1
|
van Veggel L, Voets AM, Vanmierlo T, Schreiber R. Insights from an academic endeavor into central nervous system drug discovery. Neural Regen Res 2025; 20:1717-1718. [PMID: 39104109 PMCID: PMC11688561 DOI: 10.4103/nrr.nrr-d-24-00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/08/2024] [Accepted: 05/24/2024] [Indexed: 08/07/2024] Open
Affiliation(s)
- Lieve van Veggel
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - An M. Voets
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Tim Vanmierlo
- BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Rudy Schreiber
- Section of Psychopharmacology, Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
2
|
Nair LV, Nair RV, Ahmad Lone B, Shenoy SJ, Jayasree RS. Atomically Precise Fluorescent Gold Nanocluster as a Barrier-Permeable and Brain-Specific Imaging Probe. Chem Asian J 2025; 20:e202400590. [PMID: 39380543 DOI: 10.1002/asia.202400590] [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: 05/24/2024] [Revised: 09/28/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Photonic nanomaterials play a crucial role in facilitating the necessary signal for optical brain imaging, presenting a promising avenue for early diagnosis of brain-related disorders. However, the blood-brain barrier (BBB) presents a significant challenge, blocking the entry of most molecules or materials from the bloodstream into the brain. To overcome this, photonic nanocrystals in the form of gold clusters (LAuC) with size less than 3 nm, have been developed, with Levodopa conjugated to LAuC (Dop@LAuC) for targeted brain imaging. Dop@LAuC crosses the BBB and emits in the near-infrared (NIR) wavelength, enabling real-time optical brain imaging. An in vitro BBB model using brain endothelial cells showed that 50 % of Dop@LAuC crossed the barrier within 3 hours, compared to only 10 % of LAuC, highlighting the enhanced ability of L-dopa-conjugated gold clusters to penetrate the BBB. In vivo optical imaging in healthy mice further confirmed the material's efficacy to cross BBB without compromising the barrier integrity.
Collapse
Affiliation(s)
- Lakshmi V Nair
- Division of Biophotonics and Imaging, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, 695012, India
- School of Mechanical and Materials Engineering, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Resmi V Nair
- Division of Biophotonics and Imaging, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, 695012, India
- Programmable Molecular Design Lab, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, 560064, India
| | - Bilal Ahmad Lone
- Division of Biophotonics and Imaging, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, 695012, India
| | - Sachin J Shenoy
- Division ofin vivomodels and Testing, Division of Molecular Medicine, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, 695012, India
| | - Ramapurath S Jayasree
- Division of Biophotonics and Imaging, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum, 695012, India
| |
Collapse
|
3
|
Ardiles NM, Tapia-Cuevas V, Estay SF, Alcaino A, Velásquez VB, Sotomayor-Zárate R, Chávez AE, Moya PR. Increased forebrain EAAT3 expression confers resilience to chronic stress. J Neurochem 2025; 169:e16216. [PMID: 39245629 DOI: 10.1111/jnc.16216] [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: 04/02/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024]
Abstract
Depression is a disabling and highly prevalent psychiatric illness. Multiple studies have linked glutamatergic dysfunction with the pathophysiology of depression, but the exact alterations in the glutamatergic system that contribute to depressive-like behaviors are not fully understood. Recent evidence suggests that a decreased level in neuronal glutamate transporter (EAAT3), known to control glutamate levels and limit the activation of glutamate receptors at synaptic sites, may contribute to the manifestation of a depressive phenotype. Here, we tested the possibility that increased EAAT3 expression at excitatory synapses could reduce the susceptibility of mice to develop depressive-like behaviors when challenged to a 5-week unpredictable chronic mild stress (UCMS) protocol. Mice overexpressing EAAT3 in the forebrain (EAAT3glo/CMKII) and control littermates (EAAT3glo) were assessed for depressive-like behaviors and long-term memory performance after being subjected to UCMS conditions. We found that, after UCMS, EAAT3glo/CMKII mice did not exhibit depressive-like behaviors or memory alterations observed in control mice. Moreover, we found that EAAT3glo/CMKII mice did not show alterations in phasic dopamine release in the nucleus accumbens neither in long-term synaptic plasticity in the CA1 region of the hippocampus after UCMS, as observed in control littermates. Altogether these results suggest that forebrain EAAT3 overexpression may be related to a resilient phenotype, both at behavioral and functional level, to the deleterious effect of chronic stress, highlighting the importance of neuronal EAAT3 in the pathophysiology of depressive-like behaviors.
Collapse
Affiliation(s)
- Nicolás M Ardiles
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
| | - Vissente Tapia-Cuevas
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
| | - Sebastián F Estay
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alejandro Alcaino
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Victoria B Velásquez
- Programa de Doctorado en Ciencias, Mención Neurociencia, Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurobiología y Fisipatología Integrativa (CENFI), Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Neurobiología y Fisipatología Integrativa (CENFI), Universidad de Valparaíso, Valparaíso, Chile
| | - Andrés E Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo R Moya
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Facultad de Ciencias, Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
- Centro de Estudios Traslacionales en Estrés y Salud Mental (C-ESTRES), Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
4
|
Zhang YD, Shi DD, Wang Z. Neurobiology of Obsessive-Compulsive Disorder from Genes to Circuits: Insights from Animal Models. Neurosci Bull 2024; 40:1975-1994. [PMID: 38982026 PMCID: PMC11625044 DOI: 10.1007/s12264-024-01252-9] [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: 12/14/2023] [Accepted: 03/27/2024] [Indexed: 07/11/2024] Open
Abstract
Obsessive-compulsive disorder (OCD) is a chronic, severe psychiatric disorder that has been ranked by the World Health Organization as one of the leading causes of illness-related disability, and first-line interventions are limited in efficacy and have side-effect issues. However, the exact pathophysiology underlying this complex, heterogeneous disorder remains unknown. This scenario is now rapidly changing due to the advancement of powerful technologies that can be used to verify the function of the specific gene and dissect the neural circuits underlying the neurobiology of OCD in rodents. Genetic and circuit-specific manipulation in rodents has provided important insights into the neurobiology of OCD by identifying the molecular, cellular, and circuit events that induce OCD-like behaviors. This review will highlight recent progress specifically toward classic genetic animal models and advanced neural circuit findings, which provide theoretical evidence for targeted intervention on specific molecular, cellular, and neural circuit events.
Collapse
Affiliation(s)
- Ying-Dan Zhang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Dong-Dong Shi
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| | - Zhen Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
- Shanghai Intelligent Psychological Evaluation and Intervention Engineering Technology Research Center, Shanghai, 200030, China.
| |
Collapse
|
5
|
Liu Y, Zhao Y, Ma J, Guo S, Gao X, Wang B, Gong L, Lv Z, Guo Y. Optimal glycine allowance levels in low-protein diets and the dynamic requirement model for broilers. Poult Sci 2024; 103:104255. [PMID: 39332340 PMCID: PMC11467656 DOI: 10.1016/j.psj.2024.104255] [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: 06/06/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/29/2024] Open
Abstract
This study aimed to investigate the effects of different glycine levels in low-protein diets on the growth, nitrogen deposition, and expression of intestinal amino acid and glucose transporters in broilers from 29 to 42 d of age, in order to determine the optimal glycine supplementation level. A total of 240 male broilers at 29 days old were randomly assigned to 5 groups: the control group with a crude protein level of 20%, and experimental groups with low-protein diets (LP130) containing 18% crude protein, supplemented with glycine to achieve standardized ileal digestible (SID) glycine + serine to lysine ratios of 134% (LP134), 140% (LP140), and 145% (LP145). The results showed that the LP134 group had similar growth performance and slaughter performance compared to the control group (P > 0.05), whereas other low-protein diet groups had significantly lower growth performance (P < 0.05). Regression analysis determined that the optimal ratio for SID glycine + serine to lysine was 137%. A dynamic model for glycine + serine requirements was established through binary regression analysis: y = 599.051 × BW^0.75 + 8.381 × ADG (R2 = 0.998, P < 0.001). Feeding LP134, LP140, and LP145 diets significantly improved nitrogen deposition rates in broilers (P < 0.05). Low-protein diets significantly upregulated mRNA levels of b0,+AT, EAAT3, and SGLT1 genes in the duodenum (P < 0.05). In conclusion, appropriate glycine supplementation in low-protein diets can enhance growth performance, and nitrogen deposition efficiency, and regulate the expression of intestinal amino acid and glucose transporters. The optimal ratio of SID glycine + serine to lysine in low-protein diets for broilers aged 29 to 42 d is 137%.
Collapse
Affiliation(s)
- Yongfa Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yizhu Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiran Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Siyan Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xuyang Gao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Bin Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lu Gong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zengpeng Lv
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
6
|
Camporesi S, Xin L, Golay P, Eap CB, Cleusix M, Cuenod M, Fournier M, Hashimoto K, Jenni R, Ramain J, Restellini R, Solida A, Conus P, Do KQ, Khadimallah I. Neurocognition and NMDAR co-agonists pathways in individuals with treatment resistant first-episode psychosis: a 3-year follow-up longitudinal study. Mol Psychiatry 2024; 29:3669-3679. [PMID: 38849515 PMCID: PMC11541217 DOI: 10.1038/s41380-024-02631-4] [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: 01/09/2024] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/09/2024]
Abstract
This study aims to determine whether 1) individuals with treatment-resistant schizophrenia display early cognitive impairment compared to treatment-responders and healthy controls and 2) N-methyl-D-aspartate-receptor hypofunction is an underlying mechanism of cognitive deficits in treatment-resistance. In this case‒control 3-year-follow-up longitudinal study, n = 697 patients with first-episode psychosis, aged 18 to 35, were screened for Treatment Response and Resistance in Psychosis criteria through an algorithm that assigns patients to responder, limited-response or treatment-resistant category (respectively resistant to 0, 1 or 2 antipsychotics). Assessments at baseline: MATRICS Consensus Cognitive Battery; N-methyl-D-aspartate-receptor co-agonists biomarkers in brain by MRS (prefrontal glutamate levels) and plasma (D-serine and glutamate pathways key markers). Patients were compared to age- and sex-matched healthy controls (n = 114). Results: patient mean age 23, 27% female. Treatment-resistant (n = 51) showed lower scores than responders (n = 183) in processing speed, attention/vigilance, working memory, verbal learning and visual learning. Limited responders (n = 59) displayed an intermediary phenotype. Treatment-resistant and limited responders were merged in one group for the subsequent D-serine and glutamate pathway analyses. This group showed D-serine pathway dysregulation, with lower levels of the enzymes serine racemase and serine-hydroxymethyltransferase 1, and higher levels of the glutamate-cysteine transporter 3 than in responders. Better cognition was associated with higher D-serine and lower glutamate-cysteine transporter 3 levels only in responders; this association was disrupted in the treatment resistant group. Treatment resistant patients and limited responders displayed early cognitive and persistent functioning impairment. The dysregulation of NMDAR co-agonist pathways provides underlying molecular mechanisms for cognitive deficits in treatment-resistant first-episode psychosis. If replicated, our findings would open ways to mechanistic biomarkers guiding response-based patient stratification and targeting cognitive improvement in clinical trials.
Collapse
Affiliation(s)
- Sara Camporesi
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Department of psychiatry and Emergency Department, Geneva University Hospital, Geneva, Switzerland
| | - Lijing Xin
- Center for Biomedical Imaging, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Philippe Golay
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Chin Bin Eap
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
- Center for Research and Innovation in Clinical Pharmaceutical Sciences, University of Lausanne, Lausanne, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Martine Cleusix
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Michel Cuenod
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Margot Fournier
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Raoul Jenni
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Julie Ramain
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Training and Research Institute in Mental Health (IFRSM), Neuchâtel Centre of Psychiatry, Neuchâtel, Switzerland
| | - Romeo Restellini
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Emergency medicine department, Geneva University Hospital, Geneva, Switzerland
| | - Alessandra Solida
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
- Psychiatry Department for Adults 2, Neuchâtel Centre of Psychiatry, Prefargier, Switzerland
| | - Philippe Conus
- Service of General Psychiatry, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Kim Q Do
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland
| | - Ines Khadimallah
- Center for Psychiatric Neuroscience, Department of Psychiatry, Lausanne University Hospital, Lausanne, Switzerland.
