1
|
Zhang Q, Huo Y, Zhu R, Zhang X, Zeng L, Hu Z. Molecular mechanism of METTL3 regulating hippocampal neuronal injury induced by sepsis-associated encephalopathy. Arch Physiol Biochem 2025:1-11. [PMID: 40025626 DOI: 10.1080/13813455.2025.2465337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 01/02/2025] [Accepted: 02/06/2025] [Indexed: 03/04/2025]
Abstract
OBJECTIVE This study explores the mechanism of methyltransferase like 3 (METTL3) on sepsis-associated encephalopathy (SAE)-induced hippocampal neuronal injury. METHODS A murine model of SAE was established by caecal ligation and puncture. Hippocampal cells were induced by lipopolysaccharide (LPS). The mouse survival was observed and behavioural tests evaluated cognitive function. METTL3 and glutamic-oxaloacetic transaminase 1 (GOT1) expressions were detected via RT-qPCR and Western blot. Immunofluorescence staining examined the co-localization of NeuN and METTL3. The m6A enrichment on GOT1 was determined by MeRIP. RESULTS METTL3 and GOT1 were highly expressed in SAE mice and LPS-stimulated hippocampal cells. SAE mice exhibited cognitive function impairment, reduced survival rate, and decreased neuronal cells. LPS induction increased hippocampal cell apoptosis and enhanced inflammation. Silence of METTL3 reduced hippocampal neuronal injury in SAE mice and LPS-induced hippocampal cell injury. CONCLUSION METTL3-mediated m6A modification on GOT1 mRNA elevates GOT1 expression, thereby aggravating SAE-induced hippocampal neuronal injury.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yan Huo
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Runying Zhu
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xujie Zhang
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lingwei Zeng
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenjie Hu
- Department of Intensive Care Unit, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Perrino S, Vazana U, Prager O, Schori L, Ben-Arie G, Minarik A, Chen YM, Haçariz O, Hashimoto M, Roth Y, Pell GS, Friedman A, Brodt P. Transcranial Magnetic Stimulation Enhances the Therapeutic Effect of IGF-Trap in Intracerebral Glioma Models. Pharmaceuticals (Basel) 2024; 17:1607. [PMID: 39770449 PMCID: PMC11677529 DOI: 10.3390/ph17121607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Glioblastoma multiforme is an aggressive malignancy with a dismal 5-year survival rate of 5-10%. Current therapeutic options are limited, due in part to drug exclusion by the blood-brain barrier (BBB). We have previously shown that high-amplitude repetitive transcranial magnetic stimulation (rTMS) in rats allowed the delivery across the BBB of an IGF signaling inhibitor-IGF-Trap. The objective of this study was to assess the therapeutic effect of IGF-Trap when delivered in conjunction with rTMS on the intracerebral growth of glioma. Results: We found that systemic administration of IGF-Trap without rTMS had a minimal effect on the growth of orthotopically injected glioma cells in rats and mice, compared to control animals injected with vehicle only or treated with sham rTMS. In rats treated with a combination of rTMS and IGF-Trap, we observed a growth retardation of C6 tumors for up to 14 days post-tumor cell injection, although tumors eventually progressed. In mice, tumors were detectable in all control groups by 14-17 days post-injection of glioma GL261 cells and progressed rapidly thereafter. In mice treated with rTMS prior to IGF-Trap administration, tumor growth was inhibited or delayed, although the tumors also eventually progressed. Conclusion: The results showed that rTMS could increase the anti-tumor effect of IGF-Trap during the early phases of tumor growth. Further optimization of the rTMS protocol is required to improve survival outcomes.
Collapse
Affiliation(s)
- Stephanie Perrino
- The Research Institute, The McGill University Health Center, Montreal, QC H4A 3J1, Canada; (S.P.)
| | - Udi Vazana
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zelman Center for Brain Science Research, Ben-Gurion University of the Negev, 1 Ben-Gurion Blvd., Beer-Sheva 8410501, Israel; (U.V.); (O.P.); (A.F.)
| | - Ofer Prager
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zelman Center for Brain Science Research, Ben-Gurion University of the Negev, 1 Ben-Gurion Blvd., Beer-Sheva 8410501, Israel; (U.V.); (O.P.); (A.F.)
| | - Lior Schori
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zelman Center for Brain Science Research, Ben-Gurion University of the Negev, 1 Ben-Gurion Blvd., Beer-Sheva 8410501, Israel; (U.V.); (O.P.); (A.F.)
| | - Gal Ben-Arie
- Faculty of Health Sciences, Ben-Gurion University of the Negev, 1 Ben-Gurion Blvd., Beer-Sheva 8410501, Israel
- Department of Radiology, Soroka Medical Center, Beer-Sheva 8410501, Israel
| | - Anna Minarik
- Department of Medical Neuroscience and the Brain Repair Centre, Faculty of Medicine, Dalhousie University, 5850 College St., Halifax, NS B3H 4R2, Canada
| | - Yinhsuan Michely Chen
- The Research Institute, The McGill University Health Center, Montreal, QC H4A 3J1, Canada; (S.P.)
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Orçun Haçariz
- The Research Institute, The McGill University Health Center, Montreal, QC H4A 3J1, Canada; (S.P.)
- Department of Surgery, Division of Experimental Surgery, McGill University, Montreal, QC H3A 0G4, Canada
| | - Masakazu Hashimoto
- The Research Institute, The McGill University Health Center, Montreal, QC H4A 3J1, Canada; (S.P.)
| | - Yiftach Roth
- Brainsway Ltd., 19 Hartom St., Jerusalem 9777518, Israel; (Y.R.); (G.S.P.)
| | - Gabriel S. Pell
- Brainsway Ltd., 19 Hartom St., Jerusalem 9777518, Israel; (Y.R.); (G.S.P.)
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, The Zelman Center for Brain Science Research, Ben-Gurion University of the Negev, 1 Ben-Gurion Blvd., Beer-Sheva 8410501, Israel; (U.V.); (O.P.); (A.F.)
- Department of Medical Neuroscience and the Brain Repair Centre, Faculty of Medicine, Dalhousie University, 5850 College St., Halifax, NS B3H 4R2, Canada
| | - Pnina Brodt
- The Research Institute, The McGill University Health Center, Montreal, QC H4A 3J1, Canada; (S.P.)
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
- Department of Surgery, Division of Experimental Surgery, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
3
|
Pérez-Mato M, Dopico-López A, Akkoc Y, López-Amoedo S, Correa-Paz C, Candamo-Lourido M, Iglesias-Rey R, López-Arias E, Bugallo-Casal A, da Silva-Candal A, Bravo SB, Chantada-Vázquez MDP, Arias S, Santamaría-Cadavid M, Estany-Gestal A, Zaghmi A, Gauthier MA, Gutiérrez-Fernández M, Martin A, Llop J, Rodríguez C, Almeida Á, Migliavacca M, Polo E, Pelaz B, Gozuacik D, El Yamani N, SenGupta T, Rundén-Pran E, Vivancos J, Castellanos M, Díez-Tejedor E, Sobrino T, Rabinkov A, Mirelman D, Castillo J, Campos F. Preclinical validation of human recombinant glutamate-oxaloacetate transaminase for the treatment of acute ischemic stroke. iScience 2024; 27:111108. [PMID: 39524351 PMCID: PMC11543921 DOI: 10.1016/j.isci.2024.111108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/21/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The blood enzyme glutamate-oxaloacetate transaminase (GOT) has been postulated as an effective therapeutic to protect the brain during stroke. To demonstrate its potential clinical utility, a new human recombinant form of GOT (rGOT) was produced for medical use. We tested the pharmacokinetics and evaluated the protective efficacy of rGOT in rodent and non-human primate models that reflected clinical stroke conditions. We found that continuous intravenous administration of rGOT within the first 8 h after ischemic onset significantly reduced the infarct size in both severe (30%) and mild lesions (48%). Cerebrospinal fluid and proteomics analysis, in combination with positron emission tomography imaging, indicated that rGOT can reach the brain and induce cytoprotective autophagy and induce local protection by alleviating neuronal apoptosis. Our results suggest that rGOT can be safely used immediately in patients suspected of having a stroke. This study requires further validation in clinical stroke populations.
Collapse
Affiliation(s)
- María Pérez-Mato
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Antonio Dopico-López
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Yunus Akkoc
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Sonia López-Amoedo
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María Candamo-Lourido
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Esteban López-Arias
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ana Bugallo-Casal
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Andrés da Silva-Candal
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - María del Pilar Chantada-Vázquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Research Unit, Lucus Augusti University Hospital (HULA), Servizo Galego de Saúde (SERGAS), 27002 Lugo, Spain
| | - Susana Arias
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - María Santamaría-Cadavid
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unit of Methodology of the Research, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Ahlem Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Marc A. Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC J3X 1S2, Canada
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Abraham Martin
- Achucarro Basque Center for Neuroscience, 48940 Leioa, Spain
- Ikerbasque Basque Foundation for Science, 48009 Bilbao, Spain
| | - Jordi Llop
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 San Sebastian, Spain
| | - Cristina Rodríguez
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Ángeles Almeida
- Institute of Functional Biology and Genomics (IBFG), CSIC, University of Salamanca, 37007 Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, CSIC, University of Salamanca, 37007 Salamanca, Spain
| | - Martina Migliavacca
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Ester Polo
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Center for Research in Biological Chemistry and Molecular Materials (CiQUS), University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul 34450, Turkey
- Department of Medical Biology, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Naouale El Yamani
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Tanima SenGupta
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - Elise Rundén-Pran
- Health Effects Laboratory, Department for Environmental Chemistry, NILU-Norwegian Institute for Air Research, 2027 Kjeller, Norway
| | - José Vivancos
- Stroke Unit, Department of Neurology, Hospital Universitario de La Princesa & Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain
| | - Mar Castellanos
- Neurology Service, University Hospital Complex of A Coruña, A Coruña Biomedical Research Institute, 15006 A Coruña, Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology, Neurology and Cerebrovascular Disease Group, Neuroscience Area of Hospital La Paz Institute for Health Research – IdiPAZ (La Paz University Hospital- Universidad Autónoma de Madrid), 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Aharon Rabinkov
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - David Mirelman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Francisco Campos
- Translational Stroke Laboratory Group (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
4
|
Hervella P, Sampedro-Viana A, Fernández-Rodicio S, Rodríguez-Yáñez M, López-Dequidt I, Pumar JM, Mosqueira AJ, Bazarra-Barreiros M, Abengoza-Bello MT, Ortega-Espina S, Ouro A, Pérez-Mato M, Campos F, Sobrino T, Castillo J, Alonso-Alonso ML, Iglesias-Rey R. Precision Medicine for Blood Glutamate Grabbing in Ischemic Stroke. Int J Mol Sci 2024; 25:6554. [PMID: 38928260 PMCID: PMC11204254 DOI: 10.3390/ijms25126554] [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: 05/13/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Glutamate grabbers, such as glutamate oxaloacetate transaminase (GOT), have been proposed to prevent excitotoxicity secondary to high glutamate levels in stroke patients. However, the efficacy of blood glutamate grabbing by GOT could be dependent on the extent and severity of the disruption of the blood-brain barrier (BBB). Our purpose was to analyze the relationship between GOT and glutamate concentration with the patient's functional status differentially according to BBB serum markers (soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) and leukoaraiosis based on neuroimaging). This retrospective observational study includes 906 ischemic stroke patients. We studied the presence of leukoaraiosis and the serum levels of glutamate, GOT, and sTWEAK in blood samples. Functional outcome was assessed using the modified Rankin Scale (mRS) at 3 months. A significant negative correlation between GOT and glutamate levels at admission was shown in those patients with sTWEAK levels > 2900 pg/mL (Pearson's correlation coefficient: -0.249; p < 0.0001). This correlation was also observed in patients with and without leukoaraiosis (Pearson's correlation coefficients: -0.299; p < 0.001 vs. -0.116; p = 0.024). The logistic regression model confirmed the association of higher levels of GOT with lower odds of poor outcome at 3 months when sTWEAK levels were >2900 pg/mL (OR: 0.41; CI 95%: 0.28-0.68; p < 0.0001) or with leukoaraiosis (OR: 0.75; CI 95%: 0.69-0.82; p < 0.0001). GOT levels are associated with glutamate levels and functional outcomes at 3 months, but only in those patients with leukoaraiosis and elevated sTWEAK levels. Consequently, therapies targeting glutamate grabbing might be more effective in patients with BBB dysfunction.
