1
|
Mielecki D, Bratek-Gerej E, Salińska E. Metabotropic glutamate receptors-guardians and gatekeepers in neonatal hypoxic-ischemic brain injury. Pharmacol Rep 2024:10.1007/s43440-024-00653-x. [PMID: 39289333 DOI: 10.1007/s43440-024-00653-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Injury to the developing central nervous system resulting from perinatal hypoxia-ischemia (HI) is still a clinical challenge. The only approach currently available in clinical practice for severe cases of HI is therapeutic hypothermia, initiated shortly after birth and supported by medications to regulate blood pressure, control epileptic seizures, and dialysis to support kidney function. However, these treatments are not effective enough to significantly improve infant survival or prevent brain damage. The need to create a new effective therapy has focused attention on metabotropic glutamate receptors (mGluR), which control signaling pathways involved in HI-induced neurodegeneration. The complexity of mGluR actions, considering their localization and developmental changes, and the functions of each subtype in HI-evoked brain damage, combined with difficulties in the availability of safe and effective modulators, raises the question whether modulation of mGluRs with subtype-selective ligands can become a new treatment in neonatal HI. Addressing this question, this review presents the available information concerning the role of each of the eight receptor subtypes of the three mGluR groups (group I, II, and III). Data obtained from experiments performed on in vitro and in vivo neonatal HI models show the neuroprotective potential of group I mGluR antagonists, as well as group II and III agonists. The information collected in this work indicates that the neuroprotective effects of manipulating mGluR in experimental HI models, despite the need to create more safe and selective ligands for particular receptors, provide a chance to create new therapies for the sensitive brains of infants at risk.
Collapse
Affiliation(s)
- Damian Mielecki
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Ewelina Bratek-Gerej
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland
| | - Elżbieta Salińska
- Department of Neurochemistry, Mossakowski Medical Research Institute PAS, Pawińskiego 5, 02-106, Warsaw, Poland.
| |
Collapse
|
2
|
Jiang XL, Zhang ZB, Feng CX, Lin CJ, Yang H, Tan LL, Ding X, Xu LX, Li G, Pan T, Qin ZH, Sun B, Feng X, Li M. PHLDA1 contributes to hypoxic ischemic brain injury in neonatal rats via inhibiting FUNDC1-mediated mitophagy. Acta Pharmacol Sin 2024; 45:1809-1820. [PMID: 38750074 PMCID: PMC11336168 DOI: 10.1038/s41401-024-01292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/14/2024] [Indexed: 08/22/2024] Open
Abstract
Hypoxia-ischemia (HI) is one of the main causes of neonatal brain injury. Mitophagy has been implicated in the degradation of damaged mitochondria and cell survival following neonatal brain HI injury. Pleckstrin homology-like domain family A member 1 (PHLDA1) plays vital roles in the progression of various disorders including the regulation of oxidative stress, the immune responses and apoptosis. In the present study we investigated the role of PHLDA1 in HI-induced neuronal injury and further explored the mechanisms underlying PHLDA1-regulated mitophagy in vivo and in vitro. HI model was established in newborn rats by ligation of the left common carotid artery plus exposure to an oxygen-deficient chamber with 8% O2 and 92% N2. In vitro studies were conducted in primary hippocampal neurons subjected to oxygen and glucose deprivation/-reoxygenation (OGD/R). We showed that the expression of PHLDA1 was significantly upregulated in the hippocampus of HI newborn rats and in OGD/R-treated primary neurons. Knockdown of PHLDA1 in neonatal rats via lentiviral vector not only significantly ameliorated HI-induced hippocampal neuronal injury but also markedly improved long-term cognitive function outcomes, whereas overexpression of PHLDA1 in neonatal rats via lentiviral vector aggravated these outcomes. PHLDA1 knockdown in primary neurons significantly reversed the reduction of cell viability and increase in intracellular reactive oxygen species (ROS) levels, and attenuated OGD-induced mitochondrial dysfunction, whereas overexpression of PHLDA1 decreased these parameters. In OGD/R-treated primary hippocampal neurons, we revealed that PHLDA1 knockdown enhanced mitophagy by activating FUNDC1, which was abolished by FUNDC1 knockdown or pretreatment with mitophagy inhibitor Mdivi-1 (25 μM). Notably, pretreatment with Mdivi-1 or the knockdown of FUNDC1 not only increased brain infarct volume, but also abolished the neuroprotective effect of PHLDA1 knockdown in HI newborn rats. Together, these results demonstrate that PHLDA1 contributes to neonatal HI-induced brain injury via inhibition of FUNDC1-mediated neuronal mitophagy.
