1
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane lipid nanodomains modulate HCN pacemaker channels in nociceptor DRG neurons. Nat Commun 2024; 15:9898. [PMID: 39548079 PMCID: PMC11568329 DOI: 10.1038/s41467-024-54053-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we find that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels is likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observe reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Natalie L Macchi
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Nicolas L A Dumaire
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Lyuba Salih
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Erin N Lessie
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Kyle S McCommis
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Aubin Moutal
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, Saint Louis, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, USA.
| |
Collapse
|
2
|
Handlin LJ, Macchi NL, Dumaire NLA, Salih L, Lessie EN, McCommis KS, Moutal A, Dai G. Membrane Lipid Nanodomains Modulate HCN Pacemaker Channels in Nociceptor DRG Neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.02.556056. [PMID: 37732182 PMCID: PMC10508734 DOI: 10.1101/2023.09.02.556056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Cell membranes consist of heterogeneous lipid nanodomains that influence key cellular processes. Using FRET-based fluorescent assays and fluorescence lifetime imaging microscopy (FLIM), we found that the dimension of cholesterol-enriched ordered membrane domains (OMD) varies considerably, depending on specific cell types. Particularly, nociceptor dorsal root ganglion (DRG) neurons exhibit large OMDs. Disruption of OMDs potentiated action potential firing in nociceptor DRG neurons and facilitated the opening of native hyperpolarization-activated cyclic nucleotide-gated (HCN) pacemaker channels. This increased neuronal firing is partially due to an increased open probability and altered gating kinetics of HCN channels. The gating effect on HCN channels was likely due to a direct modulation of their voltage sensors by OMDs. In animal models of neuropathic pain, we observed reduced OMD size and a loss of HCN channel localization within OMDs. Additionally, cholesterol supplementation inhibited HCN channels and reduced neuronal hyperexcitability in pain models. These findings suggest that disturbances in lipid nanodomains play a critical role in regulating HCN channels within nociceptor DRG neurons, influencing pain modulation.
Collapse
|
3
|
Hamar J, Cnaani A, Kültz D. Effects of CRISPR/Cas9 targeting of the myo-inositol biosynthesis pathway on hyper-osmotic tolerance of tilapia cells. Genomics 2024; 116:110833. [PMID: 38518899 DOI: 10.1016/j.ygeno.2024.110833] [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: 12/13/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Myo-inositol is an important compatible osmolyte in vertebrates. This osmolyte is produced by the myo-inositol biosynthesis (MIB) pathway composed of myo-inositol phosphate synthase and inositol monophosphatase. These enzymes are among the highest upregulated proteins in tissues and cell cultures from teleost fish exposed to hyperosmotic conditions indicating high importance of this pathway for tolerating this type of stress. CRISPR/Cas9 gene editing of tilapia cells produced knockout lines of MIB enzymes and control genes. Metabolic activity decreased significantly for MIB KO lines in hyperosmotic media. Trends of faster growth of the MIB knockout lines in isosmotic media and faster decline of MIB knockout lines in hyperosmotic media were also observed. These results indicate a decline in metabolic fitness but only moderate effects on cell survival when tilapia cells with disrupted MIB genes are exposed to hyperosmolality. Therefore MIB genes are required for full osmotolerance of tilapia cells.
Collapse
Affiliation(s)
- Jens Hamar
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA
| | - Avner Cnaani
- Department of Poultry and Aquaculture, Institute of Animal Sciences, Agricultural Research Organization, Volcani Center, P.O. Box 15159, Rishon LeZion 7528809, Israel
| | - Dietmar Kültz
- Department of Animal Sciences & Genome Center, University of California Davis, Meyer Hall, One Shields Avenue, Davis, CA 95616, USA.
| |
Collapse
|
4
|
Saha S, Krishnan H, Raghu P. IMPA1 dependent regulation of phosphatidylinositol 4,5-bisphosphate and calcium signalling by lithium. Life Sci Alliance 2024; 7:e202302425. [PMID: 38056909 PMCID: PMC10700560 DOI: 10.26508/lsa.202302425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023] Open
Abstract
Lithium (Li) is widely used as a mood stabilizer to treat bipolar affective disorder. However, the molecular targets of Li that underpin its therapeutic effect remain unresolved. Inositol monophosphatase (IMPA1) is an enzyme involved in phosphatidylinositol 4,5-bisphosphate (PIP2) resynthesis after PLC signaling. In vitro, Li inhibits IMPA1, but the relevance of this inhibition within neural cells remains unknown. Here, we report that treatment with therapeutic concentrations of Li reduces receptor-activated calcium release from intracellular stores and delays PIP2 resynthesis. These effects of Li are abrogated in IMPA1 deleted cells. We also observed that in human forebrain cortical neurons, treatment with Li reduced neuronal excitability and calcium signals. After Li treatment of human cortical neurons, transcriptome analyses revealed down-regulation of signaling by glutamate, a key excitatory neurotransmitter in the human brain. Collectively, our findings suggest that inhibition of IMPA1 by Li reduces receptor-activated PLC signaling and neuronal excitability.
Collapse
Affiliation(s)
- Sankhanil Saha
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| | - Harini Krishnan
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| | - Padinjat Raghu
- National Centre for Biological Sciences-TIFR GKVK Campus, Bangalore, India
| |
Collapse
|
5
|
Handlin LJ, Dai G. Direct regulation of the voltage sensor of HCN channels by membrane lipid compartmentalization. Nat Commun 2023; 14:6595. [PMID: 37852983 PMCID: PMC10584925 DOI: 10.1038/s41467-023-42363-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023] Open
Abstract
Ion channels function within a membrane environment characterized by dynamic lipid compartmentalization. Limited knowledge exists regarding the response of voltage-gated ion channels to transmembrane potential within distinct membrane compartments. By leveraging fluorescence lifetime imaging microscopy (FLIM) and Förster resonance energy transfer (FRET), we visualized the localization of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels in membrane domains. HCN4 exhibits a greater propensity for incorporation into ordered lipid domains compared to HCN1. To investigate the conformational changes of the S4 helix voltage sensor of HCN channels, we used dual stop-codon suppression to incorporate different noncanonical amino acids, orthogonal click chemistry for site-specific fluorescence labeling, and transition metal FLIM-FRET. Remarkably, altered FRET levels were observed between VSD sites within HCN channels upon disruption of membrane domains. We propose that the voltage-sensor rearrangements, directly influenced by membrane lipid domains, can explain the heightened activity of pacemaker HCN channels when localized in cholesterol-poor, disordered lipid domains, leading to membrane hyperexcitability and diseases.
Collapse
Affiliation(s)
- Lucas J Handlin
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA
| | - Gucan Dai
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, 1100 South Grand Blvd., St. Louis, MO, 63104, USA.
| |
Collapse
|
6
|
Dai G. Signaling by Ion Channels: Pathways, Dynamics and Channelopathies. MISSOURI MEDICINE 2023; 120:367-373. [PMID: 37841571 PMCID: PMC10569386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Charged ions and ion channels play a critical role in regulating the electrical activities of excitable cells. This review discusses the principles of ion channel regulation in the time domain, as well as the diseases that can arise from channel dysfunction and disturbances in ionic balance. Ion channel signaling is a dynamic process that is essential for various physiological functions, including pain sensation, motor control, and the body's response to stress, such as fight-or-flight response.
Collapse
Affiliation(s)
- Gucan Dai
- Edward A. Doisy Department of Biochemistry & Molecular Biology, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
7
|
Yoder KK, Chumin EJ, Mustafi SM, Kolleck KA, Halcomb ME, Hile KL, Plawecki MH, O'Connor SJ, Dzemidzic M, Wu YC. Effects of acute alcohol exposure and chronic alcohol use on neurite orientation dispersion and density imaging (NODDI) parameters. Psychopharmacology (Berl) 2023; 240:1465-1472. [PMID: 37209164 PMCID: PMC10594986 DOI: 10.1007/s00213-023-06380-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 04/28/2023] [Indexed: 05/22/2023]
Abstract
RATIONALE Little is known about how acute and chronic alcohol exposure may alter the in vivo membrane properties of neurons. OBJECTIVES We employed neurite orientation dispersion and density imaging (NODDI) to examine acute and chronic effects of alcohol exposure on neurite density. METHODS Twenty-one healthy social drinkers (CON) and thirteen nontreatment-seeking individuals with alcohol use disorder (AUD) underwent a baseline multi-shell diffusion magnetic resonance imaging (dMRI) scan. A subset (10 CON, 5 AUD) received dMRI during intravenous infusions of saline and alcohol during dMRI. NODDI parametric images included orientation dispersion (OD), isotropic volume fraction (ISOVF), and corrected intracellular volume fraction (cICVF). Diffusion tensor imaging metrics of fractional anisotropy and mean, axial, and radial diffusivity (FA, MD, AD, RD) were also computed. Average parameter values were extracted from white matter (WM) tracts defined by the Johns Hopkins University atlas. RESULTS There were group differences in FA, RD, MD, OD, and cICVF, primarily in the corpus callosum. Both saline and alcohol had effects on AD and cICVF in WM tracts proximal to the striatum, cingulate, and thalamus. This is the first work to indicate that acute fluid infusions may alter WM properties, which are conventionally believed to be insensitive to acute pharmacological challenges. It also suggests that the NODDI approach may be sensitive to transient changes in WM. The next steps should include determining if the effect on neurite density differs with solute or osmolality, or both, and translational studies to assess how alcohol and osmolality affect the efficiency of neurotransmission.
Collapse
Affiliation(s)
- Karmen K Yoder
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA.
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Ste. 414, Indianapolis, IN, 46202, USA.
| | - Evgeny J Chumin
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Ste. 414, Indianapolis, IN, 46202, USA
- Department of Psychological and Brain Sciences, Indiana University, 1101 E 10th St, IN, 47405, Bloomington, USA
- Indiana University Network Science Institute, Indiana University, 1015 E 11th St, Bloomington, IN, 47408, USA
| | - Sourajit M Mustafi
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
| | - Kelly A Kolleck
- Indiana University School of Medicine, 340 W. 10th St., Indianapolis, IN, 46202, USA
| | - Meredith E Halcomb
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
| | - Karen L Hile
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
| | - Martin H Plawecki
- Department of Psychiatry, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4800, Indianapolis, IN, 46202, USA
| | - Sean J O'Connor
- Department of Psychiatry, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4800, Indianapolis, IN, 46202, USA
| | - Mario Dzemidzic
- Department of Neurology, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4700, Indianapolis, IN, 46202, USA
| | - Yu-Chien Wu
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Center for Neuroimaging, Indiana Institute of Biomedical Imaging, Indiana University School of Medicine, 355 W. 16th St., GH Ste. 4100, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W. 15th Street, Ste. 414, Indianapolis, IN, 46202, USA
| |
Collapse
|
8
|
Derkaczew M, Martyniuk P, Osowski A, Wojtkiewicz J. Cyclitols: From Basic Understanding to Their Association with Neurodegeneration. Nutrients 2023; 15:2029. [PMID: 37432155 DOI: 10.3390/nu15092029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 07/12/2023] Open
Abstract
One of the most common cyclitols found in eukaryotic cells-Myo-inositol (MI) and its derivatives play a key role in many cellular processes such as ion channel physiology, signal transduction, phosphate storage, cell wall formation, membrane biogenesis and osmoregulation. The aim of this paper is to characterize the possibility of neurodegenerative disorders treatment using MI and the research of other therapeutic methods linked to MI's derivatives. Based on the reviewed literature the researchers focus on the most common neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease and Spinocerebellar ataxias, but there are also works describing other seldom encountered diseases. The use of MI, d-pinitol and other methods altering MI's metabolism, although research on this topic has been conducted for years, still needs much closer examination. The dietary supplementation of MI shows a promising effect on the treatment of neurodegenerative disorders and can be of great help in alleviating the accompanying depressive symptoms.