Collapse
Grants
- SAF2017-84267-R, PDC2021-121455-I00 Spanish Ministry of Science and Innovation
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- PI17/01103, PI22/00938, PI21/01256/, DTS23/00103, RD16/0019/0001, RD21/0006/0003, CB22/05/00067, CPII17/00027, CPII19/00020, CP22/00061, FI22/00200 Instituto de Salud Carlos III
- EAPA_791/2018_ NEUROATLANTIC, 0624_2IQBIONEURO_6_E INTERREG
Collapse
Affiliation(s)
- Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain;
| | - Iria López-Dequidt
- Department of Neurology, Hospital Clínico Universitario de Ferrol, 15405 Ferrol, Spain;
| | - José M. Pumar
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Antonio J. Mosqueira
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Marcos Bazarra-Barreiros
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - María Teresa Abengoza-Bello
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Sara Ortega-Espina
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.O.); (T.S.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Pérez-Mato
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.P.-M.); (F.C.)
| | - Francisco Campos
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.P.-M.); (F.C.)
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.O.); (T.S.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| |
Collapse
|
5
|
Liu L, Tian X, Li W. Mechanistic study of the anti-excitatory amino acid toxicity of Bushen Zhichan decoction for Parkinson's disease based on the transcriptional regulation of EAAT1 by YY1. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117857. [PMID: 38350506 DOI: 10.1016/j.jep.2024.117857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bushen Zhichan decoction (BSZCF) is derived from Liuwei Dihuang Pill, a famous Chinese herbal formula recorded in the book Key to Therapeutics of Children's Diseases. It has been widely used as a basic prescription for nourishing and tonifying the liver and kidneys to treat Parkinson's disease (PD), but its mechanism remains to be explored. AIM OF THE STUDY BSZCF, a Chinese herbal formula comprising five herbs: Rehmannia glutinosa (Gaertn.) DC., Dioscorea oppositifolia L., Cornus officinalis Siebold & Zucc., Fallopia multiflora (Thunb.) Haraldson and Cistanche tubulosa (Schenk) Wight, is used clinically to treat PD. In vivo and in vitro experiments were designed to elucidate the mechanism of BSZCF in the protection of dopamine (DA) neurons and the treatment of PD. The toxicity of excitatory amino acids (EAA) may be attenuated by inhibiting the transcription factor Yin Yang 1 (YY1) and up-regulating the expression of excitatory amino acid transporter 1 (EAAT1). MATERIALS AND METHODS IN VIVO: After 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) was intraperitoneally injected into specific pathogen free (SPF) C57BL/6J mice, model mice were intragastrically given adamantane hydrochloride tablets (AHT) or different doses of BSZCF for 14 days. Both open field and pole-climbing tests were conducted to assess behavioral changes. In vitro: 1-Methyl-4-phe-nylpyridiniumiodide (MPP+)-injured human neuroblastoma cells (SH-SY5Y) were utilized to construct PD cell models. Primary astrocytes were transfected with EAAT1 and YY1 lentiviruses for EAAT1 gene knockout and YY1 gene knockout astrocytes, respectively. The high performance liquid chromatography-mass spectrometry (HPLC-MS) analysis of BSZCF was performed to control the quality of blood drugs. The optimal concentration and time of PD cell models treated by BSZCF were determined by the use of Cell Counting Kit-8 (CCK8). Enzyme-linked immunosorbent assay (ELISA) was used for measuring glutamate (Glu) in the peripheral blood and cells of each group. Western blotting (WB) and real-time quantitative polymerase chain reaction (qPCR) were used to detect tyrosine hydroxylase (TH), dopamine transporters (DAT), EAAT1 and YY1 protein and mRNA. After the blockade of EAAT1, immunofluorescence (IF) assay was used to detect the TH protein in each group. RESULTS In vivo research showed that BSZCF improved the behavioral symptoms of PD mice, and reduced the death of DA neurons and the level of Glu. The mechanism may be related to the decrease of YY1 expression and the increase of EAAT1 levels. In vitro experiments showed that the anti-excitatory amino acid toxicity of BSZCF was achieved by inhibiting YY1 expression and regulating EAAT1. CONCLUSIONS By inhibiting YY1 to increase the expression of EAAT1 and attenuating the toxicity of Glu, BSZCF exerts the effect of protecting DA neurons and treating PD-like symptoms in mice.
Collapse
Affiliation(s)
- Leilei Liu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Xinyun Tian
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| | - Wentao Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, China.
| |
Collapse
|
6
|
Zhang C, Ma Y, Zhao Y, Guo N, Han C, Wu Q, Mu C, Zhang Y, Tan S, Zhang J, Liu X. Systematic review of melatonin in cerebral ischemia-reperfusion injury: critical role and therapeutic opportunities. Front Pharmacol 2024; 15:1356112. [PMID: 38375039 PMCID: PMC10875093 DOI: 10.3389/fphar.2024.1356112] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/22/2024] [Indexed: 02/21/2024] Open
Abstract
Cerebral ischemia-reperfusion (I/R) injury is the predominant causes for the poor prognosis of ischemic stroke patients after reperfusion therapy. Currently, potent therapeutic interventions for cerebral I/R injury are still very limited. Melatonin, an endogenous hormone, was found to be valid in preventing I/R injury in a variety of organs. However, a systematic review covering all neuroprotective effects of melatonin in cerebral I/R injury has not been reported yet. Thus, we perform a comprehensive overview of the influence of melatonin on cerebral I/R injury by collecting all available literature exploring the latent effect of melatonin on cerebral I/R injury as well as ischemic stroke. In this systematic review, we outline the extensive scientific studies and summarize the beneficial functions of melatonin, including reducing infarct volume, decreasing brain edema, improving neurological functions and attenuating blood-brain barrier breakdown, as well as its key protective mechanisms on almost every aspect of cerebral I/R injury, including inhibiting oxidative stress, neuroinflammation, apoptosis, excessive autophagy, glutamate excitotoxicity and mitochondrial dysfunction. Subsequently, we also review the predictive and therapeutic implications of melatonin on ischemic stroke reported in clinical studies. We hope that our systematic review can provide the most comprehensive introduction of current advancements on melatonin in cerebral I/R injury and new insights into personalized diagnosis and treatment of ischemic stroke.
Collapse
Affiliation(s)
- Chenguang Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yumei Ma
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yating Zhao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chen Han
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian Wu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Changqing Mu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yue Zhang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shutong Tan
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Zhang
- Department of Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Shenyang, Liaoning, China
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning, China
| | - Xu Liu
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Cheng M, Ding Y, Kim E, Geng X. Exploring the Therapeutic Potential of Peritoneal Dialysis (PD) in the Treatment of Neurological Disorders. Cell Transplant 2024; 33:9636897241236576. [PMID: 38506429 PMCID: PMC10956140 DOI: 10.1177/09636897241236576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 03/21/2024] Open
Abstract
Peritoneal dialysis (PD) is a well-established renal replacement therapy commonly employed in clinical practice. While its primary application is in the treatment of kidney disease, its potential in addressing other systemic disorders, including neurological diseases, has garnered increasing interest. This study provides a comprehensive overview of the related technologies, unique advantages, and clinical applications of PD in the context of neurological disorders. By exploring the mechanism underlying PD, its application in neurological diseases, and associated complications, we addressed the feasibility and benefits of PD as an adjunct therapy for various neurological conditions. Our study aims to highlight its role in detoxification and symptom management, as well as its advantages over other universally accepted methods of renal replacement therapy. Our goal is to bring to the spotlight the therapeutic potential of PD in neurological diseases, such as stroke, stimulate further research, and broaden the scope of its application in the clinical setting.
Collapse
Affiliation(s)
- Muyuan Cheng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Enoch Kim
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
8
|
Huang Y, Wang Z, Huang ZX, Liu Z. Biomarkers and the outcomes of ischemic stroke. Front Mol Neurosci 2023; 16:1171101. [PMID: 37342100 PMCID: PMC10277488 DOI: 10.3389/fnmol.2023.1171101] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/16/2023] [Indexed: 06/22/2023] Open
Abstract
Biomarkers are measurable substances that could be used as objective indicators for disease diagnosis, responses to treatments, and outcomes predictions. In this review, we summarized the data on a number of important biomarkers including glutamate, S100B, glial fibrillary acidic protein, receptor for advanced glycation end-products, intercellular adhesion molecule-1, von willebrand factor, matrix metalloproteinase-9, interleukin-6, tumor necrosis factor-a, activated protein C, copeptin, neuron-specific enolase, tau protein, gamma aminobutyric acid, blood glucose, endothelial progenitor cells, and circulating CD34-positive cells that could be potentially used to indicate the disease burden and/or predict clinical outcome of ischemic stroke. We examined the relationship between specific biomarkers and disease burden and outcomes and discussed the potential mechanisms underlying the relationship. The clinical significance and implications of these biomarkers were also discussed.
Collapse
Affiliation(s)
- Ying Huang
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhenzhen Wang
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhi-Xin Huang
- Department of Neurology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| |
Collapse
|
9
|
Snider S, Albano L, Gagliardi F, Comai S, Roncelli F, De Domenico P, Pompeo E, Panni P, Bens N, Calvi MR, Mortini P, Ruban A. Substantially elevated serum glutamate and CSF GOT-1 levels associated with cerebral ischemia and poor neurological outcomes in subarachnoid hemorrhage patients. Sci Rep 2023; 13:5246. [PMID: 37002262 PMCID: PMC10066256 DOI: 10.1038/s41598-023-32302-3] [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: 08/24/2022] [Accepted: 03/25/2023] [Indexed: 04/03/2023] Open
Abstract
Brain injury and cerebral vasospasm during the 14 days after the subarachnoid hemorrhage (SAH) are considered the leading causes of poor outcomes. The primary injury induces a cascade of events, including increased intracranial pressure, cerebral vasospasm and ischemia, glutamate excitotoxicity, and neuronal cell death. The objective of this study was to monitor the time course of glutamate, and associated enzymes, such as glutamate-oxaloacetate transaminase (GOT1), glutamate-pyruvate transaminase (GPT) in cerebrospinal fluid (CSF) and serum, shortly after SAH, and to assess their prognostic value. A total of 74 participants participated in this study: 45 participants with SAH and 29 controls. Serum and CSF were sampled up to 14 days after SAH. SAH participants' clinical and neurological status were assessed at hospitalization, at discharge from the hospital, and 3 months after SAH. Furthermore, a logistic regression analysis was carried out to evaluate the ability of GOT1 and glutamate levels to predict neurological outcomes. Our results demonstrated consistently elevated serum and CSF glutamate levels after SAH. Furthermore, serum glutamate level was significantly higher in patients with cerebral ischemia and poor neurological outcome. CSF GOT1 was significantly higher in patients with uncontrolled intracranial hypertension and cerebral ischemia post-SAH, and independently predicted poor neurological outcomes.
Collapse
Affiliation(s)
- Silvia Snider
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Albano
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Gagliardi
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Comai
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Francesca Roncelli
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pierfrancesco De Domenico
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Edoardo Pompeo
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Panni
- Department of Neuroradiology, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Nicole Bens
- Behavioral Neuroscience, Human Movement Science, Mathematics, Pre-Medicine, Northeastern University COS, Boston, MA, USA
| | - Maria Rosa Calvi
- Department of Neurocritical Care, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Ruban
- Sackler Faculty of Medicine, Steyer School of Health Professions, Tel Aviv University, P.O. Box 39040, 6997801, Tel-Aviv, Israel.
- Sagol School of Neuroscience, Tel-Aviv University, P.O. Box 39040, 6997801, Tel-Aviv, Israel.
| |
Collapse
|
10
|
Ahmadabad MA, Naeimi A, Keymoradzadeh A, Faghani S, Ahmadabad MA, Boroujeni NA, Mohammadpour H, Saberi A. Evaluation of De Ritis (AST/ALT), ALP/ALT, and AST/ALP ratios as prognostic factors in patients with acute ischemic stroke. BMC Neurol 2022; 22:450. [PMID: 36463106 PMCID: PMC9719238 DOI: 10.1186/s12883-022-02989-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Stroke is one of the leading causes of disability worldwide. Recently, stroke prognosis estimation has received much attention. This study investigates the prognostic role of aspartate transaminase/alanine transaminase (De Ritis, AAR), alkaline phosphatase/alanine transaminase (ALP/ALT), and aspartate transaminase/alkaline phosphatase (AST/ALP) ratios in acute ischemic stroke (AIS). METHODS This retrospective cohort study involved patients who experienced their first-ever AIS between September 2019 and June 2021. Clinical and laboratory data were collected within the first 24 hours after admission. Functional and mortality outcomes were evaluated 90 days after hospital discharge in clinical follow-up. Functional outcome was assessed by a modified Rankin Scale (mRS). The correlation between the laboratory data and study outcomes was evaluated using univariate analysis. In addition, regression models were developed to evaluate the predictive role of AST/ALP, ALP/ALT, and AAR ratios on the study outcomes. RESULTS Two hundred seventy-seven patients (mean age 69.10 ± 13.55, 53.1% female) were included. According to univariate analysis, there was a weak association between 3-months mRS, and both AST/ALT (r = 0.222, P < 0.001), and AST/ALP (r = 0.164, P = 0.008). Subsequently, higher levels of these ratios and absolute values of AST, ALT, and ALP were reported in deceased patients. Based on regression models adjusted with co-variable (age, gender, underlying disease, and history of smoking) AST/ALT and AST/ALP ratios had a significant independent association with 3-month mRS (CI:1.37-4.52, p = 0.003, and CI: 4.45-11,547.32, p = 0.007, respectively) and mortality (CI: 0.17-1.06, adjusted R2 = 0.21, p = 0.007, and CI: 0.10-2.91, p = 0.035, adjusted R2 = 0.20, respectively). CONCLUSIONS Elevated AST/ALP and AAR ratios at admission were correlated with poorer outcomes at 3 months in patients with first-ever AIS. Prospective studies in larger cohorts are required to confirm our findings and to evaluate further whether the AST/ALP and De Ritis ratios may represent a useful tool for determining the prognosis of AIS patients.