Collapse
Affiliation(s)
- Xiao-Lu Jiang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zu-Bin Zhang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsycho Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Chen-Jie Lin
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Hui Yang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lan-Lan Tan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Gen Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Tao Pan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zheng-Hong Qin
- Institute of Health Technology, Global Institute of Software Technology, Qingshan Road, Suzhou Science & Technology Tower, Hi-Tech Area, Suzhou, 215163, China
| | - Bin Sun
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xing Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
3
|
Han D, Li Z, Luo L, Jiang H. Targeting Hypoxia and HIF1α in Triple-Negative Breast Cancer: New Insights from Gene Expression Profiling and Implications for Therapy. BIOLOGY 2024; 13:577. [PMID: 39194515 DOI: 10.3390/biology13080577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024]
Abstract
Breast cancer is a complex and multifaceted disease with diverse risk factors, types, and treatment options. Triple-negative breast cancer (TNBC), which lacks the expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2), is the most aggressive subtype. Hypoxia is a common feature of tumors and is associated with poor prognosis. Hypoxia can promote tumor growth, invasion, and metastasis by stimulating the production of growth factors, inducing angiogenesis, and suppressing antitumor immune responses. In this study, we used mRNA-seq technology to systematically investigate the gene expression profile of MDA-MB-231 cells under hypoxia. We found that the hypoxia-inducible factor (HIF) signaling pathway is the primary pathway involved in the cellular response to hypoxia. The genes in which expression levels were upregulated in response to hypoxia were regulated mainly by HIF1α. In addition, hypoxia upregulated various genes, including Nim1k, Rimkla, Cpne6, Tpbgl, Kiaa11755, Pla2g4d, and Ism2, suggesting that it regulates cellular processes beyond angiogenesis, metabolism, and known processes. We also found that HIF1α was hyperactivated in MDA-MB-231 cells under normoxia. A HIF1α inhibitor effectively inhibited the invasion, migration, proliferation, and metabolism of MDA-MB-231 cells. Our findings suggest that hypoxia and the HIF signaling pathway play more complex and multifaceted roles in TNBC than previously thought. These findings have important implications for the development of new therapeutic strategies for TNBC.
Collapse
Affiliation(s)
- Delong Han
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
- Institute for Inheritance-Based Innovation of Chinese Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Zeyu Li
- Institute for Inheritance-Based Innovation of Chinese Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Lingjie Luo
- Institute for Inheritance-Based Innovation of Chinese Medicine, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
- Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Hezhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
4
|
Huo L, Fu J, Wang S, Wang H, Liu X. Emerging ferroptosis inhibitors as a novel therapeutic strategy for the treatment of neonatal hypoxic-ischemic encephalopathy. Eur J Med Chem 2024; 271:116453. [PMID: 38701713 DOI: 10.1016/j.ejmech.2024.116453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/30/2023] [Accepted: 04/24/2024] [Indexed: 05/05/2024]
Abstract
Neonatal hypoxia-ischemia encephalopathy (NHIE), an oxygen deprivation-mediated brain injury due to birth asphyxia or reduced cerebral blood perfusion, often leads to lifelong sequelae, including seizures, cerebral palsy, and mental retardation. NHIE poses a significant health challenge, as one of the leading causes of neonatal morbidity and mortality globally. Despite this, available therapies are limited. Numerous studies have recently demonstrated that ferroptosis, an iron-dependent non-apoptotic regulated form of cell death characterized by lipid peroxidation (LPO) and iron dyshomeostasis, plays a role in the genesis of NHIE. Moreover, recently discovered compounds have been shown to exert potential therapeutic effects on NHIE by inhibiting ferroptosis. This comprehensive review summarizes the fundamental mechanisms of ferroptosis contributing to NHIE. We focus on various emerging therapeutic compounds exhibiting characteristics of ferroptosis inhibition and delineate their pharmacological benefits for the treatment of NHIE. This review suggests that pharmacological inhibition of ferroptosis may be a potential therapeutic strategy for NHIE.