Collapse
Affiliation(s)
- Maria Derkaczew
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Students' Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Piotr Martyniuk
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
- Students' Scientific Club of Pathophysiologists, Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Adam Osowski
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| |
Collapse
|
9
|
Terra Machado D, Bernardes Brustolini OJ, Côrtes Martins Y, Grivet Mattoso Maia MA, Ribeiro de Vasconcelos AT. Inference of differentially expressed genes using generalized linear mixed models in a pairwise fashion. PeerJ 2023; 11:e15145. [PMID: 37033732 PMCID: PMC10078460 DOI: 10.7717/peerj.15145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/09/2023] [Indexed: 04/05/2023] Open
Abstract
Background
Technological advances involving RNA-Seq and Bioinformatics allow quantifying the transcriptional levels of genes in cells, tissues, and cell lines, permitting the identification of Differentially Expressed Genes (DEGs). DESeq2 and edgeR are well-established computational tools used for this purpose and they are based upon generalized linear models (GLMs) that consider only fixed effects in modeling. However, the inclusion of random effects reduces the risk of missing potential DEGs that may be essential in the context of the biological phenomenon under investigation. The generalized linear mixed models (GLMM) can be used to include both effects.
Methods
We present DEGRE (Differentially Expressed Genes with Random Effects), a user-friendly tool capable of inferring DEGs where fixed and random effects on individuals are considered in the experimental design of RNA-Seq research. DEGRE preprocesses the raw matrices before fitting GLMMs on the genes and the derived regression coefficients are analyzed using the Wald statistical test. DEGRE offers the Benjamini-Hochberg or Bonferroni techniques for P-value adjustment.
Results
The datasets used for DEGRE assessment were simulated with known identification of DEGs. These have fixed effects, and the random effects were estimated and inserted to measure the impact of experimental designs with high biological variability. For DEGs’ inference, preprocessing effectively prepares the data and retains overdispersed genes. The biological coefficient of variation is inferred from the counting matrices to assess variability before and after the preprocessing. The DEGRE is computationally validated through its performance by the simulation of counting matrices, which have biological variability related to fixed and random effects. DEGRE also provides improved assessment measures for detecting DEGs in cases with higher biological variability. We show that the preprocessing established here effectively removes technical variation from those matrices. This tool also detects new potential candidate DEGs in the transcriptome data of patients with bipolar disorder, presenting a promising tool to detect more relevant genes.
Conclusions
DEGRE provides data preprocessing and applies GLMMs for DEGs’ inference. The preprocessing allows efficient remotion of genes that could impact the inference. Also, the computational and biological validation of DEGRE has shown to be promising in identifying possible DEGs in experiments derived from complex experimental designs. This tool may help handle random effects on individuals in the inference of DEGs and presents a potential for discovering new interesting DEGs for further biological investigation.
Collapse
Affiliation(s)
- Douglas Terra Machado
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | - Yasmmin Côrtes Martins
- Laboratório de Bioinformática, Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
10
|
Gong T, Hui SCN, Zöllner HJ, Britton M, Song Y, Chen Y, Gudmundson AT, Hupfeld KE, Davies-Jenkins CW, Murali-Manohar S, Porges EC, Oeltzschner G, Chen W, Wang G, Edden RAE. Neurometabolic timecourse of healthy aging. Neuroimage 2022; 264:119740. [PMID: 36356822 PMCID: PMC9902072 DOI: 10.1016/j.neuroimage.2022.119740] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/20/2022] [Accepted: 11/06/2022] [Indexed: 11/09/2022] Open
Abstract
PURPOSE The neurometabolic timecourse of healthy aging is not well-established, in part due to diversity of quantification methodology. In this study, a large structured cross-sectional cohort of male and female subjects throughout adulthood was recruited to investigate neurometabolic changes as a function of age, using consensus-recommended magnetic resonance spectroscopy quantification methods. METHODS 102 healthy volunteers, with approximately equal numbers of male and female participants in each decade of age from the 20s, 30s, 40s, 50s, and 60s, were recruited with IRB approval. MR spectroscopic data were acquired on a 3T MRI scanner. Metabolite spectra were acquired using PRESS localization (TE=30 ms; 96 transients) in the centrum semiovale (CSO) and posterior cingulate cortex (PCC). Water-suppressed spectra were modeled using the Osprey algorithm, employing a basis set of 18 simulated metabolite basis functions and a cohort-mean measured macromolecular spectrum. Pearson correlations were conducted to assess relationships between metabolite concentrations and age for each voxel; Spearman correlations were conducted where metabolite distributions were non-normal. Paired t-tests were run to determine whether metabolite concentrations differed between the PCC and CSO. Finally, robust linear regressions were conducted to assess both age and sex as predictors of metabolite concentrations in the PCC and CSO and separately, to assess age, signal-noise ratio, and full width half maximum (FWHM) linewidth as predictors of metabolite concentrations. RESULTS Data from four voxels were excluded (2 ethanol; 2 unacceptably large lipid signal). Statistically-significant age*metabolite Pearson correlations were observed for tCho (r(98)=0.33, p<0.001), tCr (r(98)=0.60, p<0.001), and mI (r(98)=0.32, p=0.001) in the CSO and for NAAG (r(98)=0.26, p=0.008), tCho(r(98)=0.33, p<0.001), tCr (r(98)=0.39, p<0.001), and Gln (r(98)=0.21, p=0.034) in the PCC. Spearman correlations for non-normal variables revealed a statistically significant correlation between sI and age in the CSO (r(86)=0.26, p=0.013). No significant correlations were seen between age and tNAA, NAA, Glx, Glu, GSH, PE, Lac, or Asp in either region (all p>0.20). Age associations for tCho, tCr, mI and sI in the CSO and for NAAG, tCho, and tCr in the PCC remained when controlling for sex in robust regressions. CSO NAAG and Asp, as well as PCC tNAA, sI, and Lac were higher in women; PCC Gln was higher in men. When including an age*sex interaction term in robust regression models, a significant age*sex interaction was seen for tCho (F(1,96)=11.53, p=0.001) and GSH (F(1,96)=7.15, p=0.009) in the CSO and tCho (F(1,96)=9.17, p=0.003), tCr (F(1,96)=9.59, p=0.003), mI (F(1,96)=6.48, p=0.012), and Lac (F(1,78)=6.50, p=0.016) in the PCC. In all significant interactions, metabolite levels increased with age in females, but not males. There was a significant positive correlation between linewidth and age. Age relationships with tCho, tCr, and mI in the CSO and tCho, tCr, mI, and sI in the PCC were significant after controlling for linewidth and FWHM in robust regressions. CONCLUSION The primary (correlation) results indicated age relationships for tCho, tCr, mI, and sI in the CSO and for NAAG, tCho, tCr, and Gln in the PCC, while no age correlations were found for tNAA, NAA, Glx, Glu, GSH, PE, Lac, or Asp in either region. Our results provide a normative foundation for future work investigating the neurometabolic time course of healthy aging using MRS.
Collapse
Affiliation(s)
- Tao Gong
- Departments of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Departments of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Steve C N Hui
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Helge J Zöllner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Mark Britton
- Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, United States of America; McKnight Brain Research Foundation, University of Florida, FL, United States of America; Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States of America
| | - Yulu Song
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Yufan Chen
- Departments of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Aaron T Gudmundson
- Department of Neurobiology and Behavior, University of California, Irvine, CA, United States of America
| | - Kathleen E Hupfeld
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Christopher W Davies-Jenkins
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Saipavitra Murali-Manohar
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | - Eric C Porges
- Center for Cognitive Aging and Memory, University of Florida, Gainesville, FL, United States of America; McKnight Brain Research Foundation, University of Florida, FL, United States of America; Department of Clinical and Health Psychology, University of Florida, Gainesville, FL, United States of America
| | - Georg Oeltzschner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| | | | - Guangbin Wang
- Departments of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Departments of Radiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China.
| | - Richard A E Edden
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, United States of America
| |
Collapse
|
11
|
Brosch PK, Korsa T, Taban D, Eiring P, Kreisz P, Hildebrand S, Neubauer J, Zimmermann H, Sauer M, Shirakashi R, Djuzenova CS, Sisario D, Sukhorukov VL. Glucose and Inositol Transporters, SLC5A1 and SLC5A3, in Glioblastoma Cell Migration. Cancers (Basel) 2022; 14:5794. [PMID: 36497276 PMCID: PMC9738886 DOI: 10.3390/cancers14235794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/04/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022] Open
Abstract
(1) Background: The recurrence of glioblastoma multiforme (GBM) is mainly due to invasion of the surrounding brain tissue, where organic solutes, including glucose and inositol, are abundant. Invasive cell migration has been linked to the aberrant expression of transmembrane solute-linked carriers (SLC). Here, we explore the role of glucose (SLC5A1) and inositol transporters (SLC5A3) in GBM cell migration. (2) Methods: Using immunofluorescence microscopy, we visualized the subcellular localization of SLC5A1 and SLC5A3 in two highly motile human GBM cell lines. We also employed wound-healing assays to examine the effect of SLC inhibition on GBM cell migration and examined the chemotactic potential of inositol. (3) Results: While GBM cell migration was significantly increased by extracellular inositol and glucose, it was strongly impaired by SLC transporter inhibition. In the GBM cell monolayers, both SLCs were exclusively detected in the migrating cells at the monolayer edge. In single GBM cells, both transporters were primarily localized at the leading edge of the lamellipodium. Interestingly, in GBM cells migrating via blebbing, SLC5A1 and SLC5A3 were predominantly detected in nascent and mature blebs, respectively. (4) Conclusion: We provide several lines of evidence for the involvement of SLC5A1 and SLC5A3 in GBM cell migration, thereby complementing the migration-associated transportome. Our findings suggest that SLC inhibition is a promising approach to GBM treatment.
Collapse
Affiliation(s)
- Philippa K. Brosch
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Tessa Korsa
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Danush Taban
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Patrick Eiring
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Philipp Kreisz
- Julius-von-Sachs Institute, University of Würzburg, 97082 Würzburg, Germany;
| | - Sascha Hildebrand
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Julia Neubauer
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
| | - Heiko Zimmermann
- Fraunhofer Institute for Biomedical Engineering (IBMT), 66280 Sulzbach, Germany; (J.N.); (H.Z.)
- Department of Molecular and Cellular Biotechnology, Saarland University, 66123 Saarbrücken, Germany
- Faculty of Marine Science, Universidad Católica del Norte, Coquimbo 1281, Chile
| | - Markus Sauer
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Ryo Shirakashi
- Institute of Industrial Science, The University of Tokyo, Tokyo 153-8505, Japan;
| | - Cholpon S. Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, 97080 Würzburg, Germany;
| | - Dmitri Sisario
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| | - Vladimir L. Sukhorukov
- Department of Biotechnology & Biophysics, Biocenter, University of Würzburg, 97074 Würzburg, Germany; (P.K.B.); (T.K.); (D.T.); (P.E.); (S.H.); (M.S.)
| |
Collapse
|
12
|
Cai J, Li X, Wu S, Tian Y, Zhang Y, Wei Z, Jin Z, Li X, Chen X, Chen WX. Assessing the causal association between human blood metabolites and the risk of epilepsy. Lab Invest 2022; 20:437. [PMID: 36180952 PMCID: PMC9524049 DOI: 10.1186/s12967-022-03648-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/18/2022] [Indexed: 11/18/2022]
Abstract
Background Metabolic disturbance has been reported in patients with epilepsy. Still, the evidence about the causal role of metabolites in facilitating or preventing epilepsy is lacking. Systematically investigating the causality between blood metabolites and epilepsy would help provide novel targets for epilepsy screening and prevention. Methods We conducted two-sample Mendelian randomization (MR) analysis. Data for 486 human blood metabolites came from a genome-wide association study (GWAS) comprising 7824 participants. GWAS data for epilepsy were obtained from the International League Against Epilepsy (ILAE) consortium for primary analysis and the FinnGen consortium for replication and meta-analysis. Sensitivity analyses were conducted to evaluate heterogeneity and pleiotropy. Results 482 out of 486 metabolites were included for MR analysis following rigorous genetic variants selection. After IVW and sensitivity analysis filtration, six metabolites with causal effects on epilepsy were identified from the ILAE consortium. Only four metabolites remained significant associations with epilepsy when combined with the FinnGen consortium [uridine: odds ratio (OR) = 2.34, 95% confidence interval (CI) = 1.48–3.71, P = 0.0003; 2-hydroxystearate: OR = 1.61, 95% CI = 1.19–2.18, P = 0.002; decanoylcarnitine: OR = 0.82, 95% CI = 0.72–0.94, P = 0.004; myo-inositol: OR = 0.77, 95% CI = 0.62–0.96, P = 0.02]. Conclusion The evidence that the four metabolites mentioned above are associated with epilepsy in a causal way provides a novel insight into the underlying mechanisms of epilepsy by integrating genomics with metabolism, and has an implication for epilepsy screening and prevention. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03648-5.
