1
|
Samaiya PK, Krishnamurthy S, Kumar A. Mitochondrial dysfunction in perinatal asphyxia: role in pathogenesis and potential therapeutic interventions. Mol Cell Biochem 2021; 476:4421-4434. [PMID: 34472002 DOI: 10.1007/s11010-021-04253-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 08/20/2021] [Indexed: 01/13/2023]
Abstract
Perinatal asphyxia (PA)-induced brain injury may present as hypoxic-ischemic encephalopathy in the neonatal period, and long-term sequelae such as spastic motor deficits, intellectual disability, seizure disorders and learning disabilities. The brain injury is secondary to both the hypoxic-ischemic event and oxygenation-reperfusion following resuscitation. Following PA, a time-dependent progression of neuronal insult takes place in terms of transition of cell death from necrosis to apoptosis. This transition is the result of time-dependent progression of pathomechanisms which involve excitotoxicity, oxidative stress, and ultimately mitochondrial dysfunction in developing brain. More precisely mitochondrial respiration is suppressed and calcium signalling is dysregulated. Consequently, Bax-dependent mitochondrial permeabilization occurs leading to release of cytochrome c and activation of caspases leading to transition of cell death in developing brain. The therapeutic window lies within this transition process. At present, therapeutic hypothermia (TH) is the only clinical treatment available for treating moderate as well as severe asphyxia in new-born as it attenuates secondary loss of high-energy phosphates (ATP) (Solevåg et al. in Free Radic Biol Med 142:113-122, 2019; Gunn et al. in Pediatr Res 81:202-209, 2017), improving both short- and long-term outcomes. Mitoprotective therapies can offer a new avenue of intervention alone or in combination with therapeutic hypothermia for babies with birth asphyxia. This review will explore these mitochondrial pathways, and finally will summarize past and current efforts in targeting these pathways after PA, as a means of identifying new avenues of therapeutic intervention.
Collapse
Affiliation(s)
- Puneet K Samaiya
- Department of Pharmacy, Shri G.S. Institute of Technology and Science, Indore, MP, 452003, India.
| | - Sairam Krishnamurthy
- Neurotherapeutics Lab, Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pediatrics, Institute of Medical Sciences, BHU, Varanasi, UP, India
| |
Collapse
|
2
|
Engin A, Engin AB. N-Methyl-D-Aspartate Receptor Signaling-Protein Kinases Crosstalk in Cerebral Ischemia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1275:259-283. [PMID: 33539019 DOI: 10.1007/978-3-030-49844-3_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Although stroke is very often the cause of death worldwide, the burden of ischemic and hemorrhagic stroke varies between regions and over time regarding differences in prognosis, prevalence of risk factors, and treatment strategies. Excitotoxicity, oxidative stress, dysfunction of the blood-brain barrier, neuroinflammation, and lysosomal membrane permeabilization, sequentially lead to the progressive death of neurons. In this process, protein kinases-related checkpoints tightly regulate N-methyl-D-aspartate (NMDA) receptor signaling pathways. One of the major hallmarks of cerebral ischemia is excitotoxicity, characterized by overactivation of glutamate receptors leading to intracellular Ca2+ overload and ultimately neuronal death. Thus, reduced expression of postsynaptic density-95 protein and increased protein S-nitrosylation in neurons is responsible for neuronal vulnerability in cerebral ischemia. In this chapter death-associated protein kinases, cyclin-dependent kinase 5, endoplasmic reticulum stress-induced protein kinases, hyperhomocysteinemia-related NMDA receptor overactivation, ephrin-B-dependent amplification of NMDA-evoked neuronal excitotoxicity and lysosomocentric hypothesis have been discussed.Consequently, ample evidences have demonstrated that enhancing extrasynaptic NMDA receptor activity triggers cell death after stroke. In this context, considering the dual roles of NMDA receptors in both promoting neuronal survival and mediating neuronal damage, selective augmentation of NR2A-containing NMDA receptor activation in the presence of NR2B antagonist may constitute a promising therapy for stroke.