Collapse
Affiliation(s)
- Liang Huo
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| | - Jianhua Fu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Shimeng Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China
| | - Xueyan Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, 11004, China.
| |
Collapse
|
5
|
Ji T, Pang Y, Cheng M, Wang R, Chen X, Zhang C, Liu M, Zhang J, Zhong C. Deletion of glutamate carboxypeptidase II (GCPII), but not GCPIII, provided long-term benefits in mice with traumatic brain injury. CNS Neurosci Ther 2023; 29:3786-3801. [PMID: 37349952 PMCID: PMC10651966 DOI: 10.1111/cns.14299] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/28/2023] [Indexed: 06/24/2023] Open
Abstract
MAIN PROBLEM N-acetylaspartylglutamate (NAAG) has neuroprotective effects in traumatic brain injury (TBI) by activating metabotropic glutamate receptor 3 (mGluR3) and reducing glutamate release. Glutamate carboxypeptidase II (GCPII) is the primary enzyme responsible for the hydrolysis of NAAG. It remains unclear whether glutamate carboxypeptidase III (GCPIII), a homolog of GCPII, can partially compensate for GCPII's function. METHODS GCPII-/- , GCPIII-/- , and GCPII/III-/- mice were generated using CRISPR/Cas9 technology. Mice brain injury model was established through moderate controlled cortical impact (CCI). The relationship between GCPII and GCPIII was explored by analyzing injury response signals in the hippocampus and cortex of mice with different genotypes at the acute (1 day) and subacute (7 day) phase after TBI. RESULTS In this study, we found that deletion of GCPII reduced glutamate production, excitotoxicity, and neuronal damage and improved cognitive function, but GCPIII deletion had no significant neuroprotective effect. Additionally, there was no significant difference in the neuroprotective effect between the combination of GCPII and GCPIII deletion and GCPII deletion alone. CONCLUSION These results suggest that GCPII inhibition may be a therapeutic option for TBI, and that GCPIII may not act as a complementary enzyme to GCPII in this context.
Collapse
Affiliation(s)
- Tongjie Ji
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Ying Pang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Meng Cheng
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Rui Wang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Xu Chen
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Chunyu Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Min Liu
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| | - Jing Zhang
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Institute for Advanced StudyTongji UniversityShanghaiChina
| | - Chunlong Zhong
- Department of NeurosurgeryShanghai East Hospital, School of Medicine, Tongji UniversityShanghaiChina
| |
Collapse
|
6
|
Caramelo B, Monteiro-Alfredo T, Martins J, Sereno J, Castelhano J, Manadas B, Castelo-Branco M, Matafome P. Functional imaging and neurochemistry identify in vivo neuroprotection mechanisms counteracting excitotoxicity and neurovascular changes in the hippocampus and visual cortex of obese and type 2 diabetic animal models. J Neurochem 2023; 165:892-906. [PMID: 37026518 DOI: 10.1111/jnc.15825] [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: 01/25/2023] [Revised: 03/30/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023]
Abstract
Functional MRI (fMRI) with 1 H-MRS was combined on the hippocampus and visual cortex of animal models of obesity (high-fat diet, HFD) and type 2 diabetes (T2D) to identify the involved mechanisms and temporal evolution of neurometabolic changes in these disorders that could serve as potentially reliable clinical biomarkers. HFD rats presented elevated levels of N-acetylaspartylglutamate (NAAG) (p = 0.0365 vs. standard diet, SD) and glutathione (GSH) (p = 0.0494 vs. SD) in the hippocampus. NAAG and GSH levels in this structure proved to be correlated (r = 0.4652, p = 0.0336). This mechanism was not observed in diabetic rats. Combining MRS and fMRI-evaluated blood-oxygen-level-dependent (BOLD) response, elevated taurine (p = 0.0326 vs. HFD) and GABA type A receptor (GABAA R) (p = 0.0211 vs. SD and p = 0.0153 vs. HFD) were observed in the visual cortex of only diabetic rats, counteracting the elevated BOLD response and suggesting an adaptative mechanism against hyperexcitability observed in the primary visual cortex (V1) (p = 0.0226 vs. SD). BOLD amplitude was correlated with the glutamate levels (r = 0.4491; p = 0.0316). Therefore, here we found evidence for several biological dichotomies regarding excitotoxicity and neuroprotection in different brain regions, identifying putative markers of their different susceptibility and response to the metabolic and vascular insults of obesity and diabetes.