Collapse
Affiliation(s)
- Jiahao Cai
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoyu Li
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Shangbin Wu
- Department of Pediatrics, Guangdong Provincial Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yang Tian
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yani Zhang
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zixin Wei
- Department of Pulmonary and Critical Care Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zixiang Jin
- First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xiaojing Li
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiong Chen
- Department of Pediatric Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Wen-Xiong Chen
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
13
|
Abbott GW. Kv Channel Ancillary Subunits: Where Do We Go from Here? Physiology (Bethesda) 2022; 37:0. [PMID: 35797055 PMCID: PMC9394777 DOI: 10.1152/physiol.00005.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 01/10/2023] Open
Abstract
Voltage-gated potassium (Kv) channels each comprise four pore-forming α-subunits that orchestrate essential duties such as voltage sensing and K+ selectivity and conductance. In vivo, however, Kv channels also incorporate regulatory subunits-some Kv channel specific, others more general modifiers of protein folding, trafficking, and function. Understanding all the above is essential for a complete picture of the role of Kv channels in physiology and disease.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California
| |
Collapse
|
14
|
Jensen JB, Falkenburger BH, Dickson EJ, de la Cruz L, Dai G, Myeong J, Jung SR, Kruse M, Vivas O, Suh BC, Hille B. Biophysical physiology of phosphoinositide rapid dynamics and regulation in living cells. J Gen Physiol 2022; 154:e202113074. [PMID: 35583815 PMCID: PMC9121023 DOI: 10.1085/jgp.202113074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 04/28/2022] [Indexed: 01/07/2023] Open
Abstract
Phosphoinositide membrane lipids are ubiquitous low-abundance signaling molecules. They direct many physiological processes that involve ion channels, membrane identification, fusion of membrane vesicles, and vesicular endocytosis. Pools of these lipids are continually broken down and refilled in living cells, and the rates of some of these reactions are strongly accelerated by physiological stimuli. Recent biophysical experiments described here measure and model the kinetics and regulation of these lipid signals in intact cells. Rapid on-line monitoring of phosphoinositide metabolism is made possible by optical tools and electrophysiology. The experiments reviewed here reveal that as for other cellular second messengers, the dynamic turnover and lifetimes of membrane phosphoinositides are measured in seconds, controlling and timing rapid physiological responses, and the signaling is under strong metabolic regulation. The underlying mechanisms of this metabolic regulation remain questions for the future.
Collapse
Affiliation(s)
- Jill B. Jensen
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | | | - Eamonn J. Dickson
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA
| | - Lizbeth de la Cruz
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Gucan Dai
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO
| | - Jongyun Myeong
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO
| | | | - Martin Kruse
- Department of Biology and Program in Neuroscience, Bates College, Lewiston, ME
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Byung-Chang Suh
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Bertil Hille
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
15
|
Dai G. Neuronal KCNQ2/3 channels are recruited to lipid raft microdomains by palmitoylation of BACE1. J Gen Physiol 2022; 154:213033. [PMID: 35201266 PMCID: PMC8876601 DOI: 10.1085/jgp.202112888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 02/04/2022] [Indexed: 12/14/2022] Open
Abstract
β-Secretase 1 (β-site amyloid precursor protein [APP]-cleaving enzyme 1, BACE1) plays a crucial role in the amyloidogenesis of Alzheimer’s disease (AD). BACE1 was also discovered to act like an auxiliary subunit to modulate neuronal KCNQ2/3 channels independently of its proteolytic function. BACE1 is palmitoylated at its carboxyl-terminal region, which brings BACE1 to ordered, cholesterol-rich membrane microdomains (lipid rafts). However, the physiological consequences of this specific localization of BACE1 remain elusive. Using spectral Förster resonance energy transfer (FRET), BACE1 and KCNQ2/3 channels were confirmed to form a signaling complex, a phenomenon that was relatively independent of the palmitoylation of BACE1. Nevertheless, palmitoylation of BACE1 was required for recruitment of KCNQ2/3 channels to lipid-raft domains. Two fluorescent probes, designated L10 and S15, were used to label lipid-raft and non-raft domains of the plasma membrane, respectively. Coexpressing BACE1 substantially elevated FRET between L10 and KCNQ2/3, whereas the BACE1-4C/A quadruple mutation failed to produce this effect. In contrast, BACE1 had no significant effect on FRET between S15 probes and KCNQ2/3 channels. A reduction of BACE1-dependent FRET between raft-targeting L10 probes and KCNQ2/3 channels by applying the cholesterol-extracting reagent methyl-β-cyclodextrin (MβCD), raft-disrupting general anesthetics, or pharmacological inhibitors of palmitoylation, all supported the hypothesis of the palmitoylation-dependent and raft-specific localization of KCNQ2/3 channels. Furthermore, mutating the four carboxyl-terminal cysteines (4C/A) of BACE1 abolished the BACE1-dependent increase of FRET between KCNQ2/3 and the lipid raft–specific protein caveolin 1. Taking these data collectively, we propose that the AD-related protein BACE1 underlies the localization of a neuronal potassium channel.
Collapse
Affiliation(s)
- Gucan Dai
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| |
Collapse
|
16
|
Zhou Y, Sun M, Sun P, Gao H, Yang H, Jing Y, Hussain MA, Saxena RK, Carther FI, Wang Q, Li H. Tonoplast inositol transporters: Roles in plant abiotic stress response and crosstalk with other signals. JOURNAL OF PLANT PHYSIOLOGY 2022; 271:153660. [PMID: 35240513 DOI: 10.1016/j.jplph.2022.153660] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 02/22/2022] [Accepted: 02/22/2022] [Indexed: 06/14/2023]
Abstract
Inositol transporters (INT) are thought to be the pivotal transporters for vital metabolites, in particular lipids, minerals, and sugars. These transporters play an important role in transitional metabolism and various signaling pathways in plants through regulating the transduction of messages from hormones, neurotransmitters, and immunologic and growth factors. Extensive studies have been conducted on animal INT, with promising outcomes. However, only few recent studies have highlighted the importance and complexity of INT genes in the regulation of plant physiology stages, including growth and tolerance to stress conditions. The present review summarizes the most recent findings concerning the role of INT or inositol genes in plant metabolism and the response mechanisms triggered by external stressors. Moreover, we highlight the emerging role of vacuoles and vacuolar INT in plant molecular transition and their related roles in plant growth and development. INTs are the essential mediators of inositol uptake and its intracellular broadcasting for various metabolic pathways where they play crucial roles. Additionally, we report evidence on Na+/inositol transporters, which until now have only been characterized in animals, as well as H+/inositol symporters and their kinetic functions and physiological role and suggest their roles and operating mode in plants. A more comprehensive understanding of the INT functioning system, in particular the coordinated movement of inositol and the relation between inositol generation and other important plant signaling pathways, would greatly advance the study of plant stress adaptation.
Collapse
Affiliation(s)
- Yonggang Zhou
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - Monan Sun
- College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Pengyu Sun
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - Hongtao Gao
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - He Yang
- RDFZ Sanya School, Sanya, 572025, China.
| | - Yan Jing
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - Muhammad Azhar Hussain
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - Rachit K Saxena
- International Crops Research Institute for the Semi-Arid Tropics (ICRISAT), Hyderabad, 502324, India.
| | - Foka Idrice Carther
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| | - Qingyu Wang
- College of Plant Science, Jilin University, Changchun, 130062, China.
| | - Haiyan Li
- Sanya Nanfan Research Institute of Hainan University, Hainan Yazhou Bay Seed Laboratory, Sanya, 572025, China; College of Tropical Crops, Hainan University, Haikou, 570288, China.
| |
Collapse
|
17
|
de la Cruz L, Riquelme R, Vivas O, Barria A, Jensen JB. Dishevelled coordinates phosphoinositide kinases PI4KIIIα and PIP5KIγ for efficient PtdInsP2 synthesis. J Cell Sci 2022; 135:274231. [PMID: 34982154 PMCID: PMC8919331 DOI: 10.1242/jcs.259145] [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: 07/14/2021] [Accepted: 12/14/2021] [Indexed: 02/05/2023] Open
Abstract
Phosphatidylinositol(4,5)-bisphosphate (PtdInsP2) is an important modulator of many cellular processes, and its abundance in the plasma membrane is closely regulated. We examined the hypothesis that members of the Dishevelled scaffolding protein family can bind the lipid kinases phosphatidylinositol 4-kinase (PI4K) and phosphatidylinositol 4-phosphate 5-kinase (PIP5K), facilitating synthesis of PtdInsP2 directly from phosphatidylinositol. We used several assays for PtdInsP2 to examine the cooperative function of phosphoinositide kinases and the Dishevelled protein Dvl3 in the context of two receptor signaling cascades. Simultaneous overexpression of PI4KIIIα (also known as PI4KA) and PIP5KIγ (also known as PIP5K1C) had a synergistic effect on PtdInsP2 synthesis that was recapitulated by overexpression of Dvl3. Increasing the activity of Dvl3 by overexpression increased resting plasma membrane PtdInsP2. Knockdown of Dvl3 reduced resting plasma membrane PtdInsP2 and slowed PtdInsP2 resynthesis following receptor activation. We confirm that Dvl3 promotes coupling of PI4KIIIα and PIP5KIγ and show that this interaction is essential for efficient resynthesis of PtdInsP2 following receptor activation.
Collapse
|
18
|
Myo-Inositol Limits Kainic Acid-Induced Epileptogenesis in Rats. Int J Mol Sci 2022; 23:ijms23031198. [PMID: 35163126 PMCID: PMC8835653 DOI: 10.3390/ijms23031198] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 01/09/2023] Open
Abstract
Epilepsy is a severe neurological disease characterized by spontaneous recurrent seizures (SRS). A complex pathophysiological process referred to as epileptogenesis transforms a normal brain into an epileptic one. Prevention of epileptogenesis is a subject of intensive research. Currently, there are no clinically approved drugs that can act as preventive medication. Our previous studies have revealed highly promising antiepileptogenic properties of a compound-myo-inositol (MI) and the present research broadens previous results and demonstrates the long-term disease-modifying effect of this drug, as well as the amelioration of cognitive comorbidities. For the first time, we show that long-term treatment with MI: (i) decreases the frequency and duration of electrographic SRS in the hippocampus; (ii) has an ameliorating effect on spatial learning and memory deficit associated with epileptogenesis, and (iii) attenuates cell loss in the hippocampus. MI treatment also alters the expression of the glial fibrillary acidic protein, LRRC8A subunit of volume-regulated anion channels, and protein tyrosine phosphatase receptor type R, all expected to counteract the epileptogenesis. All these effects are still present even 4 weeks after MI treatment ceased. This suggests that MI may exert multiple actions on various epileptogenesis-associated changes in the brain and, therefore, could be considered as a candidate target for prevention of epileptogenesis.
Collapse
|
19
|
Tewari SG, Kwan B, Elahi R, Rajaram K, Reifman J, Prigge ST, Vaidya AB, Wallqvist A. Metabolic adjustments of blood-stage Plasmodium falciparum in response to sublethal pyrazoleamide exposure. Sci Rep 2022; 12:1167. [PMID: 35064153 PMCID: PMC8782945 DOI: 10.1038/s41598-022-04985-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/04/2022] [Indexed: 11/08/2022] Open
Abstract
Due to the recurring loss of antimalarial drugs to resistance, there is a need for novel targets, drugs, and combination therapies to ensure the availability of current and future countermeasures. Pyrazoleamides belong to a novel class of antimalarial drugs that disrupt sodium ion homeostasis, although the exact consequences of this disruption in Plasmodium falciparum remain under investigation. In vitro experiments demonstrated that parasites carrying mutations in the metabolic enzyme PfATP4 develop resistance to pyrazoleamide compounds. However, the underlying mechanisms that allow mutant parasites to evade pyrazoleamide treatment are unclear. Here, we first performed experiments to identify the sublethal dose of a pyrazoleamide compound (PA21A092) that caused a significant reduction in growth over one intraerythrocytic developmental cycle (IDC). At this drug concentration, we collected transcriptomic and metabolomic data at multiple time points during the IDC to quantify gene- and metabolite-level alterations in the treated parasites. To probe the effects of pyrazoleamide treatment on parasite metabolism, we coupled the time-resolved omics data with a metabolic network model of P. falciparum. We found that the drug-treated parasites adjusted carbohydrate metabolism to enhance synthesis of myoinositol-a precursor for phosphatidylinositol biosynthesis. This metabolic adaptation caused a decrease in metabolite flux through the pentose phosphate pathway, causing a decreased rate of RNA synthesis and an increase in oxidative stress. Our model analyses suggest that downstream consequences of enhanced myoinositol synthesis may underlie adjustments that could lead to resistance emergence in P. falciparum exposed to a sublethal dose of a pyrazoleamide drug.