Collapse
Affiliation(s)
- Mona Asghari Ahmadabad
- grid.411874.f0000 0004 0571 1549Neurosciences Research Center, Neurology Department, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arvin Naeimi
- grid.411874.f0000 0004 0571 1549Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arman Keymoradzadeh
- grid.411600.2Department of Neurosurgery, School of Medicine Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahriar Faghani
- grid.411705.60000 0001 0166 0922Department of Neurology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Asghari Ahmadabad
- grid.411874.f0000 0004 0571 1549Neurosciences Research Center, Neurology Department, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasim Athari Boroujeni
- grid.411874.f0000 0004 0571 1549Neurosciences Research Center, Neurology Department, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hanieh Mohammadpour
- grid.411874.f0000 0004 0571 1549Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Alia Saberi
- grid.411874.f0000 0004 0571 1549Neurosciences Research Center, Neurology Department, Poursina Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
11
|
Gruenbaum BF, Zlotnik A, Fleidervish I, Frenkel A, Boyko M. Glutamate Neurotoxicity and Destruction of the Blood–Brain Barrier: Key Pathways for the Development of Neuropsychiatric Consequences of TBI and Their Potential Treatment Strategies. Int J Mol Sci 2022; 23:ijms23179628. [PMID: 36077024 PMCID: PMC9456007 DOI: 10.3390/ijms23179628] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 11/18/2022] Open
Abstract
Traumatic brain injury (TBI) is associated with significant cognitive and psychiatric conditions. Neuropsychiatric symptoms can persist for years following brain injury, causing major disruptions in patients’ lives. In this review, we examine the role of glutamate as an aftereffect of TBI that contributes to the development of neuropsychiatric conditions. We hypothesize that TBI causes long-term blood–brain barrier (BBB) dysfunction lasting many years and even decades. We propose that dysfunction in the BBB is the central factor that modulates increased glutamate after TBI and ultimately leads to neurodegenerative processes and subsequent manifestation of neuropsychiatric conditions. Here, we have identified factors that determine the upper and lower levels of glutamate concentration in the brain after TBI. Furthermore, we consider treatments of disruptions to BBB integrity, including repairing the BBB and controlling excess glutamate, as potential therapeutic modalities for the treatment of acute and chronic neuropsychiatric conditions and symptoms. By specifically focusing on the BBB, we hypothesize that restoring BBB integrity will alleviate neurotoxicity and related neurological sequelae.
Collapse
Affiliation(s)
- Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| | - Ilya Fleidervish
- Department of Physiology and Cell Biology, Faculty of Health Sciences and Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Amit Frenkel
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion of the Negev, Beer-Sheva 84105, Israel
- Correspondence:
| |
Collapse
|
12
|
Glutamate Efflux across the Blood–Brain Barrier: New Perspectives on the Relationship between Depression and the Glutamatergic System. Metabolites 2022; 12:metabo12050459. [PMID: 35629963 PMCID: PMC9143347 DOI: 10.3390/metabo12050459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Depression is a significant cause of disability and affects millions worldwide; however, antidepressant therapies often fail or are inadequate. Current medications for treating major depressive disorder can take weeks or months to reach efficacy, have troubling side effects, and are limited in their long-term capabilities. Recent studies have identified a new set of glutamate-based approaches, such as blood glutamate scavengers, which have the potential to provide alternatives to traditional antidepressants. In this review, we hypothesize as to the involvement of the glutamate system in the development of depression. We identify the mechanisms underlying glutamate dysregulation, offering new perspectives on the therapeutic modalities of depression with a focus on its relationship to blood–brain barrier (BBB) permeability. Ultimately, we conclude that in diseases with impaired BBB permeability, such as depression following stroke or traumatic brain injury, or in neurogenerative diseases, the glutamate system should be considered as a pathway to treatment. We propose that drugs such as blood glutamate scavengers should be further studied for treatment of these conditions.
Collapse
|
13
|
Kaplan-Arabaci O, Acari A, Ciftci P, Gozuacik D. Glutamate Scavenging as a Neuroreparative Strategy in Ischemic Stroke. Front Pharmacol 2022; 13:866738. [PMID: 35401202 PMCID: PMC8984161 DOI: 10.3389/fphar.2022.866738] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/08/2022] [Indexed: 12/17/2022] Open
Abstract
Stroke is the second highest reason of death in the world and the leading cause of disability. The ischemic stroke makes up the majority of stroke cases that occur due to the blockage of blood vessels. Therapeutic applications for ischemic stroke include thrombolytic treatments that are in limited usage and only applicable to less than 10% of the total stroke patients, but there are promising new approaches. The main cause of ischemic neuronal death is glutamate excitotoxicity. There have been multiple studies focusing on neuroprotection via reduction of glutamate both in ischemic stroke and other neurodegenerative diseases that ultimately failed due to the obstacles in delivery. At that point, systemic glutamate grabbing, or scavenging is an ingenious way of decreasing glutamate levels upon ischemic stroke. The main advantage of this new therapeutic method is the scavengers working in the circulating blood so that there is no interference with the natural brain neurophysiology. In this review, we explain the molecular mechanisms of ischemic stroke, provide brief information about existing drugs and approaches, and present novel systemic glutamate scavenging methods. This review hopefully will elucidate the potential usage of the introduced therapeutic approaches in stroke patients.
Collapse
Affiliation(s)
- Oykum Kaplan-Arabaci
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Alperen Acari
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Pinar Ciftci
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Devrim Gozuacik
- Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Sabancı University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey.,School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
14
|
Zaghmi A, Pérez-Mato M, Dopico-López A, Candamo-Lourido M, Campos F, Gauthier MA. New Perspectives for Developing Therapeutic Bioconjugates of Metabolite-Depleting Enzymes: Lessons Learned Combating Glutamate Excitotoxicity. Biomacromolecules 2022; 23:1864-1872. [PMID: 35394759 DOI: 10.1021/acs.biomac.2c00117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glutamate, the main excitatory neurotransmitter in the central nervous system, plays an essential role in several cognitive activities such as memorizing and learning. Excessive glutamate release and disturbance of glutamate homeostasis participates in multiple neuronal pathologies including cerebral ischemia (inadequate blood supply), traumatic brain injury (e.g., from a fall or an accident), multiple sclerosis, epilepsy, migraine, fetal hypoxia, or Alzheimer's disease. Attenuating excitotoxicity by, for example, targeting glutamate receptors has proved to be beneficial in animal models but has largely failed in clinical trials because of toxic side effects. New therapeutic concepts have been explored to reduce the excitotoxic effect caused by the excessive glutamate release by using or stimulating glutamate-depleting enzymes in the bloodstream. These enzymes indirectly act upon the brain by depleting glutamate in the bloodstream, which is believed to siphon it out of the brain. Recent studies have shown that bioconjugate approaches applied to such enzymes exacerbate this therapeutic effect but raise additional questions for future research. This Perspective provides an overview of lessons learned by our group when exploring bioconjugate approaches for combatting glutamate excitotoxicity as an illustration of how research on therapeutic bioconjugates is evolving.
Collapse
Affiliation(s)
- Ahlem Zaghmi
- Institut National de la Recherche Scientifique, EMT Research Center, Varennes J3X 1S2, Canada
| | - María Pérez-Mato
- Neuroscience and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Center, La Paz University Hospital, Neuroscience Area of IdiPAZ Health Research Institute, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Antonio Dopico-López
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15782, Spain
| | - María Candamo-Lourido
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15782, Spain
| | - Francisco Campos
- Clinical Neuroscience Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15782, Spain
| | - Marc A Gauthier
- Institut National de la Recherche Scientifique, EMT Research Center, Varennes J3X 1S2, Canada
| |
Collapse
|
15
|
Miller MR, DiBattista A, Patel MA, Daley M, Tenn C, Nakashima A, Rhind SG, Vartanian O, Shiu MY, Caddy N, Garrett M, Saunders D, Smith I, Jetly R, Fraser DD. A Distinct Metabolite Signature in Military Personnel Exposed to Repetitive Low-Level Blasts. Front Neurol 2022; 13:831792. [PMID: 35463119 PMCID: PMC9021419 DOI: 10.3389/fneur.2022.831792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/10/2022] [Indexed: 11/30/2022] Open
Abstract
Military Breachers and Range Staff (MBRS) are subjected to repeated sub-concussive blasts, and they often report symptoms that are consistent with a mild traumatic brain injury (mTBI). Biomarkers of blast injury would potentially aid blast injury diagnosis, surveillance and avoidance. Our objective was to identify plasma metabolite biomarkers in military personnel that were exposed to repeated low-level or sub-concussive blast overpressure. A total of 37 military members were enrolled (18 MBRS and 19 controls), with MBRS having participated in 8-20 breaching courses per year, with a maximum exposure of 6 blasts per day. The two cohorts were similar except that the number of blast exposures were significantly higher in the MBRS, and the MBRS cohort suffered significantly more post-concussive symptoms and poorer health on assessment. Metabolomics profiling demonstrated significant differences between groups with 74% MBRS classification accuracy (CA). Feature reduction identified 6 metabolites that resulted in a MBRS CA of 98%, and included acetic acid (23.7%), formate (22.6%), creatine (14.8%), acetone (14.2%), methanol (12,7%), and glutamic acid (12.0%). All 6 metabolites were examined with individual receiver operating characteristic (ROC) curve analyses and demonstrated areas-under-the-curve (AUCs) of 0.82-0.91 (P ≤ 0.001) for MBRS status. Several parsimonious combinations of three metabolites increased accuracy of ROC curve analyses to AUCs of 1.00 (P < 0.001), while a combination of volatile organic compounds (VOCs; acetic acid, acetone and methanol) yielded an AUC of 0.98 (P < 0.001). Candidate biomarkers for chronic blast exposure were identified, and if validated in a larger cohort, may aid surveillance and care of military personnel. Future point-of-care screening could be developed that measures VOCs from breath, with definitive diagnoses confirmed with plasma metabolomics profiling.
Collapse
Affiliation(s)
- Michael R. Miller
- Lawson Health Research Institute, London, ON, Canada
- Department of Pediatrics, Western University, London, ON, Canada
| | - Alicia DiBattista
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
- Neurolytix Inc., Toronto, ON, Canada
| | - Maitray A. Patel
- Department of Computer Science, Western University, London, ON, Canada
| | - Mark Daley
- Department of Computer Science, Western University, London, ON, Canada
- The Vector Institute for Artificial Intelligence, Toronto, ON, Canada
| | - Catherine Tenn
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Ann Nakashima
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
| | - Shawn G. Rhind
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
- Faculty of Kinesiology & Physical Education, University of Toronto, Toronto, ON, Canada
| | - Oshin Vartanian
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
| | - Maria Y. Shiu
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
| | - Norleen Caddy
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Michelle Garrett
- Defence Research and Development Canada, Suffield Research Centre, Medicine Hat, AB, Canada
| | - Doug Saunders
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
| | - Ingrid Smith
- Defence Research and Development Canada, Toronto Research Centre, Toronto, ON, Canada
| | - Rakesh Jetly
- Canadian Forces Health Services, National Defence Headquarters, Ottawa, ON, Canada
- Department of Psychiatry, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Douglas D. Fraser
- Lawson Health Research Institute, London, ON, Canada
- Department of Pediatrics, Western University, London, ON, Canada
- Neurolytix Inc., Toronto, ON, Canada
- Clinical Neurological Sciences, Western University, London, ON, Canada
- Physiology and Pharmacology, Western University, London, ON, Canada
| |
Collapse
|
16
|
Frank D, Gruenbaum BF, Shelef I, Zvenigorodsky V, Severynovska O, Gal R, Dubilet M, Zlotnik A, Kofman O, Boyko M. Blood Glutamate Scavenging With Pyruvate as a Novel Preventative and Therapeutic Approach for Depressive-Like Behavior Following Traumatic Brain Injury in a Rat Model. Front Neurosci 2022; 16:832478. [PMID: 35237125 PMCID: PMC8883046 DOI: 10.3389/fnins.2022.832478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022] Open
Abstract
Depression is a common and serious complication following traumatic brain injury (TBI). Both depression and TBI have independently been associated with pathologically elevated extracellular brain glutamate levels. In the setting of TBI, blood glutamate scavenging with pyruvate has been widely shown as an effective method to provide neuroprotection by reducing blood glutamate and subsequent brain glutamate levels. Here we evaluate pyruvate as a novel approach in the treatment and prevention of post-TBI depression-like behavior in a rat model. Rats were divided into five groups: (1) sham-operated control with pyruvate, (2) sham-operated control with placebo, (3) post-TBI with placebo, (4) post-TBI given preventative pyruvate, and (5) post-TBI treated with pyruvate. These groups had an equal number of females and males. Rats were assessed for depressive-like behavior, neurological status, and glutamate levels in the blood and brain. Post-TBI neurological deficits with concurrent elevations in glutamate levels were demonstrated, with peak glutamate levels 24 h after TBI. Following TBI, the administration of either prophylactic or therapeutic pyruvate led to reduced glutamate levels, improved neurologic recovery, and improved depressive-like behavior. Glutamate scavenging with pyruvate may be an effective prophylactic and therapeutic option for post-TBI depression by reducing associated elevations in brain glutamate levels.