- Translational Research Center, University Hospital of Psychiatry, University of Bern, Bern, Switzerland.
| |
Collapse
|
7
|
Olukosi OA, Philippi H, Veluri S, Kasireddy B, Ajao AM, Pilevar M, Oluseyifunmi IW. Assessment of two diet types in reduced-crude protein diets with or without phytase supplementation - implications on key phenotypic responses in 21-day-old broiler chickens. Br Poult Sci 2024:1-10. [PMID: 39399981 DOI: 10.1080/00071668.2024.2412136] [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: 05/23/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
1. Two concurrent experiments were conducted to investigate the effect of using the crude protein (CP) value of supplemental amino acids (AA) in formulating reduced-crude protein (RCP) diets. The RCP diets formulated without accounting for CP values of supplemental AA (RCPN) or otherwise (RCPY) or a positive control (PC) diet were fed without (Experiment 1) or with (Experiment 2) phytase.2. Each experiment utilised 105 male broiler chicks. Birds were provided a common starter diet from d 0-7. On d 21, ileal digesta were collected from the distal half of the ileum. For mRNA expression analysis, tissues were collected from the mid-jejunum and the liver. Excreta grab samples were collected for analysis for N content.3. In Experiment 1, there was a stepwise decrease (p < 0.01) in weight gain and excreta N for birds receiving PC, RCPN and RCPY diets. The coefficients of ileal digestibility of His, Leu, Phe and Trp were greater (p < 0.05) in birds that received RCPY rather than the PC diets. The relative mRNA expression of CAT1 was greater (p < 0.05) for birds that received the PC diet.4. In Experiment 2, growth performance and excreta N were not different between the PC and RCPN diets, but weight gain, feed intake and excreta N were greater (p < 0.01) in birds receiving PC or RCPN diets. The coefficients of digestibility were greater (p < 0.01) in RCP than PC diets for Lys, Thr, Cys, Gly and Ser. The mRNA expression for S6kinase and PRKAβ2 was greater (p < 0.05) for birds fed RCPN compared to PC.5. In conclusion, accounting for the N content of supplemental AA during feed formulation for RCP diets will influence the effect of CP reduction on growth performance and ileal amino acid digestibility.
Collapse
Affiliation(s)
- O A Olukosi
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - H Philippi
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - S Veluri
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - B Kasireddy
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - A M Ajao
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - M Pilevar
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| | - I W Oluseyifunmi
- Department of Poultry Science, University of Georgia, Athens, GA, USA
| |
Collapse
|
8
|
Qiu B, Boudker O. Structural basis of the excitatory amino acid transporter 3 substrate recognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611541. [PMID: 39282329 PMCID: PMC11398500 DOI: 10.1101/2024.09.05.611541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Excitatory amino acid transporters (EAATs) reside on cell surfaces and uptake substrates, including L-glutamate, L-aspartate, and D-aspartate, using ion gradients. Among five EAATs, EAAT3 is the only isoform that can efficiently transport L-cysteine, a substrate for glutathione synthesis. Recent work suggests that EAAT3 also transports the oncometabolite R-2-hydroxyglutarate (R-2HG). Here, we examined the structural basis of substrate promiscuity by determining the cryo-EM structures of EAAT3 bound to different substrates. We found that L-cysteine binds to EAAT3 in thiolate form, and EAAT3 recognizes different substrates by fine-tuning local conformations of the coordinating residues. However, using purified human EAAT3, we could not observe R-2HG binding or transport. Imaging of EAAT3 bound to L-cysteine revealed several conformational states, including an outward-facing state with a semi-open gate and a disrupted sodium-binding site. These structures illustrate that the full gate closure, coupled with the binding of the last sodium ion, occurs after substrate binding. Furthermore, we observed that different substrates affect how the transporter distributes between a fully outward-facing conformation and intermediate occluded states on a path to the inward-facing conformation, suggesting that translocation rates are substrate-dependent.
Collapse
Affiliation(s)
- Biao Qiu
- Department of Physiology & Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
- Howard Hughes Medical Institute, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| | - Olga Boudker
- Department of Physiology & Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
- Howard Hughes Medical Institute, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| |
Collapse
|
9
|
Lin Z, Yang S, Qiu Q, Cui G, Zhang Y, Yao M, Li X, Chen C, Gu J, Wang T, Yin P, Sun L, Hao Y. Hypoxia-induced cysteine metabolism reprogramming is crucial for the tumorigenesis of colorectal cancer. Redox Biol 2024; 75:103286. [PMID: 39079386 PMCID: PMC11340627 DOI: 10.1016/j.redox.2024.103286] [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: 07/05/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Metabolic reprogramming is a hallmark of human cancer, and cancer-specific metabolism provides opportunities for cancer diagnosis, prognosis, and treatment. However, the underlying mechanisms by which metabolic pathways affect the initiation and progression of colorectal cancer (CRC) remain largely unknown. Here, we demonstrate that cysteine is highly enriched in colorectal tumors compared to adjacent non-tumor tissues, thereby promoting tumorigenesis of CRC. Synchronously importing both cysteine and cystine in colorectal cancer cells is necessary to maintain intracellular cysteine levels. Hypoxia-induced reactive oxygen species (ROS) and ER stress regulate the co-upregulation of genes encoding cystine transporters (SLC7A11, SLC3A2) and genes encoding cysteine transporters (SLC1A4, SLC1A5) through the transcription factor ATF4. Furthermore, the metabolic flux from cysteine to reduced glutathione (GSH), which is critical to support CRC growth, is increased due to overexpression of glutathione synthetase GSS in CRC. Depletion of cystine/cysteine by recombinant cyst(e)inase effectively inhibits the growth of colorectal tumors by inducing autophagy in colorectal cancer cells through mTOR-ULK signaling axis. This study demonstrates the underlying mechanisms of cysteine metabolism in tumorigenesis of CRC, and evaluates the potential of cysteine metabolism as a biomarker or a therapeutic target for CRC.
Collapse
Affiliation(s)
- Zhang Lin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Shiyi Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Qianqian Qiu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, 200433, China
| | - Gaoping Cui
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Yanhua Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Meilian Yao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Xiangyu Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Chengkun Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Jun Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Ting Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China
| | - Peng Yin
- Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research, College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, China
| | - Longci Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China; Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Yujun Hao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Biomedical Engineering, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200032, China.
| |
Collapse
|
10
|
Borba JV, Canzian J, Resmim CM, Silva RM, Duarte MCF, Mohammed KA, Schoenau W, Adedara IA, Rosemberg DB. Towards zebrafish models to unravel translational insights of obsessive-compulsive disorder: A neurobehavioral perspective. Neurosci Biobehav Rev 2024; 162:105715. [PMID: 38734195 DOI: 10.1016/j.neubiorev.2024.105715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/08/2024] [Accepted: 05/04/2024] [Indexed: 05/13/2024]
Abstract
Obsessive-compulsive disorder (OCD) is a chronic and debilitating illness that has been considered a polygenic and multifactorial disorder, challenging effective therapeutic interventions. Although invaluable advances have been obtained from human and rodent studies, several molecular and mechanistic aspects of OCD etiology are still obscure. Thus, the use of non-traditional animal models may foster innovative approaches in this field, aiming to elucidate the underlying mechanisms of disease from an evolutionary perspective. The zebrafish (Danio rerio) has been increasingly considered a powerful organism in translational neuroscience research, especially due to the intrinsic features of the species. Here, we outline target mechanisms of OCD for translational research, and discuss how zebrafish-based models can contribute to explore neurobehavioral aspects resembling those found in OCD. We also identify possible advantages and limitations of potential zebrafish-based models, as well as highlight future directions in both etiological and therapeutic research. Lastly, we reinforce the use of zebrafish as a promising tool to unravel the biological basis of OCD, as well as novel pharmacological therapies in the field.
Collapse
Affiliation(s)
- João V Borba
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil.
| | - Julia Canzian
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Cássio M Resmim
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Rossano M Silva
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Maria C F Duarte
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Khadija A Mohammed
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - William Schoenau
- Department of Physiology and Pharmacology, Health Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil
| | - Isaac A Adedara
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; Graduate Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria, 1000 Roraima Avenue, Santa Maria, RS 97105-900, Brazil; The International Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA 70458, USA.
| |
Collapse
|
11
|
Fontana ACK, Poli AN, Gour J, Srikanth YV, Anastasi N, Ashok D, Khatiwada A, Reeb KL, Cheng MH, Bahar I, Rawls SM, Salvino JM. Synthesis and Structure-Activity Relationships for Glutamate Transporter Allosteric Modulators. J Med Chem 2024; 67:6119-6143. [PMID: 38626917 PMCID: PMC11056993 DOI: 10.1021/acs.jmedchem.3c01909] [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: 10/12/2023] [Revised: 03/23/2024] [Accepted: 03/27/2024] [Indexed: 04/19/2024]
Abstract
Excitatory amino acid transporters (EAATs) are essential CNS proteins that regulate glutamate levels. Excess glutamate release and alteration in EAAT expression are associated with several CNS disorders. Previously, we identified positive allosteric modulators (PAM) of EAAT2, the main CNS transporter, and have demonstrated their neuroprotective properties in vitro. Herein, we report on the structure-activity relationships (SAR) for the analogs identified from virtual screening and from our medicinal chemistry campaign. This work identified several selective EAAT2 positive allosteric modulators (PAMs) such as compounds 4 (DA-023) and 40 (NA-014) from a library of analogs inspired by GT949, an early generation compound. This series also provides nonselective EAAT PAMs, EAAT inhibitors, and inactive compounds that may be useful for elucidating the mechanism of EAAT allosteric modulation.
Collapse
Affiliation(s)
- Andréia C. K. Fontana
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Adi N.R. Poli
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Jitendra Gour
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Yellamelli V.V. Srikanth
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Nicholas Anastasi
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Devipriya Ashok
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Apeksha Khatiwada
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Katelyn L. Reeb
- Department
of Pharmacology and Physiology, Drexel University
College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Mary Hongying Cheng
- Laufer
Center for Physical & Quantitative Biology, Stony Brook University, Stony
Brook, New York 11794, United States
| | - Ivet Bahar
- Department
of Biochemistry and Cell Biology, College of Arts & Sciences and
School of Medicine, Stony Brook University, Stony Brook, New York 11794, United States
- Laufer
Center for Physical & Quantitative Biology, Stony Brook University, Stony
Brook, New York 11794, United States
| | - Scott M. Rawls
- Center
for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania 19140United States
| | - Joseph M. Salvino
- Medicinal
Chemistry, Molecular and Cellular Oncogenesis (MCO) Program, The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
- The
Wistar
Cancer Center Molecular Screening, The Wistar
Institute, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
12
|
van Veggel L, Mocking TA, Sijben HJ, Liu R, Gorostiola González M, Dilweg MA, Royakkers J, Li A, Kumar V, Dong YY, Bullock A, Sauer DB, Diliën H, van Westen GJ, Schreiber R, Heitman LH, Vanmierlo T. Still in Search for an EAAT Activator: GT949 Does Not Activate EAAT2, nor EAAT3 in Impedance and Radioligand Uptake Assays. ACS Chem Neurosci 2024; 15:1424-1431. [PMID: 38478848 PMCID: PMC10995951 DOI: 10.1021/acschemneuro.3c00731] [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: 11/10/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Excitatory amino acid transporters (EAATs) are important regulators of amino acid transport and in particular glutamate. Recently, more interest has arisen in these transporters in the context of neurodegenerative diseases. This calls for ways to modulate these targets to drive glutamate transport, EAAT2 and EAAT3 in particular. Several inhibitors (competitive and noncompetitive) exist to block glutamate transport; however, activators remain scarce. Recently, GT949 was proposed as a selective activator of EAAT2, as tested in a radioligand uptake assay. In the presented research, we aimed to validate the use of GT949 to activate EAAT2-driven glutamate transport by applying an innovative, impedance-based, whole-cell assay (xCELLigence). A broad range of GT949 concentrations in a variety of cellular environments were tested in this assay. As expected, no activation of EAAT3 could be detected. Yet, surprisingly, no biological activation of GT949 on EAAT2 could be observed in this assay either. To validate whether the impedance-based assay was not suited to pick up increased glutamate uptake or if the compound might not induce activation in this setup, we performed radioligand uptake assays. Two setups were utilized; a novel method compared to previously published research, and in a reproducible fashion copying the methods used in the existing literature. Nonetheless, activation of neither EAAT2 nor EAAT3 could be observed in these assays. Furthermore, no evidence of GT949 binding or stabilization of purified EAAT2 could be observed in a thermal shift assay. To conclude, based on experimental evidence in the present study GT949 requires specific assay conditions, which are difficult to reproduce, and the compound cannot simply be classified as an activator of EAAT2 based on the presented evidence. Hence, further research is required to develop the tools needed to identify new EAAT modulators and use their potential as a therapeutic target.