Collapse
Affiliation(s)
- Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| |
Collapse
|
3
|
Ma X, Zhang Y, Zhu D, Chen Z, Xu M, He L, Shi T, Huang L, Zou J. Low dosage chloroquine protects retinal ganglion cells against glutamate-induced cell death. Exp Eye Res 2019; 181:285-293. [PMID: 30831085 DOI: 10.1016/j.exer.2019.02.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 01/17/2019] [Accepted: 02/25/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Xiaoyun Ma
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China.
| | - Yun Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dan Zhu
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Zufeng Chen
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China
| | - Manshan Xu
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Linping He
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Tingli Shi
- Department of Ophthalmology, Guanghua Integrative Medicine Hospital, Shanghai, China
| | - Lvzhen Huang
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Instistute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases College of Optometry, Peking University Health Science Center, China.
| | - Jun Zou
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Seyama T, Kamei Y, Iriyama T, Imada S, Ichinose M, Toshimitsu M, Fujii T, Asou H. Pretreatment with magnesium sulfate attenuates white matter damage by preventing cell death of developing oligodendrocytes. J Obstet Gynaecol Res 2018; 44:601-607. [PMID: 29363221 DOI: 10.1111/jog.13568] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 11/08/2017] [Indexed: 01/30/2023]
Abstract
AIM Antenatal maternal administration of magnesium sulfate (MgSO4 ) reduces cerebral palsy in preterm infants. However, it remains controversial as to whether it also reduces occurrence of white matter damage, or periventricular leukomalacia. We assessed the effect of MgSO4 against white matter damage induced by hypoxic-ischemic insult using a neonatal rat model and culture of premyelinating oligodendrocytes (pre-OL). METHODS Rat pups at postnatal day (P) 6 were administered either MgSO4 or vehicle intraperitoneally before hypoxic-ischemic insult (unilateral ligation of the carotid artery followed by 6% oxygen for 1 h). The population of oligodendrocyte (OL) markers and CD-68-positive microglia at P11, and TdT-mediated biotin-16-dUTP nick-end labeling (TUNEL)-positive cells at P8 were evaluated in pericallosal white matter. Primary cultures of mouse pre-OL were subjected to oxygen glucose deprivation condition, and the lactate dehydrogenase release from culture cells was evaluated to assess cell viability. RESULTS Pretreatment with MgSO4 attenuated the loss of OL markers, such as myelin basic protein and Olig2, in ipsilateral pericallosal white matter and decreased the number of CD-68-positive microglia and TUNEL-positive cells in vivo. Pretreatment with MgSO4 also inhibited lactate dehydrogenase release from pre-OL induced by oxygen glucose deprivation in vitro. CONCLUSION Pretreatment with MgSO4 attenuates white matter damage by preventing cell death of pre-OL.
Collapse
Affiliation(s)
- Takahiro Seyama
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshimasa Kamei
- Department of Obstetrics and Gynecology, Saitama Medical University Hospital, Saitama, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Shinya Imada
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Mari Ichinose
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Masatake Toshimitsu
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Tokyo, Japan
| | - Hiroaki Asou
- Center for Kampo Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Tanshinone IIA Inhibits Glutamate-Induced Oxidative Toxicity through Prevention of Mitochondrial Dysfunction and Suppression of MAPK Activation in SH-SY5Y Human Neuroblastoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4517486. [PMID: 28690763 PMCID: PMC5485345 DOI: 10.1155/2017/4517486] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/17/2017] [Accepted: 05/02/2017] [Indexed: 01/27/2023]
Abstract
Glutamate excitotoxicity is associated with many neurological diseases, including cerebral ischemia and neurodegenerative diseases. Tanshinone IIA, a diterpenoid naphthoquinone from Salvia miltiorrhiza, has been shown to suppress presynaptic glutamate release, but its protective mechanism against glutamate-induced neurotoxicity is lacking. Using SH-SY5Y human neuroblastoma cells, we show here that excessive glutamate exposure decreases cell viability and proliferation and increases LDH release. Pretreatment with tanshinone IIA, however, prevents the decrease in cell viability and proliferation and the increase in LDH release induced by glutamate. Tanshinone IIA also attenuates glutamate-induced oxidative stress by reducing reactive oxygen species level and malondialdehyde and protein carbonyl contents and by enhancing activities and protein levels of superoxide dismutase and catalase. We then show that tanshinone IIA prevents glutamate-induced mitochondrial dysfunction by increasing mitochondrial membrane potential and ATP content and by reducing mitochondrial protein carbonyl content. Moreover, tanshinone IIA can inhibit glutamate-induced apoptosis through regulation of apoptosis-related protein expression and MAPK activation, including elevation of Bcl-2 protein level, decrease in Bax and cleaved caspase-3 levels, and suppression of JNK and p38 MAPK activation. Collectively, our findings demonstrate that tanshinone IIA protects SH-SY5Y cells against glutamate toxicity by reducing oxidative stress and regulating apoptosis and MAPK pathways.
