1
|
Ozdemi̇r C, Isik B, Koca G, Inan MA. Effects of mid‑gestational sevoflurane and magnesium sulfate on maternal oxidative stress, inflammation and fetal brain histopathology. Exp Ther Med 2024; 28:286. [PMID: 38827470 PMCID: PMC11140313 DOI: 10.3892/etm.2024.12574] [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: 07/04/2023] [Accepted: 03/08/2024] [Indexed: 06/04/2024] Open
Abstract
Models of inflammation, oxidative stress, hyperoxia and hypoxia have demonstrated that magnesium sulfate (MgSO4), a commonly used drug in obstetrics, has neuroprotective potential. In the present study, the effects of MgSO4 treatment on inflammation, oxidative stress and fetal brain histopathology were evaluated in an experimental rat model following sevoflurane (Sv) exposure during the mid-gestational period. Rats were randomly divided into groups: C (control; no injections or anesthesia), Sv (exposure to 2.5% Sv for 2 h), MgSO4 (administered 270 mg/kg MgSO4 intraperitoneally) and Sv + MgSO4 (Sv administered 30 min after MgSO4 injection). Inflammatory and oxidative stress markers were measured in the serum and neurotoxicity was investigated histopathologically in fetal brain tissue. Short-term mid-gestational exposure to a 1.1 minimum alveolar concentration of Sv did not significantly increase the levels of any of the measured biochemical markers, except for TNF-α. Histopathological evaluations demonstrated no findings suggestive of pathological apoptosis, neuroinflammation or oxidative stress-induced cell damage. MgSO4 injection prior to anesthesia caused no significant differences in biochemical or histopathological marker levels compared to the C and Sv groups. The present study indicated that short-term exposure to Sv could potentially be considered a harmless external stimulus to the fetal brain.
Collapse
Affiliation(s)
- Cagri Ozdemi̇r
- Department of Anesthesiology and Reanimation, Mamak State Hospital, 06270 Ankara, Turkey
| | - Berrin Isik
- Department of Anesthesiology and Reanimation, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| | - Gulce Koca
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| | - Mehmet Arda Inan
- Department of Medical Pathology, Faculty of Medicine, Gazi University, 06560 Ankara, Turkey
| |
Collapse
|
2
|
Mesmar F, Muhsen M, Farooq I, Maxey G, Tourigny JP, Tennessen J, Bondesson M. Exposure to the pesticide tefluthrin causes developmental neurotoxicity in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.28.596249. [PMID: 38854095 PMCID: PMC11160659 DOI: 10.1101/2024.05.28.596249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
BACKGROUND The insecticide tefluthrin is widely used in agriculture, resulting in widespread pollution. Tefluthrin is a type I pyrethroid characterized by its high persistence in the environment. Understanding the mechanisms of toxicity of tefluthrin will improve its risk assessment. OBJECTIVES We aimed to decipher the molecular modes of action of tefluthrin. METHODS Phenotypic developmental toxicity was assessed by exposing zebrafish embryos and larvae to increasing concentrations of tefluthrin. Tg(mnx:mGFP) line was used to assess neurotoxicity. Multi-omics approaches including transcriptomics and lipidomics were applied to analyze RNA and lipid contents, respectively. Finally, an in-silico ligand-protein docking computational method was used to study a possible interaction between tefluthrin and a protein target. RESULTS Tefluthrin exposure caused severe morphological malformations in zebrafish larvae, including motor neuron abnormalities. The differentially expressed genes were associated with neurotoxicity and metabolic disruption. Lipidomics analysis revealed a disruption in fatty acid, phospholipid, and lysophospholipid recycling. Protein docking modeling suggested that the LPCAT3 enzyme, which recycles lysophospholipids in the Land's cycle, directly interacts with tefluthrin. CONCLUSIONS Tefluthrin exposure causes morphological and neuronal malformations in zebrafish larvae at nanomolar concentrations. Multi-omics results revealed a potential molecular initiating event i.e., inhibition of LPCAT3, and key events i.e., an altered lysophospholipid to phospholipid ratio, leading to the adverse outcomes of neurotoxicity and metabolic disruption.
Collapse
|
3
|
Wang C, Bhutta A, Zhang X, Liu F, Liu S, Latham LE, Talpos JC, Patterson TA, Slikker W. Development of a primate model to evaluate the effects of ketamine and surgical stress on the neonatal brain. Exp Biol Med (Maywood) 2023; 248:624-632. [PMID: 37208914 PMCID: PMC10350805 DOI: 10.1177/15353702231168144] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 05/21/2023] Open
Abstract
With advances in pediatric and obstetric surgery, pediatric patients are subject to complex procedures under general anesthesia. The effects of anesthetic exposure on the developing brain may be confounded by several factors including pre-existing disorders and surgery-induced stress. Ketamine, a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, is routinely used as a pediatric general anesthetic. However, controversy remains about whether ketamine exposure may be neuroprotective or induce neuronal degeneration in the developing brain. Here, we report the effects of ketamine exposure on the neonatal nonhuman primate brain under surgical stress. Eight neonatal rhesus monkeys (postnatal days 5-7) were randomly assigned to each of two groups: Group A (n = 4) received 2 mg/kg ketamine via intravenous bolus prior to surgery and a 0.5 mg/kg/h ketamine infusion during surgery in the presence of a standardized pediatric anesthetic regimen; Group B (n = 4) received volumes of normal saline equivalent to those of ketamine given to Group A animals prior to and during surgery, also in the presence of a standardized pediatric anesthetic regimen. Under anesthesia, the surgery consisted of a thoracotomy followed by closing the pleural space and tissue in layers using standard surgical techniques. Vital signs were monitored to be within normal ranges throughout anesthesia. Elevated levels of cytokines interleukin (IL)-8, IL-15, monocyte chemoattractant protein-1 (MCP-1), and macrophage inflammatory protein (MIP)-1β at 6 and 24 h after surgery were detected in ketamine-exposed animals. Fluoro-Jade C staining revealed significantly higher neuronal degeneration in the frontal cortex of ketamine-exposed animals, compared with control animals. Intravenous ketamine administration prior to and throughout surgery in a clinically relevant neonatal primate model appears to elevate cytokine levels and increase neuronal degeneration. Consistent with previous data on the effects of ketamine on the developing brain, the results from the current randomized controlled study in neonatal monkeys undergoing simulated surgery show that ketamine does not provide neuroprotective or anti-inflammatory effects.
Collapse
Affiliation(s)
- Cheng Wang
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Adnan Bhutta
- University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Riley Children’s Hospital, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xuan Zhang
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Fang Liu
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Shuliang Liu
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Leah E Latham
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - John C Talpos
- Division of Neurotoxicology, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | - Tucker A Patterson
- Office of Research, National Center for Toxicological Research, Food and Drug Administration (FDA), Jefferson, AR 72079, USA
| | | |
Collapse
|
4
|
Riviello JJ, Curry DJ, Weiner HL. An Introduction to Minimally Invasive Pediatric Epilepsy Surgery. JOURNAL OF PEDIATRIC EPILEPSY 2022. [DOI: 10.1055/s-0042-1759876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AbstractThe field of minimally invasive surgery has evolved over the past 50 years, including neurosurgery, with an evolution to “minimally invasive neurosurgery” when feasible. Epilepsy surgery has followed this trend, with a transition from standard neurosurgical techniques to minimally invasive techniques in all phases of neurosurgical involvement. These include the diagnostic intracranial electroencephalogram with a subdural exploration to stereoelectroencephalography, the actual resection from an open craniotomy to a less destructive technique, or the multiple modalities of neuromodulation instead of a destructive surgery.The influence of these minimally invasive techniques has resulted in a change in the overall philosophy of pediatric epilepsy surgery. The expectations of what is considered “successful” epilepsy surgery has changed from total seizure control, in other words, a “cure,” to palliative epilepsy surgery with a decrease in the targeted seizures, especially “disabling seizures.” This has led to an overall greater acceptance of epilepsy surgery. This article summarizes the major reasons behind the explosion of minimally invasive pediatric epilepsy surgery, which are amplified in the subsequent articles. Some of this chapter includes the authors' opinions.
Collapse
Affiliation(s)
- James J. Riviello
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, Unites States
- Department of Neurology, Texas Children's Hospital, Houston, Texas, Unites States
| | - Daniel J. Curry
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, Unites States
- Division of Pediatric Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, Unites States
| | - Howard L. Weiner
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, Unites States
- Division of Pediatric Neurosurgery, Department of Surgery, Texas Children's Hospital, Houston, Texas, Unites States
| |
Collapse
|
5
|
Prenatal exposure to benzodiazepines and Z-drugs in humans and risk of adverse neurodevelopmental outcomes in offspring: A systematic review. Neurosci Biobehav Rev 2022; 137:104647. [PMID: 35367514 DOI: 10.1016/j.neubiorev.2022.104647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
When used during pregnancy, benzodiazepines (BZDs) and related z-drugs could pass readily through the placenta and the foetal blood-brain barrier, where they can bind to γ-amino butyric acid (GABA) receptors in the developing foetal brain. Yet, data on long-term safety of prenatal BZD and z-drug use and its impact on offspring neurodevelopment are inconclusive. In this systematic review, we qualitatively synthetize the existing evidence on maternal exposure to various BZDs and z-drugs during pregnancy and offspring cognitive, emotional, behavioural, and motor skills developmental outcomes. Nineteen studies were included. We used harvest plots to visualize the directions of reported associations. Despite several associations between distinct types of BZDs and z-drugs and an increased risk of outcomes within different neurodevelopmental domains were observed, a remarkable scarcity of overall research on the topic and considerable discrepancies in methodology, particularly towards controlling for confounding by indication, precluded drawing conclusions with a reasonable degree of certainty. We outline various research strategies to mitigate methodological limitations and provide directions for future empirical studies on the topic.