Collapse
Affiliation(s)
- Dmitry Frank
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Benjamin F. Gruenbaum
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Vladislav Zvenigorodsky
- Department of Radiology, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Olena Severynovska
- Department of Physiology, Faculty of Biology, Ecology and Medicine, Dnepropetrovsk State University, Dnepropetrovsk, Ukraine
| | - Ron Gal
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Michael Dubilet
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Ora Kofman
- Department of Psychology, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Be’er Sheva, Israel
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Ben-Gurion University of the Negev, Be’er Sheva, Israel
- *Correspondence: Matthew Boyko,
| |
Collapse
|
17
|
Goldshmit Y, Perelroizen R, Yakovchuk A, Banyas E, Mayo L, David S, Benbenishty A, Blinder P, Shalom M, Ruban A. Blood glutamate scavengers increase pro-apoptotic signaling and reduce metastatic melanoma growth in-vivo. Sci Rep 2021; 11:14644. [PMID: 34282238 PMCID: PMC8290021 DOI: 10.1038/s41598-021-94183-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/06/2021] [Indexed: 11/09/2022] Open
Abstract
Inhibition of extracellular glutamate (Glu) release decreases proliferation and invasion, induces apoptosis, and inhibits melanoma metastatic abilities. Previous studies have shown that Blood-glutamate scavenging (BGS), a novel treatment approach, has been found to be beneficial in attenuating glioblastoma progression by reducing brain Glu levels. Therefore, in this study we evaluated the ability of BGS treatment to inhibit brain metastatic melanoma progression in-vivo. RET melanoma cells were implanted in C56BL/6J mice to induce brain melanoma tumors followed by treatment with BGS or vehicle administered for fourteen days. Bioluminescent imaging was conducted to evaluate tumor growth, and plasma/CSF Glu levels were monitored throughout. Immunofluorescence staining of Ki67 and 53BP1 was used to analyze tumor cell proliferation and DNA double-strand breaks. In addition, we analyzed CD8, CD68, CD206, p-STAT1 and iNOS expression to evaluate alterations in tumor micro-environment and anti-tumor immune response due to treatment. Our results show that BGS treatment reduces CSF Glu concentration and consequently melanoma growth in-vivo by decreasing tumor cell proliferation and increasing pro-apoptotic signaling in C56BL/6J mice. Furthermore, BGS treatment supported CD8+ cell recruitment and CD68+ macrophage invasion. These findings suggest that BGS can be of potential therapeutic relevance in the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Yona Goldshmit
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel.,Australian Regenerative Medicine Institute, Monash Biotechnology, 15 Innovation Walk, Clayton, VIC, 3800, Australia
| | - Rita Perelroizen
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Alex Yakovchuk
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel
| | - Evgeni Banyas
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel
| | - Lior Mayo
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Sari David
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel
| | - Amit Benbenishty
- Department of Biological Regulation, Weizmann Institute of Science, 7610001, Rehovot, Israel
| | - Pablo Blinder
- Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel.,Neurobiology Department, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Moshe Shalom
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel
| | - Angela Ruban
- Steyer School of Health Professions, Sackler Faculty of Medicine, Tel-Aviv University, P.O. Box 39040, 6997801, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
18
|
Martínez-Miguel P, Albalate M, Durán-Laforet V, Peña-Martínez C, de Sequera P, Bouarich H, Peña-Esparragoza K, López-Ongil S, Lizasoain I, Sánchez-Prieto J, Torres M, Moro MÁ, Rodríguez-Puyol D. Effective glutamate clearance from the systemic circulation by hemodialysis: Potential relevance for cerebral ischemia management. Artif Organs 2021; 45:1183-1188. [PMID: 33560549 DOI: 10.1111/aor.13933] [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: 12/25/2020] [Revised: 01/30/2021] [Accepted: 02/03/2021] [Indexed: 11/27/2022]
Abstract
High glutamate levels after head trauma or cerebral ischemia have neurotoxic effects. The objective of the present study was to evaluate the efficacy of hemodialysis to remove glutamate from the blood and to assess the behavior of this small molecule. Ten patients with end-renal disease on hemodialysis were included in the study. Glutamate clearance was evaluated within the first hour of hemodialysis on a midweek dialysis day on five patients who underwent low flux hemodialysis, whereas the other five patients underwent highly efficient hemodialysis (high flux hemodialysis on one day and online hemodiafiltration on another day). Glutamate clearance with hemodialysis was very effective and did not show any differences between the techniques (low flux: 214 [55], high flux: 204 [37], online hemodiafiltration: 202 [16], median (interquartile range), P = .7). Glutamate clearance was almost equivalent to vascular access plasma flow and it was not affected by dialyzer permeability or ultrafiltration rate. After a hemodialysis session, a significant decrease in glutamate blood level was observed (prehemodialysis: 59.7 [36.1], posthemodialysis 37.0 [49.2], P = .005). Dialysis performed under fasting condition showed higher glutamate reduction rate (60%) than that under feeding condition (20%). Hemodialysis may be an effective method to reduce glutamate blood levels, and the molecule clearance does not differ between the different techniques used. Considering previous results in experimental models, hemodialysis without hemodynamic stress, could be considered for reducing glutamate neurotoxic effects in acute ischemic strokes of patients in chronic hemodialysis programs.
Collapse
Affiliation(s)
| | - Marta Albalate
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Violeta Durán-Laforet
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Carolina Peña-Martínez
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Patricia de Sequera
- Servicio de Nefrología, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Hanane Bouarich
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain
| | - Korina Peña-Esparragoza
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain
| | - Susana López-Ongil
- Fundación de Investigación Biomédica, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain
| | - Ignacio Lizasoain
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - José Sánchez-Prieto
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - Magdalena Torres
- Departamento de Bioquímica y Biología Molecular, Facultad de Veterinaria, Universidad Complutense, Madrid, Spain
| | - María Ángeles Moro
- Departamento de Farmacología y Toxicología, Facultad de Medicina, Universidad Complutense, Madrid, Spain
| | - Diego Rodríguez-Puyol
- Servicio de Nefrología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain.,Departmento de Medicina y Especialidades Médicas, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Spain
| |
Collapse
|
19
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
20
|
Goldshmit Y, Banyas E, Bens N, Yakovchuk A, Ruban A. Blood glutamate scavengers and exercises as an effective neuroprotective treatment in mice with spinal cord injury. J Neurosurg Spine 2020; 33:692-704. [PMID: 32619986 DOI: 10.3171/2020.4.spine20302] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/16/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Excitotoxicity due to neuronal damage and glutamate release is one of the first events that leads to the progression of neuronal degeneration and functional impairment. This study is based on a paradigm shift in the therapeutic approach for treating spinal cord injury (SCI). The authors tested a new treatment targeting removal of CNS glutamate into the blood circulation by injection of the blood glutamate scavengers (BGSs) recombinant enzyme glutamate-oxaloacetate transaminase (rGOT1) and its cosubstrate oxaloacetic acid (OxAc). Their primary objective was to investigate whether BGS treatment, followed by treadmill exercises in mice with SCI, could attenuate excitotoxicity, inflammation, scarring, and axonal degeneration and, at a later time point, improve functional recovery. METHODS A pharmacokinetic experiment was done in C57BL/6 naive mice to verify rGOT1/OxAc blood activity and to characterize the time curve of glutamate reduction in the blood up to 24 hours. The reduction of glutamate in CSF after BGS administration in mice with SCI was confirmed by high-performance liquid chromatography. Next, SCI (left hemisection) was induced in the mice, and the mice were randomly assigned to one of the following groups at 1 hour postinjury: control (underwent SCI and received PBS), treadmill exercises, rGOT1/OxAc treatment, or rGOT1/OxAc treatment followed by treadmill exercises. Treatment started 1 hour postinjury with an injection of rGOT1/OxAc and continued for 5 consecutive days. Starting 1 week after SCI, the exercises and the combined treatment groups recommenced the treadmill exercise regimen 5 days a week for 3 months. Locomotor function was assessed for 3 months using the horizontal grid walking test and CatWalk. Axonal anterograde and wallerian degenerations were evaluated using tetramethylrhodamine dextran. Tissue sections were immunofluorescently stained for Iba1, GFAP, GAP-43, synaptophysin, and NeuN. RESULTS BGS treatment decreased the CSF glutamate level up to 50%, reduced axonal wallerian degeneration, and increased axonal survival and GAP-43 expression in neuronal cells. Combined treatment reduced inflammation, scarring, and lesion size. Additionally, the combination of BGS treatment and exercises increased synapses around motor neurons and enhanced axonal regeneration through the lesion site. This resulted in motor function improvement 3 months post-SCI. CONCLUSIONS As shown by biochemical, immunohistochemical, and functional analysis, BGSs exhibit a substantial neuroprotective effect by reducing excitotoxicity and secondary damage after SCI. Furthermore, in combination with exercises, they reduced axonal degeneration and scarring and resulted in improved functional recovery.
Collapse
Affiliation(s)
- Yona Goldshmit
- 1Steyer School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- 2Australian Regenerative Medicine Institute, Monash Biotechnology, Clayton, Victoria, Australia; and
| | - Evgeni Banyas
- 1Steyer School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nicole Bens
- 1Steyer School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alex Yakovchuk
- 1Steyer School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Angela Ruban
- 1Steyer School of Health Professions, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- 3Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
21
|
Vidoni ED, Choi IY, Lee P, Reed G, Zhang N, Pleen J, Mahnken JD, Clutton J, Becker A, Sherry E, Bothwell R, Anderson H, Harris RA, Brooks W, Wilkins HM, Mosconi L, Burns JM, Swerdlow RH. Safety and target engagement profile of two oxaloacetate doses in Alzheimer's patients. Alzheimers Dement 2020; 17:7-17. [PMID: 32715609 DOI: 10.1002/alz.12156] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/19/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Brain bioenergetics are defective in Alzheimer's disease (AD). Preclinical studies find oxaloacetate (OAA) enhances bioenergetics, but human safety and target engagement data are lacking. METHODS We orally administered 500 or 1000 mg OAA, twice daily for 1 month, to AD participants (n = 15 each group) and monitored safety and tolerability. To assess brain metabolism engagement, we performed fluorodeoxyglucose positron emission tomography (FDG PET) and magnetic resonance spectroscopy before and after the intervention. We also assessed pharmacokinetics and cognitive performance. RESULTS Both doses were safe and tolerated. Compared to the lower dose, the higher dose benefited FDG PET glucose uptake across multiple brain regions (P < .05), and the higher dose increased parietal and frontoparietal glutathione (P < .05). We did not demonstrate consistent blood level changes and cognitive scores did not improve. CONCLUSIONS 1000 mg OAA, taken twice daily for 1 month, is safe in AD patients and engages brain energy metabolism.