Collapse
Affiliation(s)
- Shivendra G Tewari
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, USA.
| | - Bobby Kwan
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Rubayet Elahi
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Krithika Rajaram
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Jaques Reifman
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, USA
| | - Sean T Prigge
- Department of Molecular Microbiology and Immunology, Johns Hopkins University, Baltimore, MD, USA
| | - Akhil B Vaidya
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Anders Wallqvist
- Department of Defense Biotechnology High Performance Computing Software Applications Institute, Telemedicine and Advanced Technology Research Center, U.S. Army Medical Research and Development Command, Fort Detrick, MD, USA.
| |
Collapse
|
20
|
Myeong J, de la Cruz L, Jung SR, Yeon JH, Suh BC, Koh DS, Hille B. Phosphatidylinositol 4,5-bisphosphate is regenerated by speeding of the PI 4-kinase pathway during long PLC activation. J Gen Physiol 2021; 152:211533. [PMID: 33186442 PMCID: PMC7671494 DOI: 10.1085/jgp.202012627] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 10/13/2020] [Indexed: 01/05/2023] Open
Abstract
The dynamic metabolism of membrane phosphoinositide lipids involves several cellular compartments including the ER, Golgi, and plasma membrane. There are cycles of phosphorylation and dephosphorylation and of synthesis, transfer, and breakdown. The simplified phosphoinositide cycle comprises synthesis of phosphatidylinositol in the ER, transport, and phosphorylation in the Golgi and plasma membranes to generate phosphatidylinositol 4,5-bisphosphate, followed by receptor-stimulated hydrolysis in the plasma membrane and return of the components to the ER for reassembly. Using probes for specific lipid species, we have followed and analyzed the kinetics of several of these events during stimulation of M1 muscarinic receptors coupled to the G-protein Gq. We show that during long continued agonist action, polyphosphorylated inositol lipids are initially depleted but then regenerate while agonist is still present. Experiments and kinetic modeling reveal that the regeneration results from gradual but massive up-regulation of PI 4-kinase pathways rather than from desensitization of receptors. Golgi pools of phosphatidylinositol 4-phosphate and the lipid kinase PI4KIIIα (PI4KA) contribute to this homeostatic regeneration. This powerful acceleration, which may be at the level of enzyme activity or of precursor and product delivery, reveals strong regulatory controls in the phosphoinositide cycle.
Collapse
Affiliation(s)
- Jongyun Myeong
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | - Lizbeth de la Cruz
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | | | - Jun-Hee Yeon
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Byung-Chang Suh
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, South Korea
| | - Duk-Su Koh
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| | - Bertil Hille
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
21
|
Grecco GG, Chumin EJ, Dzemidzic M, Cheng H, Finn P, Newman S, Dydak U, Yoder KK. Anterior cingulate cortex metabolites and white matter microstructure: a multimodal study of emergent alcohol use disorder. Brain Imaging Behav 2021; 15:2436-2444. [PMID: 34097282 DOI: 10.1007/s11682-020-00443-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 12/27/2022]
Abstract
Multimodal imaging is increasingly used to address neuropathology associated with alcohol use disorder (AUD). Few studies have investigated relationships between metabolite concentrations and white matter (WM) integrity; currently, there are no such data in AUD. In this preliminary study, we used complementary neuroimaging techniques, magnetic resonance spectroscopy (MRS), and diffusion weighted imaging (DWI), to study AUD neurophysiology. We tested for relationships between metabolites in the dorsal anterior cingulate cortex (dACC) and adjacent WM microstructure in young adult AUD and control (CON) subjects. Sixteen AUD and fourteen CON underwent whole-brain DWI and MRS of the dACC. Outcomes were dACC metabolites, and diffusion tensor metrics of dACC-adjacent WM. Multiple linear regression terms included WM region, group, and region × group for prediction of dACC metabolites. dACC myo-inositol was positively correlated with axial diffusivity in the left anterior corona radiata (p < 0.0001) in CON but not AUD (group effect: p < 0.001; region × group: p < 0.001; Bonferroni-corrected). In the bilateral anterior corona radiata and right genu of the corpus callosum, glutamate was negatively related to mean diffusivity in AUD, but not CON subjects (all model terms: p < 0.05, uncorrected). In AUD subjects, dACC glutamate was negatively correlated with AUD symptom severity. This is likely the first integrative study of cortical metabolites and WM integrity in young individuals with AUD. Differential relationships between dACC metabolites and adjacent WM tract integrity in AUD could represent early consequences of hazardous drinking, and/or novel biomarkers of early-stage AUD. Additional studies are required to replicate these findings, and to determine the behavioral relevance of these results.
Collapse
Affiliation(s)
- Gregory G Grecco
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, GH 4100, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Evgeny J Chumin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, GH 4100, Indianapolis, IN, 46202, USA.,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA.,Indiana University Network Science Institute, Bloomington, IN, USA
| | - Mario Dzemidzic
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, GH 4100, Indianapolis, IN, 46202, USA.,Department of Neurology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hu Cheng
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Peter Finn
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Sharlene Newman
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Ulrike Dydak
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, GH 4100, Indianapolis, IN, 46202, USA.,School of Health Sciences, Purdue University, West Lafayette, IN, USA
| | - Karmen K Yoder
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 West 16th Street, GH 4100, Indianapolis, IN, 46202, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA. .,Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA.
| |
Collapse
|
22
|
Patkee PA, Baburamani AA, Long KR, Dimitrova R, Ciarrusta J, Allsop J, Hughes E, Kangas J, McAlonan GM, Rutherford MA, De Vita E. Neurometabolite mapping highlights elevated myo-inositol profiles within the developing brain in down syndrome. Neurobiol Dis 2021; 153:105316. [PMID: 33711492 PMCID: PMC8039898 DOI: 10.1016/j.nbd.2021.105316] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 02/06/2021] [Accepted: 02/22/2021] [Indexed: 01/02/2023] Open
Abstract
The neurodevelopmental phenotype in Down Syndrome (DS), or Trisomy 21, is variable including a wide spectrum of cognitive impairment and a high risk of early-onset Alzheimer's disease (AD). A key metabolite of interest within the brain in DS is Myo-inositol (mIns). The NA+/mIns co-transporter is located on human chromosome 21 and is overexpressed in DS. In adults with DS, elevated brain mIns was previously associated with cognitive impairment and proposed as a risk marker for progression to AD. However, it is unknown if brain mIns is increased earlier in development. The aim of this study was to estimate mIns concentration levels and key brain metabolites [N-acetylaspartate (NAA), Choline (Cho) and Creatine (Cr)] in the developing brain in DS and aged-matched controls. We used in vivo magnetic resonance spectroscopy (MRS) in neonates with DS (n = 12) and age-matched controls (n = 26) scanned just after birth (36-45 weeks postmenstrual age). Moreover, we used Mass Spectrometry in early (10-20 weeks post conception) ex vivo fetal brain tissue samples from DS (n = 14) and control (n = 30) cases. Relative to [Cho] and [Cr], we report elevated ratios of [mIns] in vivo in the basal ganglia/thalamus, in neonates with DS, when compared to age-matched typically developing controls. Glycine concentration ratios [Gly]/[Cr] and [Cho]/[Cr] also appear elevated. We observed elevated [mIns] in the ex vivo fetal cortical brain tissue in DS compared with controls. In conclusion, a higher level of brain mIns was evident as early as 10 weeks post conception and was measurable in vivo from 36 weeks post-menstrual age. Future work will determine if this early difference in metabolites is linked to cognitive outcomes in childhood or has utility as a potential treatment biomarker for early intervention.
Collapse
Affiliation(s)
- Prachi A Patkee
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK
| | - Ana A Baburamani
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK
| | - Katherine R Long
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE1 1UL, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, SE1 1UL, UK
| | - Ralica Dimitrova
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK; Department of Forensic and Neurodevelopmental Science, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AB, UK
| | - Judit Ciarrusta
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK; Department of Forensic and Neurodevelopmental Science, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AB, UK
| | - Joanna Allsop
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK
| | - Emer Hughes
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK
| | - Johanna Kangas
- Department of Forensic and Neurodevelopmental Science, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AB, UK
| | - Grainne M McAlonan
- Department of Forensic and Neurodevelopmental Science, Sackler Institute for Translational Neurodevelopment, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AB, UK
| | - Mary A Rutherford
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK
| | - Enrico De Vita
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK; Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
23
|
Kruse M, Whitten RJ. Control of Neuronal Excitability by Cell Surface Receptor Density and Phosphoinositide Metabolism. Front Pharmacol 2021; 12:663840. [PMID: 33967808 PMCID: PMC8097148 DOI: 10.3389/fphar.2021.663840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/29/2021] [Indexed: 12/27/2022] Open
Abstract
Phosphoinositides are members of a family of minor phospholipids that make up about 1% of all lipids in most cell types. Despite their low abundance they have been found to be essential regulators of neuronal activities such as action potential firing, release and re-uptake of neurotransmitters, and interaction of cytoskeletal proteins with the plasma membrane. Activation of several different neurotransmitter receptors can deplete phosphoinositide levels by more than 90% in seconds, thereby profoundly altering neuronal behavior; however, despite the physiological importance of this mechanism we still lack a profound quantitative understanding of the connection between phosphoinositide metabolism and neuronal activity. Here, we present a model that describes phosphoinositide metabolism and phosphoinositide-dependent action potential firing in sympathetic neurons. The model allows for a simulation of activation of muscarinic acetylcholine receptors and its effects on phosphoinositide levels and their regulation of action potential firing in these neurons. In this paper, we describe the characteristics of the model, its calibration to experimental data, and use the model to analyze how alterations of surface density of muscarinic acetylcholine receptors or altered activity levels of a key enzyme of phosphoinositide metabolism influence action potential firing of sympathetic neurons. In conclusion, the model provides a comprehensive framework describing the connection between muscarinic acetylcholine signaling, phosphoinositide metabolism, and action potential firing in sympathetic neurons which can be used to study the role of these signaling systems in health and disease.
Collapse
Affiliation(s)
- Martin Kruse
- Department of Biology, Bates College, Lewiston, ME, United States
- Program in Neuroscience, Bates College, Lewiston, ME, United States
| | - Rayne J. Whitten
- Program in Neuroscience, Bates College, Lewiston, ME, United States
| |
Collapse
|
24
|
Thongprayoon C, Cheungpasitporn W, Yap JQ, Qian Q. Increased mortality risk associated with serum sodium variations and borderline hypo- and hypernatremia in hospitalized adults. Nephrol Dial Transplant 2021; 35:1746-1752. [PMID: 31219584 PMCID: PMC7538236 DOI: 10.1093/ndt/gfz098] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 04/23/2019] [Indexed: 12/12/2022] Open
Abstract
Background This study aimed to evaluate short-term and long-term mortalities in a cohort of unselected hospitalized patients with serum sodium concentration ([Na+]) variations within and outside of reference range. Methods All adult patients admitted to the Mayo Clinic, Rochester, MN, USA from January 2011 to December 2013 (n = 147358) were retrospectively screened. Unique patients admitted during the study period were examined. The main exposure was serum [Na+] variation. Outcome measures were hospital and 1-year all-cause mortalities. Results A total of 60944 patients, mean age 63 ± 17 years, were studied. On admission, 17% (n = 10066) and 1.4% (n = 852) had hypo- and hypernatremia, respectively. During the hospital stay, 11044 and 4128 developed hypo- and hypernatremia, respectively, accounting for 52.3 and 82.9% of the total hypo- and hypernatremic patients. Serum [Na+] variations of ≥6 mEq/L occurred in 40.6% (n = 24 740) of the 60 944 patients and were significantly associated with hospital and 1-year mortalities after adjusting potential confounders (including demographics, comorbidities, estimated glomerular filtration rate, admission serum [Na+], number of [Na+] measurements and length of hospital stay). Adjusted odds ratios for hospital and 1-year mortalities increased with increasing [Na+] variations in a dose-dependent manner, from 1.47 to 5.48 (all 95% confidence intervals >1.0). Moreover, in fully adjusted models, [Na+] variations (≥6 mEq/L) within the reference range (135–145 mEq/L) or borderline hypo- or hypernatremia (133–137 and 143–147 mEq/L, respectively) compared with 138–142 mEq/L were associated with increased hospital and 1-year mortalities. Conclusion In hospitalized adults, [Na+] fluctuation (≥6 mEq/L) irrespective of admission [Na+] and borderline hypo- or hypernatremia are independent predictors of progressively increasing short- and long-term mortality burdens.