Collapse
Affiliation(s)
- Lieve van Veggel
- Department
of Neuroscience, BIOMED Biomedical Research Institute, Faculty of
Medicine and Life Sciences, Hasselt University, 3590 Hasselt, Belgium
- Department
of Psychiatry and Neuropsychology, Division of Translational Neuroscience,
European Graduate School of Neuroscience, School for Mental Health
and Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
- University
MS Center (UMSC), 3900 Hasselt-Pelt, Belgium
| | - Tamara A.M. Mocking
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Hubert J. Sijben
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Rongfang Liu
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Marina Gorostiola González
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Majlen A. Dilweg
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Jeroen Royakkers
- Sensor
Engineering
Department, Faculty of Science and Engineering, Maastricht University, 6200 Maastricht, The Netherlands
| | - Anna Li
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, U.K.
| | - Vijay Kumar
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, U.K.
| | - Yin Yao Dong
- Nuffield
Department of Clinical Neurosciences, Weatherall Institute of Molecular
Medicine, University of Oxford, OX3 7BN Oxford, U.K.
| | - Alex Bullock
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, U.K.
| | - David B. Sauer
- Centre
for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, OX3 7BN Oxford, U.K.
| | - Hanne Diliën
- Sensor
Engineering
Department, Faculty of Science and Engineering, Maastricht University, 6200 Maastricht, The Netherlands
| | - Gerard J.P. van Westen
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
| | - Rudy Schreiber
- Section
of Psychopharmacology, Neuropsychology and Psychopharmacology, Faculty
of Psychology and Neuroscience, Maastricht
University, 6200 Maastricht, The Netherlands
| | - Laura H. Heitman
- Leiden
Academic Centre for Drug Research (LACDR), Division of Drug Discovery
and Safety, Leiden University, 2333 Leiden, The Netherlands
- Oncode
Institute, Einsteinweg
55, 2333 Leiden, The Netherlands
| | - Tim Vanmierlo
- Department
of Neuroscience, BIOMED Biomedical Research Institute, Faculty of
Medicine and Life Sciences, Hasselt University, 3590 Hasselt, Belgium
- Department
of Psychiatry and Neuropsychology, Division of Translational Neuroscience,
European Graduate School of Neuroscience, School for Mental Health
and Neuroscience, Maastricht University, 6200 Maastricht, The Netherlands
- University
MS Center (UMSC), 3900 Hasselt-Pelt, Belgium
| |
Collapse
|
13
|
Yan J, Ton H, Yan J, Dong Y, Xie Z, Jiang H. Anesthetic Sevoflurane Induces Enlargement of Dendritic Spine Heads in Mouse Neurons via Tau-Dependent Mechanisms. Anesth Analg 2024:00000539-990000000-00796. [PMID: 38507523 DOI: 10.1213/ane.0000000000006941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
BACKGROUND Sevoflurane induces neuronal dysfunction and cognitive impairment. However, the underlying mechanism remains largely to be determined. Tau, cyclophilin D, and dendritic spine contribute to cognitive function. But whether changes in dendritic spines are involved in the effects of sevoflurane and the potential association with tau and cyclophilin D is not clear. METHODS We harvested hippocampal neurons from wild-type mice, tau knockout mice, and cyclophilin D knockout mice. We treated these neurons with sevoflurane at day in vitro 7 and measured the diameter of dendritic spine head and the number of dendritic spines. Moreover, we determined the effects of sevoflurane on the expression of excitatory amino acid transporter 3 (EAAT3), extracellular glutamate levels, and miniature excitatory postsynaptic currents (mEPSCs). Finally, we used lithium, cyclosporine A, and overexpression of EAAT3 in the interaction studies. RESULTS Sevoflurane-induced tau phosphgorylation increased the diameter of dendritic spine head and decreased the number of dendritic spines in neurons harvested from wild-type and cyclophilin D knockout mice, but not tau knockout mice. Sevoflurane decreased the expression of EAAT3, increased extracellular glutamate levels, and decreased the frequency of mEPSCs in the neurons. Overexpression of EAAT3 mitigated the effects of sevoflurane on dendritic spines. Lithium, but not cyclosporine A, attenuated the effects of sevoflurane on dendritic spines. Lithium also inhibited the effects of sevoflurane on EAAT3 expression and mEPSCs. CONCLUSIONS These data suggest that sevoflurane induces a tau phosphorylation-dependent demtrimental effect on dendritic spine via decreasing EAAT3 expression and increasing extracellular glutamate levels, leading to neuronal dysfunction.
Collapse
Affiliation(s)
- Jia Yan
- From the Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Hoai Ton
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Jing Yan
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanlin Dong
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Zhongcong Xie
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts
| | - Hong Jiang
- From the Department of Anesthesiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Gao Y, Liu N, Chen J, Zheng P, Niu J, Tang S, Peng X, Wu J, Yu J, Ma L. Neuropharmacological insight into preventive intervention in posttraumatic epilepsy based on regulating glutamate homeostasis. CNS Neurosci Ther 2023; 29:2430-2444. [PMID: 37309302 PMCID: PMC10401093 DOI: 10.1111/cns.14294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 05/15/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Posttraumatic epilepsy (PTE) is one of the most critical complications of traumatic brain injury (TBI), significantly increasing TBI patients' neuropsychiatric symptoms and mortality. The abnormal accumulation of glutamate caused by TBI and its secondary excitotoxicity are essential reasons for neural network reorganization and functional neural plasticity changes, contributing to the occurrence and development of PTE. Restoring glutamate balance in the early stage of TBI is expected to play a neuroprotective role and reduce the risk of PTE. AIMS To provide a neuropharmacological insight for drug development to prevent PTE based on regulating glutamate homeostasis. METHODS We discussed how TBI affects glutamate homeostasis and its relationship with PTE. Furthermore, we also summarized the research progress of molecular pathways for regulating glutamate homeostasis after TBI and pharmacological studies aim to prevent PTE by restoring glutamate balance. RESULTS TBI can lead to the accumulation of glutamate in the brain, which increases the risk of PTE. Targeting the molecular pathways affecting glutamate homeostasis helps restore normal glutamate levels and is neuroprotective. DISCUSSION Taking glutamate homeostasis regulation as a means for new drug development can avoid the side effects caused by direct inhibition of glutamate receptors, expecting to alleviate the diseases related to abnormal glutamate levels in the brain, such as PTE, Parkinson's disease, depression, and cognitive impairment. CONCLUSION It is a promising strategy to regulate glutamate homeostasis through pharmacological methods after TBI, thereby decreasing nerve injury and preventing PTE.
Collapse
Affiliation(s)
- Yuan Gao
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Ning Liu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Juan Chen
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Ping Zheng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianguo Niu
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody‐Based Drug and Intelligent Delivery System, School of Pharmaceutical SciencesHunan University of MedicineHuaihuaChina
| | - Xiaodong Peng
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jing Wu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Jianqiang Yu
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
| | - Lin Ma
- Department of PharmacologyNingxia Medical UniversityYinchuanChina
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous RegionNingxia Medical UniversityYinchuanChina
| |
Collapse
|
15
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
16
|
Piepgras J, Rohrbeck A, Just I, Bittner S, Ahnert-Hilger G, Höltje M. Enhancement of Phosphorylation and Transport Activity of the Neuronal Glutamate Transporter Excitatory Amino Acid Transporter 3 by C3bot and a 26mer C3bot Peptide. Front Cell Neurosci 2022; 16:860823. [PMID: 35783090 PMCID: PMC9240211 DOI: 10.3389/fncel.2022.860823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
In primary murine hippocampal neurons we investigated the regulation of EAAT3-mediated glutamate transport by the Clostridium botulinum C3 transferase C3bot and a 26mer peptide derived from full length protein. Incubation with either enzyme-competent C3bot or enzyme-deficient C3bot156–181 peptide resulted in the upregulation of glutamate uptake by up to 22% compared to untreated cells. A similar enhancement of glutamate transport was also achieved by the classical phorbol-ester-mediated activation of protein kinase C subtypes. Yet comparable, effects elicited by C3 preparations seemed not to rely on PKCα, γ, ε, or ζ activation. Blocking of tyrosine phosphorylation by tyrosine kinase inhibitors prevented the observed effect mediated by C3bot and C3bot 26mer. By using biochemical and molecular biological assays we could rule out that the observed C3bot and C3bot 26mer-mediated effects solely resulted from enhanced transporter expression or translocation to the neuronal surface but was rather mediated by transporter phosphorylation at tyrosine residues that was found to be significantly enhanced following incubation with either full length protein or the 26mer C3 peptide.
Collapse
Affiliation(s)
- Johannes Piepgras
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Astrid Rohrbeck
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Ingo Just
- Institute of Toxicology, Hannover Medical School, Hanover, Germany
| | - Stefan Bittner
- Department of Neurology, Focus Program Translational Neuroscience and Immunotherapy, Rhine-Main Neuroscience Network, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gudrun Ahnert-Hilger
- Department of Neurobiology, Max Planck Institute for Biophysical Chemistry, University of Göttingen, Göttingen, Germany
| | - Markus Höltje
- Institut für Integrative Neuroanatomie, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- *Correspondence: Markus Höltje,
| |
Collapse
|
17
|
Whulanza Y, Arafat Y, Rahman S, Utomo M, Kassegne S. On-chip testing of a carbon-based platform for electro-adsorption of glutamate. Heliyon 2022; 8:e09445. [PMID: 35647339 PMCID: PMC9133582 DOI: 10.1016/j.heliyon.2022.e09445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/30/2022] [Accepted: 05/11/2022] [Indexed: 11/25/2022] Open
Abstract
It is known that excessive concentrations of glutamate in the brain can cause neurotoxicity. A common approach to neutralizing this phenomenon is the use of suppressant drugs. However, excessive dependence on suppressant drugs could potentially lead to adversarial side effects, such as drug addiction. Here, we propose an alternative approach to this problem by controlling excessive amounts of glutamate ions through carbon-based, neural implant–mediated uptake. In this study, we introduce a microfluidic system that enables us to emulate the uptake of glutamate into the carbon matrix. The uptake is controlled using electrical pulses to incorporate glutamate ions into the carbon matrix through electro-adsorption. The effect of electric potential on glutamate ion uptake to control the amount of glutamate released into the microfluidic system was observed. The glutamate concentration was measured using a Ultra Violet-Visible spectrophotometer. The current setup demonstrated that a low pulsatile electric potential (0.5–1.5 V) was able to effectively govern the uptake of glutamate ions. The stimulated carbon matrix was able to decrease glutamate concentration by up to 40%. Furthermore, our study shows that these “entrapped” glutamate molecules can be effectively released upon electrical stimulation, thereby reversing the carbon electrical charge through a process called reverse uptake. A release model was used to study the profile of glutamate release from the carbon matrix at a potential of 0–1.5 V. This study showed that a burst release of glutamate was evident at an applied voltage higher than 0.5 V. Ultimately, the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) test for cytotoxicity indicated a cell viability of more than 80% for the carbon matrix. This test demonstrates that the carbon matrix can support the proliferation of cells and has a nontoxic composition; thus, it could be accepted as a candidate material for use as neural implants.
Collapse
|
18
|
Zhang Y, Chu JMT, Wong GTC. Cerebral Glutamate Regulation and Receptor Changes in Perioperative Neuroinflammation and Cognitive Dysfunction. Biomolecules 2022; 12:biom12040597. [PMID: 35454185 PMCID: PMC9029551 DOI: 10.3390/biom12040597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system and is intricately linked to learning and memory. Its activity depends on the expression of AMPA and NMDA receptors and excitatory amino transporters on neurons and glial cells. Glutamate transporters prevent the excess accumulation of glutamate in synapses, which can lead to aberrant synaptic signaling, excitotoxicity, or cell death. Neuroinflammation can occur acutely after surgical trauma and contributes to the development of perioperative neurocognitive disorders, which are characterized by impairment in multiple cognitive domains. In this review, we aim to examine how glutamate handling and glutamatergic function are affected by neuroinflammation and their contribution to cognitive impairment. We will first summarize the current data regarding glutamate in neurotransmission, its receptors, and their regulation and trafficking. We will then examine the impact of inflammation on glutamate handling and neurotransmission, focusing on changes in glial cells and the effect of cytokines. Finally, we will discuss these changes in the context of perioperative neuroinflammation and the implications they have for perioperative neurocognitive disorders.