Collapse
Affiliation(s)
- Eric D Vidoni
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - In-Young Choi
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Phil Lee
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Radiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Gregory Reed
- Department of Pharmacology and Toxicology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Na Zhang
- Department of Pharmacology and Toxicology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Joseph Pleen
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Jonathan D Mahnken
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Biostatistics, Kansas University Medical Center, Kansas City, Kansas, USA
| | | | - Annette Becker
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | - Erica Sherry
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | | | - Heidi Anderson
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
| | - Robert A Harris
- Biochemistry and Molecular Biology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - William Brooks
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Hoglund Biomedical Imaging Center, Kansas City, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Jeffrey M Burns
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Center, Fairway, Kansas, USA
- Department of Neurology, Kansas University Medical Center, Kansas City, Kansas, USA
- Department of Molecular and Integrative Physiology, Kansas University Medical Center, Kansas City, Kansas, USA
- Biochemistry and Molecular Biology, Kansas University Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
22
|
Brain to blood efflux as a mechanism underlying the neuroprotection mediated by rapid remote preconditioning in brain ischemia. Mol Biol Rep 2020; 47:5385-5395. [DOI: 10.1007/s11033-020-05626-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 06/26/2020] [Indexed: 12/21/2022]
|
23
|
Obrenovich M, Jaworski H, Tadimalla T, Mistry A, Sykes L, Perry G, Bonomo RA. The Role of the Microbiota-Gut-Brain Axis and Antibiotics in ALS and Neurodegenerative Diseases. Microorganisms 2020; 8:E784. [PMID: 32456229 PMCID: PMC7285349 DOI: 10.3390/microorganisms8050784] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
: The human gut hosts a wide and diverse ecosystem of microorganisms termed the microbiota, which line the walls of the digestive tract and colon where they co-metabolize digestible and indigestible food to contribute a plethora of biochemical compounds with diverse biological functions. The influence gut microbes have on neurological processes is largely yet unexplored. However, recent data regarding the so-called leaky gut, leaky brain syndrome suggests a potential link between the gut microbiota, inflammation and host co-metabolism that may affect neuropathology both locally and distally from sites where microorganisms are found. The focus of this manuscript is to draw connection between the microbiota-gut-brain (MGB) axis, antibiotics and the use of "BUGS AS DRUGS" for neurodegenerative diseases, their treatment, diagnoses and management and to compare the effect of current and past pharmaceuticals and antibiotics for alternative mechanisms of action for brain and neuronal disorders, such as Alzheimer disease (AD), Amyotrophic Lateral Sclerosis (ALS), mood disorders, schizophrenia, autism spectrum disorders and others. It is a paradigm shift to suggest these diseases can be largely affected by unknown aspects of the microbiota. Therefore, a future exists for applying microbial, chemobiotic and chemotherapeutic approaches to enhance translational and personalized medical outcomes. Microbial modifying applications, such as CRISPR technology and recombinant DNA technology, among others, echo a theme in shifting paradigms, which involve the gut microbiota (GM) and mycobiota and will lead to potential gut-driven treatments for refractory neurologic diseases.
Collapse
Affiliation(s)
- Mark Obrenovich
- Research Service, Louis Stokes Cleveland, Department of Veteran’s Affairs Medical Center, Cleveland, OH 44106, USA; (H.J.); (T.T.); (R.A.B.)
- Departments of Chemistry, Biochemistry, Pathology and Molecular Biology, Case Western Reserve University, Cleveland, OH 44106, USA
- The Gilgamesh Foundation for Medical Science and Research, Cleveland, OH 44116, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA
- Cleveland State University Departments of Chemistry and Engineering, Cleveland, OH 44115, USA;
| | - Hayden Jaworski
- Research Service, Louis Stokes Cleveland, Department of Veteran’s Affairs Medical Center, Cleveland, OH 44106, USA; (H.J.); (T.T.); (R.A.B.)
- Cleveland State University Departments of Chemistry and Engineering, Cleveland, OH 44115, USA;
| | - Tara Tadimalla
- Research Service, Louis Stokes Cleveland, Department of Veteran’s Affairs Medical Center, Cleveland, OH 44106, USA; (H.J.); (T.T.); (R.A.B.)
- Departments of Chemistry, Biochemistry, Pathology and Molecular Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Adil Mistry
- Cleveland State University Departments of Chemistry and Engineering, Cleveland, OH 44115, USA;
| | - Lorraine Sykes
- Department of Laboratory Medicine, Metro Health Medical Center, Cleveland, OH 44109, USA;
| | - George Perry
- Department of Biology University of Texas San Antonio, San Antonio, TX 78249, USA;
| | - Robert A. Bonomo
- Research Service, Louis Stokes Cleveland, Department of Veteran’s Affairs Medical Center, Cleveland, OH 44106, USA; (H.J.); (T.T.); (R.A.B.)
- Departments of Chemistry, Biochemistry, Pathology and Molecular Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
24
|
Conway ME. Alzheimer's disease: targeting the glutamatergic system. Biogerontology 2020; 21:257-274. [PMID: 32048098 PMCID: PMC7196085 DOI: 10.1007/s10522-020-09860-4] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disease that causes a progressive decline in memory, language and problem solving. For decades mechanism-based therapies have primarily focused on amyloid β (Aβ) processing and pathways that govern neurofibrillary tangle generation. With the potential exception to Aducanumab, a monotherapy to target Aβ, clinical trials in these areas have been challenging and have failed to demonstrate efficacy. Currently, the prescribed therapies for AD are those that target the cholinesterase and glutamatergic systems that can moderately reduce cognitive decline, dependent on the individual. In the brain, over 40% of neuronal synapses are glutamatergic, where the glutamate level is tightly regulated through metabolite exchange in neuronal, astrocytic and endothelial cells. In AD brain, Aβ can interrupt effective glutamate uptake by astrocytes, which evokes a cascade of events that leads to neuronal swelling, destruction of membrane integrity and ultimately cell death. Much work has focussed on the post-synaptic response with little insight into how glutamate is regulated more broadly in the brain and the influence of anaplerotic pathways that finely tune these mechanisms. The role of blood branched chain amino acids (BCAA) in regulating neurotransmitter profiles under disease conditions also warrant discussion. Here, we review the importance of the branched chain aminotransferase proteins in regulating brain glutamate and the potential consequence of dysregulated metabolism in the context of BCAA or glutamate accumulation. We explore how the reported benefits of BCAA supplementation or restriction in improving cognitive function in other neurological diseases may have potential application in AD. Given that memantine, the glutamate receptor agonist, shows clinical relevance it is now timely to research related pathways, an understanding of which could identify novel approaches to treatment of AD.
Collapse
Affiliation(s)
- Myra E Conway
- Faculty of Health and Applied Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK. .,Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
25
|
Gruenbaum BF, Kutz R, Zlotnik A, Boyko M. Blood glutamate scavenging as a novel glutamate-based therapeutic approach for post-stroke depression. Ther Adv Psychopharmacol 2020; 10:2045125320903951. [PMID: 32110376 PMCID: PMC7026819 DOI: 10.1177/2045125320903951] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/31/2019] [Indexed: 12/16/2022] Open
Abstract
Post-stroke depression (PSD) is a major complication of stroke that significantly impacts functional recovery and quality of life. While the exact mechanism of PSD is unknown, recent attention has focused on the association of the glutamatergic system in its etiology and treatment. Minimizing secondary brain damage and neuropsychiatric consequences associated with excess glutamate concentrations is a vital part of stroke management. The blood glutamate scavengers, oxaloacetate and pyruvate, degrade glutamate in the blood to its inactive metabolite, 2-ketoglutarate, by the coenzymes glutamate-oxaloacetate transaminase (GOT) and glutamate-pyruvate transaminase (GPT), respectively. This reduction in blood glutamate concentrations leads to a subsequent shift of glutamate down its concentration gradient from the blood to the brain, thereby decreasing brain glutamate levels. Although there are not yet any human trials that support blood glutamate scavengers for clinical use, there is increasing evidence from animal research of their efficacy as a promising new therapeutic approach for PSD. In this review, we present recent evidence in the literature of the potential therapeutic benefits of blood glutamate scavengers for reducing PSD and other related neuropsychiatric conditions. The evidence reviewed here should be useful in guiding future clinical trials.
Collapse
Affiliation(s)
- Benjamin F Gruenbaum
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruslan Kutz
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexander Zlotnik
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthew Boyko
- Division of Anesthesiology and Critical Care, Soroka University Medical Center and the Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84101, Israel
| |
Collapse
|
26
|
Dimopoulos C, Papadakis M, Perrea D, Nikiteas N, Kontzoglou K. The Effect of Cyclosporine and the Consequences in Hepatic and Renal Function Following Ischemic Stroke in a Rats' Model. J Stroke Cerebrovasc Dis 2019; 29:104562. [PMID: 31836361 DOI: 10.1016/j.jstrokecerebrovasdis.2019.104562] [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: 06/27/2019] [Revised: 11/01/2019] [Accepted: 11/21/2019] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Recent studies have indicated that the damaging effects of stroke are not only limited to the brain. We sought to examine the changes of liver and renal enzymes in the acute phase of ischemic stroke and to investigate possible explanations and therapeutic options, concerning in particular the functional alterations of peripheral organs after administration of an anti-inflammatory agent. MATERIAL/METHODS Twelve-week-old Wistar male rats were randomly divided into control and Cyclosporine groups (n = 10 each). Cyclosporine was given orally by gavage for 5 days prior to cerebral ischemia at a total volume of 15 mg/kg/day. All animals were subjected to 60 minutes focal ischemia by filament occlusion of the middle cerebral artery. Serum concentrations of Creatinine, Urea, SGOT, SGPT, and γGT were determined at the time before surgery and after 60 minutes brain ischemia. RESULTS Comparing data of 2 time-points, in both groups the serum liver enzyme levels increased progressively during the ischemic period. The liver enzymes and Urea were significantly lower in the Cyclosporine group than in the control group and the levels of Creatinine were slightly higher in the Cyclosporine group, in both time-points. CONCLUSIONS The detection of high liver enzyme serum levels in the acute phase of ischemic stroke implies the secondary effect of cerebral infraction on the peripheral organs and particularly on the liver function. Cyclosporine seems to exhibit a protective activity and to affect both liver and renal function after ischemic stroke.
Collapse
Affiliation(s)
- Christos Dimopoulos
- Laboratory for Experimental Surgery and Surgical Research "N.S Christeas", Medical School of Athens, Athens, Greece; Department of Vascular and Endovascular Surgery, Heinrich-Heine-University Medical Center Düsseldorf, Düsseldorf, Germany.
| | - Marios Papadakis
- Department of Plastic Surgery, Helios Clinic Wuppertal, University Hospital Witten-Herdecke, Wuppertal, Germany
| | - Despina Perrea
- Laboratory for Experimental Surgery and Surgical Research "N.S Christeas", Medical School of Athens, Athens, Greece
| | - Nikolaos Nikiteas
- Laboratory for Experimental Surgery and Surgical Research "N.S Christeas", Medical School of Athens, Athens, Greece; Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Kontzoglou
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School of Athens, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
27
|
AY O, OI O, FO Y, AM A, IO A, OJ O. Oral Monosodium Glutamate Differentially Affects Open-Field Behaviours, Behavioural Despair and Place Preference in Male and Female Mice. ACTA ACUST UNITED AC 2019. [DOI: 10.2174/2211556008666181213160527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background:
Monosodium glutamate (MSG) is a flavour enhancer which induces
behavioural changes in animals. However the influence of sex on the behavioural response
to MSG has not been investigated.
Objective:
The sex-differential effects of MSG on open-field behaviours, anxiety-related
behaviour, behavioural despair, place-preference, and plasma/brain glutamate levels in
adult mice were assessed.
Methods:
Mice were assigned to three groups (1-3), based on the models used to assess
behaviours. Animals in group 1 were for the elevated-plus maze and tail-suspension paradigms,
group 2 for the open-field and forced-swim paradigms, while mice in group 3 were
for observation in the conditioned place preference paradigm. Mice in all groups were further
assigned into five subgroups (10 males and 10 females), and administered vehicle (distilled
water at 10 ml/kg) or one of four doses of MSG (20, 40, 80 and 160 mg/kg) daily for
6 weeks, following which they were exposed to the behavioural paradigms. At the end of
the behavioural tests, the animals were sacrificed, and blood was taken for estimation of
glutamate levels. The brains were also homogenised for estimation of glutamate levels.
Results:
MSG was associated with a reduction in locomotion in males and females (except
at 160 mg/kg, male), an anxiolytic response in females, an anxiogenic response in males,
and decreased behavioural despair in both sexes (females more responsive). Postconditioning
MSG-associated place-preference was significantly higher in females. Plasma/
brain glutamate was not significantly different between sexes.
Conclusion:
Repeated MSG administration alters a range of behaviours in a sex-dependent
manner in mice.