Collapse
Affiliation(s)
- Beatriz Caramelo
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
| | - Tamaeh Monteiro-Alfredo
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
| | - João Martins
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - José Sereno
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Coimbra Chemistry Centre (CQC), Chemistry Department, University of Coimbra, Coimbra, Portugal
| | - João Castelhano
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
- Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Centre for Neurosciences and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Institute for Nuclear Sciences Applied to Health (ICNAS), University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Institute of Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center, Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| |
Collapse
|
7
|
Xiong F, Jiang K, Wu Y, Lou C, Ding C, Zhang W, Zhang X, Li C, Zheng H, Gao H. Intermittent fasting alleviates type 1 diabetes-induced cognitive dysfunction by improving the frontal cortical metabolic disorder. Biochim Biophys Acta Mol Basis Dis 2023:166725. [PMID: 37127173 DOI: 10.1016/j.bbadis.2023.166725] [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: 01/20/2023] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Intermittent fasting (IF) is an ecological strategy to control various metabolic disorder symptoms, but its protective effect on type 1 diabetes (T1D)-induced cognitive dysfunction and the underlying mechanisms remain poorly defined. Herein, we examined the efficacy of IF in altering the behaviors and brain metabolome in T1D mice and investigated the potential molecular mechanisms. We demonstrated that IF remarkably improved frontal cortical-dependent memory in T1D mice and reduced the loss of neuronal cells. Metabolomics and targeted mass spectrometry assay showed that IF reprogrammed the frontal cortical metabolome composition, including activated the aspartate and glutamate pathway and reversed glycerophospholipid and sphingolipid depositions in T1D mice. Mechanistically, IF attenuated the levels of oxidative stress proteins, such as NOX2, NOX4, 8-OHdG, 4-HNE, and inhibited the levels of pro-apoptotic factors Bax and cleaved Caspase-3, finally improved the memory ability of T1D mice. In vitro studies confirmed the protective effect of the supplemented N-acetylaspartate, a pivotal metabolite involved in IF-regulated T1D-induced cognitive dysfunction, in high glucose-stimulated SH-SY5Y cells by eliminating toxic lipids accumulation, oxidative stress and apoptosis. To conclude, the frontal cortical metabolites mediated the protective effects of IF against T1D-induced cognitive dysfunction by attenuating oxidative stress and apoptotic signaling. Thus, IF can be a potential therapeutic strategy for T1D-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Fen Xiong
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Kaiyuan Jiang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yali Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Cong Lou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chengjie Ding
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wenli Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xi Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Chen Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hong Zheng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Hongchang Gao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Metabonomics & Medical NMR, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Key Laboratory of Efficacy Evaluation of Traditional Chinese Medicine and Encephalopathy Research of Zhejiang Province, Wenzhou 325035, China.