Collapse
Affiliation(s)
- Charat Thongprayoon
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Wisit Cheungpasitporn
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - John Q Yap
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Qi Qian
- Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| |
Collapse
|
25
|
López-Gambero AJ, Sanjuan C, Serrano-Castro PJ, Suárez J, Rodríguez de Fonseca F. The Biomedical Uses of Inositols: A Nutraceutical Approach to Metabolic Dysfunction in Aging and Neurodegenerative Diseases. Biomedicines 2020; 8:biomedicines8090295. [PMID: 32825356 PMCID: PMC7554709 DOI: 10.3390/biomedicines8090295] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/05/2023] Open
Abstract
Inositols are sugar-like compounds that are widely distributed in nature and are a part of membrane molecules, participating as second messengers in several cell-signaling processes. Isolation and characterization of inositol phosphoglycans containing myo- or d-chiro-inositol have been milestones for understanding the physiological regulation of insulin signaling. Other functions of inositols have been derived from the existence of multiple stereoisomers, which may confer antioxidant properties. In the brain, fluctuation of inositols in extracellular and intracellular compartments regulates neuronal and glial activity. Myo-inositol imbalance is observed in psychiatric diseases and its use shows efficacy for treatment of depression, anxiety, and compulsive disorders. Epi- and scyllo-inositol isomers are capable of stabilizing non-toxic forms of β-amyloid proteins, which are characteristic of Alzheimer’s disease and cognitive dementia in Down’s syndrome, both associated with brain insulin resistance. However, uncertainties of the intrinsic mechanisms of inositols regarding their biology are still unsolved. This work presents a critical review of inositol actions on insulin signaling, oxidative stress, and endothelial dysfunction, and its potential for either preventing or delaying cognitive impairment in aging and neurodegenerative diseases. The biomedical uses of inositols may represent a paradigm in the industrial approach perspective, which has generated growing interest for two decades, accompanied by clinical trials for Alzheimer’s disease.
Collapse
Affiliation(s)
- Antonio J. López-Gambero
- Departamento de Biología Celular, Genética y Fisiología, Campus de Teatinos s/n, Universidad de Málaga, Andalucia Tech, 29071 Málaga, Spain;
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
| | | | - Pedro Jesús Serrano-Castro
- UGC Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain;
| | - Juan Suárez
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (F.R.d.F.); Tel.: +34-952614012 (J.S.)
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (F.R.d.F.); Tel.: +34-952614012 (J.S.)
| |
Collapse
|
26
|
Barrese V, Stott JB, Baldwin SN, Mondejar-Parreño G, Greenwood IA. SMIT (Sodium-Myo-Inositol Transporter) 1 Regulates Arterial Contractility Through the Modulation of Vascular Kv7 Channels. Arterioscler Thromb Vasc Biol 2020; 40:2468-2480. [PMID: 32787517 PMCID: PMC7505149 DOI: 10.1161/atvbaha.120.315096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Objective: The SMIT1 (sodium:myo-inositol transporter 1) regulates myo-inositol movement into cells and responses to hypertonic stimuli. Alteration of myo-inositol levels has been associated with several diseases, including hypertension, but there is no evidence of a functional role of SMIT1 in the vasculature. Recent evidence showed that in the nervous system SMIT1 interacted and modulated the function of members of the Kv7 family of voltage-gated potassium channels, which are also expressed in the vasculature where they regulate arterial contractility. Therefore, in this study, we evaluated whether SMIT1 was functionally relevant in arterial smooth muscle. Approach and Results: Immunofluorescence and polymerase chain reaction experiments revealed that SMIT1 was expressed in rat renal and mesenteric vascular smooth muscle cells. Isometric tension recordings showed that incubation of renal arteries with raffinose and myo-inositol (which increases SMIT1 expression) reduced the contractile responses to methoxamine, an effect that was abolished by preincubation with the pan-Kv7 blocker linopirdine and by molecular knockdown of Kv7.4 and Kv7.5. Knockdown of SMIT1 increased the contraction of renal arteries induced by methoxamine, impaired the response to the Kv7.2–Kv7.5 activator ML213 but did not interfere with the relaxant responses induced by openers of other potassium channels. Proximity ligation assay showed that SMIT1 interacted with heteromeric channels formed by Kv7.4 and Kv7.5 proteins in both renal and mesenteric vascular smooth muscle cells. Patch-clamp experiments showed that incubation with raffinose plus myo-inositol increased Kv7 currents in vascular smooth muscle cells. Conclusions: SMIT1 protein is expressed in vascular smooth muscle cells where it modulates arterial contractility through an association with Kv7.4/Kv7.5 heteromers.
Collapse
Affiliation(s)
- Vincenzo Barrese
- Vascular Research Centre, Institute of Molecular & Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., S.N.B., I.A.G.).,Department of Neuroscience, Reproductive Science and Dentistry, University of Naples Federico II, Italy (V.B.)
| | - Jennifer B Stott
- Vascular Research Centre, Institute of Molecular & Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., S.N.B., I.A.G.)
| | - Samuel N Baldwin
- Vascular Research Centre, Institute of Molecular & Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., S.N.B., I.A.G.)
| | - Gema Mondejar-Parreño
- Department of Pharmacology and Toxicology. School of Medicine, Universidad Complutense de Madrid, Spain (G.M.-P.)
| | - Iain A Greenwood
- Vascular Research Centre, Institute of Molecular & Clinical Sciences, St George's, University of London, United Kingdom (V.B., J.B.S., S.N.B., I.A.G.)
| |
Collapse
|
27
|
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
|
28
|
Chewcharat A, Thongprayoon C, Cheungpasitporn W, Mao MA, Thirunavukkarasu S, Kashani KB. Trajectories of Serum Sodium on In-Hospital and 1-Year Survival among Hospitalized Patients. Clin J Am Soc Nephrol 2020; 15:600-607. [PMID: 32213501 PMCID: PMC7269204 DOI: 10.2215/cjn.12281019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/07/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND AND OBJECTIVES This study aimed to investigate the association between in-hospital trajectories of serum sodium and risk of in-hospital and 1-year mortality in patients in hospital. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS This is a single-center cohort study. All adult patients who were hospitalized from years 2011 through 2013 who had available admission serum sodium and at least three serum sodium measurements during hospitalization were included. The trend of serum sodium during hospitalization was analyzed using group-based trajectory modeling; the five main trajectories were grouped as follows: (1) stable normonatremia, (2) uncorrected hyponatremia, (3) borderline high serum sodium, (4) corrected hyponatremia, and (5) fluctuating serum sodium. The outcome of interest was in-hospital mortality and 1-year mortality. Stable normonatremia was used as the reference group for outcome comparison. RESULTS A total of 43,539 patients were analyzed. Of these, 47% had stable normonatremia, 15% had uncorrected hyponatremia, 31% had borderline high serum sodium, 3% had corrected hyponatremia, and 5% had fluctuating serum sodium trajectory. In adjusted analysis, there was a higher in-hospital mortality among those with uncorrected hyponatremia (odds ratio [OR], 1.33; 95% CI, 1.06 to 1.67), borderline high serum sodium (OR, 1.66; 95% CI, 1.38 to 2.00), corrected hyponatremia (OR, 1.50; 95% CI, 1.02 to 2.20), and fluctuating serum sodium (OR, 4.61; 95% CI, 3.61 to 5.88), compared with those with the normonatremia trajectory. One-year mortality was higher among those with uncorrected hyponatremia (hazard ratio [HR], 1.28; 95% CI, 1.19 to 1.38), borderline high serum sodium (HR, 1.18; 95% CI, 1.11 to 1.26), corrected hyponatremia (HR, 1.24; 95% CI, 1.08 to 1.42), and fluctuating serum sodium (HR, 2.10; 95% CI, 1.89 to 2.33) compared with those with the normonatremia trajectory. CONCLUSIONS More than half of patients who had been hospitalized had an abnormal serum sodium trajectory during hospitalization. This study demonstrated that not only the absolute serum sodium levels but also their in-hospital trajectories were significantly associated with in-hospital and 1-year mortality. The highest in-hospital and 1-year mortality risk was associated with the fluctuating serum sodium trajectory. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2020_03_25_CJN.12281019.mp3.
Collapse
Affiliation(s)
- Api Chewcharat
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Charat Thongprayoon
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Internal Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael A Mao
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida; and
| | - Sorkko Thirunavukkarasu
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Kianoush B Kashani
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota; .,Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
29
|
Cabrera-Cruz H, Oróstica L, Plaza-Parrochia F, Torres-Pinto I, Romero C, Vega M. The insulin-sensitizing mechanism of myo-inositol is associated with AMPK activation and GLUT-4 expression in human endometrial cells exposed to a PCOS environment. Am J Physiol Endocrinol Metab 2020; 318:E237-E248. [PMID: 31874063 DOI: 10.1152/ajpendo.00162.2019] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine-metabolic disorder characterized by hyperandrogenism and ovulatory dysfunction but also obesity and hyperinsulinemia. These characteristics induce an insulin-resistant state in tissues such as the endometrium, affecting its reproductive functions. Myo-inositol (MYO) is an insulin-sensitizing compound used in PCOS patients; however, its insulin-sensitizing mechanism is unclear. To understand the relationship of MYO with insulin action in endometrial cells, sodium/myo-inositol transporter 1 (SMIT-1) (MYO-transporter), and MYO effects on protein levels related to the insulin pathway were evaluated. SMIT-1 was assessed in endometrial tissue from women with normal weight, obesity, insulin resistance, and PCOS; additionally, using an in vitro model of human endometrial cells exposed to an environment resembling hyperinsulinemic-obese-PCOS, MYO effect was evaluated on p-AMPK and GLUT-4 levels and glucose uptake by Western blot, immunocytochemistry, and confocal microscopy, respectively. SMIT-1 was detected in endometrial tissue from all groups and decreased in PCOS and obesity (P < 0.05 vs. normal weight). In the in vitro model, PCOS conditions decreased p-AMPK levels, while they were restored with MYO (P < 0.05). The diminished GLUT-4 protein levels promoted by PCOS environment were restored by MYO through SMIT-1 and p-AMPK-dependent mechanism (P < 0.05). Also, MYO restored glucose uptake in cells under PCOS condition through a p-AMPK-dependent mechanism. Finally, these results were similar to those obtained with metformin treatment in the same in vitro conditions. Consequently, MYO could be a potential insulin sensitizer through its positive effects on insulin-resistant tissues as PCOS-endometrium, acting through SMIT-1, provoking AMPK activation and elevated GLUT-4 levels and, consequently, increase glucose uptake by human endometrial cells. Therefore, MYO may be used as an effective treatment option in insulin-resistant PCOS women.