Collapse
|
6
|
Zhang T, Ji D, Sun J, Song J, Nie L, Sun N. NPAS4 suppresses propofol-induced neurotoxicity by inhibiting autophagy in hippocampal neuronal cells. Arch Biochem Biophys 2021; 711:109018. [PMID: 34418347 DOI: 10.1016/j.abb.2021.109018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/16/2021] [Accepted: 08/16/2021] [Indexed: 01/15/2023]
Abstract
Propofol, a general intravenous anesthetic, has been demonstrated to cause a profound neuroapoptosis in the developing brain followed by long-term neurocognitive impairment. Our study aimed to examine the neuroprotective effect of neuronal PAS domain protein 4 (NPAS4), an activity-dependent neuron-specific transcription factor, on propofol-induced neurotoxicity in hippocampal neuronal HT22 cells. The differentially expressed genes in HT22 cells after treatment with propofol were screened from Gene Expression Omnibus dataset GSE106799. NPAS4 expression in HT22 cells treated with different doses of propofol was investigated by qRT-PCR and Western blot analysis. Cell viability, lactate dehydrogenase (LDH) release, caspase-3 activity, and apoptosis were evaluated by MTT, a LDH-Cytotoxicity Assay Kit, a Caspase-3 Colorimetric Assay Kit, and TUNEL assay, respectively. The protein levels of LC3-I, LC3-II, Beclin 1, p62 and NPAS4 were detected using Western blot analysis. Propofol treatment concentration-dependently decreased NPAS4 expression in HT22 cells. Propofol treatment inhibited cell viability, increased LDH release and caspase-3 activity, and induced apoptosis and autophagy in HT22 cells. NPAS4 overexpression suppressed propofol-induced cell injury and autophagy in HT22 cells. Mechanistically, autophagy agonist rapamycin attenuated the neuroprotective effect of NPAS4 in propofol-treated HT22 cells. In conclusion, NAPS4 overexpression protected hippocampal neuronal HT22 cells against propofol-induced neurotoxicity by reducing autophagy.
Collapse
Affiliation(s)
- Tongyin Zhang
- Department of Anesthesiology, Nanshi Hospital Affiliated to Henan University, Nanyang, 473065, China
| | - Daofei Ji
- Department of Neurosurgery, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Junyi Sun
- Department of Anesthesiology, Nanshi Hospital Affiliated to Henan University, Nanyang, 473065, China
| | - Jiangling Song
- Department of Anesthesiology, Nanshi Hospital Affiliated to Henan University, Nanyang, 473065, China
| | - Limin Nie
- Department of Anesthesiology, Nanshi Hospital Affiliated to Henan University, Nanyang, 473065, China
| | - Na Sun
- Catheterization Room, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, 223002, China.
| |
Collapse
|
7
|
Hyperoxia Inhibits Proliferation of Retinal Endothelial Cells in a Myc-Dependent Manner. Life (Basel) 2021; 11:life11070614. [PMID: 34202240 PMCID: PMC8304924 DOI: 10.3390/life11070614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/21/2021] [Indexed: 01/03/2023] Open
Abstract
Oxygen supplementation is necessary to prevent mortality in severely premature infants. However, the supraphysiological concentration of oxygen utilized in these infants simultaneously creates retinovascular growth attenuation and vasoobliteration that induces the retinopathy of prematurity. Here, we report that hyperoxia regulates the cell cycle and retinal endothelial cell proliferation in a previously unknown Myc-dependent manner, which contributes to oxygen-induced retinopathy.
Collapse
|
8
|
Uema S, Horita M, Takadera T. Protective effects of calcium ions via L-type calcium channels and NMDA receptors on prostaglandin E 2-induced apoptosis in rat cortical cells. Mol Biol Rep 2021; 48:4517-4525. [PMID: 34089465 DOI: 10.1007/s11033-021-06472-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/01/2021] [Indexed: 11/24/2022]
Abstract
Calcium ions mediate a variety of physiological responses of developing neurons including survival. The purpose of this study was to examine the effect of calcium influx through L-type calcium channels (LTCCs) or NMDA receptors on prostaglandin E2 (PGE2)-induced apoptosis in rat cortical cells. Cultures of rat cortical cells were prepared from an embryonic day 18 rat neocortex. After culturing for 2 or 8 days in vitro (DIV), the cells were subjected to PGE2 treatment for 48 h. FPL64176, an LTCC agonist, protected the cells at 2 and 8 DIV from PGE2-induced apoptosis. On the other hand, N-methyl-D-aspartate (NMDA), an agonist of NMDA receptor, protected the cells from PGE2-induced apoptosis only at 8 DIV. FPL64176 increased the calcium levels at 2 and 8 DIV, whereas NMDA increased the calcium levels only at 8 DIV. The protective effects of the LTCC agonist and NMDA on PGE2-induced apoptosis were blocked following treatment of the cells with protein kinase C inhibitors. Our results suggest that LTCCs and NMDA receptors modulate the cell death of developing cortical neurons possibly through a protein kinase C pathway.
Collapse
Affiliation(s)
- Shota Uema
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan
| | - Mizue Horita
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan
| | - Tsuneo Takadera
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan.
| |
Collapse
|
9
|
Falsaperla R, Scalia B, Giugno A, Pavone P, Motta M, Caccamo M, Ruggieri M. Treating the symptom or treating the disease in neonatal seizures: a systematic review of the literature. Ital J Pediatr 2021; 47:85. [PMID: 33827647 PMCID: PMC8028713 DOI: 10.1186/s13052-021-01027-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
Aim The existing treatment options for neonatal seizures have expanded over the last few decades, but no consensus has been reached regarding the optimal therapeutic protocols. We systematically reviewed the available literature examining neonatal seizure treatments to clarify which drugs are the most effective for the treatment of specific neurologic disorders in newborns. Method We reviewed all available, published, literature, identified using PubMed (published between August 1949 and November 2020), that focused on the pharmacological treatment of electroencephalogram (EEG)-confirmed neonatal seizures. Results Our search identified 427 articles, of which 67 were included in this review. Current knowledge allowed us to highlight the good clinical and electrographic responses of genetic early-onset epilepsies to sodium channel blockers and the overall good response to levetiracetam, whose administration has also been demonstrated to be safe in both full-term and preterm newborns. Interpretation Our work contributes by confirming the limited availability of evidence that can be used to guide the use of anticonvulsants to treat newborns in clinical practice and examining the efficacy and potentially harmful side effects of currently available drugs when used to treat the developing newborn brain; therefore, our work might also serve as a clinical reference for future studies.
Collapse
Affiliation(s)
- Raffaele Falsaperla
- Neonatal Intensive Care Unit, A.O.U. San Marco-Policlinico, University of Catania, Via Carlo Azeglio Ciampi, 95121, Catania, Italy
| | - Bruna Scalia
- Neonatal Intensive Care Unit, A.O.U. San Marco-Policlinico, University of Catania, Via Carlo Azeglio Ciampi, 95121, Catania, Italy.
| | - Andrea Giugno
- Post graduate programme in Pediatrics, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Piero Pavone
- Unit of Clinical Pediatrics, A.O.U. "Policlinico", P.O. "G. Rodolico", University of Catania, Catania, Italy
| | - Milena Motta
- Neonatal Intensive Care Unit, A.O.U. San Marco-Policlinico, University of Catania, Via Carlo Azeglio Ciampi, 95121, Catania, Italy
| | - Martina Caccamo
- Neonatal Intensive Care Unit, A.O.U. San Marco-Policlinico, University of Catania, Via Carlo Azeglio Ciampi, 95121, Catania, Italy
| | - Martino Ruggieri
- Department of Clinical and Experimental Medicine Section of Pediatrics and Child Neuropsychiatry, A.O.U. San Marco- Policlinico, University of Catania, Catania, Italy
| |
Collapse
|
10
|
Pharmacodynamic Effects of Standard versus High Caffeine Doses in the Developing Brain of Neonatal Rats Exposed to Intermittent Hypoxia. Int J Mol Sci 2021; 22:ijms22073473. [PMID: 33801707 PMCID: PMC8037517 DOI: 10.3390/ijms22073473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/11/2021] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Caffeine citrate, at standard doses, is effective for reducing the incidence of apnea of prematurity (AOP) and may confer neuroprotection and decrease neonatal morbidities in extremely low gestational age neonates (ELGANs) requiring oxygen therapy. We tested the hypothesis that high-dose caffeine (HiC) has no adverse effects on the neonatal brain. (2) Methods: Newborn rat pups were randomized to room air (RA), hyperoxia (Hx) or neonatal intermittent hypoxia (IH), from birth (P0) to P14 during which they received intraperitoneal injections of LoC (20 mg/kg on P0; 5 mg/kg/day on P1-P14), HiC (80 mg/kg; 20 mg/kg), or equivalent volume saline. Blood gases, histopathology, myelin and neuronal integrity, and adenosine receptor reactivity were assessed. (3) Results: Caffeine treatment in Hx influenced blood gases more than treatment in neonatal IH. Exposure to neonatal IH resulted in hemorrhage and higher brain width, particularly in layer 2 of the cerebral cortex. Both caffeine doses increased brain width in RA, but layer 2 was increased only with HiC. HiC decreased oxidative stress more effectively than LoC, and both doses reduced apoptosis biomarkers. In RA, both caffeine doses improved myelination, but the effect was abolished in Hx and neonatal IH. Similarly, both doses inhibited adenosine 1A receptor in all oxygen environments, but adenosine 2A receptor was inhibited only in RA and Hx. (4) Conclusions: Caffeine, even at high doses, when administered in normoxia, can confer neuroprotection, evidenced by reductions in oxidative stress, hypermyelination, and increased Golgi bodies. However, varying oxygen environments, such as Hx or neonatal IH, may alter and modify pharmacodynamic actions of caffeine and may even override the benefits caffeine.
Collapse
|
11
|
Ulmke PA, Xie Y, Sokpor G, Pham L, Shomroni O, Berulava T, Rosenbusch J, Basu U, Fischer A, Nguyen HP, Staiger JF, Tuoc T. Post-transcriptional regulation by the exosome complex is required for cell survival and forebrain development via repression of P53 signaling. Development 2021; 148:dev.188276. [PMID: 33462115 DOI: 10.1242/dev.188276] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022]
Abstract
Fine-tuned gene expression is crucial for neurodevelopment. The gene expression program is tightly controlled at different levels, including RNA decay. N6-methyladenosine (m6A) methylation-mediated degradation of RNA is essential for brain development. However, m6A methylation impacts not only RNA stability, but also other RNA metabolism processes. How RNA decay contributes to brain development is largely unknown. Here, we show that Exosc10, a RNA exonuclease subunit of the RNA exosome complex, is indispensable for forebrain development. We report that cortical cells undergo overt apoptosis, culminating in cortical agenesis upon conditional deletion of Exosc10 in mouse cortex. Mechanistically, Exosc10 directly binds and degrades transcripts of the P53 signaling-related genes, such as Aen and Bbc3. Overall, our findings suggest a crucial role for Exosc10 in suppressing the P53 pathway, in which the rapid turnover of the apoptosis effectors Aen and Bbc3 mRNAs is essential for cell survival and normal cortical histogenesis.