Collapse
|
19
|
Wang XY, Liu WG, Hou AS, Song YX, Ma YL, Wu XD, Cao JB, Mi WD. Dysfunction of EAAT3 Aggravates LPS-Induced Post-Operative Cognitive Dysfunction. MEMBRANES 2022; 12:membranes12030317. [PMID: 35323793 PMCID: PMC8951453 DOI: 10.3390/membranes12030317] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 11/23/2022]
Abstract
Numerous results have revealed an association between inhibited function of excitatory amino acid transporter 3 (EAAT3) and several neurodegenerative diseases. This was also corroborated by our previous studies which showed that the EAAT3 function was intimately linked to learning and memory. With this premise, we examined the role of EAAT3 in post-operative cognitive dysfunction (POCD) and explored the potential benefit of riluzole in countering POCD in the present study. We first established a recombinant adeno-associated-viral (rAAV)-mediated shRNA to knockdown SLC1A1/EAAT3 expression in the hippocampus of adult male mice. The mice then received an intracerebroventricular microinjection of 2 μg lipopolysaccharide (LPS) to construct the POCD model. In addition, for old male mice, 4 mg/kg of riluzole was intraperitoneally injected for three consecutive days, with the last injection administered 2 h before the LPS microinjection. Cognitive function was assessed using the Morris water maze 24 h following the LPS microinjection. Animal behavioral tests, as well as pathological and biochemical assays, were performed to clarify the role of EAAT3 function in POCD and evaluate the effect of activating the EAAT3 function by riluzole. In the present study, we established a mouse model with hippocampal SLC1A1/EAAT3 knockdown and found that hippocampal SLC1A1/EAAT3 knockdown aggravated LPS-induced learning and memory deficits in adult male mice. Meanwhile, LPS significantly inhibited the expression of EAAT3 membrane protein and the phosphorylation level of GluA1 protein in the hippocampus of adult male mice. Moreover, riluzole pretreatment significantly increased the expression of hippocampal EAAT3 membrane protein and also ameliorated LPS-induced cognitive impairment in elderly male mice. Taken together, our results demonstrated that the dysfunction of EAAT3 is an important risk factor for POCD susceptibility and therefore, it may become a promising target for POCD treatment.
Collapse
Affiliation(s)
- Xiao-Yan Wang
- Chinese PLA Medical School, Beijing 100853, China; (X.-Y.W.); (W.-G.L.)
- Department of Anesthesiology, The Fourth Medical Center of Chinese PLA General Hospital, Beijing 100037, China
| | - Wen-Gang Liu
- Chinese PLA Medical School, Beijing 100853, China; (X.-Y.W.); (W.-G.L.)
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
| | - Ai-Sheng Hou
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
| | - Yu-Xiang Song
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
| | - Yu-Long Ma
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
| | - Xiao-Dong Wu
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
| | - Jiang-Bei Cao
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
- Correspondence: (J.-B.C.); (W.-D.M.)
| | - Wei-Dong Mi
- Department of Anesthesiology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, China; (A.-S.H.); (Y.-X.S.); (Y.-L.M.); (X.-D.W.)
- Correspondence: (J.-B.C.); (W.-D.M.)
| |
Collapse
|
20
|
Boo YC. Metabolic Basis and Clinical Evidence for Skin Lightening Effects of Thiol Compounds. Antioxidants (Basel) 2022; 11:antiox11030503. [PMID: 35326153 PMCID: PMC8944565 DOI: 10.3390/antiox11030503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 12/17/2022] Open
Abstract
Melanin pigment is a major factor in determining the color of the skin, and its abnormal increase or decrease can cause serious pigmentation disorders. The melanin pigment of the skin is divided into light pheomelanin and dark eumelanin, and a big difference between them is whether they contain sulfur. Melanin synthesis starts from a common reaction in which tyrosine or dihydroxyphenylalanine (DOPA) is oxidized by tyrosinase (TYR) to produce dopaquinone (DQ). DQ is spontaneously converted to leukodopachrome and then oxidized to dopachrome, which enters the eumelanin synthesis pathway. When DQ reacts with cysteine, cysteinyl dopa is generated, which is oxidized to cysteinyl DQ and enters the pheomelanin synthesis pathway. Therefore, thiol compounds can influence the relative synthesis of eumelanin and pheomelanin. In addition, thiol compounds can inhibit enzymatic activity by binding to copper ions at the active site of TYR, and act as an antioxidant scavenging reactive oxygen species and free radicals or as a modulator of redox balance, thereby inhibiting overall melanin synthesis. This review will cover the metabolic aspects of thiol compounds, the role of thiol compounds in melanin synthesis, comparison of the antimelanogenic effects of various thiol compounds, and clinical trials on the skin lightening efficacy of thiol compounds. We hope that this review will help identify the advantages and disadvantages of various thiol compounds as modulators of skin pigmentation and contribute to the development of safer and more effective strategies for the treatment of pigmentation disorders.
Collapse
Affiliation(s)
- Yong Chool Boo
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea;
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Korea
| |
Collapse
|
21
|
Guerrero-Molina MP, Morales-Conejo M, Delmiro A, Morán M, Domínguez-González C, Arranz-Canales E, Ramos-González A, Arenas J, Martín MA, González de la Aleja J. Elevated glutamate and decreased glutamine levels in the cerebrospinal fluid of patients with MELAS syndrome. J Neurol 2022; 269:3238-3248. [PMID: 35088140 PMCID: PMC8794606 DOI: 10.1007/s00415-021-10942-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
Background Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is a genetically heterogeneous disorder caused by mitochondrial DNA (mtDNA) mutations in the MT-TL1 gene. The pathophysiology of neurological manifestations is still unclear, but neuronal hyperexcitability and neuron–astrocyte uncoupling have been suggested. Glutamatergic neurotransmission is linked to glucose oxidation and mitochondrial metabolism in astrocytes and neurons. Given the relevance of neuron–astrocyte metabolic coupling and astrocyte function regulating energetic metabolism, we aimed to assess glutamate and glutamine CSF levels in MELAS patients. Methods This prospective observational case–control study determined glutamate and glutamine CSF levels in patients with MELAS syndrome and compared them with controls. The plasma and CSF levels of the remaining amino acids and lactate were also determined. Results Nine adult patients with MELAS syndrome (66.7% females mean age 35.8 ± 3.2 years) and 19 controls (63.2% females mean age 42.7 ± 3.8 years) were included. The CSF glutamate levels were significantly higher in patients with MELAS than in controls (18.48 ± 1.34 vs. 5.31 ± 1.09 μmol/L, p < 0.001). Significantly lower glutamine concentrations in patients with MELAS than controls were shown in CSF (336.31 ± 12.92 vs. 407.06 ± 15.74 μmol/L, p = 0.017). Moreover, the CSF levels of alanine, the branched-chain amino acids (BCAAs) and lactate were significantly higher in patients with MELAS. Conclusions Our results suggest the glutamate–glutamine cycle is altered probably due to metabolic imbalance, and as a result, the lactate–alanine and BCAA–glutamate cycles are upregulated. These findings might have therapeutic implications in MELAS syndrome. Supplementary Information The online version contains supplementary material available at 10.1007/s00415-021-10942-7.
Collapse
Affiliation(s)
- María Paz Guerrero-Molina
- Neuromuscular Disorders Unit, Neurology Department, University Hospital, 12 de Octubre, Madrid, Spain.
| | - Montserrat Morales-Conejo
- Department of Internal Medicine, University Hospital, 12 de Octubre, Madrid, Spain.,National Reference Center for Congenital Errors of Metabolism (CSUR) an European Reference Center for Inherited Metabolic Disease (MetabERN), University Hospital, 12 de Octubre, Madrid, Spain.,Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain
| | - Aitor Delmiro
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain.,Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), Madrid, Spain.,Research Institute ('imas12'), University Hospital, 12 de Octubre, Madrid, Spain
| | - María Morán
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain.,Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), Madrid, Spain.,Research Institute ('imas12'), University Hospital, 12 de Octubre, Madrid, Spain
| | - Cristina Domínguez-González
- Neuromuscular Disorders Unit, Neurology Department, University Hospital, 12 de Octubre, Madrid, Spain.,Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain.,Research Institute ('imas12'), University Hospital, 12 de Octubre, Madrid, Spain
| | - Elena Arranz-Canales
- Department of Internal Medicine, University Hospital, 12 de Octubre, Madrid, Spain.,National Reference Center for Congenital Errors of Metabolism (CSUR) an European Reference Center for Inherited Metabolic Disease (MetabERN), University Hospital, 12 de Octubre, Madrid, Spain
| | - Ana Ramos-González
- Department of Neuroradiology, University Hospital, 12 de Octubre, Madrid, Spain
| | - Joaquín Arenas
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain.,Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), Madrid, Spain.,Research Institute ('imas12'), University Hospital, 12 de Octubre, Madrid, Spain
| | - Miguel A Martín
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, Madrid, Spain.,Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), Madrid, Spain.,Research Institute ('imas12'), University Hospital, 12 de Octubre, Madrid, Spain
| | - Jesús González de la Aleja
- National Reference Center for Congenital Errors of Metabolism (CSUR) an European Reference Center for Inherited Metabolic Disease (MetabERN), University Hospital, 12 de Octubre, Madrid, Spain.,Epilepsy Unit, Neurology Department, University Hospital, 12 de Octubre, Madrid, Spain
| |
Collapse
|
22
|
Saha K, Yang JW, Hofmaier T, Venkatesan S, Steinkellner T, Kudlacek O, Sucic S, Freissmuth M, Sitte HH. Constitutive Endocytosis of the Neuronal Glutamate Transporter Excitatory Amino Acid Transporter-3 Requires ARFGAP1. Front Physiol 2021; 12:671034. [PMID: 34040545 PMCID: PMC8141794 DOI: 10.3389/fphys.2021.671034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
The eukaryotic endocytic pathway regulates protein levels available at the plasma membrane by recycling them into specific endosomal compartments. ARFGAP1 is a component of the coat protein I (COPI) complex but it also plays a role in promoting adapter protein-2 (AP-2) mediated endocytosis. The excitatory amino acid transporter-3 (EAAT3) mediates the reuptake of glutamate from the synaptic cleft to achieve rapid termination of synaptic transmission at glutamatergic synapses. In this study, we identified two interacting proteins of EAAT3 by mass spectrometry (MS) ARFGAP1 and ARF6. We explored the role of ARFGAP1 and ARF6 in the endocytosis of EAAT3. Our data revealed that ARFGAP1 plays a role in the recycling of EAAT3, by utilizing its GTPase activating protein (GAP) activity and ARF6 acting as the substrate. ARFGAP1 promotes cargo sorting of EAAT3 via a single phenylalanine residue (F508) located at the C-terminus of the transporter. ARFGAP1-promoted AP-2 dependent endocytosis is abolished upon neutralizing F508. We utilized a heterologous expression system to identify an additional motif in the C-terminus of EAAT3 that regulates its endocytosis. Impairment in endocytosis did not affect somatodendritic targeting in cultured hippocampal neurons. Our findings support a model where endocytosis of EAAT3 is a multifactorial event regulated by ARFGAP1, occurring via the C-terminus of the transporter, and is the first study to examine the role of ARFGAP1 in the endocytosis of a transport protein.
Collapse
Affiliation(s)
- Kusumika Saha
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria.,Institut Cochin, INSERM U1016, CNRS UMR8104, Université Paris Descartes, Paris, France
| | - Jae-Won Yang
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Tina Hofmaier
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - SanthoshKannan Venkatesan
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Steinkellner
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Kudlacek
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Sonja Sucic
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Freissmuth
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Harald H Sitte
- Institute of Pharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
23
|
Barahimi P, Karimian M, Nejati M, Azami Tameh A, Atlasi MA. Oxytocin improves ischemic stroke by reducing expression of excitatory amino acid transporter 3 in rat MCAO model. J Immunoassay Immunochem 2021; 42:513-524. [PMID: 33788672 DOI: 10.1080/15321819.2021.1906270] [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] [Indexed: 01/28/2023]
Abstract
Various molecular mechanisms are activated in neurons during ischemic stroke. Extracellular glutamate secretion into brain tissue causes neurotoxicity and brain damage. Excitatory amino acid transporter 3 (EAAT3) could remove the extracellular glutamate. Neuroprotective activity of oxytocin (OT) in ischemia of various tissues has been reported. This study investigates the neuroprotective effect of OT in an animal model of middle cerebral artery occlusion (MCAO) and the possible role of EAAT3. Transient MCAO was performed as a model of ischemic stroke in male rats and then OT was administrated intra-nasally. Infarct volume was measured by 2, 3, 5-triphenyl tetrazolium chloride staining. Nissl staining method was performed for the evaluation of neuronal cell morphology. Immunohistochemistry assay was performed to analyze the EAAT3 expression in the ischemic region. OT significantly reduced the infarct volume in the cerebral cortex and striatum after ischemia (P< .05). In addition, OT reduces the number of neurons with pyknotic nuclei that are significantly increased in the ischemic region (P< .01) Immunohistochemistry results showed that although EAAT3 expression increased after ischemia, OT therapy increased EAAT3 expression further (P< .05). Therefore, increased EAAT3 expression could be one of the neuroprotective mechanisms of OT after MCAO.