Collapse
|
6
|
Samaiya PK, Narayan G, Kumar A, Krishnamurthy S. Tempol (4 hydroxy-tempo) inhibits anoxia-induced progression of mitochondrial dysfunction and associated neurobehavioral impairment in neonatal rats. J Neurol Sci 2017; 375:58-67. [DOI: 10.1016/j.jns.2017.01.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/20/2016] [Accepted: 01/06/2017] [Indexed: 10/20/2022]
|
7
|
D'Orsi B, Mateyka J, Prehn JHM. Control of mitochondrial physiology and cell death by the Bcl-2 family proteins Bax and Bok. Neurochem Int 2017; 109:162-170. [PMID: 28315370 DOI: 10.1016/j.neuint.2017.03.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/07/2017] [Accepted: 03/09/2017] [Indexed: 01/14/2023]
Abstract
Neuronal cell death is often triggered by events that involve intracellular increases in Ca2+. Under resting conditions, the intracellular Ca2+ concentration is tightly controlled by a number of extrusion and sequestering mechanisms involving the plasma membrane, mitochondria, and ER. These mechanisms act to prevent a disruption of neuronal ion homeostasis. As these processes require ATP, excessive Ca2+ overloading may cause energy depletion, mitochondrial dysfunction, and may eventually lead to Ca2+-dependent cell death. Excessive Ca2+ entry though glutamate receptors (excitotoxicity) has been implicated in several neurologic and chronic neurodegenerative diseases, including ischemic stroke, epilepsy, and Alzheimer's disease. Recent evidence has revealed that excitotoxic cell death is regulated by the B-cell lymphoma-2 (Bcl-2) family of proteins. Bcl-2 proteins, comprising of both pro-apoptotic and anti-apoptotic members, have been shown to not only mediate the intrinsic apoptosis pathway by controlling mitochondrial outer membrane (MOM) integrity, but to also control neuronal Ca2+ homeostasis and energetics. In this review, the role of Bcl-2 family proteins in the regulation of apoptosis, their expression in the central nervous system and how they control Ca2+-dependent neuronal injury are summarized. We review the current knowledge on Bcl-2 family proteins in the regulation of mitochondrial function and bioenergetics, including the fusion and fission machinery, and their role in Ca2+ homeostasis regulation at the mitochondria and ER. Specifically, we discuss how the 'pro-apoptotic' Bcl-2 family proteins, Bax and Bok, physiologically expressed in the nervous system, regulate such 'non-apoptotic/daytime' functions.
Collapse
Affiliation(s)
- Beatrice D'Orsi
- Department of Physiology & Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Julia Mateyka
- Department of Physiology & Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, Centre for the Study of Neurological Disorders, Royal College of Surgeons in Ireland, Dublin 2, Ireland.
| |
Collapse
|
8
|
Glutamate input in the dorsal raphe nucleus as a determinant of escalated aggression in male mice. J Neurosci 2015; 35:6452-63. [PMID: 25904796 DOI: 10.1523/jneurosci.2450-14.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although the dorsal raphe nucleus (DRN) has long been linked to neural control of aggression, little is known about the regulatory influences of the DRN when an animal engages in either adaptive species-typical aggressive behavior or escalated aggression. Therefore it is important to explore which neurotransmitter inputs into the DRN determine the escalation of aggression in male mice. Previously, we observed that microinjection of the GABAB receptor agonist baclofen into the DRN escalates aggressive behavior in male mice. Here, we used a serotonin (5-HT) neuron-specific GABAB receptor knock-out mouse to demonstrate that baclofen acts on nonserotonergic neurons to escalate aggression. Intra-DRN baclofen administration increased glutamate release, but did not alter GABA release, within the DRN. Microinjection of l-glutamate into the DRN escalated dose-dependently attack bites toward an intruder. In vivo microdialysis showed that glutamate release increased in the DRN during an aggressive encounter, and the level of glutamate was further increased when the animal was engaged in escalated aggressive behavior after social instigation. Finally, 5-HT release was increased within the DRN and also in the medial prefrontal cortex when animals were provoked by social instigation, and during escalated aggression after social instigation, but this increase in 5-HT release was not observed when animals were engaged in species-typical aggression. In summary, glutamate input into the DRN is enhanced during escalated aggression, which causes a phasic increase of 5-HT release from the DRN 5-HT neurons.