Collapse
Affiliation(s)
- Pauline Antonie Ulmke
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Yuanbin Xie
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Gannan Medical University, 341000 Ganzhou, The People's Republic of China
| | - Godwin Sokpor
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Linh Pham
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Orr Shomroni
- Microarray and Deep-Sequencing Core Facility, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Tea Berulava
- German Center for Neurodegenerative Diseases, Goettingen 37075, Germany
| | - Joachim Rosenbusch
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Uttiya Basu
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Goettingen 37075, Germany.,Department for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, Goettingen 37075, Germany.,Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Goettingen, Goettingen 37075, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Jochen F Staiger
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Tran Tuoc
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany .,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| |
Collapse
|
12
|
Primary cilia safeguard cortical neurons in neonatal mouse forebrain from environmental stress-induced dendritic degeneration. Proc Natl Acad Sci U S A 2020; 118:2012482118. [PMID: 33443207 DOI: 10.1073/pnas.2012482118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The developing brain is under the risk of exposure to a multitude of environmental stressors. While perinatal exposure to excessive levels of environmental stress is responsible for a wide spectrum of neurological and psychiatric conditions, the developing brain is equipped with intrinsic cell protection, the mechanisms of which remain unknown. Here we show, using neonatal mouse as a model system, that primary cilia, hair-like protrusions from the neuronal cell body, play an essential role in protecting immature neurons from the negative impacts of exposure to environmental stress. More specifically, we found that primary cilia prevent the degeneration of dendritic arbors upon exposure to alcohol and ketamine, two major cell stressors, by activating cilia-localized insulin-like growth factor 1 receptor and downstream Akt signaling. We also found that activation of this pathway inhibits Caspase-3 activation and caspase-mediated cleavage/fragmentation of cytoskeletal proteins in stress-exposed neurons. These results indicate that primary cilia play an integral role in mitigating adverse impacts of environmental stressors such as drugs on perinatal brain development.
Collapse
|
13
|
Johne M, Römermann K, Hampel P, Gailus B, Theilmann W, Ala-Kurikka T, Kaila K, Löscher W. Phenobarbital and midazolam suppress neonatal seizures in a noninvasive rat model of birth asphyxia, whereas bumetanide is ineffective. Epilepsia 2020; 62:920-934. [PMID: 33258158 DOI: 10.1111/epi.16778] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Neonatal seizures are the most frequent type of neurological emergency in newborn infants, often being a consequence of prolonged perinatal asphyxia. Phenobarbital is currently the most widely used antiseizure drug for treatment of neonatal seizures, but fails to stop them in ~50% of cases. In a neonatal hypoxia-only model based on 11-day-old (P11) rats, the NKCC1 inhibitor bumetanide was reported to potentiate the antiseizure activity of phenobarbital, whereas it was ineffective in a human trial in neonates. The aim of this study was to evaluate the effect of clinically relevant doses of bumetanide as add-on to phenobarbital on neonatal seizures in a noninvasive model of birth asphyxia in P11 rats, designed for better translation to the human term neonate. METHODS Intermittent asphyxia was induced for 30 minutes by exposing the rat pups to three 7 + 3-minute cycles of 9% and 5% O2 at constant 20% CO2 . Drug treatments were administered intraperitoneally either before or immediately after asphyxia. RESULTS All untreated rat pups had seizures within 10 minutes after termination of asphyxia. Phenobarbital significantly blocked seizures when applied before asphyxia at 30 mg/kg but not 15 mg/kg. Administration of phenobarbital after asphyxia was ineffective, whereas midazolam (0.3 or 1 mg/kg) exerted significant antiseizure effects when administered before or after asphyxia. In general, focal seizures were more resistant to treatment than generalized convulsive seizures. Bumetanide (0.3 mg/kg) alone or in combination with phenobarbital (15 or 30 mg/kg) exerted no significant effect on seizure occurrence. SIGNIFICANCE The data demonstrate that bumetanide does not increase the efficacy of phenobarbital in a model of birth asphyxia, which is consistent with the negative data of the recent human trial. The translational data obtained with the novel rat model of birth asphyxia indicate that it is a useful tool to evaluate novel treatments for neonatal seizures.
Collapse
Affiliation(s)
- Marie Johne
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Philip Hampel
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Björn Gailus
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Wiebke Theilmann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany
| | - Tommi Ala-Kurikka
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
14
|
Usiello A, Di Fiore MM, De Rosa A, Falvo S, Errico F, Santillo A, Nuzzo T, Chieffi Baccari G. New Evidence on the Role of D-Aspartate Metabolism in Regulating Brain and Endocrine System Physiology: From Preclinical Observations to Clinical Applications. Int J Mol Sci 2020; 21:E8718. [PMID: 33218144 PMCID: PMC7698810 DOI: 10.3390/ijms21228718] [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: 10/14/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 11/16/2022] Open
Abstract
The endogenous amino acids serine and aspartate occur at high concentrations in free D-form in mammalian organs, including the central nervous system and endocrine glands. D-serine (D-Ser) is largely localized in the forebrain structures throughout pre and postnatal life. Pharmacologically, D-Ser plays a functional role by acting as an endogenous coagonist at N-methyl-D-aspartate receptors (NMDARs). Less is known about the role of free D-aspartate (D-Asp) in mammals. Notably, D-Asp has a specific temporal pattern of occurrence. In fact, free D-Asp is abundant during prenatal life and decreases greatly after birth in concomitance with the postnatal onset of D-Asp oxidase expression, which is the only enzyme known to control endogenous levels of this molecule. Conversely, in the endocrine system, D-Asp concentrations enhance after birth during its functional development, thereby suggesting an involvement of the amino acid in the regulation of hormone biosynthesis. The substantial binding affinity for the NMDAR glutamate site has led us to investigate the in vivo implications of D-Asp on NMDAR-mediated responses. Herein we review the physiological function of free D-Asp and of its metabolizing enzyme in regulating the functions of the brain and of the neuroendocrine system based on recent genetic and pharmacological human and animal studies.
Collapse
Affiliation(s)
- Alessandro Usiello
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Napoli, Italy;
| | - Maria Maddalena Di Fiore
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
| | - Arianna De Rosa
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Napoli, Italy;
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Sara Falvo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
| | - Francesco Errico
- Dipartimento di Agraria, Università degli Studi di Napoli Federico II, Via Università, 100, 80055 Portici, Italy;
| | - Alessandra Santillo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
| | - Tommaso Nuzzo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
- CEINGE Biotecnologie Avanzate, Via Gaetano Salvatore 486, 80145 Napoli, Italy;
| | - Gabriella Chieffi Baccari
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università della Campania «L. Vanvitelli», Via Vivaldi 43, 81100 Caserta, Italy; (M.M.D.F.); (S.F.); (A.S.); (T.N.)
| |
Collapse
|
15
|
Errico F, Cuomo M, Canu N, Caputo V, Usiello A. New insights on the influence of free d-aspartate metabolism in the mammalian brain during prenatal and postnatal life. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140471. [PMID: 32561430 DOI: 10.1016/j.bbapap.2020.140471] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 01/08/2023]
Abstract
Free d-aspartate is abundant in the mammalian embryonic brain. However, following the postnatal onset of the catabolic d-aspartate oxidase (DDO) activity, cerebral d-aspartate levels drastically decrease, remaining constantly low throughout life. d-Aspartate stimulates both glutamatergic NMDA receptors (NMDARs) and metabotropic Glu5 receptors. In rodents, short-term d-aspartate exposure increases spine density and synaptic plasticity, and improves cognition. Conversely, persistently high d-Asp levels produce NMDAR-dependent neurotoxic effects, leading to precocious neuroinflammation and cell death. These pieces of evidence highlight the dichotomous impact of d-aspartate signaling on NMDAR-dependent processes and, in turn, unveil a neuroprotective role for DDO in preventing the detrimental effects of excessive d-aspartate stimulation during aging. Here, we will focus on the in vivo influence of altered d-aspartate metabolism on the modulation of glutamatergic functions and its involvement in translational studies. Finally, preliminary data on the role of embryonic d-aspartate in the mouse brain will also be reviewed.
Collapse
Affiliation(s)
- Francesco Errico
- Department of Agricultural Sciences, University of Naples "Federico II", 80055 Portici, Italy.
| | - Mariella Cuomo
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", 80131 Naples, Italy
| | - Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata", 00133 Rome, Italy; Institute of Biochemistry and Cell Biology, National Research Council (CNR), 00015, Monterotondo Scalo, Rome, Italy
| | - Viviana Caputo
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; Department of Environmental, Biological and Pharmaceutical Science and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", 81100 Caserta, Italy
| |
Collapse
|
16
|
Regions of the basal ganglia and primary olfactory system are most sensitive to neurodegeneration after extended sevoflurane anesthesia in the perinatal rat. Neurotoxicol Teratol 2020; 80:106890. [PMID: 32413489 DOI: 10.1016/j.ntt.2020.106890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/10/2020] [Accepted: 04/29/2020] [Indexed: 01/22/2023]
Abstract
Extended general anesthesia early in life is neurotoxic in multiple species. However, little is known about the temporal progression of neurodegeneration after general anesthesia. It is also unknown if a reduction in natural cell death, or an increase in cell creation, occurs as a form of compensation after perinatal anesthesia exposure. The goal of this study was to evaluate markers of neurodegeneration and cellular division at 2, 24, or 72 h after sevoflurane (Sevo) exposure (6 h) in fully oxygenated postnatal day (PND) 7 rats. Neurodegeneration was observed in areas throughout the forebrain, while the largest changes (fold increase above vehicle) were observed in areas associated with either the primary olfactory learning pathways or the basal ganglia. These regions included the indusium griseum (IG, 25-fold), the posterior dorso medial hippocampal CA1 (17-fold), bed nucleus of the stria terminalis (Bed Nuclei STM, 5-fold), the shell of the nucleus accumbens (Acb, 5-fold), caudate/putamen (CPu, 5-fold), globus pallidus (GP, 9-fold) and associated thalamic (11-fold) and cortical regions (5-fold). Sevo neurodegeneration was minimal or undetectable in the ventral tegmentum, substantia nigra, and most of the hypothalamus and frontal cortex. In most brain regions where neurodegeneration was increased 2 h post Sevo exposure, the levels returned to <4-fold above control levels by 24 h. However, in the IG, CA1, GP, anterior thalamus, medial preoptic nucleus of the hypothalamus (MPO), anterior hypothalamic area (AHP), and the amygdaloid nuclei, neurodegeneration at 24 h was double or more than that at 2 h post exposure. Anesthesia exposure causes either a prolonged period of neurodegeneration in certain brain regions, or a distinct secondary degenerative event occurs after the initial insult. Moreover, regions most sensitive to Sevo neurodegeneration did not necessarily coincide with areas of new cell birth, and new cell birth was not consistently affected by Sevo. The profile of anesthesia related neurotoxicity changes with time, and multiple mechanisms of toxicity may exist in a time-dependent fashion.