Collapse
Affiliation(s)
- Parinaz Barahimi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Majid Nejati
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.,Core Research Laboratory, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
24
|
Qiu B, Matthies D, Fortea E, Yu Z, Boudker O. Cryo-EM structures of excitatory amino acid transporter 3 visualize coupled substrate, sodium, and proton binding and transport. SCIENCE ADVANCES 2021; 7:7/10/eabf5814. [PMID: 33658209 PMCID: PMC7929514 DOI: 10.1126/sciadv.abf5814] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 01/19/2021] [Indexed: 05/16/2023]
Abstract
Human excitatory amino acid transporter 3 (hEAAT3) mediates glutamate uptake in neurons, intestine, and kidney. Here, we report cryo-EM structures of hEAAT3 in several functional states where the transporter is empty, bound to coupled sodium ions only, or fully loaded with three sodium ions, a proton, and the substrate aspartate. The structures suggest that hEAAT3 operates by an elevator mechanism involving three functionally independent subunits. When the substrate-binding site is near the cytoplasm, it has a remarkably low affinity for the substrate, perhaps facilitating its release and allowing the rapid transport turnover. The mechanism of the coupled uptake of the sodium ions and the substrate is conserved across evolutionarily distant families and is augmented by coupling to protons in EAATs. The structures further suggest a mechanism by which a conserved glutamate residue mediates proton symport.
Collapse
Affiliation(s)
- Biao Qiu
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| | - Doreen Matthies
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Eva Fortea
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| | - Zhiheng Yu
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Olga Boudker
- Department of Physiology and Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA.
- Howard Hughes Medical Institute, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| |
Collapse
|
25
|
Brymer KJ, Barnes JR, Parsons MP. Entering a new era of quantifying glutamate clearance in health and disease. J Neurosci Res 2021; 99:1598-1617. [PMID: 33618436 DOI: 10.1002/jnr.24810] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/22/2021] [Accepted: 02/01/2021] [Indexed: 12/21/2022]
Abstract
Glutamate transporter proteins, expressed on both neurons and glia, serve as the main gatekeepers that dictate the spatial and temporal actions of extracellular glutamate. Glutamate is essential to the function of the healthy brain yet paradoxically contributes to the toxicity associated with many neurodegenerative diseases. Rapid transporter-mediated glutamate uptake, primarily occurring at astrocytic processes, tightens the efficiency of excitatory network activity and prevents toxic glutamate build-up in the extracellular space. Glutamate transporter dysfunction is thought to underlie myriad central nervous system (CNS) diseases including Alzheimer and Huntington disease. Over the past few decades, techniques such as biochemical uptake assays and electrophysiological recordings of transporter currents from individual astrocytes have revealed the remarkable ability of the CNS to efficiently clear extracellular glutamate. In more recent years, the rapidly evolving glutamate-sensing "sniffers" now allow researchers to visualize real-time glutamate transients on a millisecond time scale with single synapse spatial resolution in defined cell populations. As we transition to an increased reliance on optical-based methods of glutamate visualization and quantification, it is of utmost importance to understand not only the advantages that glutamate biosensors bring to the table but also the associated caveats and their implications for data interpretation. In this review, we summarize the strengths and limitations of the commonly used methods to quantify glutamate uptake. We then discuss what these techniques, when viewed as a complementary whole, have told us about the brain's ability to regulate glutamate levels, in both health and in the context of neurodegenerative disease.
Collapse
Affiliation(s)
- Kyle J Brymer
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jocelyn R Barnes
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Matthew P Parsons
- Faculty of Medicine, Division of Biomedical Sciences, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
26
|
Safari MS, Obexer D, Baier-Bitterlich G, Zur Nedden S. PKN1 Is a Novel Regulator of Hippocampal GluA1 Levels. Front Synaptic Neurosci 2021; 13:640495. [PMID: 33613259 PMCID: PMC7892898 DOI: 10.3389/fnsyn.2021.640495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Alterations in the processes that control α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) expression, assembly and trafficking are closely linked to psychiatric and neurodegenerative disorders. We have recently shown that the serine/threonine kinase Protein kinase N1 (PKN1) is a developmentally active regulator of cerebellar synaptic maturation by inhibiting AKT and the neurogenic transcription factor neurogenic differentiation factor-2 (NeuroD2). NeuroD2 is involved in glutamatergic synaptic maturation by regulating expression levels of various synaptic proteins. Here we aimed to study the effect of Pkn1 knockout on AKT phosphorylation and NeuroD2 levels in the hippocampus and the subsequent expression levels of the NeuroD2 targets and AMPAR subunits: glutamate receptor 1 (GluA1) and GluA2/3. We show that PKN1 is expressed throughout the hippocampus. Interestingly, not only postnatal but also adult hippocampal phospho-AKT and NeuroD2 levels were significantly elevated upon Pkn1 knockout. Postnatal and adult Pkn1 -/- hippocampi showed enhanced expression of the AMPAR subunit GluA1, particularly in area CA1. Surprisingly, GluA2/3 levels were not different between both genotypes. In addition to higher protein levels, we also found an enhanced GluA1 content in the membrane fraction of postnatal and adult Pkn1 -/- animals, while GluA2/3 levels remained unchanged. This points toward a very specific regulation of GluA1 expression and/or trafficking by the novel PKN1-AKT-NeuroD2 axis. Considering the important role of GluA1 in hippocampal development as well as the pathophysiology of several disorders, ranging from Alzheimer's, to depression and schizophrenia, our results validate PKN1 for future studies into neurological disorders related to altered AMPAR subunit expression in the hippocampus.
Collapse
Affiliation(s)
- Motahareh Solina Safari
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | - Dido Obexer
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Stephanie Zur Nedden
- CCB-Biocenter, Institute of Neurobiochemistry, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Arkhipova V, Fu H, Hoorens MWH, Trinco G, Lameijer LN, Marin E, Feringa BL, Poelarends GJ, Szymanski W, Slotboom DJ, Guskov A. Structural Aspects of Photopharmacology: Insight into the Binding of Photoswitchable and Photocaged Inhibitors to the Glutamate Transporter Homologue. J Am Chem Soc 2021; 143:1513-1520. [PMID: 33449695 PMCID: PMC7844824 DOI: 10.1021/jacs.0c11336] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Photopharmacology addresses the challenge of drug selectivity and
side effects through creation of photoresponsive molecules activated
with light with high spatiotemporal precision. This is achieved through
incorporation of molecular photoswitches and photocages into the pharmacophore.
However, the structural basis for the light-induced modulation of
inhibitory potency in general is still missing, which poses a major
design challenge for this emerging field of research. Here we solved
crystal structures of the glutamate transporter homologue GltTk in complex with photoresponsive transport inhibitors—azobenzene
derivative of TBOA (both in trans and cis configuration) and with the photocaged compound ONB-hydroxyaspartate.
The essential role of glutamate transporters in the functioning of
the central nervous system renders them potential therapeutic targets
in the treatment of neurodegenerative diseases. The obtained structures
provide a clear structural insight into the origins of photocontrol
in photopharmacology and lay the foundation for application of photocontrolled
ligands to study the transporter dynamics by using time-resolved X-ray
crystallography.
Collapse
Affiliation(s)
- Valentina Arkhipova
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.,Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Haigen Fu
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Mark W H Hoorens
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.,Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Gianluca Trinco
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Lucien N Lameijer
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.,Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Egor Marin
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands.,Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Ben L Feringa
- Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Gerrit J Poelarends
- Department of Chemical and Pharmaceutical Biology, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Wiktor Szymanski
- University Medical Center Groningen, Department of Radiology, Medical Imaging Center, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.,Stratingh Institute for Chemistry, Faculty of Science and Engineering, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Dirk J Slotboom
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Albert Guskov
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands.,Research Center for Molecular Mechanisms of Aging and Age-related Diseases, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| |
Collapse
|
28
|
Abstract
OCD has lagged behind other psychiatric illnesses in the identification of molecular treatment targets, due in part to a lack of significant findings in genome-wide association studies. However, while progress in this area is being made, OCD's symptoms of obsessions, compulsions, and anxiety can be deconstructed into distinct neural functions that can be dissected in animal models. Studies in rodents and non-human primates have highlighted the importance of cortico-basal ganglia-thalamic circuits in OCD pathophysiology, and emerging studies in human post-mortem brain tissue point to glutamatergic synapse abnormalities as a potential cellular substrate for observed dysfunctional behaviors. In addition, accumulated evidence points to a potential role for neuromodulators including serotonin and dopamine in both OCD pathology and treatment. Here, we review current efforts to use animal models for the identification of molecules, cell types, and circuits relevant to OCD pathophysiology. We start by describing features of OCD that can be modeled in animals, including circuit abnormalities and genetic findings. We then review different strategies that have been used to study OCD using animal model systems, including transgenic models, circuit manipulations, and dissection of OCD-relevant neural constructs. Finally, we discuss how these findings may ultimately help to develop new treatment strategies for OCD and other related disorders.
Collapse
Affiliation(s)
- Brittany L Chamberlain
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA.,Center for Neuroscience Program and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susanne E Ahmari
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh, Pittsburgh, PA, USA. .,Center for Neuroscience Program and Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
29
|
Chuang WY, Shih HD, Shy YM, Chang SC, Lee TT. Evaluation of mushroom waste compost on broiler body composition, nutrient absorption and adipose metabolism. ITALIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1080/1828051x.2020.1808861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Wen Yang Chuang
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan
| | - Hsin Der Shih
- Taiwan Agricultural Research Institute, Council of Agriculture, Executive Yuan, Taichung, Taiwan
| | - Yi Ming Shy
- Hsinchu Branch, Livestock Research Institute, Council of Agriculture, Executive Yuan, Miaoli, Taiwan
| | - Sheng Chang Chang
- Kaohsiung Animal Propagation Station, Livestock Research Institute, Council of Agriculture, Kaohsiung, Taiwan
| | - Tzu Tai Lee
- Department of Animal Science, National Chung Hsing University, Taichung, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
30
|
Neurobiology, Functions, and Relevance of Excitatory Amino Acid Transporters (EAATs) to Treatment of Refractory Epilepsy. CNS Drugs 2020; 34:1089-1103. [PMID: 32926322 DOI: 10.1007/s40263-020-00764-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epilepsy is one of the most prevalent and devastating neurological disorders characterized by episodes of unusual sensations, loss of awareness, and reoccurring seizures. The frequency and intensity of epileptic fits can vary to a great degree, with almost a third of all cases resistant to available therapies. At present, there is a major unmet need for effective and specific therapeutic intervention. Impairments of the exquisite balance between excitatory and inhibitory synaptic processes in the brain are considered key in the onset and pathophysiology of the disease. As the primary excitatory neurotransmitter in the central nervous system, glutamate has been implicated in the process, with the glutamatergic system holding center stage in the pathobiology as well as in developing disease-modifying therapies. Emerging data pinpoint impairments of glutamate clearance as one of the key causative factors in drug-resistant disease forms. Reinstatement of glutamate homeostasis using pharmacological and genetic modulation of glutamate clearance is therefore considered to be of major translational relevance. In this article, we review the neurobiological and clinical evidence suggesting complex aberrations in the activity and functions of excitatory amino acid transporters (EAATs) in epilepsy, with knock-on effects on glutamate homeostasis as a leading cause for the development of refractory forms. We consider the emerging data on pharmacological and genetic manipulations of EAATs, with reference to seizures and glutamate dyshomeostasis, and review their fundamental and translational relevance. We discuss the most recent advances in the EAATs research in human and animal models, along with numerous questions that remain open for debate and critical appraisal. Contrary to the widely held view on EAATs as a promising therapeutic target for management of refractory epilepsy as well as other neurological and psychiatric conditions related to glutamatergic hyperactivity and glutamate-induced cytotoxicity, we stress that the true relevance of EAAT2 as a target for medical intervention remains to be fully appreciated and verified. Despite decades of research, the emerging properties and functional characteristics of glutamate transporters and their relationship with neurophysiological and behavioral correlates of epilepsy challenge the current perception of this disease and fit unambiguously in neither EAATs functional deficit nor in reversal models. We stress the pressing need for new approaches and models for research and restoration of the physiological activity of glutamate transporters and synaptic transmission to achieve much needed therapeutic effects. The complex mechanism of EAATs regulation by multiple factors, including changes in the electrochemical environment and ionic gradients related to epileptic hyperactivity, impose major therapeutic challenges. As a final note, we consider the evolving views and present a cautious perspective on the key areas of future progress in the field towards better management and treatment of refractory disease forms.