Collapse
|
9
|
Liu J, Li J, Yang Y, Wang X, Zhang Z, Zhang L. Neuronal apoptosis in cerebral ischemia/reperfusion area following electrical stimulation of fastigial nucleus. Neural Regen Res 2014; 9:727-34. [PMID: 25206880 PMCID: PMC4146268 DOI: 10.4103/1673-5374.131577] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2014] [Indexed: 11/26/2022] Open
Abstract
Previous studies have indicated that electrical stimulation of the cerebellar fastigial nucleus in rats may reduce brain infarct size, increase the expression of Ku70 in cerebral ischemia/reperfusion area, and decrease the number of apoptotic neurons. However, the anti-apoptotic mechanism of Ku70 remains unclear. In this study, fastigial nucleus stimulation was given to rats 24, 48, and 72 hours before cerebral ischemia/reperfusion injury. Results from the electrical stimulation group revealed that rats exhibited a reduction in brain infarct size, a significant increase in the expression of Ku70 in cerebral ischemia/reperfusion regions, and a decreased number of terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells. Double immunofluorescence staining revealed no co-localization of Ku70 with TUNEL-positive cells. However, Ku70 partly co-localized with Bax protein in the cytoplasm of rats with cerebral ischemia/reperfusion injury. These findings suggest an involvement of Ku70 with Bax in the cytoplasm of rats exposed to electrical stimulation of the cerebellar fastigial nucleus, and may thus provide an understanding into the anti-apoptotic activity of Ku70 in cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jingli Liu
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jingpin Li
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yi Yang
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Xiaoling Wang
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zhaoxia Zhang
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Lei Zhang
- Department of Neurology, the First Hospital Affiliated to Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
10
|
Mousa AM, Al-Fadhli AS, Rao MS, Kilarkaje N. Gestational lead exposure induces developmental abnormalities and up-regulates apoptosis of fetal cerebellar cells in rats. Drug Chem Toxicol 2014; 38:73-83. [PMID: 24724870 DOI: 10.3109/01480545.2014.907578] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Lead (Pb), a known environmental toxicant, adversely affects almost all organ systems. In this study, we investigated the effects of maternal lead exposure on fetal rat cerebellum. Female Sprague-Dawley rats were given lead nitrate in drinking water (0, 0.5, and 1%) for two weeks before conception, and during pregnancy. Fetuses were collected by caesarian section on gestational day 21 and observed for developmental abnormalities. The fetal cerebellar sections from control and 1% lead group were stained with cresyl violet. Immunohistochemical expressions of p53, Bax, Bcl-2, and caspase 3 were quantified by AnalySIS image analyzer (Life Science, Germany). Lead exposure induced developmental abnormalities of eyes, ear, limbs, neck and ventral abdominal wall; however, these abnormalities were commonly seen in the 1% lead-treated group. In addition, lead also caused fetal mortality and reduced body growth in both dose groups and reduced brain weight in the 1% lead-treated group. The fetal cerebella from the 1% lead-treated group showed unorganized cerebellar cortical layers, and degenerative changes in granule and Purkinje cells such as the formation of clumps of Nissl granules. An increase in Bax and caspase 3, and a decrease in Bcl-2 (p < 0.05), but not in p53, showed apoptosis of the neurons. In conclusion, gestational lead exposure in rats induces fetal toxicity and developmental abnormalities. The lead exposure also impairs development of cerebellar layers, induces structural changes, and apoptosis in the fetal cerebellar cortex. These results suggest that lead exposure during gestation is extremely toxic to developing cerebellum in rats.