Collapse
|
17
|
Prenatal expression of d-aspartate oxidase causes early cerebral d-aspartate depletion and influences brain morphology and cognitive functions at adulthood. Amino Acids 2020; 52:597-617. [DOI: 10.1007/s00726-020-02839-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/06/2020] [Indexed: 12/25/2022]
|
18
|
González-Maciel A, Romero-Velázquez RM, Alfaro-Rodríguez A, Sanchez Aparicio P, Reynoso-Robles R. Prenatal exposure to oxcarbazepine increases hippocampal apoptosis in rat offspring. J Chem Neuroanat 2019; 103:101729. [PMID: 31794794 DOI: 10.1016/j.jchemneu.2019.101729] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/29/2019] [Accepted: 11/29/2019] [Indexed: 01/18/2023]
Abstract
This study assessed apoptosis in the offspring of rats exposed to oxcarbazepine (OXC) from day 7 to 15 of gestation. Three groups of pregnant Wistar rats were used: 1) Control, treated with saline solution; 2) treated with 100 mg/kg OXC; 3) treated with 100 mg/kg of carbamazepine (CBZ, as a positive control for apoptosis); the route of administration was intragastric. Apoptosis was detected at three postnatal ages using the TUNEL technique in the CA1, and CA3 regions of the hippocampus and in the dentate gyrus (DG); neurogenesis was assessed in the DG using an antibody against doublecortin. The litter characteristics were recorded. OXC increased apoptosis in all regions (p < 0.01) at the three ages evaluated. Lamination disruption occurred in CA1 and CA3 due to the neuron absence and to ectopic neurons; there were also malformations in the dorsal lamina of the DG in 38% and 25% of the pups born from rats treated with OXC and CBZ respectively. CBZ also increased apoptosis. No clear effect on neurogenesis in the DG was observed. The size of the litter was smaller (p < 0.01) in the experimental groups. Nineteen-day OXC fetuses had low weight (p < 0.01), but 21 and 30 postnatal days old CBZ and OXC pups were overweight (p < 0.01). The results demonstrate that OXC administered during gestation is pro-apoptotic, alters the cytoarchitecture of the hippocampus, reduces litter size, and probably influences postnatal weight. We provide evidence of the proapoptotic effect of CBZ when administered early in gestation.
Collapse
Affiliation(s)
- A González-Maciel
- Laboratory of Cell and Tissue Morphology, Instituto Nacional de Pediatría, Secretaría de Salud, Insurgentes Sur No. 3700-C, Mexico City, C. P. 04530, Mexico.
| | - R M Romero-Velázquez
- Laboratory of Cell and Tissue Morphology, Instituto Nacional de Pediatría, Secretaría de Salud, Insurgentes Sur No. 3700-C, Mexico City, C. P. 04530, Mexico.
| | - A Alfaro-Rodríguez
- Division of Neurosciences, Instituto Nacional de Rehabilitación, "Luis Guillermo Ibarra Ibarra", Secretaría de Salud, Col. Arenal de Guadalupe, Mexico City, C.P. 14389, Mexico.
| | - P Sanchez Aparicio
- Faculty of Veterinary Medicine, Department of Pharmacology, Universidad Autónoma del Estado de México, Mexico
| | - R Reynoso-Robles
- Laboratory of Cell and Tissue Morphology, Instituto Nacional de Pediatría, Secretaría de Salud, Insurgentes Sur No. 3700-C, Mexico City, C. P. 04530, Mexico.
| |
Collapse
|
19
|
Signaling network between the dysregulated expression of microRNAs and mRNAs in propofol-induced developmental neurotoxicity in mice. Sci Rep 2018; 8:14172. [PMID: 30242182 PMCID: PMC6155049 DOI: 10.1038/s41598-018-32474-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
Mounting evidence has demonstrated that general anesthetics could induce acute neuroapoptosis in developing animals followed by long-term cognitive dysfunction, with the mechanisms remaining largely unknown. The aim of this study was to investigate the effect of the intravenous anesthetic propofol on the profiles of microRNAs (miRNAs) and messenger RNAs (mRNAs), and their interactive signaling networks in the developing mouse hippocampus. Postnatal day 7 (P7) mice were exposed to propofol for 3 hours. Hippocampi were harvested from both P7 (3 hours after exposure) and P60 mice for the analysis of the expression of 726 miRNAs and 24,881 mRNAs, and apoptosis. Long-term memory ability of P60 mice was analyzed using the Morris Water Maze. Propofol induced acute apoptosis in the hippocampus, and impaired memory function of mice. There were 100 altered mRNAs and 18 dysregulated miRNAs in the propofol-treated hippocampi compared with the intralipid-treated control tissues on P7. Bioinformatics analysis of these abnormally expressed genes on P7 indicated that 34 dysregulated miRNA-mRNA target pairs were related to pathological neurological and developmental disorder processes such as cell viability, cell morphology and migration, neural stem cell proliferation and neurogenesis, oligodendrocyte myelination, reactive oxygen species, and calcium signaling. Neonatal propofol exposure also resulted in the abnormal expression of 49 mRNAs and 4 miRNAs in P60 mouse hippocampi. Specifically, bioinformatics analysis indicates that among these dysregulated mRNAs and miRNAs, there were 2 dysregulated miRNA-mRNA targets pairs (Fam46a/miR-363-3p and Rgs3/miR-363-3p) that might be related to the effect of propofol on long-term cognitive function. Collectively, our novel investigation indicates that acute and long-term dysregulated miRNA-mRNA signaling networks potentially participate in propofol-induced developmental neurotoxicity.
Collapse
|
20
|
Neuroprotective Action of the CB1/2 Receptor Agonist, WIN 55,212-2, against DMSO but Not Phenobarbital-Induced Neurotoxicity in Immature Rats. Neurotox Res 2018; 35:173-182. [PMID: 30141144 DOI: 10.1007/s12640-018-9944-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/30/2018] [Accepted: 08/02/2018] [Indexed: 01/14/2023]
Abstract
The developing brain is uniquely susceptible to drug-induced increases in programmed cell death or apoptosis. Many compounds, including anticonvulsant drugs, anesthetic agents, and ethanol, when administered in a narrow postnatal window in rodents, result in increased pruning of neurons. Here, we report that dimethyl sulfoxide (DMSO) triggers widespread neurodegeneration in the immature (postnatal day, P7) rat brain, an effect consistent with a prior report in neonatal mice. We found that the synthetic cannabinoid receptor agonist WIN 55,212-2 (WIN) exerts a neuroprotective effect against DMSO-induced cell death. We extended these findings to determine if WIN is neuroprotective against another drug class known to increase developmental cell death, namely antiseizure drugs. The antiseizure drug phenobarbital (PB) remains the primary treatment for neonatal seizures, despite significantly increasing cell death in the developing rodent brain. WIN exerts antiseizure effects in immature rodent seizure models, but increases the toxicity associated with neonatal ethanol exposure. We thus sought to determine if WIN would protect against or exacerbate PB-induced cell death. Unlike either the prior report with ethanol or our present findings with DMSO, WIN was largely without effect on PB-induced cell death. WIN alone did not increase cell death over levels observed in vehicle-treated rats. These data suggest that WIN has a favorable safety profile in the developing brain and could potentially serve as an adjunct therapy with phenobarbital (albeit one that does not attenuate PB-induced toxicity).
Collapse
|
21
|
Han JY, Moon CJ, Youn YA, Sung IK, Lee IG. Efficacy of levetiracetam for neonatal seizures in preterm infants. BMC Pediatr 2018; 18:131. [PMID: 29636029 PMCID: PMC5892045 DOI: 10.1186/s12887-018-1103-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/27/2018] [Indexed: 11/13/2022] Open
Abstract
Background Neonatal seizures remain a significant clinical problem, and therapeutic options are still not diverse with limited efficacy. Levetiracetam (LEV) is a relatively new and wide spectrum anti-seizure medication with favorable pharmacokinetics and safety profile. In the recent decades, LEV has been increasingly used for the treatment of neonatal seizures. The aim of this study was to describe the experience of using LEV as the first line anti-seizure medication for preterm infants. Methods A retrospective analysis of 37 preterm infants who were treated with LEV as the first-line anti-seizure medication was performed. Results Mean gestational age of the 37 preterm infants was 31.5 ± 1.9 weeks (range, 26 to 36+ 6 weeks). Twenty-one infants (57%) were seizure-free while given LEV at the end of the first week, and no additional anti-seizure medication was required. Loading doses of LEV ranged from 40 to 60 mg/kg (mean 56 mg/kg) and the maintenance dose ranged from 20 to 30 mg/kg (mean 23 mg/kg). No adverse effect was observed. Conclusions Levetiracetam can be a good and safe choice for treatment of neonatal seizures in preterm infants. Prospective double blind controlled studies are needed in the future.