| |
Collapse
|
8
|
Sah N, Zhang Z, Chime A, Fowler A, Mendez-Trendler A, Sharma A, Kannan RM, Slusher B, Kannan S. Dendrimer-Conjugated Glutamate Carboxypeptidase II Inhibitor Restores Microglial Changes in a Rabbit Model of Cerebral Palsy. Dev Neurosci 2023; 45:268-275. [PMID: 36990069 PMCID: PMC10614263 DOI: 10.1159/000530389] [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: 10/21/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
We have previously shown that maternal endotoxin exposure leads to a phenotype of cerebral palsy and pro-inflammatory microglia in the brain in neonatal rabbits. "Activated" microglia overexpress the enzyme glutamate carboxypeptidase II (GCPII) that hydrolyzes N-acetylaspartylglutamate to N-acetylaspartate and glutamate, and we have shown previously that inhibiting microglial GCPII is neuroprotective. Glutamate-induced injury and associated immune signaling can alter microglial responses including microglial process movements for surveillance and phagocytosis. We hypothesize that inhibition of GCPII activity could alter microglial phenotype and normalize microglial process movement/dynamics. Newborn rabbit kits exposed to endotoxin in utero, when treated with dendrimer-conjugated 2-(phosphonomethyl)-pentanedioic acid (D-2PMPA), a potent and selective inhibitor of microglial GCPII, showed profound changes in microglial phenotype within 48 h of treatment. Live imaging of hippocampal microglia in ex vivo brain slice preparations revealed larger cell body and phagocytic cup sizes with less stable microglia processes in CP kits compared to healthy controls. D-2PMPA treatment led to significant reversal of microglial process stability to healthy control levels. Our results emphasize the importance of microglial process dynamics in determining the state of microglial function in the developing brain and demonstrate how GCPII inhibition specifically in microglia can effectively change the microglial process motility to healthy control levels, potentially impacting migration, phagocytosis, and inflammatory functions.
Collapse
Affiliation(s)
- Nirnath Sah
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhi Zhang
- Department of Natural Sciences, University of Michigan-Dearborn, Dearborn, MI, USA
| | - Alicia Chime
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amanda Fowler
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Antonio Mendez-Trendler
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Chemistry, Washington State University, Pullman, WA, USA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Slusher
- Johns Hopkins Drug Discovery Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sujatha Kannan
- Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
9
|
Yıldızhan K, Nazıroğlu M. NMDA Receptor Activation Stimulates Hypoxia-Induced TRPM2 Channel Activation, Mitochondrial Oxidative Stress, and Apoptosis in Neuronal Cell Line: Modular Role of Memantine. Brain Res 2023; 1803:148232. [PMID: 36610553 DOI: 10.1016/j.brainres.2023.148232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/19/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023]
Abstract
TRPM2 channel is activated by the increase of hypoxia (HYP)-mediated excessive mitochondrial (mROS) and cytosolic (cROS) free reactive oxygen species generation and intracellular free Ca2+ ([Ca2+]i) influx. The stimulations of the N-methyl-d-aspartate(NMDA) receptor and TRPM2 channel induce mROS and apoptosis in the neurons, although their inhibitions via the treatments of memantine (MEM) and MK-801 decrease mROS and apoptosis. However, the molecular mechanisms underlying MEM treatment and NMDA inhibition' neuroprotection via TRPM2 inhibition in the HYP remain elusive. We investigated the modulator role of MEM and NMDA via the modulation of TRPM2 on oxidative neurodegeneration and apoptosis in SH-SY5Y neuronal cells. Six groups were induced in the SH-SY5Y and HEK293 cells as follows: Control, MEM, NMDA blocker (MK-801), HYP (CoCl2), HYP + MEM, and HYP + MK-801. The HYP caused to the increases of TRPM2 and PARP-1 expressions, and TRPM2 agonist (H2O2 and ADP-ribose)-induced TRPM2 current density and [Ca2+]i concentration via the upregulation of mitochondrial membrane potential, cROS, and mROS generations. The alterations were not observed in the absence of TRPM2 in the HEK293 cells. The increase of cROS, mROS, lipid peroxidation, cell death (propidium iodide/Hoechst) rate, apoptosis, caspase -3, caspase -8, and caspase -9 were restored via upregulation of glutathione and glutathione peroxidase by the treatments of TRPM2 antagonists (ACA or 2-APB), MEM, and MK-801. In conclusion, the inhibition of NMDA receptor via MEM treatment modulated HYP-mediated mROS, apoptosis, and TRPM2-induced excessive [Ca2+]i and may provide an avenue for protecting HYP-mediated neurodegenerative diseases associated with the increase of mROS, [Ca2+]i, and apoptosis.