Collapse
Affiliation(s)
- Heidy Cabrera-Cruz
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
- Department of Bioanalysis and Immunology, Faculty of Sciences, National Autonomous University of Honduras, Tegucigalpa, Honduras
| | - Lorena Oróstica
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad Diego Portales, Santiago, Chile
| | - Francisca Plaza-Parrochia
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
| | - Ignacio Torres-Pinto
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
| | - Carmen Romero
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
- Department of Obstetrics and Gynecology, Clinical Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Margarita Vega
- Laboratory of Endocrinology and Reproductive Biology, Clinical Hospital, University of Chile, Santiago, Chile
- Department of Obstetrics and Gynecology, Clinical Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
30
|
de la Cruz L, Traynor-Kaplan A, Vivas O, Hille B, Jensen JB. Plasma membrane processes are differentially regulated by type I phosphatidylinositol phosphate 5-kinases and RASSF4. J Cell Sci 2020; 133:jcs.233254. [PMID: 31831523 DOI: 10.1242/jcs.233254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Phosphoinositide lipids regulate many cellular processes and are synthesized by lipid kinases. Type I phosphatidylinositol phosphate 5-kinases (PIP5KIs) generate phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P 2]. Several phosphoinositide-sensitive readouts revealed the nonequivalence of overexpressing PIP5KIβ, PIP5KIγ or Ras association domain family 4 (RASSF4), believed to activate PIP5KIs. Mass spectrometry showed that each of these three proteins increased total cellular phosphatidylinositol bisphosphates (PtdInsP 2) and trisphosphates (PtdInsP 3) at the expense of phosphatidylinositol phosphate (PtdInsP) without changing lipid acyl chains. Analysis of KCNQ2/3 channels and PH domains confirmed an increase in plasma membrane PtdIns(4,5)P 2 in response to PIP5KIβ or PIP5KIγ overexpression, but RASSF4 required coexpression with PIP5KIγ to increase plasma membrane PtdIns(4,5)P 2 Effects on the several steps of store-operated calcium entry (SOCE) were not explained by plasma membrane phosphoinositide increases alone. PIP5KIβ and RASSF4 increased STIM1 proximity to the plasma membrane, accelerated STIM1 mobilization and speeded onset of SOCE; however, PIP5KIγ reduced STIM1 recruitment but did not change induced Ca2+ entry. These differences imply actions through different segregated pools of phosphoinositides and specific protein-protein interactions and targeting.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lizbeth de la Cruz
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA
| | - Alexis Traynor-Kaplan
- ATK Innovation, Analytics and Discovery, North Bend, WA 98045, USA.,Department of Medicine/Gastroenterology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA
| | - Bertil Hille
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA
| | - Jill B Jensen
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA 98195-7290, USA
| |
Collapse
|
31
|
Gonzalez-Uarquin F, Rodehutscord M, Huber K. Myo-inositol: its metabolism and potential implications for poultry nutrition-a review. Poult Sci 2019; 99:893-905. [PMID: 32036985 PMCID: PMC7587644 DOI: 10.1016/j.psj.2019.10.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/26/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
Myo-inositol (MI) has gained relevance in physiology research during the last decade. As a constituent of animal cells, MI was proven to be crucial in several metabolic and regulatory processes. Myo-inositol is involved in lipid signaling, osmolarity, glucose, and insulin metabolism. In humans and rodents, dietary MI was assessed to be important for health so that MI supplementation appeared to be a valuable alternative for treatment of several diseases as well as for improvements in metabolic performance. In poultry, there is a lack of evidence not only related to specific species-linked metabolic processes but also about the effects of dietary MI on performance and health. This review intends to provide information about the meaning of dietary MI in animal metabolism as well as to discuss potential implications of dietary MI in poultry health and performance with the aim to identify open questions in poultry research.
Collapse
Affiliation(s)
| | - Markus Rodehutscord
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Korinna Huber
- Institute of Animal Science, University of Hohenheim, 70599 Stuttgart, Germany.
| |
Collapse
|
32
|
Concentration- and time-dependent effects of myo-inositol on evoked epileptic afterdischarge in the hippocampus in vivo. Neuroreport 2019; 30:1129-1134. [PMID: 31568207 DOI: 10.1097/wnr.0000000000001341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Epilepsy is one of the most widespread neurological diseases characterized by spontaneous recurrent seizures. There is no cure for epilepsy, and available pharmacological treatments with anti-seizure drugs are only symptomatic. Moreover, about third of epilepsy patients are resistant to the anti-seizure drugs. Thus, it is essential to discover new anti-epilepsy drugs. Recently, myo-inositol has been identified as a promising antiepileptic compound. In the present study, using electrophysiological method, we examined for the first time, the effect of myo-inositol on the generation of epileptic afterdischarges in the hippocampus evoked by a local electrical stimulation. This was achieved by implanting two electrodes with a cannula into the same dorsal hippocampus, which allowed for simultaneous local injection of myo-inositol or saline and afterdischarges induction and recording from the same hippocampus. We found that myo-inositol has time- and concentration-dependent effects on the evoked afterdischarges. Specifically, 5 minutes after 1 M myo-inositol infusion, the afterdischarges duration was significantly decreased as compared to preinjection durations in the same animals and also as compared to preinjection level durations in saline injected or contralateral hippocampus myo-inositol infused animals. Further, 0.055 M myo-inositol significantly decreased afterdischarges duration at 5 minutes as compared to 40 minutes post-injection. At both concentrations, the afterdischarges duration recovered to the pre-injection value at 40 minutes after the myo-inositol injection. The present data, taken together with our previous results, strongly suggest that myo-inositol has significant local seizure-suppressant effect.
Collapse
|
33
|
Chhetri DR. Myo-Inositol and Its Derivatives: Their Emerging Role in the Treatment of Human Diseases. Front Pharmacol 2019; 10:1172. [PMID: 31680956 PMCID: PMC6798087 DOI: 10.3389/fphar.2019.01172] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Myo-inositol has been established as an important growth-promoting factor of mammalian cells and animals. The role of myo-inositol as a lipotropic factor has been proven, in addition to its involvement as co-factors of enzymes and as messenger molecules in signal transduction. Myo-inositol deficiency leads to intestinal lipodystrophy in animals and "inositol-less death" in some fungi. Of late, diverse uses of myo-inositol and its derivatives have been discovered in medicinal research. These compounds are used in the treatment of a variety of ailments from diabetes to cancer, and continued research in this direction promises a new future in therapeutics. In different diseases, inositols implement different strategies for therapeutic actions such as tissue specific increase or decrease in inositol products, production of inositol phosphoglycans (IPGs), conversion of myo-inositol (MI) to D-chiro-inositol (DCI), modulation of signal transduction, regulation of reactive oxygen species (ROS) production, etc. Though inositol pharmacology is a relatively lesser-known field, recent years of research has generated a critical mass of information on the subject. This review aims to summarize our current understanding on the role of inositol derivatives in ameliorating the symptoms of different diseases.
Collapse
Affiliation(s)
- Dhani Raj Chhetri
- Department of Botany, School of Life Sciences, Sikkim University, Gangtok, India
| |
Collapse
|
34
|
Li M, Xi P, Xu Y, Wang Z, Han X, Ren W, Phouthapane V, Miao J. Taurine Attenuates Streptococcus uberis-Induced Bovine Mammary Epithelial Cells Inflammation via Phosphoinositides/Ca 2+ Signaling. Front Immunol 2019; 10:1825. [PMID: 31447841 PMCID: PMC6692464 DOI: 10.3389/fimmu.2019.01825] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/18/2019] [Indexed: 12/18/2022] Open
Abstract
Taurine may alleviate the inflammatory injury induced by Streptococcus uberis (S. uberis) infection by regulating intracellular Ca2+ levels. However, the underlying mechanisms remain unclear. Infection leads to subversion of phosphoinositides (PIs) which are closely related to Ca2+ signaling. In order to investigate whether taurine regulates inflammation by means of PIs/ Ca2+ systems, competitive inhibitors of taurine (β-alanine) siTauT, siPAT1, siPLC, siCaN, siPKC, and inhibitors of PLC (U73122), PKC (RO31-8220), and CaN (FK 506) were used. The results indicate that taurine transfers the extracellular nutrient signal for intercellular innate immunity to phosphoinositides without a need to enter the cytoplasm while regulating intracellular Ca2+ levels during inflammation. Both the Ca2+-PKCα-NF-κB, and Ca2+-CaM-CaN-NFAT signaling pathways of S. uberis infection and the regulatory roles of taurine follow activation of PIs/Ca2+ systems. These data increase our understanding on the mechanisms of multifunctional nutrient, taurine attenuated inflammatory responses caused by S. uberis infection, and provide theoretical support for the prevention of this disease.
Collapse
Affiliation(s)
- Ming Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Panpan Xi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuanyuan Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhenglei Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiangan Han
- Chinese Academy of Agricultural Sciences, Shanghai Veterinary Research Institute, Shanghai, China
| | - Wenkai Ren
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, College of Animal Science, Subtropical Institute of Animal Nutrition and Feed, South China Agricultural University, Guangzhou, China
| | - Vanhnaseng Phouthapane
- Biotechnology and Ecology Institute, Ministry of Science and Technology, Vientiane, Laos
| | - Jinfeng Miao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
35
|
Cannizzaro M, Jarošová J, De Paepe B. Relevance of solute carrier family 5 transporter defects to inherited and acquired human disease. J Appl Genet 2019; 60:305-317. [PMID: 31286439 DOI: 10.1007/s13353-019-00502-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 06/21/2019] [Accepted: 06/30/2019] [Indexed: 01/12/2023]
Abstract
The solute carrier (SLC) group of membrane transport proteins is crucial for cells via their control of import and export of vital molecules across the cellular membrane. Defects in these transporters with narrow substrate specificities cause monogenic disorders, giving us essential clues of their precise roles in cellular functioning. The SLC5 family in particular has been linked to various human diseases, of mild and severe phenotype as well as high and low prevalence. In this review, we describe the effects on health of SLC5 dysfunction and dysregulation by summarizing findings in patients with transporter gene defects. Patients display a plethora of pathologies which include glucose/galactose malabsorption, familiar renal glycosuria, thyroid dyshormonogenesis, and distal hereditary motor neuronopathies. In addition, the therapeutic potential of intervening in transporter activities for treating common diseases such as diabetes and cancer is explored.