Collapse
Affiliation(s)
- Alyaa M Mousa
- Department of Anatomy, Faculty of Medicine, Kuwait University , Kuwait
| | | | | | | |
Collapse
|
11
|
Gu Q, Zhai L, Feng X, Chen J, Miao Z, Ren L, Qian X, Yu J, Li Y, Xu X, Liu CF. Apelin-36, a potent peptide, protects against ischemic brain injury by activating the PI3K/Akt pathway. Neurochem Int 2013; 63:535-40. [DOI: 10.1016/j.neuint.2013.09.017] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 08/17/2013] [Accepted: 09/22/2013] [Indexed: 01/08/2023]
|
12
|
Gliyazova NS, Huh EY, Ibeanu GC. A novel phenoxy thiophene sulphonamide molecule protects against glutamate evoked oxidative injury in a neuronal cell model. BMC Neurosci 2013; 14:93. [PMID: 24004478 PMCID: PMC3846642 DOI: 10.1186/1471-2202-14-93] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 08/29/2013] [Indexed: 11/13/2022] Open
Abstract
Background Glutamate is one of the major neurotransmitters in the central nervous system. It is a potent neurotoxin capable of neuronal destruction through numerous signal pathways when present in high concentration. Glutamate-evoked excitotoxicity has been implicated in the etiology of many neurodegenerative diseases including Alzheimer’s disease (AD), Parkinson’s disease (PD), and ischemic stroke. Increasing evidence has shown that reactive oxygen species (ROS) provoked by glutamate-linked oxidative stress plays a crucial role in the pathogenesis of these disorders. We previously reported the discovery of an aryl thiophene compound, 4-chloro-N-(naphthalen-1-ylmethyl)-5-(3-(piperazin-1-yl)phenoxy)thiophene-2-sulfonamide (B355252) from a proprietary library of small molecules. We showed that this compound was capable of potentiating nerve growth factor (NGF)-primed neurite outgrowth in neuronal cell models in a low NGF environment. In the present study we investigated the neuroprotective effects and signaling pathways of B355252 on glutamate-evoked excitotoxicity in HT-22, a murine hippocampal neuronal cell line. Results Glutamate significantly decreased HT-22 neuronal cell viability in a concentration-dependent manner as measured by the MTT assay. Co-treatment with 2, 4, and 8 μM B355252 protected against cell death caused by glutamate-induced toxicity by 9.1% (p<0.01), 26.0% (p<0.001), and 61.9% (p<0.001) respectively, compared to glutamate-treated control group. B355252 at a concentration of 8 μM fully rescued HT-22 from the neurototoxic effects of glutamate, and by itself increased cell viability by 16% (p<0.001) above untreated control. Glutamate enhanced reduction in glutathione (GSH) synthesis was reversed by 15% (p<0.01) in the presence of B355252. B355252 reduced the expression of apoptosis inducing factor (AIF) by 27%, while the proapoptotic Bcl-2 associated X protein (Bax) was strongly attenuated 3-fold. Glutamate-evoked increase in intracellular calcium (Ca2+) load and subsequent ROS production was inhibited by 71% (p<0.001) and 40% (p<0.001) respectively, to comparable level as untreated control in the presence of B355252. Glutamate significantly upregulated the phosphorylation of extracellular signal regulated kinase Erk1/2 (pERK1/2), while decreasing Erk3. In contrast, B355252 potently attenuated the glutamate-dependent activation of Erk1/2 and robustly increased the level of ERK3 in HT-22. Conclusions A novel phenoxy thiophene small molecule, B355252, suppresses glutamate-evoked oxidative stress in HT-22 neurons by blocking Ca2+ and ROS production, and altering the expression or phosphorylation states of Erk kinases. This molecule previously reported to enhance neurite outgrowth in the presence of sub-physiological concentrations of NGF appears to be a promising drug candidate for development as a potential therapeutic and neuroprotective agent for various neurodegenerative disorders.
Collapse
Affiliation(s)
- Nailya S Gliyazova
- BRITE, North Carolina Central University, 1801 Fayetteville Street, Durham, NC 27707, USA.