Collapse
Affiliation(s)
- Kenan Yıldızhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Mustafa Nazıroğlu
- Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey; Drug Discovery Unit, BSN Health, Analyses, Innov., Consult., Org., Agricul., Trade Ltd, Isparta, Turkey; Department of Biophysics, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.
| |
Collapse
|
10
|
Group II Metabotropic Glutamate Receptors Reduce Apoptosis and Regulate BDNF and GDNF Levels in Hypoxic-Ischemic Injury in Neonatal Rats. Int J Mol Sci 2022; 23:ijms23137000. [PMID: 35806000 PMCID: PMC9266366 DOI: 10.3390/ijms23137000] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Birth asphyxia causes brain injury in neonates, but a fully successful treatment has yet to be developed. This study aimed to investigate the effect of group II mGlu receptors activation after experimental birth asphyxia (hypoxia-ischemia) on the expression of factors involved in apoptosis and neuroprotective neurotrophins. Hypoxia-ischemia (HI) on 7-day-old rats was used as an experimental model. The effects of intraperitoneal application of mGluR2 agonist LY379268 (5 mg/kg) and the specific mGluR3 agonist NAAG (5 mg/kg) (1 h or 6 h after HI) on apoptotic processes and initiation of the neuroprotective mechanism were investigated. LY379268 and NAAG applied shortly after HI prevented brain damage and significantly decreased pro-apoptotic Bax and HtrA2/Omi expression, increasing expression of anti-apoptotic Bcl-2. NAAG or LY379268 applied at both times also decreased HIF-1α formation. HI caused a significant decrease in BDNF concentration, which was restored after LY379268 or NAAG administration. HI-induced increase in GDNF concentration was decreased after administration of LY379268 or NAAG. Our results show that activation of mGluR2/3 receptors shortly after HI prevents brain damage by the inhibition of excessive glutamate release and apoptotic damage decrease. mGluR2 and mGluR3 agonists produced comparable results, indicating that both receptors may be a potential target for early treatment in neonatal HI.
Collapse
|
11
|
Resitoglu B, Yalcın C, Komur M, Polat A, Erdogan S, Beydagi H. Relationship of ozone application and time in rats with hypoxic-ischemic brain injury. Hippokratia 2021; 25:56-62. [PMID: 35937511 PMCID: PMC9347343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND Despite the important advances in pregnancy and newborn follow-up, hypoxic-ischemic encephalopathy is still one of the prominent causes of newborn mortality and disability worldwide, and there is no sufficiently effective treatment for it yet. This study aimed to investigate whether the ozone injection, administered in a single-dose as a preconditioning agent before the hypoxia and in single and repeated doses on different days following the hypoxia, would affect the spatial memory performance of the rats in the Morris water maze test or on their apoptotic cell numbers. METHODS The study consisted of 102 seven-day-old male Wistar baby rats randomly divided into five groups. Rats in all groups were induced with hypoxic-ischemic brain injury (HIBI) except for the Sham group, and 1.2 mg/kg ozone was administered intraperitoneally. For the apoptosis evaluation, eight rats from each of the first four groups were decapitated by cervical dislocation. Terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) assay was used for immunohistochemical quantification of apoptosis in the excised brains. Blood malondialdehyde (MDA) and superoxide dismutase (SOD) levels were measured in the blood samples collected through cardiac puncture. Fourteen-week-old rats underwent the Morris water maze test to test their long-term spatial memory. RESULTS On apoptotic quantification in the right hemisphere using the TUNEL assay, the numbers of apoptotic neurons in the ozone preconditioning group (Group 3) and the group given ozone on the day of hypoxia (Group 4) were found to be significantly higher than the Sham group (Group 1), but significantly lower than the non-treatment group (Group 2) (p <0.001; p <0.001, respectively). Group 3 rats had the highest mean MDA level and SOD activity. Considering the platform finding times in the first four days of the tests, Group 4 had the shortest times after Group 1; and on Day 4, Group 4 found the platforms significantly sooner than Groups 2, 3, and 5 (p <0.001). Comparison of Groups 1 and 4 revealed significantly shorter times for Group 1 for each day except for Day 2. CONCLUSIONS Other studies have shown that controlled application of ozone would result in oxidative preconditioning and reduce the damage induced by reactive oxygen species through enabling adaptation to oxidative stress. Our study obtained remarkable and encouraging findings for ozone administration in HIBI by examining Group 4's performance in the first four days and the difference in its platform finding times between Day 1 and Day 4. HIPPOKRATIA 2021, 25 (2):56-62.
