intestine, skin, and developing bone. Because these gene products mediate the transport of a broad range of metabolites and toxins, it seems likely that, apart from their known functions, these transporters play a role in transport of organic molecules, perhaps including those with morphogenetic activity. These genes could also play important developmental roles independent of transport function.
Collapse
Affiliation(s)
- A Pavlova
- Renal, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
28
|
Mizuno N, Kitayama S, Saishin Y, Shimada S, Morita K, Mitsuhata C, Kurihara H, Dohi T. Molecular cloning and characterization of rat trp homologues from brain. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1999; 64:41-51. [PMID: 9889314 DOI: 10.1016/s0169-328x(98)00296-4] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Identification of trp (transient receptor potential) gene from Drosophila photoreceptor and subsequent molecular cloning of the human cDNA homologues suggest its participation in capacitative calcium entry (CCE) or so called store-operated Ca2+ channel (SOC). We identified five different trp-related amplifications of reverse-transcription-polymerase chain reaction (RT-PCR) from rat brain; these corresponded to mouse trp homologues, mtrp1,3,4,5,6 and were distributed in various tissues with multiple expression levels. Two cDNAs, homologous to Drosophila trp from rat brain, designated rtrp3 and rtrp6, were isolated and characterized. By RT-PCR analysis, mRNAs of rtrp3 and rtrp6 were found to be expressed differently in brain and other tissues. In situ hybridization analysis revealed that rtrp6 mRNA was preferentially expressed in hippocampal dentate gyrus and cortical layers II and III. Expression of rat TRP3 and TRP6 in COS cells revealed an increase in CCE, as compared to that in the mock-transfected COS cells of the control. Isolation of cDNAs of rat trp gene family provides a useful model for studying mechanism of CCE.
Collapse
Affiliation(s)
- N Mizuno
- Department of Pharmacology, Hiroshima University School of Dentistry, Kasumi 1-2-3, Minami-ku, Hiroshima, 734-8553, Japan
| | | | | | | | | | | | | | | |
Collapse
|