Collapse
Affiliation(s)
- Onaolapo AY
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Olawore OI
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Yusuf FO
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adeyemo AM
- Behavioural Neuroscience/Neurobiology Unit, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| | - Adewole IO
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Onaolapo OJ
- Behavioural Neuroscience/Neuropharmacology Unit, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| |
Collapse
|
28
|
Neuroprotection mediated by remote preconditioning is associated with a decrease in systemic oxidative stress and changes in brain and blood glutamate concentration. Neurochem Int 2019; 129:104461. [DOI: 10.1016/j.neuint.2019.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/30/2019] [Accepted: 05/07/2019] [Indexed: 11/20/2022]
|
29
|
Frank D, Kuts R, Tsenter P, Gruenbaum BF, Grinshpun Y, Zvenigorodsky V, Shelef I, Natanel D, Brotfain E, Zlotnik A, Boyko M. The effect of pyruvate on the development and progression of post-stroke depression: A new therapeutic approach. Neuropharmacology 2019; 155:173-184. [PMID: 31153808 DOI: 10.1016/j.neuropharm.2019.05.035] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 12/27/2022]
Abstract
Post-stroke depression (PSD) is a common and serious complication following stroke. Both stroke and depression have independently been associated with pathologically elevated glutamate levels in the brain's extra-cerebral fluid (ECF). Here we evaluate an alternative therapeutic approach to PSD with pyruvate. Rats were randomly assigned into one of 3 groups: Middle Cerebral Artery Occlusion (MCAO) plus pyruvate treatment, MCAO plus placebo treatment, and sham operated rats. Post-MCAO depressive and anxiety-like behavior was assessed, along with neurological status, brain infarct zone, brain edema, blood brain barrier (BBB) breakdown, cerebrospinal fluid and blood glutamate levels. Anxiety-like behavior and levels of blood alanine and α-ketoglutarate were measured in naïve rats treated with pyruvate, as a control. Post-stroke neurological deficit with concurrent elevation in glutamate levels were demonstrated, with peak glutamate levels 24 h after MCAO. Treatment with pyruvate led to reduced glutamate levels 24 h after MCAO and improved neurologic recovery. Pyruvate treatment reduced lesion volume, brain edema and the extent of BBB permeability 24 h post-MCAO. Naïve rats treated with pyruvate showed increased levels of α-ketoglutarate. Rats demonstrated post-stroke depressive behavior that was improved by the administration of pyruvate. There was less anxiety-like behavior in post-stroke rats treated with placebo in comparison to the post-stroke rats treated with pyruvate or sham operated rats. Glutamate scavenging with pyruvate appears to be an effective as a method in providing neuroprotection following stroke and as a therapeutic option for the treatment of PSD by reducing the consequent elevations in CNS glutamate levels.
Collapse
Affiliation(s)
- Dmitry Frank
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ruslan Kuts
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Philip Tsenter
- Division of Internal Medicine, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Benjamin F Gruenbaum
- Department of Anesthesiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yulia Grinshpun
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vladislav Zvenigorodsky
- Department of Radiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ilan Shelef
- Department of Radiology, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Dmitry Natanel
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Evgeny Brotfain
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexander Zlotnik
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Matthew Boyko
- Department of Anesthesiology and Critical Care, Soroka University Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
30
|
Mohamed LA, Markandaiah SS, Bonanno S, Pasinelli P, Trotti D. Excess glutamate secreted from astrocytes drives upregulation of P-glycoprotein in endothelial cells in amyotrophic lateral sclerosis. Exp Neurol 2019; 316:27-38. [PMID: 30974102 DOI: 10.1016/j.expneurol.2019.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/09/2019] [Accepted: 04/06/2019] [Indexed: 12/13/2022]
Abstract
In amyotrophic lateral sclerosis (ALS), upregulation in expression and activity of the ABC transporter P-glycoprotein (P-gp) driven by disease advancement progressively reduces CNS penetration and efficacy of the ALS drug, riluzole. Post-mortem spinal cord tissues from ALS patients revealed elevated P-gp expression levels in endothelial cells of the blood-spinal cord barrier compared to levels measured in control, non-diseased individuals. We recently found that astrocytes expressing familial ALS-linked SOD1 mutations regulate expression levels of P-gp in endothelial cells, which also exhibit a concomitant, significant increase in reactive oxygen species production and NFκB nuclear translocation when exposed to mutant SOD1 astrocyte conditioned media. In this study, we found that glutamate, which is abnormally secreted by mutant SOD1 and sporadic ALS astrocytes, drives upregulation of P-gp expression and activity levels in endothelial cells via activation of N-Methyl-D-Aspartic acid (NMDA) receptors. Surprisingly, astrocyte-secreted glutamate regulation of endothelial P-gp levels is not a mechanism shared by all forms of ALS. C9orf72-ALS astrocytes had no effect on endothelial cell P-gp expression and did not display increased glutamate secretion. Utilizing an optimized in vitro human BBB model consisting of patient-derived induced pluripotent stem cells, we showed that co-culture of endothelial cells with patient-derived astrocytes increased P-gp expression levels and transport activity, which was significantly reduced when endothelial cells were incubated with the NMDAR antagonist, MK801. Overall, our findings unraveled a complex molecular interplay between astrocytes of different ALS genotypes and endothelial cells potentially occurring in disease that could differentially impact ALS prognosis and efficacy of pharmacotherapies.
Collapse
Affiliation(s)
- Loqman A Mohamed
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Shashirekha S Markandaiah
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Silvia Bonanno
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Piera Pasinelli
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA
| | - Davide Trotti
- Jefferson Weinberg ALS Center, Vickie and Jack Farber Institute for Neuroscience, Department of Neuroscience, Thomas Jefferson University, 900 Walnut Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
31
|
Pérez-Mato M, Iglesias-Rey R, Vieites-Prado A, Dopico-López A, Argibay B, Fernández-Susavila H, da Silva-Candal A, Pérez-Díaz A, Correa-Paz C, Günther A, Ávila-Gómez P, Isabel Loza M, Baumann A, Castillo J, Sobrino T, Campos F. Blood glutamate EAAT 2-cell grabbing therapy in cerebral ischemia. EBioMedicine 2018; 39:118-131. [PMID: 30555045 PMCID: PMC6354443 DOI: 10.1016/j.ebiom.2018.11.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/13/2018] [Accepted: 11/13/2018] [Indexed: 01/10/2023] Open
Abstract
Background Excitatory amino acid transporter 2 (EAAT2) plays a pivotal role in glutamate clearance in the adult brain, thereby preventing excitotoxic effects. Considering the high efficacy of EAAT2 for glutamate uptake, we hypothesized that the expression of this transporter in mesenchymal stem cells (MSCs) for systemic administration could yield a cell-based glutamate-grabbing therapy, combining the intrinsic properties of these cells with excitotoxic protection. Methods To address this hypothesis, EAAT2-encoding cDNA was introduced into MSCs and human embryonic kidney 293 cells (HEK cells) as the control cell line. EAAT2 expression and functionality were evaluated by in vitro assays. Blood glutamate-grabbing activity was tested in healthy and ischemic rat models treated with 3 × 106 and 9 × 106 cells/animal. Findings The expression of EAAT2 in both cell types conferred the expected glutamate-grabbing activity in in vitro and in vivo studies. The functional improvement observed in ischemic rats treated with EAAT2–HEK at low dose, confirmed that this effect was indeed mediated by the glutamate-grabbing activity associated with EAAT2 functionality. Unexpectedly, both cell doses of non-transfected MSCs induced higher protection than transfected EAAT2–MSCs by another mechanism independent of the glutamate-grabbing capacity. Interpretation Although the transfection procedure most likely interferes with some of the intrinsic protective mechanisms of mesenchymal cells, the results show that the induced expression of EAAT2 in cells represents a novel alternative to mitigate the excitotoxic effects of glutamate and paves the way to combine this strategy with current cell therapies for cerebral ischemia.
Collapse
Affiliation(s)
- María Pérez-Mato
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Alba Vieites-Prado
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Antonio Dopico-López
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Bárbara Argibay
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Héctor Fernández-Susavila
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Andrés da Silva-Candal
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Amparo Pérez-Díaz
- Drug Screening Platform/Biofarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Anne Günther
- Institute of Complex Systems-Cellular Biophysics (ICS-4), Forschungszentrum Jülich, Jülich, Germany
| | - Paulo Ávila-Gómez
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - M Isabel Loza
- Drug Screening Platform/Biofarma Research Group, Centro Singular de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Arnd Baumann
- Institute of Complex Systems-Cellular Biophysics (ICS-4), Forschungszentrum Jülich, Jülich, Germany
| | - José Castillo
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain.
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory (LINC), Department of Neurology, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital, Santiago de Compostela, Spain.
| |
Collapse
|
32
|
Fiandaca MS, Gross TJ, Johnson TM, Hu MT, Evetts S, Wade-Martins R, Merchant-Borna K, Bazarian J, Cheema AK, Mapstone M, Federoff HJ. Potential Metabolomic Linkage in Blood between Parkinson's Disease and Traumatic Brain Injury. Metabolites 2018; 8:metabo8030050. [PMID: 30205491 PMCID: PMC6161135 DOI: 10.3390/metabo8030050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/01/2018] [Accepted: 09/04/2018] [Indexed: 12/17/2022] Open
Abstract
The etiologic basis for sporadic forms of neurodegenerative diseases has been elusive but likely represents the product of genetic predisposition and various environmental factors. Specific gene-environment interactions have become more salient owing, in part, to the elucidation of epigenetic mechanisms and their impact on health and disease. The linkage between traumatic brain injury (TBI) and Parkinson's disease (PD) is one such association that currently lacks a mechanistic basis. Herein, we present preliminary blood-based metabolomic evidence in support of potential association between TBI and PD. Using untargeted and targeted high-performance liquid chromatography-mass spectrometry we identified metabolomic biomarker profiles in a cohort of symptomatic mild TBI (mTBI) subjects (n = 75) 3⁻12 months following injury (subacute) and TBI controls (n = 20), and a PD cohort with known PD (n = 20) or PD dementia (PDD) (n = 20) and PD controls (n = 20). Surprisingly, blood glutamic acid levels in both the subacute mTBI (increased) and PD/PDD (decreased) groups were notably altered from control levels. The observed changes in blood glutamic acid levels in mTBI and PD/PDD are discussed in relation to other metabolite profiling studies. Should our preliminary results be replicated in comparable metabolomic investigations of TBI and PD cohorts, they may contribute to an "excitotoxic" linkage between TBI and PD/PDD.
Collapse
Affiliation(s)
- Massimo S Fiandaca
- Translational Laboratory and Biorepository, Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
- Department of Neurological Surgery, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
- Department of Anatomy & Neurobiology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
| | - Thomas J Gross
- Translational Laboratory and Biorepository, Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
- Department of Anatomy & Neurobiology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
| | - Thomas M Johnson
- Intrepid Spirit Concussion Recovery Center, Naval Medical Center Camp Lejeune, Jacksonville, NC 28540, USA.
| | - Michele T Hu
- Nuffield Department of Clinical Neurosciences, University of Oxford, 01865 Oxford, UK.
- Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals Trust, Oxford 01865, UK.
| | - Samuel Evetts
- Nuffield Department of Clinical Neurosciences, University of Oxford, 01865 Oxford, UK.
| | - Richard Wade-Martins
- Department of Physiology, Anatomy and Genetics, Oxford Parkinson's Disease Centre, University of Oxford, Oxford 01865, UK.
| | - Kian Merchant-Borna
- Department of Emergency Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14604, USA.
| | - Jeffrey Bazarian
- Department of Emergency Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14604, USA.
| | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20001, USA.
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC 20001, USA.
| | - Mark Mapstone
- Translational Laboratory and Biorepository, Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
| | - Howard J Federoff
- Translational Laboratory and Biorepository, Department of Neurology, University of California Irvine School of Medicine, Irvine, CA 92697-3910, USA.
| |
Collapse
|
33
|
da Silva-Candal A, Pérez-Díaz A, Santamaría M, Correa-Paz C, Rodríguez-Yáñez M, Ardá A, Pérez-Mato M, Iglesias-Rey R, Brea J, Azuaje J, Sotelo E, Sobrino T, Loza MI, Castillo J, Campos F. Clinical validation of blood/brain glutamate grabbing in acute ischemic stroke. Ann Neurol 2018; 84:260-273. [DOI: 10.1002/ana.25286] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 01/28/2023]
Affiliation(s)
- Andrés da Silva-Candal
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Amparo Pérez-Díaz
- Drug Screening Platform/Biofarma Research Group, Molecular Medicine and Chronic Diseases Research Center; University of Santiago de Compostela; Santiago de Compostela Spain
| | - María Santamaría
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Clara Correa-Paz
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Manuel Rodríguez-Yáñez
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Ana Ardá
- Molecular Recognition and Host-Pathogen Interactions Unit, CIC bioGUNE; Derio Spain
| | - María Pérez-Mato
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Ramón Iglesias-Rey
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - José Brea
- Drug Screening Platform/Biofarma Research Group, Molecular Medicine and Chronic Diseases Research Center; University of Santiago de Compostela; Santiago de Compostela Spain
| | - Jhonny Azuaje
- Center for Research in Biological Chemistry and Molecular Materials (CIQUS); University of Santiago de Compostela; Santiago de Compostela Spain
| | - Eddy Sotelo
- Center for Research in Biological Chemistry and Molecular Materials (CIQUS); University of Santiago de Compostela; Santiago de Compostela Spain
| | - Tomás Sobrino
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - M. Isabel Loza
- Drug Screening Platform/Biofarma Research Group, Molecular Medicine and Chronic Diseases Research Center; University of Santiago de Compostela; Santiago de Compostela Spain
| | - José Castillo
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| | - Francisco Campos
- Clinical Neurosciences Research Laboratory, Department of Neurology; Clinical University Hospital, Health Research Institute of Santiago de Compostela; Santiago de Compostela Spain
| |
Collapse
|
34
|
Wickens MM, Bangasser DA, Briand LA. Sex Differences in Psychiatric Disease: A Focus on the Glutamate System. Front Mol Neurosci 2018; 11:197. [PMID: 29922129 PMCID: PMC5996114 DOI: 10.3389/fnmol.2018.00197] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022] Open
Abstract
Alterations in glutamate, the primary excitatory neurotransmitter in the brain, are implicated in several psychiatric diseases. Many of these psychiatric diseases display epidemiological sex differences, with either males or females exhibiting different symptoms or disease prevalence. However, little work has considered the interaction of disrupted glutamatergic transmission and sex on disease states. This review describes the clinical and preclinical evidence for these sex differences with a focus on two conditions that are more prevalent in women: Alzheimer's disease and major depressive disorder, and three conditions that are more prevalent in men: schizophrenia, autism spectrum disorder, and attention deficit hyperactivity disorder. These studies reveal sex differences at multiple levels in the glutamate system including metabolic markers, receptor levels, genetic interactions, and therapeutic responses to glutamatergic drugs. Our survey of the current literature revealed a considerable need for more evaluations of sex differences in future studies examining the role of the glutamate system in psychiatric disease. Gaining a more thorough understanding of how sex differences in the glutamate system contribute to psychiatric disease could provide novel avenues for the development of sex-specific pharmacotherapies.