Collapse
Affiliation(s)
- B Resitoglu
- Department of Anesthesia, Vocational School of Health Services, School of Medicine, Mersin University, Mersin, Turkey
| | - C Yalcın
- Department of Neonatal Intensive Care Unit, School of Medicine, Mersin University, Mersin, Turkey
| | - M Komur
- Department of Pediatric Neurology, School of Medicine, Mersin University, Mersin, Turkey
| | - A Polat
- Department of Pathology, School of Medicine, Mersin University, Mersin, Turkey
| | - S Erdogan
- Department of Biostatistics, School of Medicine, Mersin University, Mersin, Turkey
| | - H Beydagi
- Department of Physiology, School of Medicine, Mersin University, Mersin, Turkey
| |
Collapse
|
12
|
Bratek - Gerej E, Bronisz A, Ziembowicz A, Salinska E. Pretreatment with mGluR2 or mGluR3 Agonists Reduces Apoptosis Induced by Hypoxia-Ischemia in Neonatal Rat Brains. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8848015. [PMID: 33763176 PMCID: PMC7963909 DOI: 10.1155/2021/8848015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/04/2021] [Accepted: 02/23/2021] [Indexed: 11/17/2022]
Abstract
Hypoxia-ischemia (HI) in an immature brain results in energy depletion and excessive glutamate release resulting in excitotoxicity and oxidative stress. An increase in reactive oxygen species (ROS) production induces apoptotic processes resulting in neuronal death. Activation of group II mGluR was shown to prevent neuronal damage after HI. The application of agonists of mGluR3 (N-acetylaspartylglutamate; NAAG) or mGluR2 (LY379268) inhibits the release of glutamate and reduces neurodegeneration in a neonatal rat model of HI, although the exact mechanism is not fully recognized. In the present study, the effects of NAAG (5 mg/kg) and LY379268 (5 mg/kg) application (24 h or 1 h before experimental birth asphyxia) on apoptotic processes as the potential mechanism of neuroprotection in 7-day-old rats were investigated. Intraperitoneal application of NAAG or LY379268 at either time point before HI significantly reduced the number of TUNEL-positive cells in the CA1 region of the ischemic brain hemisphere. Both agonists reduced expression of the proapoptotic Bax protein and increased expression of Bcl-2. Decreases in HI-induced caspase-9 and caspase-3 activity were also observed. Application of NAAG or LY379268 24 h or 1 h before HI reduced HIF-1α formation likely by reducing ROS levels. It was shown that LY379268 concentration remains at a level that is required for activation of mGluR2 for up to 24 h; however, NAAG is quickly metabolized by glutamate carboxypeptidase II (GCPII) into glutamate and N-acetyl-aspartate. The observed effect of LY379268 application 24 h or 1 h before HI is connected with direct activation of mGluR2 and inhibition of glutamate release. Based on the data presented in this study and on our previous findings, we conclude that the neuroprotective effect of NAAG applied 1 h before HI is most likely the result of a combination of mGluR3 and NMDA receptor activation, whereas the beneficial effects of NAAG pretreatment 24 h before HI can be explained by the activation of NMDA receptors and induction of the antioxidative/antiapoptotic defense system triggered by mild excitotoxicity in neurons. This response to NAAG pretreatment is consistent with the commonly accepted mechanism of preconditioning.