Collapse
Affiliation(s)
- Ji Yoon Han
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seochogu, Seoul, 137-701, South Korea
| | - Chung Joon Moon
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seochogu, Seoul, 137-701, South Korea
| | - Young Ah Youn
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seochogu, Seoul, 137-701, South Korea
| | - In Kyung Sung
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seochogu, Seoul, 137-701, South Korea
| | - In Goo Lee
- Department of Pediatrics, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpodaero, Seochogu, Seoul, 137-701, South Korea.
| |
Collapse
|
22
|
Gao L, Han J, Bai J, Dong J, Zhang S, Zhang M, Zheng J. Nicotinic Acetylcholine Receptors are Associated with Ketamine-induced Neuronal Apoptosis in the Developing Rat Retina. Neuroscience 2018; 376:1-12. [DOI: 10.1016/j.neuroscience.2018.01.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/30/2017] [Accepted: 01/29/2018] [Indexed: 12/17/2022]
|
23
|
Memantine Can Reduce Ethanol-Induced Caspase-3 Activity and Apoptosis in H4 Cells by Decreasing Intracellular Calcium. J Mol Neurosci 2017; 62:402-411. [DOI: 10.1007/s12031-017-0948-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/12/2017] [Indexed: 01/07/2023]
|
24
|
Lotfullina N, Khazipov R. Ethanol and the Developing Brain: Inhibition of Neuronal Activity and Neuroapoptosis. Neuroscientist 2017; 24:130-141. [PMID: 28580823 DOI: 10.1177/1073858417712667] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Ethanol induces massive neuroapoptosis in the developing brain. One of the main hypotheses that has been put forward to explain the deleterious actions of ethanol in the immature brain involves an inhibition of neuronal activity. Here, we review recent evidence for this hypothesis obtained in the somatosensory cortex and hippocampus of neonatal rodents. In both structures, ethanol strongly inhibits brain activity. At the doses inducing massive neuroapoptosis, ethanol completely suppresses the early activity patterns of spindle-bursts and gamma oscillations in the neocortex and the early sharp-waves in the hippocampus. The inhibitory effects of ethanol decrease with age and in adult animals, ethanol only mildly depresses neuronal firing and induces delta-wave activity. Suppression of cortical activity in neonatal animals likely involves inhibition of the myoclonic twitches, an important physiological trigger for the early activity bursts, and inhibition of the thalamocortical and intracortical circuits through a potentiation of GABAergic transmission and an inhibition of N-methyl-d-aspartate (NMDA) receptors, that is in keeping with the neuroapoptotic effects of other agents acting on GABA and NMDA receptors. These findings provide support for the hypothesis that the ethanol-induced inhibition of cortical activity is an important pathophysiological mechanism underlying massive neuroapoptosis induced by ethanol in the developing brain.
Collapse
Affiliation(s)
- Nailya Lotfullina
- 1 INMED-INSERM, Aix-Marseille University, Marseille, France.,2 Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| | - Roustem Khazipov
- 1 INMED-INSERM, Aix-Marseille University, Marseille, France.,2 Laboratory of Neurobiology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
25
|
Serotonin transporter and receptor ligands with antidepressant activity as neuroprotective and proapoptotic agents. Pharmacol Rep 2017; 69:469-478. [DOI: 10.1016/j.pharep.2017.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 01/11/2017] [Accepted: 01/18/2017] [Indexed: 12/23/2022]
|
26
|
Exome chip analyses in adult attention deficit hyperactivity disorder. Transl Psychiatry 2016; 6:e923. [PMID: 27754487 PMCID: PMC5315553 DOI: 10.1038/tp.2016.196] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Revised: 08/05/2016] [Accepted: 08/17/2016] [Indexed: 11/25/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly heritable childhood-onset neuropsychiatric condition, often persisting into adulthood. The genetic architecture of ADHD, particularly in adults, is largely unknown. We performed an exome-wide scan of adult ADHD using the Illumina Human Exome Bead Chip, which interrogates over 250 000 common and rare variants. Participants were recruited by the International Multicenter persistent ADHD CollaboraTion (IMpACT). Statistical analyses were divided into 3 steps: (1) gene-level analysis of rare variants (minor allele frequency (MAF)<1%); (2) single marker association tests of common variants (MAF⩾1%), with replication of the top signals; and (3) pathway analyses. In total, 9365 individuals (1846 cases and 7519 controls) were examined. Replication of the most associated common variants was attempted in 9847 individuals (2077 cases and 7770 controls) using fixed-effects inverse variance meta-analysis. With a Bonferroni-corrected significance level of 1.82E-06, our analyses of rare coding variants revealed four study-wide significant loci: 6q22.1 locus (P=4.46E-08), where NT5DC1 and COL10A1 reside; the SEC23IP locus (P=6.47E-07); the PSD locus (P=7.58E-08) and ZCCHC4 locus (P=1.79E-06). No genome-wide significant association was observed among the common variants. The strongest signal was noted at rs9325032 in PPP2R2B (odds ratio=0.81, P=1.61E-05). Taken together, our data add to the growing evidence of general signal transduction molecules (NT5DC1, PSD, SEC23IP and ZCCHC4) having an important role in the etiology of ADHD. Although the biological implications of these findings need to be further explored, they highlight the possible role of cellular communication as a potential core component in the development of both adult and childhood forms of ADHD.
Collapse
|
27
|
|
28
|
Costa AP, Lopes MW, Rieger DK, Barbosa SGR, Gonçalves FM, Xikota JC, Walz R, Leal RB. Differential Activation of Mitogen-Activated Protein Kinases, ERK 1/2, p38(MAPK) and JNK p54/p46 During Postnatal Development of Rat Hippocampus. Neurochem Res 2015; 41:1160-9. [PMID: 26700434 DOI: 10.1007/s11064-015-1810-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 12/12/2015] [Accepted: 12/16/2015] [Indexed: 12/11/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) are a group of serine-threonine kinases, including p38(MAPK), ERK 1/2 and JNK p54/p46, activated by phosphorylation in response to extracellular stimuli. The early postnatal period is characterized by significant changes in brain structure as well as intracellular signaling. In the hippocampus MAPKs have been involved in the modulation of development and neural plasticity. However, the temporal profile of MAPK activation throughout the early postnatal development is incomplete. An understanding of this profile is important since slight changes in the activity of these enzymes, in response to environmental stress in specific developmental windows, might alter the course of development. The present study was undertaken to investigate the hippocampal differential activation of MAPK during postnatal period. MAPK activation and total content were evaluated by Western blotting of hippocampal tissue obtained from male Wistar rats at postnatal days (P) 1, 4, 7, 10, 14, 21, 30 and 60. The total content and phosphorylation of each MAPK was expressed as mean ± SEM and then calculates as a percentile compared to P1 (set at 100 %). The results showed: (1) phosphorylation peaks of p38(MAPK) at PN4 (p = 0.036) and PN10 to PN60; (2) phosphorylation of ERK1 and ERK2 were increased with age (ERK1 p = 0.0000005 and ERK2 p = 0.003); (3) phosphorylation profile of JNK p54/p46 was not changed during the period analyzed (JNKp56 p = 0.716 and JNKp46 p = 0.192). Therefore, the activity profile of ERK 1/2 and p38(MAPK) during postnatal development of rat hippocampus are differentially regulated. Our results demonstrate that ERK 1/2 and p38(MAPK) are dynamically regulated during postnatal neurodevelopment, suggesting temporal correlation of MAPK activity with critical periods when programmed cell death and synaptogenesis are occurring. This suggests an important role for these MAPKs in postnatal development of rat hippocampus.
Collapse
Affiliation(s)
- Ana Paula Costa
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Mark William Lopes
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Débora K Rieger
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Sabrina Giovana Rocha Barbosa
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Filipe Marques Gonçalves
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - João Carlos Xikota
- Departamento de Pediatria, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Roger Walz
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
- Departamento de Clínica Médica, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil
| | - Rodrigo B Leal
- Programa de Pós-Graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
29
|
Oliveira-Pinto J, Paes-Branco D, Cristina-Rodrigues F, Krahe TE, Manhães AC, Abreu-Villaça Y, Filgueiras CC. GABAA overactivation potentiates the effects of NMDA blockade during the brain growth spurt in eliciting locomotor hyperactivity in juvenile mice. Neurotoxicol Teratol 2015; 50:43-52. [PMID: 26056730 DOI: 10.1016/j.ntt.2015.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 05/25/2015] [Accepted: 05/31/2015] [Indexed: 10/23/2022]
Abstract
Both NMDA receptor blockade and GABAA receptor overactivation during the brain growth spurt may contribute to the hyperactivity phenotype reminiscent of attention-deficit/hyperactivity disorder. Here, we evaluated the effects of exposure to MK801 (a NMDA antagonist) and/or to muscimol (a GABAA agonist) during the brain growth spurt on locomotor activity of juvenile Swiss mice. This study was carried out in two separate experiments. In the first experiment, pups received a single i.p. injection of either saline solution (SAL), MK801 (MK, 0.1, 0.3 or 0.5 mg/kg) or muscimol (MU, 0.02, 0.1 or 0.5 mg/kg) at the second postnatal day (PND2), and PNDs 4, 6 and 8. In the second experiment, we investigated the effects of a combined injection of MK (0.1 mg/kg) and MU (doses: 0.02, 0.1 or 0.5 mg/kg) following the same injection schedule of the first experiment. In both experiments, locomotor activity was assessed for 15 min at PND25. While MK promoted a dose-dependent increase in locomotor activity, exposure to MU failed to elicit significant effects. The combined exposure to the highest dose of MU and the lowest dose of MK induced marked hyperactivity. Moreover, the combination of the low dose of MK and the high dose of MU resulted in a reduced activity in the center of the open field, suggesting an increased anxiety-like behavior. These findings suggest that, during the brain growth spurt, the blockade of NMDA receptors induces juvenile locomotor hyperactivity whereas hyperactivation of GABAA receptors does not. However, GABAA overactivation during this period potentiates the effects of NMDA blockade in inducing locomotor hyperactivity.
Collapse
Affiliation(s)
- Juliana Oliveira-Pinto
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Danielle Paes-Branco
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Fabiana Cristina-Rodrigues
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Thomas E Krahe
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Cláudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar, Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil.
| |
Collapse
|
30
|
Abstract
Phenobarbital and phenytoin have been the mainstay treatment modalities for neonatal seizures. Studies have revealed these agents control seizures in less than half of neonates, can cause neuronal apoptosis in vitro, and have highly variable pharmacokinetics in neonates. In contrast, there have been no reports of levetiracetam causing these neurotoxic effects. Due to its favorable side effect and pharmacokinetic profiles and positive efficacy outcomes in neonatal studies to date, there is great interest in the use of levetiracetam for neonatal seizures. This article reviews the literature regarding the safety of levetiracetam in neonates and its efficacy in neonatal seizures.
Collapse
Affiliation(s)
- Allison L Mruk
- Lucile Packard Children's Hospital Stanford, Palo Alto, California
| | | | | |
Collapse
|
31
|
Synaptic NMDA receptor activity is coupled to the transcriptional control of the glutathione system. Nat Commun 2015; 6:6761. [PMID: 25854456 PMCID: PMC4403319 DOI: 10.1038/ncomms7761] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/25/2015] [Indexed: 02/07/2023] Open
Abstract
How the brain’s antioxidant defenses adapt to changing demand is incompletely understood. Here we show that synaptic activity is coupled, via the NMDA receptor (NMDAR), to control of the glutathione antioxidant system. This tunes antioxidant capacity to reflect the elevated needs of an active neuron, guards against future increased demand and maintains redox balance in the brain. This control is mediated via a programme of gene expression changes that boosts the synthesis, recycling and utilization of glutathione, facilitating ROS detoxification and preventing Puma-dependent neuronal apoptosis. Of particular importance to the developing brain is the direct NMDAR-dependent transcriptional control of glutathione biosynthesis, disruption of which can lead to degeneration. Notably, these activity-dependent cell-autonomous mechanisms were found to cooperate with non-cell-autonomous Nrf2-driven support from astrocytes to maintain neuronal GSH levels in the face of oxidative insults. Thus, developmental NMDAR hypofunction and glutathione system deficits, separately implicated in several neurodevelopmental disorders, are mechanistically linked. How the brain’s antioxidant defenses adapt to changing demand is not well understood. Here the authors demonstrate that synaptic activity is coupled to transcriptional control of the glutathione antioxidant system via NMDA receptors, enabling neurons to tune their antioxidant defenses.