Collapse
Affiliation(s)
- Megan M Wickens
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Lisa A Briand
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
35
|
Zhang D, Mably AJ, Walsh DM, Rowan MJ. Peripheral Interventions Enhancing Brain Glutamate Homeostasis Relieve Amyloid β- and TNFα- Mediated Synaptic Plasticity Disruption in the Rat Hippocampus. Cereb Cortex 2018; 27:3724-3735. [PMID: 27390019 DOI: 10.1093/cercor/bhw193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of glutamate homeostasis in the interstitial fluid of the brain is strongly implicated in causing synaptic dysfunction in many neurological and psychiatric illnesses. In the case of Alzheimer's disease (AD), amyloid β (Aβ)-mediated disruption of synaptic plasticity and memory can be alleviated by interventions that directly remove glutamate or block certain glutamate receptors. An alternative strategy is to facilitate the removal of excess glutamate from the nervous system by activating peripheral glutamate clearance systems. One such blood-based system, glutamate oxaloacetate transaminase (GOT), is activated by oxaloacetate, which acts as a co-substrate. We report here that synthetic and AD brain-derived Aβ-mediated inhibition of synaptic long-term potentiation in the hippocampus is alleviated by oxaloacetate. Moreover the effect of oxaloacetate was GOT-dependent. The disruptive effects of a general inhibitor of excitatory amino acid transport or TNFα, a pro-inflammatory mediator of Aβ action, were also reversed by oxaloacetate. Furthermore, another intervention that increases peripheral glutamate clearance, peritoneal dialysis, mimicked the beneficial effect of oxaloacetate. These findings lend support to the promotion of the peripheral clearance of glutamate as a means to alleviate synaptic dysfunction that is caused by impaired glutamate homeostasis in the brain.
Collapse
Affiliation(s)
- Dainan Zhang
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Alexandra J Mably
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
36
|
Distribution of the branched-chain α-ketoacid dehydrogenase complex E1α subunit and glutamate dehydrogenase in the human brain and their role in neuro-metabolism. Neurochem Int 2018; 112:49-58. [DOI: 10.1016/j.neuint.2017.10.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 10/18/2017] [Accepted: 10/18/2017] [Indexed: 11/17/2022]
|
37
|
Bai W, Zhou YG. Homeostasis of the Intraparenchymal-Blood Glutamate Concentration Gradient: Maintenance, Imbalance, and Regulation. Front Mol Neurosci 2017; 10:400. [PMID: 29259540 PMCID: PMC5723322 DOI: 10.3389/fnmol.2017.00400] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 11/20/2017] [Indexed: 12/25/2022] Open
Abstract
It is widely accepted that glutamate is the most important excitatory neurotransmitter in the central nervous system (CNS). However, there is also a large amount of glutamate in the blood. Generally, the concentration gradient of glutamate between intraparenchymal and blood environments is stable. However, this gradient is dramatically disrupted under a variety of pathological conditions, resulting in an amplifying cascade that causes a series of pathological reactions in the CNS and peripheral organs. This eventually seriously worsens a patient’s prognosis. These two “isolated” systems are rarely considered as a whole even though they mutually influence each other. In this review, we summarize what is currently known regarding the maintenance, imbalance and regulatory mechanisms that control the intraparenchymal-blood glutamate concentration gradient, discuss the interrelationships between these systems and further explore their significance in clinical practice.
Collapse
Affiliation(s)
- Wei Bai
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuan-Guo Zhou
- Molecular Biology Center, State Key Laboratory of Trauma, Burn, and Combined Injury, Research Institute of Surgery and Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
38
|
Blood Glutamate Reducing Effect of Hemofiltration in Critically Ill Patients. Neurotox Res 2017; 33:300-308. [PMID: 28836163 DOI: 10.1007/s12640-017-9791-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 07/12/2017] [Accepted: 07/28/2017] [Indexed: 10/19/2022]
Abstract
Glutamate toxicity plays a well-established role in secondary brain damage following acute and chronic brain insults. Previous studies have demonstrated the efficacy of hemodialysis and peritoneal dialysis in reducing blood glutamate levels. However, these methods are not viable options for hemodynamically unstable patients. Given more favorable hemodynamics, longer treatment, and less needed anticoagulation, we investigated whether hemofiltration could be effective in lowering blood glutamate levels. Blood samples were taken from 10 critically ill patients immediately before initiation of hemofiltration and after 1, 2, 4, 6, and 12 h, for a total of 6 blood samples. Samples were sent for determination of glutamate, glutamate oxaloacetate transaminase (GOT), glutamate pyruvate transaminase (GPT), hemoglobin, hematocrit, urea, creatinine, glucose, sodium, potassium, platelet, and white blood cell (WBC) levels. There was a statistically significant reduction in blood glutamate levels at all time points compared to baseline levels. There was no difference in levels of GOT or GPT. Hemofiltration can be a promising method of reducing blood glutamate levels, especially in critically ill patients where hemodialysis and peritoneal dialysis may be contraindicated.
Collapse
|
39
|
Popova I, Malkov A, Ivanov AI, Samokhina E, Buldakova S, Gubkina O, Osypov A, Muhammadiev RS, Zilberter T, Molchanov M, Paskevich S, Zilberter M, Zilberter Y. Metabolic correction by pyruvate halts acquired epilepsy in multiple rodent models. Neurobiol Dis 2017; 106:244-254. [PMID: 28709994 DOI: 10.1016/j.nbd.2017.07.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 07/03/2017] [Accepted: 07/10/2017] [Indexed: 01/22/2023] Open
Abstract
Metabolic intervention strategy of epilepsy treatment has been gaining broader attention due to accumulated evidence that hypometabolism, manifested in humans as reduced brain glucose consumption, is a principal factor in acquired epilepsy. Therefore, targeting deficient energy metabolism may be an effective approach for treating epilepsy. To confront this pathology we utilized pyruvate, which besides being an anaplerotic mitochondrial fuel possesses a unique set of neuroprotective properties as it: (i) is a potent reactive oxygen species scavenger; (ii) abates overactivation of Poly [ADP-ribose] polymerase 1 (PARP-1); (iii) facilitates glutamate efflux from the brain; (iv) augments brain glycogen stores; (v) is anti-inflammatory; (vi) prevents neuronal hyperexcitability; and (vii) normalizes the cytosolic redox state. In vivo, chronic oral pyruvate administration completely abolished established epileptic phenotypes in three accepted and fundamentally different rodent acquired epilepsy models. Our study reports metabolic correction by pyruvate as a potentially highly effective treatment of acquired epilepsies.
Collapse
Affiliation(s)
- I Popova
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - A Malkov
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France; Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - A I Ivanov
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France
| | - E Samokhina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - S Buldakova
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France
| | - O Gubkina
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France
| | - A Osypov
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| | - R S Muhammadiev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | | | - M Molchanov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - S Paskevich
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - M Zilberter
- Neuronal Oscillations Lab, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Y Zilberter
- Aix Marseille Université, Inserm, INS UMR_S 1106, 13005 Marseille, France.
| |
Collapse
|
40
|
Interplay between mitochondrial metabolism and oxidative stress in ischemic stroke: An epigenetic connection. Mol Cell Neurosci 2017; 82:176-194. [DOI: 10.1016/j.mcn.2017.05.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/26/2017] [Accepted: 05/24/2017] [Indexed: 12/18/2022] Open
|
41
|
Gao F, Chen C, Lu J, Zheng J, Ma XC, Yuan XY, Huo K, Han JF. De Ritis ratio (AST/ALT) as an independent predictor of poor outcome in patients with acute ischemic stroke. Neuropsychiatr Dis Treat 2017; 13:1551-1557. [PMID: 28670124 PMCID: PMC5478276 DOI: 10.2147/ndt.s139316] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The aspartate transaminase/alanine transaminase ratio (De Ritis ratio, AAR) was reported to be associated with patients' prognosis in certain diseases recently. The objective of the current study was to determine the association between the AAR at admission and poor outcome at 3 months in acute ischemic stroke (AIS) patients. PATIENTS AND METHODS This retrospective cohort study included patients who experienced their first-ever AIS between June 2015 and March 2016. The primary outcome measure was a poor outcome at 3 months (modified Rankin Scale score >2). Multivariate logistic regression models were used to assess the relationship between AAR quartiles and clinical outcomes among the AIS patients. Receiver operating characteristic curve analysis was applied to identify the optimal cutoff for AAR in predicting the prognosis of AIS. RESULTS In terms of the relationship between poor outcome and AAR, the adjusted odds ratio comparing the highest and lowest AAR quartiles was 2.15 (95% confidence interval =1.14-4.05). An AAR of 1.53 was identified as the optimal cutoff. In a prespecified subgroup analysis according to the time from symptom onset to treatment (>24 vs ≤24 hours), there was no significant difference in the effect of AAR >1.53 between the two groups. CONCLUSION An increased AAR at admission is significantly associated with a poor outcome at 3 months in AIS patients.
Collapse
Affiliation(s)
| | | | | | | | - Xian-Cang Ma
- Clinical Research Center
- Department of Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | | | | | | |
Collapse
|
42
|
Levite M. Glutamate, T cells and multiple sclerosis. J Neural Transm (Vienna) 2017; 124:775-798. [PMID: 28236206 DOI: 10.1007/s00702-016-1661-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/25/2016] [Indexed: 12/18/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, where it induces multiple beneficial and essential effects. Yet, excess glutamate, evident in a kaleidoscope of acute and chronic pathologies, is absolutely catastrophic, since it induces excitotoxicity and massive loss of brain function. Both the beneficial and the detrimental effects of glutamate are mediated by a large family of glutamate receptors (GluRs): the ionotropic glutamate receptors (iGluRs) and the metabotropic glutamate receptors (mGluRs), expressed by most/all cells of the nervous system, and also by many non-neural cells in various peripheral organs and tissues. T cells express on their cell surface several types of functional GluRs, and so do few other immune cells. Furthermore, glutamate by itself activates resting normal human T cells, and induces/elevates key T cell functions, among them: T cell adhesion, chemotactic migration, cytokine secretion, gene expression and more. Glutamate has also potent effects on antigen/mitogen/cytokine-activated T cells. Furthermore, T cells can even produce and release glutamate, and affect other cells and themselves via their own glutamate. Multiple sclerosis (MS) and its animal model Experimental Autoimmune Encephalomyelitis (EAE) are mediated by autoimmune T cells. In MS and EAE, there are excess glutamate levels, and multiple abnormalities in glutamate degrading enzymes, glutamate transporters, glutamate receptors and glutamate signaling. Some GluR antagonists block EAE. Enhancer of mGluR4 protects from EAE via regulatory T cells (Tregs), while mGluR4 deficiency exacerbates EAE. The protective effect of mGluR4 on EAE calls for testing GluR4 enhancers in MS patients. Oral MS therapeutics, namely Fingolimod, dimethyl fumarate and their respective metabolites Fingolimod-phosphate and monomethyl fumarate, can protect neurons against acute glutamatergic excitotoxic damage. Furthermore, Fingolimod reduce glutamate-mediated intracortical excitability in relapsing-remitting MS. Glatiramer acetate -COPAXONE®, an immunomodulator drug for MS, reverses TNF-α-induced alterations of striatal glutamate-mediated excitatory postsynaptic currents in EAE-afflicted mice. With regard to T cells of MS patients: (1) The cell surface expression of a specific GluR: the AMPA GluR3 is elevated in T cells of MS patients during relapse and with active disease, (2) Glutamate and AMPA (a selective agonist for glutamate/AMPA iGluRs) augment chemotactic migration of T cells of MS patients, (3) Glutamate augments proliferation of T cells of MS patients in response to myelin-derived proteins: MBP and MOG, (4) T cells of MS patients respond abnormally to glutamate, (5) Significantly higher proliferation values in response to glutamate were found in MS patients assessed during relapse, and in those with gadolinium (Gd)+ enhancing lesions on MRI. Furthermore, glutamate released from autoreactive T cells induces excitotoxic cell death of neurons. Taken together, the evidences accumulated thus far indicate that abnormal glutamate levels and signaling in the nervous system, direct activation of T cells by glutamate, and glutamate release by T cells, can all contribute to MS. This may be true also to other neurological diseases. It is postulated herein that the detrimental activation of autoimmune T cells by glutamate in MS could lead to: (1) Cytotoxicity in the CNS: T cell-mediated killing of neurons and glia cells, which would subsequently increase the extracellular glutamate levels, and by doing so increase the excitotoxicity mediated by excess glutamate, (2) Release of proinflammatory cytokines, e.g., TNFα and IFNγ that increase neuroinflammation. Finally, if excess glutamate, abnormal neuronal signaling, glutamate-induced activation of T cells, and glutamate release by T cells are indeed all playing a key detrimental role in MS, then optional therapeutic tolls include GluR antagonists, although these may have various side effects. In addition, an especially attractive therapeutic strategy is the novel and entirely different therapeutic approach to minimize excess glutamate and excitotoxicity, titled: 'brain to blood glutamate scavenging', designed to lower excess glutamate levels in the CNS by 'pumping it out' from the brain to the blood. The glutamate scavanging is achieved by lowering glutamate levels in the blood by intravenous injection of the blood enzyme glutamate oxaloacetate transaminase (GOT). The glutamate-scavenging technology, which is still experimental, validated so far for other brain pathologies, but not tested on MS or EAE yet, may be beneficial for MS too, since it could decrease both the deleterious effects of excess glutamate on neural cells, and the activation of autoimmune T cells by glutamate in the brain. The topic of glutamate scavenging, and also its potential benefit for MS, are discussed towards the end of the review, and call for research in this direction.