Collapse
|
31
|
Ma B, Zhang L, Li J, Xing T, Jiang Y, Gao F. Heat stress alters muscle protein and amino acid metabolism and accelerates liver gluconeogenesis for energy supply in broilers. Poult Sci 2020; 100:215-223. [PMID: 33357684 PMCID: PMC7772709 DOI: 10.1016/j.psj.2020.09.090] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 12/30/2022] Open
Abstract
Heat stress impairs growth performance and alters body protein and amino acid metabolism. This study was investigated to explore how body protein and amino acid metabolism changed under heat stress (HS) and the stress adaptation mechanism. A total of 144 broilers (28 d old) were divided into 3 treatment groups for 1 wk: HS group (32°C), normal control group (22°C), and pair-feeding group (22°C). We found that HS elevated the feed-to-gain ratio, reduced the ADFI and ADG, decreased breast muscle mass and plasma levels of several amino acids (glycine, lysine, threonine, and tyrosine), and increased serum glutamic oxaloacetic transaminase (GOT) activity and corticosterone (CORT) level and liver GOT and glutamic pyruvic transaminase activities. Heat stress elevated muscle atrophy F-box mRNA expression and reduced mRNA expression of the 70-kD ribosomal protein S6 kinase in the breast muscle of broilers. Broilers in the HS group exhibited striking increases of mRNA expressions of solute carrier family 1 member 1, family 3 member 1, family 7 member 1, and family 7 member-like in the liver and liver gluconeogenesis genes (PCKc, PCKm, PC, and FBP1) in comparison with the other 2 groups. In conclusion, HS increased the circulating CORT level and subsequently caused muscle protein breakdown to provide amino acid substrates to liver gluconeogenesis responsible for energy supply.
Collapse
Affiliation(s)
- Bingbing Ma
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yun Jiang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, PR China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
32
|
Forster YM, Green JL, Khatiwada A, Liberato JL, Narayana Reddy PA, Salvino JM, Bienz S, Bigler L, dos Santos WF, Karklin Fontana AC. Elucidation of the Structure and Synthesis of Neuroprotective Low Molecular Mass Components of the Parawixia bistriata Spider Venom. ACS Chem Neurosci 2020; 11:1573-1596. [PMID: 32343555 DOI: 10.1021/acschemneuro.0c00007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The South American social spider Parawixia bistriata produces a venom containing complex organic compounds with intriguing biological activities. The crude venom leads to paralysis in termites and stimulates l-glutamate uptake and inhibits GABA uptake in rat brain synaptosomes. Glutamate is the major neurotransmitter at the insect neuromuscular junction and at the mammalian central nervous system, suggesting a modulation of the glutamatergic system by the venom. Parawixin1, 2, and 10 (Pwx1, 2 and 10) are HPLC fractions that demonstrate this bioactivity. Pwx1 stimulates l-glutamate uptake through the main transporter in the brain, EAAT2, and is neuroprotective in in vivo glaucoma models. Pxw2 inhibits GABA and glycine uptake in synaptosomes and inhibits seizures and neurodegeneration, and Pwx10 increases l-glutamate uptake in synaptosomes and is neuroprotective and anticonvulsant, shown in in vivo epilepsy models. Herein, we investigated the low molecular mass compounds in this venom and have found over 20 small compounds and 36 unique acylpolyamines with and without amino acid linkers. The active substances in fractions Pwx1 and Pwx2 require further investigation. We elucidated and confirmed the structure of the active acylpolyamine in Pwx10. Both fraction Pwx10 and the synthesized component enhance the activity of transporters EAAT1 and EAAT2, and, importantly, offer in vitro neuroprotection against excitotoxicity in primary cultures. These data suggest that compounds with this mechanism could be developed into therapies for disorders in which l-glutamate excitotoxicity is involved.
Collapse
Affiliation(s)
- Yvonne M. Forster
- Department of Chemistry, University of Zurich, Zurich, CH 8057, Switzerland
| | - Jennifer Leigh Green
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - Apeksha Khatiwada
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| | - José Luiz Liberato
- Department of Biology, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil
| | | | - Joseph M. Salvino
- The Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Stefan Bienz
- Department of Chemistry, University of Zurich, Zurich, CH 8057, Switzerland
| | - Laurent Bigler
- Department of Chemistry, University of Zurich, Zurich, CH 8057, Switzerland
| | | | - Andréia Cristina Karklin Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, United States
| |
Collapse
|
33
|
Fontana IC, Zimmer AR, Rocha AS, Gosmann G, Souza DO, Lourenco MV, Ferreira ST, Zimmer ER. Amyloid-β oligomers in cellular models of Alzheimer's disease. J Neurochem 2020; 155:348-369. [PMID: 32320074 DOI: 10.1111/jnc.15030] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/21/2020] [Accepted: 04/17/2020] [Indexed: 12/22/2022]
Abstract
Amyloid-β (Aβ) dysmetabolism is tightly associated with pathological processes in Alzheimer's disease (AD). Currently, it is thought that, in addition to Aβ fibrils that give rise to plaque formation, Aβ aggregates into non-fibrillar soluble oligomers (AβOs). Soluble AβOs have been extensively studied for their synaptotoxic and neurotoxic properties. In this review, we discuss physicochemical properties of AβOs and their impact on different brain cell types in AD. Additionally, we summarize three decades of studies with AβOs, providing a compelling bulk of evidence regarding cell-specific mechanisms of toxicity. Cellular models may lead us to a deeper understanding of the detrimental effects of AβOs in neurons and glial cells, putatively shedding light on the development of innovative therapies for AD.
Collapse
Affiliation(s)
- Igor C Fontana
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Aline R Zimmer
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Andreia S Rocha
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Grace Gosmann
- Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Diogo O Souza
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil.,Department of Biochemistry, UFRGS, Porto Alegre, Brazil
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eduardo R Zimmer
- Graduate Program in Biological Sciences: Biochemistry, UFRGS, Porto Alegre, Brazil.,Department of Pharmacology, UFRGS, Porto Alegre, Brazil.,Graduate Program in Biological Sciences: Pharmacology and Therapeutics,, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
34
|
Lengel D, Huh JW, Barson JR, Raghupathi R. Progesterone treatment following traumatic brain injury in the 11-day-old rat attenuates cognitive deficits and neuronal hyperexcitability in adolescence. Exp Neurol 2020; 330:113329. [PMID: 32335121 DOI: 10.1016/j.expneurol.2020.113329] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/13/2020] [Accepted: 04/22/2020] [Indexed: 12/18/2022]
Abstract
Traumatic brain injury (TBI) in children younger than 4 years old results in cognitive and psychosocial deficits in adolescence and adulthood. At 4 weeks following closed head injury on postnatal day 11, male and female rats exhibited impairment in novel object recognition memory (NOR) along with an increase in open arm time in the elevated plus maze (EPM), suggestive of risk-taking behaviors. This was accompanied by an increase in intrinsic excitability and frequency of spontaneous excitatory post-synaptic currents (EPSCs), and a decrease in the frequency of spontaneous inhibitory post-synaptic currents in layer 2/3 neurons within the medial prefrontal cortex (PFC), a region that is implicated in both object recognition and risk-taking behaviors. Treatment with progesterone for the first week after brain injury improved NOR memory at the 4-week time point in both sham and brain-injured rats and additionally attenuated the injury-induced increase in the excitability of neurons and the frequency of spontaneous EPSCs. The effect of progesterone on cellular excitability changes after injury may be related to its ability to decrease the mRNA expression of the β3 subunit of the voltage-gated sodium channel and increase the expression of the neuronal excitatory amino acid transporter 3 in the medial PFC in sham- and brain-injured animals and also increase glutamic acid decarboxylase mRNA expression in sham- but not brain-injured animals. Progesterone treatment did not affect injury-induced changes in the EPM test. These results demonstrate that administration of progesterone immediately after TBI in 11-day-old rats reduces cognitive deficits in adolescence, which may be mediated by progesterone-mediated regulation of excitatory signaling mechanisms within the medial PFC.
Collapse
Affiliation(s)
- Dana Lengel
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America
| | - Jimmy W Huh
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Jessica R Barson
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America
| | - Ramesh Raghupathi
- Program in Neuroscience, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, PA United States of America; Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States of America.
| |
Collapse
|
35
|
Blacker CJ, Millischer V, Webb LM, Ho AM, Schalling M, Frye MA, Veldic M. EAAT2 as a Research Target in Bipolar Disorder and Unipolar Depression: A Systematic Review. MOLECULAR NEUROPSYCHIATRY 2020; 5:44-59. [PMID: 32399469 PMCID: PMC7206595 DOI: 10.1159/000501885] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022]
Abstract
Glutamate is implicated in the neuropathology of both major depressive disorder and bipolar disorder. Excitatory amino acid transporter 2 (EAAT2) is the major glutamate transporter in the mammalian brain, removing glutamate from the synaptic cleft and transporting it into glia for recycling. It is thereby the principal regulator of extracellular glutamate levels and prevents neuronal excitotoxicity. EAAT2 is a promising target for elucidating the mechanisms by which the glutamate-glutamine cycle interacts with neuronal systems in mood disorders. Forty EAAT2 studies (published January 1992-January 2018) were identified via a systematic literature search. The studies demonstrated that chronic stress/steroids were most commonly associated with decreased EAAT2. In rodents, EAAT2 inhibition worsened depressive behaviors. Human EAAT2 expression usually decreased in depression, with some regional brain differences. Fewer data have been collected regarding the roles and regulation of EAAT2 in bipolar disorder. Future directions for research include correlating EAAT2 and glutamate levels in vivo, elucidating genetic variability and epigenetic regulation, clarifying intracellular protein and pharmacologic interactions, and examining EAAT2 in different bipolar mood states. As part of a macromolecular complex within glia, EAAT2 may contribute significantly to intracellular signaling, energy regulation, and cellular homeostasis. An enhanced understanding of this system is needed.