Collapse
Affiliation(s)
- Miryam Cannizzaro
- Department of Neurology & Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Jana Jarošová
- Department of Neurology & Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Boel De Paepe
- Department of Neurology & Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
36
|
Ferrucci V, de Antonellis P, Pennino FP, Asadzadeh F, Virgilio A, Montanaro D, Galeone A, Boffa I, Pisano I, Scognamiglio I, Navas L, Diana D, Pedone E, Gargiulo S, Gramanzini M, Brunetti A, Danielson L, Carotenuto M, Liguori L, Verrico A, Quaglietta L, Errico ME, Del Monaco V, D'Argenio V, Tirone F, Mastronuzzi A, Donofrio V, Giangaspero F, Picard D, Remke M, Garzia L, Daniels C, Delattre O, Swartling FJ, Weiss WA, Salvatore F, Fattorusso R, Chesler L, Taylor MD, Cinalli G, Zollo M. Metastatic group 3 medulloblastoma is driven by PRUNE1 targeting NME1-TGF-β-OTX2-SNAIL via PTEN inhibition. Brain 2019; 141:1300-1319. [PMID: 29490009 DOI: 10.1093/brain/awy039] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/16/2018] [Indexed: 01/23/2023] Open
Abstract
Genetic modifications during development of paediatric groups 3 and 4 medulloblastoma are responsible for their highly metastatic properties and poor patient survival rates. PRUNE1 is highly expressed in metastatic medulloblastoma group 3, which is characterized by TGF-β signalling activation, c-MYC amplification, and OTX2 expression. We describe the process of activation of the PRUNE1 signalling pathway that includes its binding to NME1, TGF-β activation, OTX2 upregulation, SNAIL (SNAI1) upregulation, and PTEN inhibition. The newly identified small molecule pyrimido-pyrimidine derivative AA7.1 enhances PRUNE1 degradation, inhibits this activation network, and augments PTEN expression. Both AA7.1 and a competitive permeable peptide that impairs PRUNE1/NME1 complex formation, impair tumour growth and metastatic dissemination in orthotopic xenograft models with a metastatic medulloblastoma group 3 cell line (D425-Med cells). Using whole exome sequencing technology in metastatic medulloblastoma primary tumour cells, we also define 23 common 'non-synonymous homozygous' deleterious gene variants as part of the protein molecular network of relevance for metastatic processes. This PRUNE1/TGF-β/OTX2/PTEN axis, together with the medulloblastoma-driver mutations, is of relevance for future rational and targeted therapies for metastatic medulloblastoma group 3.10.1093/brain/awy039_video1awy039media15742053534001.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,European School of Molecular Medicine (SEMM), Milan, Italy
| | - Pasqualino de Antonellis
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Ontario, Canada
| | - Francesco Paolo Pennino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | | | - Antonella Virgilio
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Naples, Italy
| | | | - Aldo Galeone
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Naples, Italy
| | | | - Ida Pisano
- CEINGE Biotecnologie Avanzate, Naples, Italy
| | | | - Luigi Navas
- Department of Veterinary Medicine and Animal Productions, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Donatella Diana
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale della Ricerca, Naples, Italy
| | - Emilia Pedone
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale della Ricerca, Naples, Italy
| | - Sara Gargiulo
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale della Ricerca, Naples, Italy
| | - Matteo Gramanzini
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale della Ricerca, Naples, Italy
| | - Arturo Brunetti
- CEINGE Biotecnologie Avanzate, Naples, Italy.,Dipartimento di Scienze Biomediche Avanzate, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Laura Danielson
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK
| | - Marianeve Carotenuto
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | | | - Antonio Verrico
- Paediatric Neurosurgery, Ospedale Santobono-Pausilipon, Naples, Italy
| | - Lucia Quaglietta
- Paediatric Neurosurgery, Ospedale Santobono-Pausilipon, Naples, Italy
| | | | | | - Valeria D'Argenio
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Felice Tirone
- Genetic Control of Development-URT, Institute of Cell Biology and Neurobiology, National Research Council, Fondazione Santa Lucia, Rome, Italy
| | - Angela Mastronuzzi
- Dipartimento di Onco-Ematologia, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Felice Giangaspero
- Dipartimento di Scienze Radiologiche, Oncologiche e Anatomo Patologiche, Università La Sapienza, Rome, Italy.,IRCCS Neuromed, Pozzilli, Italy
| | - Daniel Picard
- German Cancer Consortium (DKTK), Department of Paediatric Oncology, Haematology, and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marc Remke
- German Cancer Consortium (DKTK), Department of Paediatric Oncology, Haematology, and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Livia Garzia
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON, Canada
| | - Craig Daniels
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Ontario, Canada
| | - Olivier Delattre
- PSL Research University, Inserm U830, Equipe Labellisée Ligue contre le Cancer, Institut Curie, Paris, France
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Francesco Salvatore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy
| | - Roberto Fattorusso
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Caserta, Italy
| | - Louis Chesler
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON, Canada
| | - Giuseppe Cinalli
- Paediatric Neurosurgery, Ospedale Santobono-Pausilipon, Naples, Italy
| | - Massimo Zollo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, Naples, Italy.,CEINGE Biotecnologie Avanzate, Naples, Italy.,European School of Molecular Medicine (SEMM), Milan, Italy.,DAI-Medicina Trasfusionale-Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| |
Collapse
|
37
|
Tzingounis AV. SMITten for KCNQ Channels. Biophys J 2019; 113:503-505. [PMID: 28793205 DOI: 10.1016/j.bpj.2017.06.056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 06/25/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022] Open
|
38
|
Abstract
Voltage-gated potassium (Kv) channels open in response to changes in membrane potential to permit passage of K+ ions across the cell membrane, down their electrochemical gradient. Sodium-coupled solute transporters utilize the downhill sodium gradient to co-transport solutes, ranging from ions to sugars to neurotransmitters, into the cell. A variety of recent studies have uncovered cooperation between these two structurally and functionally unrelated classes of protein, revealing previously unnoticed functional crosstalk and in many cases physical interaction to form channel-transporter (chansporter) complexes. Adding to this field, Bartolomé-Martín and colleagues now report that the heteromeric KCNQ2/KCNQ3 (Kv7.2/7.3) potassium channel - the primary molecular correlate of the neuronal M-current - can physically interact with two sodium-coupled neurotransmitter transporters expressed in the brain, DAT and GLT1 (dopamine and glutamate transporters, respectively). The authors provide evidence that the interactions may enhance transporter activity while dampening the depolarizing effects of sodium influx. Cumulative evidence discussed here suggests that chansporter complexes represent a widespread form of cellular signaling hub, in the CNS and other tissues. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
39
|
Abstract
Polyphosphoinositides (PPIn) are essential signaling phospholipids that make remarkable contributions to the identity of all cellular membranes and signaling cascades in mammalian cells. They exert regulatory control over membrane homeostasis via selective interactions with cellular proteins at the membrane–cytoplasm interface. This review article briefly summarizes our current understanding of the key roles that PPIn play in orchestrating and regulating crucial electrical and chemical signaling events in mammalian neurons and the significant neuro-pathophysiological conditions that arise following alterations in their metabolism.
Collapse
Affiliation(s)
- Eamonn James Dickson
- Department Physiology and Membrane Biology, University of California, Davis, CA, 95616, USA
| |
Collapse
|
40
|
Voevodskaya O, Poulakis K, Sundgren P, van Westen D, Palmqvist S, Wahlund LO, Stomrud E, Hansson O, Westman E. Brain myoinositol as a potential marker of amyloid-related pathology: A longitudinal study. Neurology 2019; 92:e395-e405. [PMID: 30610093 PMCID: PMC6369900 DOI: 10.1212/wnl.0000000000006852] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 09/18/2018] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To investigate the association between longitudinal changes in proton magnetic resonance spectroscopy (MRS) metabolites and amyloid pathology in individuals without dementia, and to explore the relationship between MRS and cognitive decline. METHODS In this longitudinal multiple time point study (a subset of the Swedish BioFINDER), we included cognitively healthy participants, individuals with subjective cognitive decline, and individuals with mild cognitive impairment. MRS was acquired serially in 294 participants (670 individual spectra) from the posterior cingulate/precuneus. Using mixed-effects models, we assessed the association between MRS and baseline β-amyloid (Aβ), and between MRS and the longitudinal Mini-Mental State Examination, accounting for APOE, age, and sex. RESULTS While baseline MRS metabolites were similar in Aβ positive (Aβ+) and negative (Aβ-) individuals, in the Aβ+ group, the estimated rate of change was +1.9%/y for myo-inositol (mI)/creatine (Cr) and -2.0%/y for N-acetylaspartate (NAA)/mI. In the Aβ- group, mI/Cr and NAA/mI yearly change was -0.05% and +1.2%; however, this was not significant across time points. The mild cognitive impairment Aβ+ group showed the steepest MRS changes, with an estimated rate of +2.93%/y (p = 0.07) for mI/Cr and -3.55%/y (p < 0.01) for NAA/mI. Furthermore, in the entire cohort, we found that Aβ+ individuals with low baseline NAA/mI had a significantly higher rate of cognitive decline than Aβ+ individuals with high baseline NAA/mI. CONCLUSION We demonstrate that the longitudinal change in mI/Cr and NAA/mI is associated with underlying amyloid pathology. MRS may be a useful noninvasive marker of Aβ-related processes over time. In addition, we show that in Aβ+ individuals, baseline NAA/mI may predict the rate of future cognitive decline.
Collapse
Affiliation(s)
- Olga Voevodskaya
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK.
| | - Konstantinos Poulakis
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Pia Sundgren
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Danielle van Westen
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Sebastian Palmqvist
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Lars-Olof Wahlund
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Erik Stomrud
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Oskar Hansson
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Eric Westman
- From the Division of Clinical Geriatrics (O.V., K.P., L.-O.W., E.W.), Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm; Department of Diagnostic Radiology (P.S., D.v.W.), Lund University; Imaging and Function (D.v.W.), Skåne University Health Care, Lund; Clinical Memory Research Unit (S.P., E.S., O.H.), Department of Clinical Sciences, Malmö, Lund University; Memory Clinic (E.S., O.H.), Skåne University Hospital, Malmö, Sweden; and Department of Neuroimaging (E.W.), Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| |
Collapse
|
41
|
Hosseiniyan Khatibi SM, Zununi Vahed F, Sharifi S, Ardalan M, Mohajel Shoja M, Zununi Vahed S. Osmolytes resist against harsh osmolarity: Something old something new. Biochimie 2019; 158:156-164. [PMID: 30629975 DOI: 10.1016/j.biochi.2019.01.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/03/2019] [Indexed: 12/14/2022]
Abstract
From the halophilic bacteria to human, cells have to survive under the stresses of harsh environments. Hyperosmotic stress is a process that triggers cell shrinkage, oxidative stress, DNA damage, and apoptosis and it potentially contributes to a number of human diseases. Remarkably, by high salts and organic solutes concentrations, a variety of organisms struggle with these conditions. Different strategies have been developed for cellular osmotic adaptations among which organic osmolyte synthesis/accumulation is a conserved once. Osmolytes are naturally occurring solutes used by cells of several halophilic (micro) organisms to preserve cell volume and function. In this review, the osmolytes diversity and their protective roles in harsh hyperosmolar environments from bacteria to human cells are highlighted. Moreover, it provides a close look at mammalian kidney osmoregulation at a molecular level. This review provides a concise view on the recent developments and advancements on the applications of osmolytes. Identification of disease-related osmolytes and their targeted-delivery may be used as a therapeutic measurement for treatment of the pathological conditions and the inherited diseases related to protein misfolding and aggregation. The molecular and cellular aspects of cell adaptation against harsh environmental osmolarity will benefit the development of effective drugs for many diseases.
Collapse
Affiliation(s)
| | | | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|
42
|
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
|
43
|
Cheng L, Wang X, Liu T, Tse G, Fu H, Li G. Modulation of Ion Channels in the Superior Cervical Ganglion Neurons by Myocardial Ischemia and Fluvastatin Treatment. Front Physiol 2018; 9:1157. [PMID: 30246810 PMCID: PMC6139347 DOI: 10.3389/fphys.2018.01157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/02/2018] [Indexed: 01/08/2023] Open
Abstract
Background: The superior cervical ganglion (SCG) of the autonomic nervous system plays an important role in different cardiovascular diseases. In this study, we investigated the effects of ischemia and fluvastatin treatment on the ion channel characteristics of SCG neurons in a rabbit myocardial ischemia (MI) model. Methods: MI was induced by abdominal subcutaneous injections of isoproterenol (ISO). The properties of the delayed rectifier potassium channel current (IK), sodium channel current (INa), and action potential (APs) on isolated SCG neurons in the control, MI-7d, MI-14d, fluvastatin-7d (fluvastatin pretreated 14 days and treated 7 days after ISO-induced MI), and fluvastatin-14d (fluvastatin pretreated 14 days and treated 14 days after ISO-induced MI) groups were studied. In addition, the RNA expressions of KCNQ3 and SCN9A in the SCG tissue were determined by performing real-time PCR. Intracellular calcium concentration was monitored using laser scanning confocal microscopy. Results: Compared with the control group, the current amplitude of IK and INa were increased in the MI-7d and MI-14d groups. KCNQ3 RNA (corresponding to channel proteins of IK) expression and SCN9A RNA (corresponding to channel proteins of INa) expression were also increased in MI groups. Activation and inactivation curves for INa in the two MI groups shifted negatively compared with the control group. These changes were reversed by fluvastatin treatment. Intracellular calcium concentration in SCG neurons was not altered significantly by MI or fluvastatin treatment. By contrast, increased AP amplitude and shortened APD90 were observed in the MI-7d and MI-14d groups. These changes were reversed in the fluvastatin-treated MI group. Conclusion: Fluvastatin treatment partly reversed the characteristics of SCG neurons in MI. The ion channel of SCG neurons could be one of the potential targets of fluvastatin in treating coronary heart diseases.
Collapse
Affiliation(s)
- Lijun Cheng
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Xinghua Wang
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Tong Liu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Gary Tse
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Huaying Fu
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Department of Cardiology, Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
44
|
Hilgemann DW, Dai G, Collins A, Lariccia V, Magi S, Deisl C, Fine M. Lipid signaling to membrane proteins: From second messengers to membrane domains and adapter-free endocytosis. J Gen Physiol 2018; 150:211-224. [PMID: 29326133 PMCID: PMC5806671 DOI: 10.1085/jgp.201711875] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Hilgemann et al. explain how lipid signaling to membrane proteins involves a hierarchy of mechanisms from lipid binding to membrane domain coalescence. Lipids influence powerfully the function of ion channels and transporters in two well-documented ways. A few lipids act as bona fide second messengers by binding to specific sites that control channel and transporter gating. Other lipids act nonspecifically by modifying the physical environment of channels and transporters, in particular the protein–membrane interface. In this short review, we first consider lipid signaling from this traditional viewpoint, highlighting innumerable Journal of General Physiology publications that have contributed to our present understanding. We then switch to our own emerging view that much important lipid signaling occurs via the formation of membrane domains that influence the function of channels and transporters within them, promote selected protein–protein interactions, and control the turnover of surface membrane.