Collapse
|
32
|
Wang X, Yang Z, Sun Y, Zhou H, Chu G, Zhang J, Meng X. Ethanol Activation of PKA Mediates Single-Minded 2 Expression in Neuronal Cells. Mol Neurobiol 2014; 52:1234-1244. [PMID: 25319570 DOI: 10.1007/s12035-014-8924-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/06/2014] [Indexed: 10/24/2022]
Abstract
Prenatal ethanol exposure can cause extensive apoptotic neurodegeneration throughout the developing central nervous system (CNS), which results in cognitive deficits and memory decline. However, the underlying mechanisms need further study. Single-minded 2 (Sim2), a transcriptional repressor, is reportedly involved in diseases that impair learning and memory, such as Down syndrome (DS) and Alzheimer's disease. It is still unknown whether Sim2 is involved in regulating ethanol-mediated neuronal injury that might ultimately lead to neuronal dysfunction and subsequent learning and memory deficits. To study the effects of ethanol on Sim2 expression and neuronal injury, we used animal models and cell culture experiments. Our results indicated that in SH-SY5Y cells, ethanol exposure increased Sim2 expression and levels of cleaved caspase 3, which is a marker for cells undergoing apoptosis. Silencing Sim2 expression attenuated caspase 3 activation and cellular apoptosis. We also found that protein kinase A (PKA) activation induced Sim2 expression, as did ethanol. Inhibiting the PKA signaling pathway with H-89 decreased Sim2 expression and cleavage of caspase 3 that was induced by ethanol in vivo and in vitro. We further found that PKA regulated Sim2 expression at the transcriptional level. These results demonstrate that ethanol leads to increased Sim2 expression via the PKA pathway, ultimately resulting in apoptotic cell death.
Collapse
Affiliation(s)
- Xiaolan Wang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.,The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhihua Yang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yinan Sun
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hanjing Zhou
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Guangpin Chu
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Zhang
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xianfang Meng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
33
|
Lecointre M, Vézier C, Bénard M, Ramdani Y, Dupré N, Brasse-Lagnel C, Henry VJ, Roy V, Marret S, Gonzalez BJ, Jégou S, Leroux-Nicollet I. Age-dependent alterations of the NMDA receptor developmental profile and adult behavior in postnatally ketamine-treated mice. Dev Neurobiol 2014; 75:315-33. [PMID: 25220981 DOI: 10.1002/dneu.22232] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 08/22/2014] [Accepted: 09/11/2014] [Indexed: 01/13/2023]
Abstract
Ketamine is a NMDA receptor (NMDAR) antagonist used in pediatric anesthesia. Given the role of glutamatergic signaling during brain maturation, we studied the effects of a single ketamine injection (40 mg/kg s.c) in mouse neonates depending on postnatal age at injection (P2, P5, or P10) on cortical NMDAR subunits expression and association with Membrane-Associated Guanylate Kinases PSD95 and SAP102. The effects of ketamine injection at P2, P5, or P10 on motor activity were compared in adulthood. Ketamine increased GluN2A and GluN2B mRNA levels in P2-treated mice without change in proteins, while it decreased GluN2B protein in P10-treated mice without change in mRNA. Ketamine reduced GluN2A mRNA and protein levels in P5-treated mice without change in GluN2B and GluN1. Ketamine affected the GluN2A/PSD95 association regardless of the age at injection, while GluN2B/PSD95 association was enhanced only in P5-treated mice. Microdissection of ketamine-treated mouse cortex showed a decrease in GluN2A mRNA level in superficial layers (I-IV) and an increase in all subunit expressions in deep layers (V-VI) in P5- and P10-treated mice, respectively. Our data suggest that ketamine impairs cortical NMDAR subunit developmental profile and delays the synaptic targeting of GluN2A-enriched NMDAR. Ketamine injection at P2 or P10 resulted in hyperlocomotion in adult male mice in an open field, without change in females. Voluntary running-wheel exercise showed age- and sex-dependent alterations of the mouse activity, especially during the dark phase. Overall, a single neonatal ketamine exposure led to short-term NMDAR cortical developmental profile impairments and long-term motor activity alterations persisting in adulthood.
Collapse
Affiliation(s)
- Maryline Lecointre
- ERI28 "Neovasc", Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Faculty of Medicine and Pharmacy, Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Aberrant Behavioral and Neurobiologic Profiles in Rodents Exposed to Ethanol or Red Wine Early in Development. CURRENT DEVELOPMENTAL DISORDERS REPORTS 2014. [DOI: 10.1007/s40474-014-0023-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
35
|
Liu X, Holtze M, Powell SB, Terrando N, Larsson MK, Persson A, Olsson SK, Orhan F, Kegel M, Asp L, Goiny M, Schwieler L, Engberg G, Karlsson H, Erhardt S. Behavioral disturbances in adult mice following neonatal virus infection or kynurenine treatment--role of brain kynurenic acid. Brain Behav Immun 2014; 36:80-9. [PMID: 24140727 PMCID: PMC3947209 DOI: 10.1016/j.bbi.2013.10.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 09/25/2013] [Accepted: 10/11/2013] [Indexed: 12/31/2022] Open
Abstract
Exposure to infections in early life is considered a risk-factor for developing schizophrenia. Recently we reported that a neonatal CNS infection with influenza A virus in mice resulted in a transient induction of the brain kynurenine pathway, and subsequent behavioral disturbances in immune-deficient adult mice. The aim of the present study was to investigate a potential role in this regard of kynurenic acid (KYNA), an endogenous antagonist at the glycine site of the N-methyl-D-aspartic acid (NMDA) receptor and at the cholinergic α7 nicotinic receptor. C57BL/6 mice were injected i.p. with neurotropic influenza A/WSN/33 virus (2400 plaque-forming units) at postnatal day (P) 3 or with L-kynurenine (2×200 mg/kg/day) at P7-16. In mice neonatally treated with L-kynurenine prepulse inhibition of the acoustic startle, anxiety, and learning and memory were also assessed. Neonatally infected mice showed enhanced sensitivity to D-amphetamine-induced (5 mg/kg i.p.) increase in locomotor activity as adults. Neonatally L-kynurenine treated mice showed enhanced sensitivity to D-amphetamine-induced (5 mg/kg i.p.) increase in locomotor activity as well as mild impairments in prepulse inhibition and memory. Also, D-amphetamine tended to potentiate dopamine release in the striatum in kynurenine-treated mice. These long-lasting behavioral and neurochemical alterations suggest that the kynurenine pathway can link early-life infection with the development of neuropsychiatric disturbances in adulthood.
Collapse
Affiliation(s)
- Xicong Liu
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Holtze
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Susan B Powell
- Department of Psychiatry, University of California San Diego, La Jolla, California, USA
| | - Niccolò Terrando
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Markus K. Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Persson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sara K. Olsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Funda Orhan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Magdalena Kegel
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Linnea Asp
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Michel Goiny
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lilly Schwieler
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Göran Engberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Håkan Karlsson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sophie Erhardt
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
36
|
Yang YHC, Vilin YY, Roberge M, Kurata HT, Johnson JD. Multiparameter screening reveals a role for Na+ channels in cytokine-induced β-cell death. Mol Endocrinol 2014; 28:406-17. [PMID: 24438339 DOI: 10.1210/me.2013-1257] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cell death plays a role in both type 1 and type 2 diabetes, but clinical treatments that specifically target β-cell survival have not yet been developed. We have recently developed live-cell imaging-based, high-throughput screening methods capable of identifying factors that modulate pancreatic β-cell death, with the hope of finding drugs that can intervene in this process. In the present study, we used a high-content screen and the Prestwick Chemical Library of small molecules to identify drugs that block cell death resulting from exposure to a cocktail of cytotoxic cytokines (25 ng/mL TNF-α, 10 ng/mL IL-1β, and 10 ng/mL IFN-γ). Data analysis with self-organizing maps revealed that 19 drugs had profiles similar to that of the no cytokine condition, indicating protection. Carbamazepine, an antiepileptic Na(+) channel inhibitor, was particularly interesting because Na(+) channels are not generally considered targets for antiapoptotic therapy in diabetes and because the function of these channels in β-cells has not been well studied. We analyzed the expression and characteristics of Na(+) currents in mature β-cells from MIP-GFP mice. We confirmed the dose-dependent protective effects of carbamazepine and another use-dependent Na(+) channel blocker in cytokine-treated mouse islet cells. Carbamazepine down-regulated the proapoptotic and endoplasmic reticulum stress signaling induced by cytokines. Together, these studies point to Na(+) channels as a novel therapeutic target in diabetes.
Collapse
Affiliation(s)
- Yu Hsuan Carol Yang
- Department of Cellular and Physiological Sciences (Y.H.C.Y., J.D.J.), Department of Anesthesiology, Pharmacology, and Therapeutics (Y.Y.V., H.T.K.), and Department of Biochemistry and Molecular Biology (M.R.), University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | |
Collapse
|
37
|
Alimov A, Wang H, Liu M, Frank JA, Xu M, Ou X, Luo J. Expression of autophagy and UPR genes in the developing brain during ethanol-sensitive and resistant periods. Metab Brain Dis 2013; 28:667-76. [PMID: 23979425 PMCID: PMC3809151 DOI: 10.1007/s11011-013-9430-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/13/2013] [Indexed: 12/23/2022]
Abstract
Fetal alcohol spectrum disorders (FASD) results from ethanol exposure to the developing fetus and is the leading cause of mental retardation. FASD is associated with a broad range of neurobehavioral deficits which may be mediated by ethanol-induced neurodegeneration in the developing brain. An immature brain is more susceptible to ethanol neurotoxicity. We hypothesize that the enhanced sensitivity of the immature brain to ethanol is due to a limited capacity to alleviate cellular stress. Using a third trimester equivalent mouse model of ethanol exposure, we demonstrated that subcutaneous injection of ethanol induced a wide-spread neuroapoptosis in postnatal day 4 (PD4) C57BL/6 mice, but had little effect on the brain of PD12 mice. We analyzed the expression profile of genes regulating apoptosis, and the pathways of ER stress response (also known as unfolded protein response, UPR) and autophagy during these ethanol-sensitive and resistant periods (PD4 versus PD12) using PCR microarray. The expression of pro-apoptotic genes, such as caspase-3, was much higher on PD4 than PD12; in contrast, the expression of genes that regulate UPR and autophagy, such as atf6, atg4, atg9, atg10, beclin1, bnip3, cebpb, ctsb, ctsd, ctss, grp78, ire1α, lamp, lc3 perk, pik3c3, and sqstm1 was significantly higher on PD12 than PD4. These results suggest that the vulnerability of the immature brain to ethanol could result from high expression of pro-apoptotic proteins and a deficiency in the stress responsive system, such as UPR and autophagy.