Collapse
Affiliation(s)
- Caren J. Blacker
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lauren M. Webb
- Mayo Medical School, Mayo Clinic, Rochester, Minnesota, USA
| | - Ada M.C. Ho
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Martin Schalling
- Department of Molecular Medicine and Surgery (MMK), Karolinska Institutet, Stockholm, Sweden
- Neurogenetics Unit, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark A. Frye
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, USA
| | - Marin Veldic
- Department of Psychiatry and Psychology, Mayo Clinic Depression Center, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
36
|
Amino Acid Transporters and Exchangers from the SLC1A Family: Structure, Mechanism and Roles in Physiology and Cancer. Neurochem Res 2020; 45:1268-1286. [DOI: 10.1007/s11064-019-02934-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/10/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
|
37
|
Pajarillo E, Rizor A, Lee J, Aschner M, Lee E. The role of astrocytic glutamate transporters GLT-1 and GLAST in neurological disorders: Potential targets for neurotherapeutics. Neuropharmacology 2019; 161:107559. [PMID: 30851309 PMCID: PMC6731169 DOI: 10.1016/j.neuropharm.2019.03.002] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 12/12/2022]
Abstract
Glutamate is the primary excitatory neurotransmitter in the central nervous system (CNS) which initiates rapid signal transmission in the synapse before its re-uptake into the surrounding glia, specifically astrocytes. The astrocytic glutamate transporters glutamate-aspartate transporter (GLAST) and glutamate transporter-1 (GLT-1) and their human homologs excitatory amino acid transporter 1 (EAAT1) and 2 (EAAT2), respectively, are the major transporters which take up synaptic glutamate to maintain optimal extracellular glutamic levels, thus preventing accumulation in the synaptic cleft and ensuing excitotoxicity. Growing evidence has shown that excitotoxicity is associated with various neurological disorders, including amyotrophic lateral sclerosis (ALS), Alzheimer's disease (AD), Parkinson's disease (PD), manganism, ischemia, schizophrenia, epilepsy, and autism. While the mechanisms of neurological disorders are not well understood, the dysregulation of GLAST/GLT-1 may play a significant role in excitotoxicity and associated neuropathogenesis. The expression and function of GLAST/GLT-1 may be dysregulated at the genetic, epigenetic, transcriptional or translational levels, leading to high levels of extracellular glutamate and excitotoxicity. Consequently, understanding the regulatory mechanisms of GLAST/GLT-1 has been an area of interest in developing therapeutics for the treatment of neurological disorders. Pharmacological agents including β-lactam antibiotics, estrogen/selective estrogen receptor modulators (SERMs), growth factors, histone deacetylase inhibitors (HDACi), and translational activators have shown significant efficacy in enhancing the expression and function of GLAST/GLT-1 and glutamate uptake both in vitro and in vivo. This comprehensive review will discuss the regulatory mechanisms of GLAST/GLT-1, their association with neurological disorders, and the pharmacological agents which mediate their expression and function. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
Affiliation(s)
- Edward Pajarillo
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Asha Rizor
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA
| | - Jayden Lee
- Department of Speech, Language & Hearing Sciences, Boston University, Boston, MA, 02215, USA
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Eunsook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL, 32301, USA.
| |
Collapse
|
38
|
Ceftriaxone Treatment Affects EAAT2 Expression and Glutamatergic Neurotransmission and Exerts a Weak Anticonvulsant Effect in Young Rats. Int J Mol Sci 2019; 20:ijms20235852. [PMID: 31766528 PMCID: PMC6928884 DOI: 10.3390/ijms20235852] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 01/03/2023] Open
Abstract
Epilepsy is a common neurological disorder. Despite the availability of a wide range of antiepileptic drugs, these are unsuccessful in preventing seizures in 20–30% of patients. Therefore, new pharmacological strategies are urgently required to control seizures. Modulation of glutamate uptake may have potential in the treatment of pharmacoresistant forms of epilepsy. Previous research showed that the antibiotic ceftriaxone (CTX) increased the expression and functional activity of excitatory amino acid transporter 2 (EAAT2) and exerted considerable anticonvulsant effects. However, other studies did not confirm a significant anticonvulsant effect of CTX administration. We investigated the impacts of CTX treatment on EAAT expression and glutamatergic neurotransmission, as well its anticonvulsant action, in young male Wistar rats. As shown by a quantitative real-time polymerase chain reaction (qPCR) assay and a Western blot analysis, the mRNA but not the protein level of EAAT2 increased in the hippocampus following CTX treatment. Repetitive CTX administration had only a mild anticonvulsant effect on pentylenetetrazol (PTZ)-induced convulsions in a maximal electroshock threshold test (MEST). CTX treatment did not affect the glutamatergic neurotransmission, including synaptic efficacy, short-term facilitation, or the summation of excitatory postsynaptic potentials (EPSPs) in the hippocampus and temporal cortex. However, it decreased the field EPSP (fEPSP) amplitudes evoked by intense electrical stimulation. In conclusion, in young rats, CTX treatment did not induce overexpression of EAAT2, therefore exerting only a weak antiseizure effect. Our data provide new insight into the effects of modulation of EAAT2 expression on brain functioning.
Collapse
|
39
|
Escobar AP, Wendland JR, Chávez AE, Moya PR. The Neuronal Glutamate Transporter EAAT3 in Obsessive-Compulsive Disorder. Front Pharmacol 2019; 10:1362. [PMID: 31803055 PMCID: PMC6872633 DOI: 10.3389/fphar.2019.01362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/28/2019] [Indexed: 01/03/2023] Open
Abstract
Obsessive compulsive disorder (OCD) is a heterogeneous psychiatric disorder affecting 1%–3% of the population worldwide. About half of OCD afflicted individuals do not respond to currently available pharmacotherapy, which is mainly based on serotonin reuptake inhibition. Therefore, there is a critical need to search novel and improved therapeutic targets to treat this devastating disorder. In recent years, accumulating evidence has supported the glutamatergic hypothesis of OCD, and particularly pointing a potential role for the neuronal glutamate transporter EAAT3. This mini-review summarizes recent findings regarding the neurobiological basis of OCD, with an emphasis on the glutamatergic neurotransmission and EAAT3 as a key player in OCD etiology.
Collapse
Affiliation(s)
- Angélica P Escobar
- Centro Interdisciplinario de Neurociencia de Valparaíso CINV, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jens R Wendland
- Centro Interdisciplinario de Neurociencia de Valparaíso CINV, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Andrés E Chávez
- Centro Interdisciplinario de Neurociencia de Valparaíso CINV, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo R Moya
- Centro Interdisciplinario de Neurociencia de Valparaíso CINV, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaiso, Chile
| |
Collapse
|
40
|
Wu P, Bjørn-Yoshimoto WE, Staudt M, Jensen AA, Bunch L. Identification and Structure-Activity Relationship Study of Imidazo[1,2- a]pyridine-3-amines as First Selective Inhibitors of Excitatory Amino Acid Transporter Subtype 3 (EAAT3). ACS Chem Neurosci 2019; 10:4414-4429. [PMID: 31573179 DOI: 10.1021/acschemneuro.9b00447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In the present study, screening of a library of 49,087 compounds at the excitatory amino acid transporter subtype 3 (EAAT3) led to the identification of 2-(furan-2-yl)-8-methyl-N-(o-tolyl)imidazo[1,2-a]pyridin-3-amine (3a) which showed a >20-fold preference for inhibition of EAAT3 (IC50 = 13 μM) over EAAT1,2,4 (EAAT1: IC50 ∼ 250 μM; EAAT2,4: IC50 > 250 μM). It was shown that a small lipophilic substituent (methyl or bromine) at the 7- and/or 8-position was essential for activity. Furthermore, the substitution pattern of the o-tolyl group (compound 5b) and the chemical nature of the substituent in the 2-position (compound 7b) were shown to be essential for the selectivity toward EAAT3 over EAAT1,2. The most prominent analogues to come out of this study are 3a and 3e that display ∼35-fold selectivity for EAAT3 (IC50 = 7.2 μM) over EAAT1,2,4 (IC50 ∼ 250 μM).
Collapse
Affiliation(s)
- Peng Wu
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Walden E. Bjørn-Yoshimoto
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Markus Staudt
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Anders A. Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2100, Denmark
| |
Collapse
|
41
|
McKee SE, Reyes TM. Effect of supplementation with methyl-donor nutrients on neurodevelopment and cognition: considerations for future research. Nutr Rev 2019; 76:497-511. [PMID: 29701796 DOI: 10.1093/nutrit/nuy007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pregnancy represents a critical period in fetal development, such that the prenatal environment can, in part, establish a lifelong trajectory of health or disease for the offspring. Poor nutrition (macro- or micronutrient deficiencies) can adversely affect brain development and significantly increase offspring risk for metabolic and neurological disease development. The concentration of dietary methyl-donor nutrients is known to alter DNA methylation in the brain, and alterations in DNA methylation can have long-lasting effects on gene expression and neuronal function. The decreased availability of methyl-donor nutrients to the developing fetus in models of poor maternal nutrition is one mechanism hypothesized to link maternal malnutrition and disease risk in offspring. Animal studies indicate that supplementation of both maternal and postnatal (early- and later-life) diets with methyl-donor nutrients can attenuate disease risk in offspring; however, clinical research is more equivocal. The objective of this review is to summarize how specific methyl-donor nutrient deficiencies and excesses during pre- and postnatal life alter neurodevelopment and cognition. Emphasis is placed on reviewing the current literature, highlighting challenges within nutrient supplementation research, and considering potential strategies to ensure robust findings in future studies.
Collapse
Affiliation(s)
- Sarah E McKee
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa M Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Combs JA, DeNicola GM. The Non-Essential Amino Acid Cysteine Becomes Essential for Tumor Proliferation and Survival. Cancers (Basel) 2019; 11:cancers11050678. [PMID: 31100816 PMCID: PMC6562400 DOI: 10.3390/cancers11050678] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/07/2023] Open
Abstract
The non-essential amino acid cysteine is used within cells for multiple processes that rely on the chemistry of its thiol group. Under physiological conditions, many non-transformed tissues rely on glutathione, circulating cysteine, and the de novo cysteine synthesis (transsulfuration) pathway as sources of intracellular cysteine to support cellular processes. In contrast, many cancers require exogeneous cystine for proliferation and viability. Herein, we review how the cystine transporter, xCT, and exogenous cystine fuel cancer cell proliferation and the mechanisms that regulate xCT expression and activity. Further, we discuss the potential contribution of additional sources of cysteine to the cysteine pool and what is known about the essentiality of these processes in cancer cells. Finally, we discuss whether cyst(e)ine dependency and associated metabolic alterations represent therapeutically targetable metabolic vulnerabilities.
Collapse
Affiliation(s)
- Joseph A Combs
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| | - Gina M DeNicola
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
43
|
Zhou X, Li G, Zhang S, Wu J. 5-HT1A Receptor Agonist Promotes Retinal Ganglion Cell Function by Inhibiting OFF-Type Presynaptic Glutamatergic Activity in a Chronic Glaucoma Model. Front Cell Neurosci 2019; 13:167. [PMID: 31130845 PMCID: PMC6509153 DOI: 10.3389/fncel.2019.00167] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/08/2019] [Indexed: 01/30/2023] Open
Abstract
Serotonin receptors are potential neuroprotective agents in degenerative diseases of the central nervous system. The protective effects of serotonin receptor (5-HT1A) agonists on the survival and function of retinal ganglion cells (RGCs) by regulating the release of the presynaptic neurotransmitter γ-aminobutyric acid (GABA) were confirmed in our previous study of a chronic glaucoma rat model. However, the roles of excitatory amino acids and their interactions with the 5-HT1A receptor in glaucoma remain unknown. Here, we found that ocular hypertension increased glutamine synthetase (GS) and excitatory amino acid transporter 2 (EAAT2) expression in rat retinas. In addition, the high expression of GS and EAAT2 induced by glaucoma was downregulated by the 5-HT1A receptor agonist 8-OH-DPAT and the 5-HT1A receptor antagonist WAY-100635, respectively. Patch-clamp techniques were used to record glutamate receptor-mediated spontaneous and miniature glutamatergic excitatory post-synaptic currents (sEPSCs and mEPSCs) as well as L-glutamate-induced current in OFF-type and ON-type RGCs in rat retinal slices. Although there were no significant differences in the frequency and amplitude of sEPSC and mEPSC release between normal and glaucoma OFF- and ON-type RGCs, exogenous 8-OH-DPAT administration specifically reduced the frequency, but not the amplitude, of sEPSC and mEPSC release in glaucoma OFF-type rather than ON-type RGCs; these effects were completely blocked by WAY-100635. In summary, 8-OH-DPAT decreases and increases GS and EAAT2 expression of glaucomatous retina, respectively, while decreasing sEPSC and mEPSC frequency. In contrast, WAY-100635 increases and decreases GS and EAAT2 expression of glaucomatous retina, respectively, while increasing sEPSC and mEPSC frequency. The reduction of glutamatergic presynaptic transmission by 8-OH-DPAT deactivates RGCs at the neural network level and reduces the excitotoxic damage in the pathological process of chronic glaucoma.
Collapse
Affiliation(s)
- Xujiao Zhou
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Gang Li
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Shenghai Zhang
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Jihong Wu
- Eye Institute, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Brain Science and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China.,NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China.,Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University, Shanghai, China
| |
Collapse
|
44
|
Allen JD, Bishop JR. A systematic review of genome-wide association studies of antipsychotic response. Pharmacogenomics 2019; 20:291-306. [PMID: 30883267 PMCID: PMC6563266 DOI: 10.2217/pgs-2018-0163] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022] Open
Abstract
Clinical symptom response to antipsychotic medications is highly variable. Genome-wide association studies (GWAS) provide a 'hypothesis-free' method of interrogating the genome for biomarkers of antipsychotic response. We performed a systematic review of GWAS findings for antipsychotic efficacy or effectiveness. 14 studies met our inclusion criteria, ten of which examined antipsychotic response using quantitative rating scales to measure symptom improvement. 15 genome-wide significant loci were identified, seven of which were replicated in other antipsychotic GWAS publications: CNTNAP5, GRID2, GRM7, 8q24 (KCNK9), PCDH7, SLC1A1 and TNIK. Notably, four replicated loci are involved in glutamatergic pathways. Additional validation and evaluation of the biological significance of these markers is warranted. These markers should also be evaluated for clinical utility, especially in the context of other validated pharmacogenomic variants (e.g., CYP450 genes). These findings may generate new avenues for development of novel antipsychotic treatments.