Collapse
Affiliation(s)
- Ewelina Bratek - Gerej
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Bronisz
- Tumor Microenvironment Laboratory, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Apolonia Ziembowicz
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| | - Elzbieta Salinska
- Department of Neurochemistry, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
13
|
Arteaga Cabeza O, Zhang Z, Smith Khoury E, Sheldon RA, Sharma A, Zhang F, Slusher BS, Kannan RM, Kannan S, Ferriero DM. Neuroprotective effects of a dendrimer-based glutamate carboxypeptidase inhibitor on superoxide dismutase transgenic mice after neonatal hypoxic-ischemic brain injury. Neurobiol Dis 2020; 148:105201. [PMID: 33271328 PMCID: PMC8351403 DOI: 10.1016/j.nbd.2020.105201] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 01/05/2023] Open
Abstract
The result of a deprivation of oxygen and glucose to the brain, hypoxic-ischemic encephalopathy (HIE), remains the most common cause of death and disability in human neonates globally and is mediated by glutamate toxicity and inflammation. We have previously shown that the enzyme glutamate carboxypeptidase (GCPII) is overexpressed in activated microglia in the presence of inflammation in fetal/newborn rabbit brain. We assessed the therapeutic utility of a GCPII enzyme inhibitor called 2-(3-Mercaptopropyl) pentanedioic acid (2MPPA) attached to a dendrimer (D-2MPPA), in order to target activated microglia in an experimental neonatal hypoxia-ischemia (HI) model using superoxide dismutase transgenic (SOD) mice that are often more injured after hypoxia-ischemia than wildtype animals. SOD overexpressing and wild type (WT) mice underwent permanent ligation of the left common carotid artery followed by 50 min of asphyxiation (10% O2) to induce HI injury on postnatal day 9 (P9). Cy5-labeled dendrimers were administered to the mice at 6 h, 24 h or 72 h after HI and brains were evaluated by immunofluorescence analysis 24 h after the injection to visualize microglial localization and uptake over time. Expression of GCPII enzyme was analyzed in microglia 24 h after the HI injury. The expression of pro- and anti-inflammatory cytokines were analyzed 24 h and 72 h post-HI. Brain damage was analyzed histologically 7 days post-HI in the three randomly assigned groups: control (C); hypoxic-ischemic (HI); and HI mice who received a single dose of D-2MPPA 6 h post-HI (HI+D-2MPPA). First, we found that GCPII was overexpressed in activated microglia 24 h after HI in the SOD overexpressing mice. Also, there was an increase in microglial activation 24 h after HI in the ipsilateral hippocampus which was most visible in the SOD+HI group. Dendrimers were mostly taken up by microglia by 24 h post-HI; uptake was more prominent in the SOD+HI mice than in the WT+HI. The inflammatory profile showed significant increase in expression of KC/GRO following injury in SOD mice compared to WT at 24 and 72 h. A greater and significant decrease in KC/GRO was seen in the SOD mice following treatment with D-2MPPA. Seven days after HI, D-2MPPA treatment decreased brain injury in the SOD+HI group, but not in WT+HI. This reduced damage was mainly seen in hippocampus and cortex. Our data indicate that the best time point to administer D-2MPPA is 6 h post-HI in order to suppress the expression of GCPII by 24 h after the damage since dendrimer localization in microglia is seen as early as 6 h with the peak of GCPII upregulation in activated microglia seen at 24 h post-HI. Ultimately, treatment with D-2MPPA at 6 h post-HI leads to a decrease in inflammatory profiles by 24 h and reduction in brain injury in the SOD overexpressing mice.
Collapse
Affiliation(s)
- O Arteaga Cabeza
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA
| | - Z Zhang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - E Smith Khoury
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R A Sheldon
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - A Sharma
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - F Zhang
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - B S Slusher
- Department of Neurology, Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - R M Kannan
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - S Kannan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - D M Ferriero
- Departments of Pediatrics, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Neurology, University of California San Francisco, San Francisco, CA 94158, USA; Departments of Newborn Brain Research Institute, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|