Collapse
Affiliation(s)
- Mia Levite
- Faculty of Medicine, School of Pharmacy, The Hebrew University, Jerusalem, Israel. .,Institute of Gene Therapy, Hadassah Medical Center, 91120, Ein Karem, Jerusalem, Israel.
| |
Collapse
|
43
|
Nashed MG, Ungard RG, Young K, Zacal NJ, Seidlitz EP, Fazzari J, Frey BN, Singh G. Behavioural Effects of Using Sulfasalazine to Inhibit Glutamate Released by Cancer Cells: A Novel target for Cancer-Induced Depression. Sci Rep 2017; 7:41382. [PMID: 28120908 PMCID: PMC5264609 DOI: 10.1038/srep41382] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
Despite the lack of robust evidence of effectiveness, current treatment options for cancer-induced depression (CID) are limited to those developed for non-cancer related depression. Here, anhedonia-like and coping behaviours were assessed in female BALB/c mice inoculated with 4T1 mammary carcinoma cells. The behavioural effects of orally administered sulfasalazine (SSZ), a system xc− inhibitor, were compared with fluoxetine (FLX). FLX and SSZ prevented the development of anhedonia-like behaviour on the sucrose preference test (SPT) and passive coping behaviour on the forced swim test (FST). The SSZ metabolites 5-aminosalicylic acid (5-ASA) and sulfapyridine (SP) exerted an effect on the SPT but not on the FST. Although 5-ASA is a known anti-inflammatory agent, neither treatment with SSZ nor 5-ASA/SP prevented tumour-induced increases in serum levels of interleukin-1β (IL-1β) and IL-6, which are indicated in depressive disorders. Thus, the observed antidepressant-like effect of SSZ may primarily be attributable to the intact form of the drug, which inhibits system xc−. This study represents the first attempt at targeting cancer cells as a therapeutic strategy for CID, rather than targeting downstream effects of tumour burden on the central nervous system. In doing so, we have also begun to characterize the molecular pathways of CID.
Collapse
Affiliation(s)
- Mina G Nashed
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Robert G Ungard
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Kimberly Young
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Natalie J Zacal
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Eric P Seidlitz
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Jennifer Fazzari
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, L8N 3K7, Canada.,Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare Hamilton, ON, L8P 3K7, Canada
| | - Gurmit Singh
- Department of Pathology &Molecular Medicine, McMaster University, Hamilton, ON, Canada.,Michael G. DeGroote Institute for Pain Research and Care, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
44
|
Brotfain E, Gruenbaum SE, Boyko M, Kutz R, Zlotnik A, Klein M. Neuroprotection by Estrogen and Progesterone in Traumatic Brain Injury and Spinal Cord Injury. Curr Neuropharmacol 2017; 14:641-53. [PMID: 26955967 PMCID: PMC4981744 DOI: 10.2174/1570159x14666160309123554] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 12/31/2015] [Accepted: 02/25/2016] [Indexed: 12/25/2022] Open
Abstract
In recent years there has been a growing body of clinical and laboratory evidence demonstrating the neuroprotective effects of estrogen and progesterone after traumatic brain injury (TBI) and spinal cord injury (SCI). In humans, women have been shown to have a lower incidence of morbidity and mortality after TBI compared with age-matched men. Similarly, numerous laboratory studies have demonstrated that estrogen and progesterone administration is associated with a mortality reduction, improvement in neurological outcomes, and a reduction in neuronal apoptosis after TBI and SCI. Here, we review the evidence that supports hormone-related neuroprotection and discuss possible underlying mechanisms. Estrogen and progesterone-mediated neuroprotection are thought to be related to their effects on hormone receptors, signaling systems, direct antioxidant effects, effects on astrocytes and microglia, modulation of the inflammatory response, effects on cerebral blood flow and metabolism, and effects on mediating glutamate excitotoxicity. Future laboratory research is needed to better determine the mechanisms underlying the hormones' neuroprotective effects, which will allow for more clinical studies. Furthermore, large randomized clinical control trials are needed to better assess their role in human neurodegenerative conditions.
Collapse
Affiliation(s)
- Evgeni Brotfain
- Department of Anesthesiology and Critical Care, Soroka Medical Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Goldsmith M, Ashani Y, Margalit R, Nyska A, Mirelman D, Tawfik DS. A new post-intoxication treatment of paraoxon and parathion poisonings using an evolved PON1 variant and recombinant GOT1. Chem Biol Interact 2016; 259:242-251. [DOI: 10.1016/j.cbi.2016.05.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/04/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
|
47
|
Castillo J, Loza MI, Mirelman D, Brea J, Blanco M, Sobrino T, Campos F. A novel mechanism of neuroprotection: Blood glutamate grabber. J Cereb Blood Flow Metab 2016; 36:292-301. [PMID: 26661174 PMCID: PMC4759671 DOI: 10.1177/0271678x15606721] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/19/2015] [Indexed: 12/22/2022]
Abstract
Glutamate excitotoxicity is a primary contributor of ischemic neuronal death and other cellular components of the neurovascular unit. Several strategies have been developed against glutamate excitotoxicity, however none of them have not shown positive results in the clinical practice so far. Nowadays, the concept of blood/brain glutamate grabbing or scavenging is well recognized as a novel and attractive protective strategy to reduce the excitotoxic effect of excess extracellular glutamate that accumulates in the brain following an ischemic stroke. The main advantage of this novel therapeutic strategy is that it occurs in the blood circulation and therefore does not affect the normal brain neurophysiology, as it has been described for other drug treatments used against glutamate excitotoxicity. In this work we report all experimental data from the beginning of our studies, focused on stroke pathology, and we describe new findings about the potential application of this therapy. Future clinical trials will allow to know the real efficacy of this novel therapeutic strategy in stroke patients.
Collapse
Affiliation(s)
- José Castillo
- Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - María Isabel Loza
- Department of Pharmacology, Universidade de Santiago de Compostela, Discovery group BioFarma, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - David Mirelman
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - José Brea
- Department of Pharmacology, Universidade de Santiago de Compostela, Discovery group BioFarma, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain BioFarma Research Group, Centro de Investigación en Medicina Molecular y Enfermedades Crónicas (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Blanco
- Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Tomás Sobrino
- Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| | - Francisco Campos
- Department of Neurology, Clinical University Hospital, Universidade de Santiago de Compostela, Health Research Institute of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
48
|
Blood glutamate grabbing does not reduce the hematoma in an intracerebral hemorrhage model but it is a safe excitotoxic treatment modality. J Cereb Blood Flow Metab 2015; 35:1206-12. [PMID: 25735920 PMCID: PMC4640266 DOI: 10.1038/jcbfm.2015.28] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/14/2015] [Accepted: 01/22/2015] [Indexed: 01/01/2023]
Abstract
Recent studies have shown that blood glutamate grabbing is an effective strategy to reduce the excitotoxic effect of extracellular glutamate released during ischemic brain injury. The purpose of the study was to investigate the effect of two of the most efficient blood glutamate grabbers (oxaloacetate and recombinant glutamate oxaloacetate transaminase 1: rGOT1) in a rat model of intracerebral hemorrhage (ICH). Intracerebral hemorrhage was produced by injecting collagenase into the basal ganglia. Three treatment groups were developed: a control group treated with saline, a group treated with oxaloacetate, and a final group treated with human rGOT1. Treatments were given 1 hour after hemorrhage. Hematoma volume (analyzed by magnetic resonance imaging (MRI)), neurologic deficit, and blood glutamate and GOT levels were quantified over a period of 14 days after surgery. The results observed showed that the treatments used induced a significant reduction of blood glutamate levels; however, they did not reduce the hematoma, nor did they improve the neurologic deficit. In the present experimental study, we have shown that this novel therapeutic strategy is not effective in case of ICH pathology. More importantly, these findings suggest that blood glutamate grabbers are a safe treatment modality that can be given in cases of suspected ischemic stroke without previous neuroimaging.
Collapse
|
49
|
Meng XE, Li N, Guo DZ, Pan SY, Li H, Yang C. High plasma glutamate levels are associated with poor functional outcome in acute ischemic stroke. Cell Mol Neurobiol 2015; 35:159-65. [PMID: 25190005 PMCID: PMC11486313 DOI: 10.1007/s10571-014-0107-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2014] [Accepted: 08/26/2014] [Indexed: 12/18/2022]
Abstract
The aim of the present study was to investigate the relationship between acute ischemic stroke and glutamate levels and to determine the prognosis value of plasma glutamate levels to predict the functional outcome. Two hundred and forty-two patients with acute ischemic stroke and 100 sex- and age-matched controls were included in the study. Plasma glutamate levels were determined by HPLC at admission in both groups. Stroke severity was assessed using the National Institutes of Health Stroke Scale (NIHSS). The modified Rankin Scale (mRS) scores at 3 months was determined to outcomes, and unfavorable outcomes were defined as mRS at 3-6. The prognostic value analyzed by logistic regression analysis, after adjusting for the possible confounders. In the 94 patients with an unfavorable functional outcome, plasma glutamate levels were higher compared with those in patients with a favorable outcome [221(IQR, 152-321) μM; 176(IQR, 112-226) μM, respectively; P < 0.0001). In multivariate logistic regression analysis, glutamate was an independent predictor of functional outcome, with an adjusted OR of 6.99 (95 % confidence interval [CI] 2.21-21.23). Receiver operating characteristics to predict functional outcome demonstrated areas under the curve of glutamate of 0.821 (95 % CI 0.733-0.878; P < 0.0001) and combined model (glutamate and NIHSS) improved the NIHSS score alone. Plasma glutamate levels can be seen as an independent short-term prognostic marker of functional outcome in Chinese patients with acute ischemic stroke even after correcting for possible confounding factors.
Collapse
Affiliation(s)
- Xiang-en Meng
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| | - Na Li
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| | - Da-Zhi Guo
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| | - Shu-Yi Pan
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| | - Hang Li
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| | - Chen Yang
- Department of Hyperbaric Oxygen, Navy General Hospital, No. 6, Fucheng road, Beijing, 100048 People’s Republic of China
| |
Collapse
|
50
|
Zilberter Y, Gubkina O, Ivanov AI. A unique array of neuroprotective effects of pyruvate in neuropathology. Front Neurosci 2015; 9:17. [PMID: 25741230 PMCID: PMC4330789 DOI: 10.3389/fnins.2015.00017] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/12/2015] [Indexed: 12/03/2022] Open
Affiliation(s)
- Yuri Zilberter
- Institut de Neurosciences des Systèmes, Aix Marseille Université, Inserm UMR_S 1106 Marseille, France
| | - Olena Gubkina
- Institut de Neurosciences des Systèmes, Aix Marseille Université, Inserm UMR_S 1106 Marseille, France
| | - Anton I Ivanov
- Institut de Neurosciences des Systèmes, Aix Marseille Université, Inserm UMR_S 1106 Marseille, France
| |
Collapse
|