Collapse
Affiliation(s)
- Josiah D Allen
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
- Medigenics Consulting LLC, Minneapolis, MN 55407, USA
| | - Jeffrey R Bishop
- Department of Experimental & Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, MN 55455, USA
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Mahan VL. Neurointegrity and neurophysiology: astrocyte, glutamate, and carbon monoxide interactions. Med Gas Res 2019; 9:24-45. [PMID: 30950417 PMCID: PMC6463446 DOI: 10.4103/2045-9912.254639] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/15/2019] [Indexed: 12/27/2022] Open
Abstract
Astrocyte contributions to brain function and prevention of neuropathologies are as extensive as that of neurons. Astroglial regulation of glutamate, a primary neurotransmitter, is through uptake, release through vesicular and non-vesicular pathways, and catabolism to intermediates. Homeostasis by astrocytes is considered to be of primary importance in determining normal central nervous system health and central nervous system physiology - glutamate is central to dynamic physiologic changes and central nervous system stability. Gasotransmitters may affect diverse glutamate interactions positively or negatively. The effect of carbon monoxide, an intrinsic central nervous system gasotransmitter, in the complex astrocyte homeostasis of glutamate may offer insights to normal brain development, protection, and its use as a neuromodulator and neurotherapeutic. In this article, we will review the effects of carbon monoxide on astrocyte homeostasis of glutamate.
Collapse
Affiliation(s)
- Vicki L. Mahan
- Division of Pediatric Cardiothoracic Surgery in the Department of Surgery, St. Christopher's Hospital for Children/Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
46
|
Liu C, Zhang Y, Liu Q, Jiang L, Li M, Wang S, Long T, He W, Kong X, Qin G, Chen L, Zhang Y, Zhou J. P2X4-receptor participates in EAAT3 regulation via BDNF-TrkB signaling in a model of trigeminal allodynia. Mol Pain 2018; 14:1744806918795930. [PMID: 30146940 PMCID: PMC6111400 DOI: 10.1177/1744806918795930] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective Previous studies of neuropathic pain have suggested that the P2X4
purinoceptor (P2X4R) in spinal microglia is essential for
maintaining allodynia following nerve injury. However, little is
known about its role in inflammatory soup-induced trigeminal
allodynia, which closely mimics chronic migraine status. Here,
we determined the contributions of P2X4R and related signaling
pathways in an inflammatory soup-induced trigeminal allodynia
model. Methods P2X4R gene and protein levels in the trigeminal nucleus caudalis
were analyzed following repeated dural inflammatory soup
infusions. p38, brain-derived neurotrophic factor, excitatory
amino acid transporter 3, c-Fos, and calcitonin gene-related
peptide protein levels in the trigeminal nucleus caudalis, as
well as trigeminal sensitivity, were assessed among the
different groups. Immunofluorescence staining was used to detect
protein localization and expression in the trigeminal nucleus
caudalis. Results Repeated inflammatory dural stimulation induced trigeminal
hyperalgesia and the upregulation of P2X4R. Immunofluorescence
revealed that P2X4R was expressed in trigeminal nucleus caudalis
microglial cells. Blockage of P2X4R produced an anti-nociceptive
effect, which was associated with an inhibition of inflammatory
soup-induced increases in p38, brain-derived neurotrophic
factor, excitatory amino acid transporter 3, c-Fos, and
calcitonin gene-related peptide protein levels. The tyrosine
receptor kinase B antagonist ANA-12 reversed trigeminal
allodynia and the upregulation of excitatory amino acid
transporter 3, c-Fos, and calcitonin gene-related peptide,
whereas the agonist 7,8-dihydroxyflavone exacerbated these
effects. Double immunostaining indicated that p38 and
brain-derived neurotrophic factor were mainly expressed in
microglial cells, whereas excitatory amino acid transporter 3
was primarily expressed in trigeminal nucleus caudalis
neurons. Conclusions These data indicate that microglial P2X4R is involved in the
regulation of excitatory amino acid transporter 3 via
brain-derived neurotrophic factor-tyrosine receptor kinase B
signaling following repeated inflammatory dural stimulation.
Microglial P2X4R activation and microglia–neuron interactions in
the trigeminal nucleus caudalis may play a role in the
pathogenesis of migraine chronicity, and the modulation of P2X4R
activation might be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Chaoyang Liu
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yixin Zhang
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qing Liu
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Li Jiang
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Maolin Li
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Sha Wang
- 2 Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Ting Long
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Wei He
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xueying Kong
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guangcheng Qin
- 2 Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Lixue Chen
- 2 Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuhong Zhang
- 3 The Clinical Molecular Diagnostics Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Jiying Zhou
- 1 Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
47
|
Liang X, Han H, Zhao X, Cao X, Yang M, Tao D, Wu R, Yue X. Quantitative analysis of amino acids in human and bovine colostrum milk samples through iTRAQ labeling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2018; 98:5157-5163. [PMID: 29577310 DOI: 10.1002/jsfa.9032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND The types and quantity of proteins vary widely between bovine and human milk, with corresponding differences in free and hydrolytic amino acids. In this study, the free and hydrolytic amino acids of bovine and human colostrum were for the first time qualitatively and quantitatively determined using isobaric tags for relative and absolute quantification technology combined with liquid chromatography tandem mass spectrometry detection. RESULTS Total free amino acid content was 0.32 g L-1 and 0.63 g L-1 in bovine and human colostrum respectively, with free amino acid content in human colostrum twice that of bovine colostrum. However, total hydrolytic amino acid content was 4.2 g L-1 and 2.2 g L-1 in bovine and human colostrum respectively. We found that the hydrolytic amino acid content in bovine colostrum was higher than that in human colostrum; however, the amount of free amino acids and the overall amino acid content in human colostrum were respectively substantially higher and more varied than in bovine colostrum. CONCLUSION Our findings revealed differences between bovine and human colostrum, with these data providing the basis for further research into amino acid metabolomics and infant formula. © 2018 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaona Liang
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Hongjiao Han
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xue Zhao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xueyan Cao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Mei Yang
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Dongbing Tao
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Rina Wu
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| | - Xiqing Yue
- College of Food Science, ShenYang Agricultural University, ShenYang, P. R. China
| |
Collapse
|
48
|
Eshraghi RS, Deth RC, Mittal R, Aranke M, Kay SIS, Moshiree B, Eshraghi AA. Early Disruption of the Microbiome Leading to Decreased Antioxidant Capacity and Epigenetic Changes: Implications for the Rise in Autism. Front Cell Neurosci 2018; 12:256. [PMID: 30158857 PMCID: PMC6104136 DOI: 10.3389/fncel.2018.00256] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Currently, 1 out of every 59 children in the United States is diagnosed with autism. While initial research to find the possible causes for autism were mostly focused on the genome, more recent studies indicate a significant role for epigenetic regulation of gene expression and the microbiome. In this review article, we examine the connections between early disruption of the developing microbiome and gastrointestinal tract function, with particular regard to susceptibility to autism. The biological mechanisms that accompany individuals with autism are reviewed in this manuscript including immune system dysregulation, inflammation, oxidative stress, metabolic and methylation abnormalities as well as gastrointestinal distress. We propose that these autism-associated biological mechanisms may be caused and/or sustained by dysbiosis, an alteration to the composition of resident commensal communities relative to the community found in healthy individuals and its redox and epigenetic consequences, changes that in part can be due to early use and over-use of antibiotics across generations. Further studies are warranted to clarify the contribution of oxidative stress and gut microbiome in the pathophysiology of autism. A better understanding of the microbiome and gastrointestinal tract in relation to autism will provide promising new opportunities to develop novel treatment modalities.
Collapse
Affiliation(s)
- Rebecca S. Eshraghi
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Richard C. Deth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Rahul Mittal
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Mayank Aranke
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Sae-In S. Kay
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Baharak Moshiree
- Division of Gastroenterology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Adrien A. Eshraghi
- Department of Otolaryngology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
49
|
Glutamate as a potential "survival factor" in an in vitro model of neuronal hypoxia/reoxygenation injury: leading role of the Na +/Ca 2+ exchanger. Cell Death Dis 2018; 9:731. [PMID: 29955038 PMCID: PMC6023866 DOI: 10.1038/s41419-018-0784-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/04/2018] [Accepted: 06/11/2018] [Indexed: 12/23/2022]
Abstract
In brain ischemia, reduction in oxygen and substrates affects mitochondrial respiratory chain and aerobic metabolism, culminating in ATP production impairment, ionic imbalance, and cell death. The restoration of blood flow and reoxygenation are frequently associated with exacerbation of tissue injury, giving rise to ischemia/reperfusion (I/R) injury. In this setting, the imbalance of brain bioenergetics induces important metabolic adaptations, including utilization of alternative energy sources, such as glutamate. Although glutamate has long been considered as a neurotoxin, it can also be used as intermediary metabolite for ATP synthesis, and both the Na+/Ca2+ exchanger (NCX) and the Na+-dependent excitatory amino-acid transporters (EAATs) are essential in this pathway. Here we analyzed the role of NCX in the potential of glutamate to improve metabolism and survival of neuronal cells subjected to hypoxia/reoxygenation (H/R). In SH-SY5Y neuroblastoma cells differentiated into a neuron-like state, H/R produced a significant cell damage, a decrease in ATP cellular content, and intracellular Ca2+ alterations. Exposure to glutamate at the onset of the reoxygenation phase attenuated H/R-induced cell damage and evoked a significant raise in intracellular ATP levels. Furthermore, we found that in H/R cells NCX reverse-mode activity was reduced, and that glutamate limited such reduction. All the effects induced by glutamate supplementation were lost when cells were transfected with small interfering RNA against NCX1 and EAAT3, suggesting the need of a specific functional interplay between these proteins for glutamate-induced protection. Collectively, our results revealed the potential beneficial effect of glutamate in an in vitro model of H/R injury and focused on the essential role exerted by NCX1. Although preliminary, these findings could be a starting point to further investigate in in vivo systems such protective effect in ischemic settings, shedding a new light on the classical view of glutamate as detrimental factor.
Collapse
|
50
|
McKee SE, Zhang S, Chen L, Rabinowitz JD, Reyes TM. Perinatal high fat diet and early life methyl donor supplementation alter one carbon metabolism and DNA methylation in the brain. J Neurochem 2018; 145:362-373. [PMID: 29423909 PMCID: PMC6288810 DOI: 10.1111/jnc.14319] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/05/2018] [Accepted: 01/26/2018] [Indexed: 12/13/2022]
Abstract
One carbon metabolism is regulated by the availability of nutrients known as methyl donors, and disruption of this pathway can affect multiple physiological systems. DNA methylation, critical for the regulation of gene expression, is linked to one carbon metabolism, and can be altered by perinatal diet. In this study, dams (n = 12/group) were fed HF or standard control (SC) diet through pregnancy and lactation, and male and female offspring were then fed either SC or methyl donor-supplemented diet (MDS) between 3 and 6 weeks of age (n = 20-26/group). Concentration of one carbon intermediates and other related metabolites were assessed within brain tissue (prefrontal cortex, PFC) through the use of mass spectrometry at 6 weeks of age. In addition, the expression of target genes and enzymes that participate in DNA methylation or are relevant to one carbon metabolism were measured. We found that MDS increases the concentration of folate intermediates in the PFC, and that this increase is blunted in male offspring from dams fed a HF diet. In addition, perinatal HF diet increased the concentration of cysteine in the PFC of both male and female offspring, consistent with oxidative stress. Furthermore, both maternal HF diet and postnatal MDS altered global DNA methylation in the PFC in males but not females. Collectively, these data demonstrate sex differences in changes in one carbon metabolites in the prefrontal cortex in response to early life high fat diet and methyl donor supplementation. Read the Editorial Highlight for this article on page 358.
Collapse
Affiliation(s)
- SE McKee
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - S Zhang
- Department of Chemistry and Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - L Chen
- Department of Chemistry and Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - JD Rabinowitz
- Department of Chemistry and Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - TM Reyes
- Department of Psychiatry and Behavioral Neurosciences, College of Medicine, University of Cincinnati, Cincinnati, OH 45237, United States
| |
Collapse
|