Collapse
Affiliation(s)
- Alexander Alimov
- Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Haiping Wang
- Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Mei Liu
- Department of Anatomy and Neurobiology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jacqueline A. Frank
- Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Mei Xu
- Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Xiaoming Ou
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | - Jia Luo
- Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536, USA
- Correspondence author: Dr. Jia Luo, Department of Molecular and Biochemical Pharmacology, University of Kentucky College of Medicine, 132 Health Sciences Research Building, 1095 Veterans Drive, Lexington, Kentucky 40536. ; Tel: 859-323-3036; Fax: 859-257-0199
| |
Collapse
|
38
|
Halbsgut LR, Fahim E, Kapoor K, Hong H, Friedman LK. Certain secondary antiepileptic drugs can rescue hippocampal injury following a critical growth period despite poor anticonvulsant activity and cognitive deficits. Epilepsy Behav 2013; 29:466-77. [PMID: 24103817 DOI: 10.1016/j.yebeh.2013.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 08/19/2013] [Accepted: 08/22/2013] [Indexed: 12/23/2022]
Abstract
Clinical and experimental studies have shown that many common secondary antiepileptic drugs (AEDs) are ineffective at blocking seizures in adulthood; however, some afford neuroprotection. In early development, certain AEDs cause apoptosis; however, it is unknown whether these drugs are neurotoxic to the juvenile brain following a developmentally regulated proapoptotic period and whether they alter the seizure threshold, seizure-induced neuronal vulnerability, and/or cognitive function. Lamotrigine (LTG), carbamazepine (CBZ), phenytoin (PHT), valproate (VPA), and topiramate (TPM) were systemically administered to rat pups for 7days beginning on postnatal (P) day 14 (P14), then half the animals were injected with kainate (KA) to trigger seizures, an age when the CA1 subregion becomes preferentially sensitive to status epilepticus. Histological outcome, seizure severity, and learning and memory were determined with an electroencephalograph (EEG), silver impregnation, and a water-maze swim task. None of the AEDs tested significantly attenuated behavioral or electrographic seizures. Phenytoin increased mortality, identifying a detrimental side effect of this drug. The other drugs (LTG, VPA, TPM, and CBZ) afforded different amounts of protection to the CA1 subregion but not to the CA3 subregion or extrahippocampal structures. With the exception of VPA, AED-treated animals lagged behind during swim task acquisition. All groups improved in the water-maze swim task over time, particularly on the last trials; however, the average escape latency was still impaired for TPM-treated animals and all AED+KA-treated groups. Thus, while certain AEDs demonstrated some neuroprotective effects, poor antiepileptic activity, memory impairment, and other deleterious side effects were observed with these drugs suggesting that the search for potentially more effective and tolerated agents is essential for improving clinical outcome in children and adolescents with epilepsy.
Collapse
|
39
|
Affiliation(s)
- A Machotta
- Department of Anesthesiology, Sophia Children's Hospital, Erasmus MC Rotterdam, Dr. Molewaterplein 60, P.O. Box 2040, 3000 CA, Rotterdam, Niederlande.
| | | |
Collapse
|
40
|
Jin J, Gong K, Zou X, Wang R, Lin Q, Chen J. The blockade of NMDA receptor ion channels by ketamine is enhanced in developing rat cortical neurons. Neurosci Lett 2013; 539:11-5. [PMID: 23395831 DOI: 10.1016/j.neulet.2013.01.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 12/03/2012] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
Abstract
Ketamine is a non-competitive antagonist of NMDA receptors (NMDARs) commonly used as a dissociative anesthetic in many pediatric procedures. Ketamine acts primarily by blocking NMDA ligand-gated channels. Experimental studies indicate that ketamine administration used for inducing clinically relevant anesthesia can lead to neurotoxic effects, such as apoptosis, selectively on immature brain neurons. However, the underlying mechanisms remain unclear. This study used whole-cell patch-clamp recordings in an in vitro preparation of forebrain slices to analyze pharmacologically the differences in the effects of ketamine administration on the NMDAR channel activity between immature and mature neurons. NMDAR channel activity was recorded in the form of evoked NMDAR-mediated excitatory postsynaptic currents (eEPSCs) from the forebrain of both neonatal and adult rats. Results show that ketamine inhibited eEPSCs in a dose-dependent manner in both immature and mature neurons. However, at each concentration of ketamine applied to the brain slice, a more extensive inhibition could be seen in neonatal neurons than in adult neurons. Further, the blocking effect of ketamine on eEPSCs was measured during the period of 1, 3, and 6h after ketamine washout. Inhibition of eEPSCs in immature neurons was still evident 6h after washout. In contrast, the blockade of eEPSCs in mature neurons recovered completely from the inhibition by ketamine in a time-dependent manner. These results indicate that ketamine produces a greater and longer blocking effect on NMDAR channels in immature neurons than in mature neurons. This differential effect is likely to be a critical link to the higher vulnerability to ketamine-induced neurotoxicity in neurons of the developing brain.
Collapse
Affiliation(s)
- Jianhui Jin
- Institute for Biomedical Sciences of Pain, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
41
|
Liu S, Paule MG, Zhang X, Newport GD, Apana SM, Berridge MS, Patterson TA, Ali SF, Slikker W, Wang C. The Evaluation of Sevoflurane-Induced Apoptotic Neurodegeneration with MicroPET Using [18F]-DFNSH in the Developing Rat Brain. ACTA ACUST UNITED AC 2013. [DOI: 10.4303/jdar/235679] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
42
|
Abstract
The brain of the infant and young child is a developing, dynamic, structure subject to functional remodelling under the influence of factors responsible for optimal neuronal development and synaptogenesis. It exhibits age dependent variation in metabolic rate, blood flow, and ability to tolerate oxidative stress. It is also characterized by an exuberance of neurotransmitter activity, particularly in the first few years of life. The dynamic evolution and adaptability of early brain function permits the organization of neuronal networks to be influenced by environmental stimulation, and, to reduce the functional impact of injury. However, these same processes may also exacerbate the harm sustained by the brain following an acquired brain injury (ABI). The developing neurons are susceptible to excitotoxicity, oxidative stress, and, inflammation, often leading to cellular necrosis and apoptosis. Despite being immunologically privileged via the blood brain barrier, the developing brain is susceptible to injury from systemic inflammation through alteration of normally protective cerebrovascular endothelial cell function. Finally, many of the therapeutic agents currently employed in post-ABI hospital care may also compromise ABI outcome via non-intended pharmacological effects. These agents include analgesic, sedative and anti-convulsant medications. This review emphasizes those physiological considerations in the developing brain which may impact the outcome after ABI, including, the cellular mechanisms of neuronal and cerebrovascular endothelial cell injury, ABI outcome and future therapeutic directions.
Collapse
|
43
|
Li J, Yu S, Lu Z, Mohamad O, Wei L. N-methyl-D-aspartate receptor subtype 3A promotes apoptosis in developing mouse brain exposed to hyperoxia. Neural Regen Res 2012; 7:273-7. [PMID: 25806068 PMCID: PMC4353099 DOI: 10.3969/j.issn.1673-5374.2012.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2011] [Accepted: 12/22/2011] [Indexed: 11/18/2022] Open
Abstract
In the present study, 7 day postnatal C57/BL6 wild-type mice (hyperoxia group) and 7 day postnatal N-methyl-D-aspartate receptor subtype 3A knockout mice (NR3A KO group) were exposed to 75% oxygen and 15% nitrogen in a closed container for 5 days. Wild-type mice raised in normoxia served as controls. TdT-mediated dUTP nick end labeling (TUNEL)/neuron-specific nuclear protein (NeuN) and 5-bromo-2'-deoxyuridine (BrdU)/NeuN immunofluorescence staining showed that the number of apoptotic cells and the number of proliferative cells in the dentate subgranular zone significantly increased in the hyperoxia group compared with the control group. However, in the same hyperoxia environment, the number of apoptotic cells and the number of proliferative cells significantly decreased in the NR3A KO group compared with hyperoxia group. TUNEL(+)/NeuN(+) and BrdU(+)/NeuN(+) cells were observed in the NR3A KO and the hyperoxia groups. These results demonstrated that the NR3A gene can promote cell apoptosis and mediate the potential damage in the developing brain induced by exposure to non-physiologically high concentrations of oxygen.
Collapse
Affiliation(s)
- Jimei Li
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China ; Departments of Pathology and Laboratory Medicine, Medical University of South Carolina, SC 29425, USA
| | - Shanping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhongyang Lu
- Departments of Pathology and Laboratory Medicine, Medical University of South Carolina, SC 29425, USA
| | - Osama Mohamad
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ling Wei
- Departments of Pathology and Laboratory Medicine, Medical University of South Carolina, SC 29425, USA ; Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
44
|
Griesmaier E, Keller M. Glutamate receptors — Prenatal insults, long-term consequences. Pharmacol Biochem Behav 2012; 100:835-40. [DOI: 10.1016/j.pbb.2011.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 03/23/2011] [Accepted: 04/15/2011] [Indexed: 10/18/2022]
|
45
|
Rakotomamonjy J, Levenes C, Baulieu EE, Schumacher M, Ghoumari AM. Novel protective effect of mifepristone on detrimental GABAA receptor activity to immature Purkinje neurons. FASEB J 2011; 25:3999-4010. [PMID: 21795502 DOI: 10.1096/fj.11-183384] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immature Purkinje neurons are particularly vulnerable cells. They survive in cerebellar slice cultures under treatment by the synthetic steroid mifepristone (RU486) that depolarizes them at this age. The present study aims at understanding the mechanism underlying this neuroprotective effect. In the developing cerebellum, the role of γ-aminobutyric acid (GABA) in neuron survival is unknown. In 3-d-old mouse cerebellar slice cultures, we show that GABA(A) receptor activation is depolarizing and excitatory. Antagonists of GABA(A) receptors rescue Purkinje neurons, demonstrating that GABA is endogenously released in this preparation and is toxic. Mifepristone likely protects these neurons by reversing GABA(A) receptor-mediated chloride fluxes and reducing their driving force. Neuroprotection by mifepristone is dose-dependently decreased by the agonist of GABA(A) receptors muscimol and by caffeine, an agonist of internal calcium store release. Moreover, the survival induced by neomycin, an inhibitor of calcium release, is partially reversed by muscimol. The p38 mitogen-activated protein kinase (MAPK) inhibitor SB239063 also rescues Purkinje neurons. In summary, we propose that when GABA is depolarizing, mifepristone protects Purkinje neurons by shunting GABA responses and probably chloride fluxes, by inhibiting p38 MAPK activity and likely internal calcium store release. A new and nonhormonal effect of mifepristone is thus revealed.