Collapse
Affiliation(s)
- Donald W Hilgemann
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Gucan Dai
- Department of Physiology and Biophysics, University of Washington, Seattle, WA
| | - Anthony Collins
- Saba University School of Medicine, The Bottom, Saba, Dutch Caribbean
| | - Vincenzo Lariccia
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche," Ancona, Italy
| | - Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University "Politecnica delle Marche," Ancona, Italy
| | - Christine Deisl
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Michael Fine
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
45
|
SMIT1 Modifies KCNQ Channel Function and Pharmacology by Physical Interaction with the Pore. Biophys J 2017; 113:613-626. [PMID: 28793216 DOI: 10.1016/j.bpj.2017.06.055] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/26/2017] [Accepted: 06/12/2017] [Indexed: 11/22/2022] Open
Abstract
Voltage-gated potassium channels of the KCNQ (Kv7) subfamily are essential for control of cellular excitability and repolarization in a wide range of cell types. Recently, we and others found that some KCNQ channels functionally and physically interact with sodium-dependent solute transporters, including myo-inositol transporters SMIT1 and SMIT2, potentially facilitating various modes of channel-transporter signal integration. In contrast to indirect effects such as channel regulation by SMIT-transported, myo-inositol-derived phosphatidylinositol 4,5-bisphosphate (PIP2), the mechanisms and functional consequences of the physical interaction of channels with transporters have been little studied. Here, using co-immunoprecipitation with different channel domains, we found that SMIT1 binds to the KCNQ2 pore module. We next tested the effects of SMIT1 co-expression, in the absence of extracellular myo-inositol or other SMIT1 substrates, on fundamental functional attributes of KCNQ2, KCNQ2/3, KCNQ1, and KCNQ1-KCNE1 channels. Without exception, SMIT1 altered KCNQ ion selectivity, sensitivity to extracellular K+, and pharmacology, consistent with an impact on conformation of the KCNQ pore. SMIT1 also altered the gating kinetics and/or voltage dependence of KCNQ2, KCNQ2/3, and KCNQ1-KCNE1. In contrast, SMIT1 had no effect on Kv1.1 (KCNA1) gating, ion selectivity, or pharmacology. We conclude that, independent of its transport activity and indirect regulatory mechanisms involving inositol-derived increases in PIP2, SMIT1, and likely other related sodium-dependent solute transporters, regulates KCNQ channel ion selectivity, gating, and pharmacology by direct physical interaction with the pore module.
Collapse
|
46
|
Lacin E, Aryal P, Glaaser IW, Bodhinathan K, Tsai E, Marsh N, Tucker SJ, Sansom MSP, Slesinger PA. Dynamic role of the tether helix in PIP 2-dependent gating of a G protein-gated potassium channel. J Gen Physiol 2017; 149:799-811. [PMID: 28720589 PMCID: PMC5560777 DOI: 10.1085/jgp.201711801] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/06/2017] [Accepted: 06/21/2017] [Indexed: 01/21/2023] Open
Abstract
G protein–gated inwardly rectifying potassium (GIRK) channels are activated by the phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2). Using functional and computational experiments, Lacin et al. reveal that PIP2 interacts with the tether helix of the neuronal GIRK channel in a dynamic way. G protein–gated inwardly rectifying potassium (GIRK) channels control neuronal excitability in the brain and are implicated in several different neurological diseases. The anionic phospholipid phosphatidylinositol 4,5 bisphosphate (PIP2) is an essential cofactor for GIRK channel gating, but the precise mechanism by which PIP2 opens GIRK channels remains poorly understood. Previous structural studies have revealed several highly conserved, positively charged residues in the “tether helix” (C-linker) that interact with the negatively charged PIP2. However, these crystal structures of neuronal GIRK channels in complex with PIP2 provide only snapshots of PIP2’s interaction with the channel and thus lack details about the gating transitions triggered by PIP2 binding. Here, our functional studies reveal that one of these conserved basic residues in GIRK2, Lys200 (6′K), supports a complex and dynamic interaction with PIP2. When Lys200 is mutated to an uncharged amino acid, it activates the channel by enhancing the interaction with PIP2. Atomistic molecular dynamic simulations of neuronal GIRK2 with the same 6′ substitution reveal an open GIRK2 channel with PIP2 molecules adopting novel positions. This dynamic interaction with PIP2 may explain the intrinsic low open probability of GIRK channels and the mechanism underlying activation by G protein Gβγ subunits and ethanol.
Collapse
Affiliation(s)
- Emre Lacin
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Prafulla Aryal
- Department of Biochemistry, University of Oxford, Oxford, England, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK
| | - Ian W Glaaser
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Eric Tsai
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nidaa Marsh
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stephen J Tucker
- OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK.,Department of Physics, University of Oxford, Oxford, England, UK
| | - Mark S P Sansom
- Department of Biochemistry, University of Oxford, Oxford, England, UK.,OXION Initiative in Ion Channels and Disease, University of Oxford, Oxford, England, UK
| | - Paul A Slesinger
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY .,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
47
|
Abbott GW. Chansporter complexes in cell signaling. FEBS Lett 2017; 591:2556-2576. [PMID: 28718502 DOI: 10.1002/1873-3468.12755] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 12/11/2022]
Abstract
Ion channels facilitate diffusion of ions across cell membranes for such diverse purposes as neuronal signaling, muscular contraction, and fluid homeostasis. Solute transporters often utilize ionic gradients to move aqueous solutes up their concentration gradient, also fulfilling a wide variety of tasks. Recently, an increasing number of ion channel-transporter ('chansporter') complexes have been discovered. Chansporter complex formation may overcome what could otherwise be considerable spatial barriers to rapid signal integration and feedback between channels and transporters, the ions and other substrates they transport, and environmental factors to which they must respond. Here, current knowledge in this field is summarized, covering both heterologous expression structure/function findings and potential mechanisms by which chansporter complexes fulfill contrasting roles in cell signaling in vivo.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
48
|
Cárdenas AM, Fernández-Olivares P, Díaz-Franulic I, González-Jamett AM, Shimahara T, Segura-Aguilar J, Caviedes R, Caviedes P. Knockdown of Myo-Inositol Transporter SMIT1 Normalizes Cholinergic and Glutamatergic Function in an Immortalized Cell Line Established from the Cerebral Cortex of a Trisomy 16 Fetal Mouse, an Animal Model of Human Trisomy 21 (Down Syndrome). Neurotox Res 2017; 32:614-623. [PMID: 28695546 DOI: 10.1007/s12640-017-9775-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022]
Abstract
The Na+/myo-inositol cotransporter (SMIT1) is overexpressed in human Down syndrome (DS) and in trisomy 16 fetal mice (Ts16), an animal model of the human condition. SMIT1 overexpression determines increased levels of intracellular myo-inositol, a precursor of phophoinositide synthesis. SMIT1 is overexpressed in CTb cells, an immortalized cell line established from the cerebral cortex of a Ts16 mouse fetus. CTb cells exhibit impaired cytosolic Ca2+ signals in response to glutamatergic and cholinergic stimuli (increased amplitude and delayed time-dependent kinetics in the decay post-stimulation), compared to our CNh cell line, derived from the cerebral cortex of a euploid animal. Considering the role of myo-inositol in intracellular signaling, we normalized SMIT1 expression in CTb cells using specific mRNA antisenses. Forty-eight hours post-transfection, SMIT1 levels in CTb cells reached values comparable to those of CNh cells. At this time, decay kinetics of Ca2+ signals induced by either glutamate, nicotine, or muscarine were accelerated in transfected CTb cells, to values similar to those of CNh cells. The amplitude of glutamate-induced cytosolic Ca2+ signals in CTb cells was also normalized. The results suggest that SMIT1 overexpression contributes to abnormal cholinergic and glutamatergic Ca2+ signals in the trisomic condition, and knockdown of DS-related genes in our Ts16-derived cell line could constitute a relevant tool to study DS-related neuronal dysfunction.
Collapse
Affiliation(s)
- Ana María Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Paola Fernández-Olivares
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Clasificador 7, Independencia, 1027, Santiago, Chile
| | - Ignacio Díaz-Franulic
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Center for Bioinformatics and Integrative Biology, Universidad Andrés Bello, Santiago, Chile
- Fundación Fraunhofer Chile, Las Condes, Chile
| | - Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | | | - Juan Segura-Aguilar
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Clasificador 7, Independencia, 1027, Santiago, Chile
| | - Raúl Caviedes
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Clasificador 7, Independencia, 1027, Santiago, Chile
| | - Pablo Caviedes
- Program of Molecular and Clinical Pharmacology, ICBM, Faculty of Medicine, University of Chile, Clasificador 7, Independencia, 1027, Santiago, Chile.
| |
Collapse
|
49
|
Li SYT, Cheng STW, Zhang D, Leung PS. Identification and Functional Implications of Sodium/ Myo-Inositol Cotransporter 1 in Pancreatic β-Cells and Type 2 Diabetes. Diabetes 2017; 66:1258-1271. [PMID: 28202581 DOI: 10.2337/db16-0880] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 02/07/2017] [Indexed: 11/13/2022]
Abstract
Myo-inositol (MI), the precursor of the second messenger phosphoinositide (PI), mediates multiple cellular events. Rat islets exhibit active transport of MI, although the mechanism involved remains elusive. Here, we report, for the first time, the expression of sodium/myo-inositol cotransporter 1 (SMIT1) in rat islets and, specifically, β-cells. Genetic or pharmacological inhibition of SMIT1 impaired glucose-stimulated insulin secretion by INS-1E cells, probably via downregulation of PI signaling. In addition, SMIT1 expression in INS-1E cells and isolated islets was augmented by acute high-glucose exposure and reduced in chronic hyperglycemia conditions. In corroboration, chronic MI treatment improved the disease phenotypes of diabetic rats and islets. On the basis of our results, we postulate that the MI transporter SMIT1 is required to maintain a stable PI pool in β-cells in order that PI remains available despite its rapid turnover.
Collapse
Affiliation(s)
- Stephen Yu Ting Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Sam Tsz Wai Cheng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dan Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Po Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
50
|
Traynor-Kaplan A, Kruse M, Dickson EJ, Dai G, Vivas O, Yu H, Whittington D, Hille B. Fatty-acyl chain profiles of cellular phosphoinositides. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:513-522. [PMID: 28189644 PMCID: PMC5392126 DOI: 10.1016/j.bbalip.2017.02.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/31/2017] [Accepted: 02/07/2017] [Indexed: 12/24/2022]
Abstract
Phosphoinositides are rapidly turning-over phospholipids that play key roles in intracellular signaling and modulation of membrane effectors. Through technical refinements we have improved sensitivity in the analysis of the phosphoinositide PI, PIP, and PIP2 pools from living cells using mass spectrometry. This has permitted further resolution in phosphoinositide lipidomics from cell cultures and small samples of tissue. The technique includes butanol extraction, derivatization of the lipids, post-column infusion of sodium to stabilize formation of sodiated adducts, and electrospray ionization mass spectrometry in multiple reaction monitoring mode, achieving a detection limit of 20pg. We describe the spectrum of fatty-acyl chains in the cellular phosphoinositides. Consistent with previous work in other mammalian primary cells, the 38:4 fatty-acyl chains dominate in the phosphoinositides of the pineal gland and of superior cervical ganglia, and many additional fatty acid combinations are found at low abundance. However, Chinese hamster ovary cells and human embryonic kidney cells (tsA201) in culture have different fatty-acyl chain profiles that change with growth state. Their 38:4 lipids lose their dominance as cultures approach confluence. The method has good time resolution and follows well the depletion in <20s of both PIP2 and PIP that results from strong activation of Gq-coupled receptors. The receptor-activated phospholipase C exhibits no substrate selectivity among the various fatty-acyl chain combinations.
Collapse
Affiliation(s)
- Alexis Traynor-Kaplan
- ATK Innovation, Analytics and Discovery, North Bend, WA 98045, USA; Department of Medicine/Gastroenterology, University of Washington School of Medicine, Seattle, WA, USA.
| | - Martin Kruse
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Eamonn J Dickson
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Gucan Dai
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Oscar Vivas
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Haijie Yu
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington School of Medicine, Seattle, WA, USA
| | - Bertil Hille
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|