Collapse
Affiliation(s)
- Jennifer Rakotomamonjy
- Unité Mixte de Recherche 788, Institut National de Santé et de Recherche Médicale (INSERM) and University Paris-Sud 11, Kremlin-Bicêtre, France
| | | | | | | | | |
Collapse
|
46
|
Schaz U, Föhr KJ, Liebau S, Fulda S, Koelch M, Fegert JM, Boeckers TM, Ludolph AG. Dose-dependent modulation of apoptotic processes by fluoxetine in maturing neuronal cells: an in vitro study. World J Biol Psychiatry 2011; 12:89-98. [PMID: 20735156 DOI: 10.3109/15622975.2010.506927] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVES Recent studies indicate that the selective serotonin reuptake inhibitor (SSRI) fluoxetine is not solely effective by the instant inhibition of the serotonin transporter (SERT) but also by its influence on mitotic and/or apoptotic processes. METHODS To investigate the effects of the compound in vitro, we treated neurons from different brain areas with increasing concentrations of fluoxetine. Additionally, human embryonic kidney (HEK-293) cells and HEK-293 cells stably expressing the SERT were used. Cell viability was quantified by MTT-assay and apoptosis via fluorescence-activated cell-sorting analyses. Fluoxetine's effect on the γ-aminobutyric acid (GABA) receptor was electrophysiologically investigated to test the hypothesis if a GABA-mimetic effect exists that might lead - additionally to the well-known N-methyl-D-aspartate (NMDA)-antagonism - to increased apoptosis in immature neurons. RESULTS In hippocampal, cortical, and both types of HEK-293 cells, viability decreased and apoptosis increased in a dose-dependent manner (0.5-75 μM). In contrast, in mesencephalic and striatal cells the viability was unchanged or even slightly stimulated up to 20 μM fluoxetine. An anti-apoptotic effect of concentrations below 10 μM was observed in these cells. The GABA(A) receptor was directly activated by fluoxetine. CONCLUSIONS We conclude that fluoxetine affects apoptotic processes independently from SERT expression. Since especially the combined GABA-mimetic and NMDA-antagonistic effects increase apoptosis in developing neuronal cells, whereas both effects are neuroprotective in adult neurons we hypothesise that these mechanisms explain the discrepancy of in vitro and in vivo studies.
Collapse
Affiliation(s)
- Ulrike Schaz
- Institute of Anatomy and Cell Biology, University of Ulm, Ulm, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhou ZW, Shu Y, Li M, Guo X, Pac-Soo C, Maze M, Ma D. The glutaminergic, GABAergic, dopaminergic but not cholinergic neurons are susceptible to anaesthesia-induced cell death in the rat developing brain. Neuroscience 2010; 174:64-70. [PMID: 21056635 DOI: 10.1016/j.neuroscience.2010.10.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Revised: 09/10/2010] [Accepted: 10/02/2010] [Indexed: 11/28/2022]
Abstract
Neuronal cell death induced by anaesthetics in the developing brain was evident in previous pre-clinical studies. However, the neuronal cell types involved in anaesthesia-induced neuronal cell death remains elusive. The aim of this study was to investigate glutamatergic, GABAergic, cholinergic and dopaminergic neuronal cell apoptosis induced by anaesthetic exposure in specific brain regions in rats. Separate cohorts of 7-day-old Sprague Dawley (SD) rat pups were randomly assigned to two groups: Naive and anaesthetics alone (70% nitrous oxide and 0.75% isoflurane exposure for 6 h). The brains were sectioned and the slices that contained the basal forebrain, substantia nigra, cornu ammonis area 1 (CA1) subarea of hippocampus or cingulate cortex were selected and subsequently subjected to double-labelled fluorescent immunohistochemistry for choline acetyltransferase, dopamine, vesicular glutamate transporter 1 (vGLUT1) or glutamic acid decarboxylase 67 (GAD67) together with caspase 3, respectively. Compared to the naive control, anaesthetic exposure significantly increased the number of caspase-3 positive cells in the CA1 subarea of hippocampus, cingulate cortex, and substantia nigra, but not in the basal forebrain. 54% and 14% of apoptotic cells in the CA1 subarea of hippocampus were GABAergic and glutamatergic neurons respectively. In the cingulate cortex, 30% and 37% of apoptotic cells were GABAergic and glutamatergic neurons respectively. In the substantia nigra, 22% of apoptotic cells were dopaminergic neurons. Our data suggests, anaesthetic exposure significantly increases neuroapoptosis of glutamatergic, GABAergic and dopaminergic neurons in the developing brain but not that of the cholinergic neurons in the basal forebrain.
Collapse
Affiliation(s)
- Z-W Zhou
- Section of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea and Westminster Hospital, London SW10 9NH, UK
| | | | | | | | | | | | | |
Collapse
|
48
|
O'Driscoll C, Bressler JP. Hairless expression attenuates apoptosis in a mouse model and the COS cell line; involvement of p53. PLoS One 2010; 5:e12911. [PMID: 20886113 PMCID: PMC2944824 DOI: 10.1371/journal.pone.0012911] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Accepted: 08/04/2010] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Neurons are more likely to die through apoptosis in the immature brain after injury whereas adult neurons in the mature brain die by necrosis. Several studies have suggested that this maturational change in the mechanism of cell death is regulated, in part, by thyroid hormone. We examined the involvement of the hairless (Hr) gene which has been suspected of having a role in cell cycle regulation and apoptosis in the hair follicle and is strongly regulated by the thyroid hormone in the brain. METHODOLOGY Forced expression of Hr by transfection decreased the number of apoptotic nuclei, levels of caspase-3 activity, and cytosolic cytochrome C in COS cells exposed to staurosporine and tunicamycin. Similarly, caspase-3 activity was lower and the decrease in mitochondrial membrane potential was smaller in cultures of adult cerebellar granule neurons from wild type mice compared to Hr knockout mice induced to undergo apoptosis. In vivo, apoptosis as detected by positive TUNEL labeling and caspase 3 activity was lower in wild-type mice compared to Hr knockouts after exposure to trimethyltin. Hr expression lowered levels of p53, p53 mediated reporter gene activity, and lower levels of the pro-apoptotic Bcl2 family member Bax in COS cells. Finally, Hr expression did not attenuate apoptosis in mouse embryonic fibroblasts from p53 knockout mice but was effective in mouse embryonic fibroblasts from wild type mice. CONCLUSIONS/SIGNIFICANCE Overall, our studies demonstrate that Hr evokes an anti-apoptotic response by repressing expression of p53 and pro-apoptotic events regulated by p53.
Collapse
Affiliation(s)
- Cliona O'Driscoll
- Division of Toxicology, Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, and Hugo Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States of America
| | - Joseph P. Bressler
- Division of Toxicology, Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University, and Hugo Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
49
|
Progressive NKCC1-dependent neuronal chloride accumulation during neonatal seizures. J Neurosci 2010; 30:11745-61. [PMID: 20810895 DOI: 10.1523/jneurosci.1769-10.2010] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Seizures induce excitatory shifts in the reversal potential for GABA(A)-receptor-mediated responses, which may contribute to the intractability of electro-encephalographic seizures and preclude the efficacy of widely used GABAergic anticonvulsants such as phenobarbital. We now report that, in intact hippocampi prepared from neonatal rats and transgenic mice expressing Clomeleon, recurrent seizures progressively increase the intracellular chloride concentration ([Cl(-)](i)) assayed by Clomeleon imaging and invert the net effect of GABA(A) receptor activation from inhibition to excitation assayed by the frequency of action potentials and intracellular Ca(2+) transients. These changes correlate with increasing frequency of seizure-like events and reduction in phenobarbital efficacy. The Na(+)-K(+)-2Cl(-) (NKCC1) cotransporter blocker bumetanide inhibited seizure-induced neuronal Cl(-) accumulation and the consequent facilitation of recurrent seizures. Our results demonstrate a novel mechanism by which seizure activity leads to [Cl(-)](i) accumulation, thereby increasing the probability of subsequent seizures. This provides a potential mechanism for the early crescendo phase of neonatal seizures.
Collapse
|
50
|
Archer T. Effects of exogenous agents on brain development: stress, abuse and therapeutic compounds. CNS Neurosci Ther 2010; 17:470-89. [PMID: 20553311 DOI: 10.1111/j.1755-5949.2010.00171.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The range of exogenous agents likely to affect, generally detrimentally, the normal development of the brain and central nervous system defies estimation although the amount of accumulated evidence is enormous. The present review is limited to certain types of chemotherapeutic and "use-and-abuse" compounds and environmental agents, exemplified by anesthetic, antiepileptic, sleep-inducing and anxiolytic compounds, nicotine and alcohol, and stress as well as agents of infection; each of these agents have been investigated quite extensively and have been shown to contribute to the etiopathogenesis of serious neuropsychiatric disorders. To greater or lesser extent, all of the exogenous agents discussed in the present treatise have been investigated for their influence upon neurodevelopmental processes during the period of the brain growth spurt and during other phases uptill adulthood, thereby maintaining the notion of critical phases for the outcome of treatment whether prenatal, postnatal, or adolescent. Several of these agents have contributed to the developmental disruptions underlying structural and functional brain abnormalities that are observed in the symptom and biomarker profiles of the schizophrenia spectrum disorders and the fetal alcohol spectrum disorders. In each case, the effects of the exogenous agents upon the status of the affected brain, within defined parameters and conditions, is generally permanent and irreversible.
Collapse
Affiliation(s)
- Trevor Archer
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|