| | | | | |
Collapse
|
13
|
Lukanowska M, Howl J, Jones S. Bioportides: Bioactive cell-penetrating peptides that modulate cellular dynamics. Biotechnol J 2013; 8:918-30. [DOI: 10.1002/biot.201200335] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Revised: 06/10/2013] [Accepted: 06/21/2013] [Indexed: 11/10/2022]
|
14
|
Heimfarth L, Loureiro SO, Dutra MF, Petenuzzo L, de Lima BO, Fernandes CG, da Rocha JBT, Pessoa-Pureur R. Disrupted cytoskeletal homeostasis, astrogliosis and apoptotic cell death in the cerebellum of preweaning rats injected with diphenyl ditelluride. Neurotoxicology 2013. [DOI: 10.1016/j.neuro.2012.10.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
15
|
Hirobe T, Terunuma E. Reduced Proliferative and Differentiative Activity of Mouse Pink-Eyed Dilution Melanoblasts is Related to Apoptosis. Zoolog Sci 2012; 29:725-32. [DOI: 10.2108/zsj.29.725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Post-treatment of Bax-inhibiting peptide reduces neuronal death and behavioral deficits following global cerebral ischemia. Neurochem Int 2011; 58:224-33. [DOI: 10.1016/j.neuint.2010.12.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Revised: 11/15/2010] [Accepted: 12/01/2010] [Indexed: 01/27/2023]
|
17
|
Gomez J, Matsuyama S. Cell-penetrating penta-peptides and Bax-inhibiting peptides: protocol for their application. Methods Mol Biol 2011; 683:465-71. [PMID: 21053150 PMCID: PMC3703514 DOI: 10.1007/978-1-60761-919-2_33] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The first series of cell-penetrating penta-peptides (CPP5s) were discovered as cytoprotective penta-peptides designed from the Bax-inhibiting domain of Ku70. Bax is an inducer of programmed cell death, and Ku70 is a multifunctional protein maintaining genomic stability and protecting cells from death by inhibiting the cytotoxic activity of Bax. Since these peptides bind and inhibit Bax, they are named Bax-inhibiting peptides (BIPs). The second series of CPP5s were developed by mutating BIP's amino acid sequences to abolish the Bax-binding activity. These peptides were used as negative control peptides to evaluate the Bax-inhibiting activity of BIPs. CPP5s are able to enter cells when they are added to the culture medium. The mechanism of cell entry of CPP5s is not yet understood. Numerous studies showed that BIP rescued cells from cytotoxic stresses both in cell culture and animal model, suggesting the therapeutic potential of BIP. Both BIPs and noncytoprotective CPP5s did not show significant toxicity even at 1.6 mM concentration in cell culture. Our recent study suggests that CPP5s has the protein transduction activity, though only green fluorescent protein (GFP) has been tested as a cargo protein. If CPP5s can deliver wide range of cargo molecules into the cell, CPP5s may be utilized as nontoxic drug delivery tool. In this article, we describe our laboratory's protocols of how to synthesize, store, and apply CPP5s for the examination of their activities of cell penetration and cytoprotection.
Collapse
Affiliation(s)
| | - Shigemi Matsuyama
- Correspondence should be addressed to Shigemi Matsuyama TEL: 216-368-5832 (USA),
| |
Collapse
|
18
|
Xu B, Xu ZF, Deng Y. Protective effects of MK-801 on manganese-induced glutamate metabolism disorder in rat striatum. ACTA ACUST UNITED AC 2010; 62:381-90. [DOI: 10.1016/j.etp.2009.05.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2009] [Revised: 05/17/2009] [Accepted: 05/19/2009] [Indexed: 12/11/2022]
|
19
|
Xu B, Xu ZF, Deng Y. Manganese exposure alters the expression ofN-methyl-d-aspartate receptor subunit mRNAs and proteins in rat striatum. J Biochem Mol Toxicol 2010; 24:1-9. [DOI: 10.1002/jbt.20306] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
20
|
Vishnudas VK, Miller JB. Ku70 regulates Bax-mediated pathogenesis in laminin-alpha2-deficient human muscle cells and mouse models of congenital muscular dystrophy. Hum Mol Genet 2009; 18:4467-77. [PMID: 19692349 DOI: 10.1093/hmg/ddp399] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The severely debilitating disease Congenital Muscular Dystrophy Type 1A (MDC1A) is caused by mutations in the gene encoding laminin-alpha2. Bax-mediated muscle cell death is a significant contributor to the severe neuromuscular pathology seen in the Lama2-null mouse model of MDC1A. To extend our understanding of pathogenesis due to laminin-alpha2-deficiency, we have now analyzed molecular mechanisms of Bax regulation in normal and laminin-alpha2-deficient muscles and cells, including myogenic cells obtained from patients with a clinical diagnosis of MDC1A. In mouse myogenic cells, we found that, as in non-muscle cells, Bax co-immunoprecipitated with the multifunctional protein Ku70. In addition, cell permeable pentapeptides designed from Ku70, termed Bax-inhibiting peptides (BIPs), inhibited staurosporine-induced Bax translocation and cell death in mouse myogenic cells. We also found that acetylation of Ku70, which can inhibit binding to Bax and can be an indicator of increased susceptibility to cell death, was more abundant in Lama2-null than in normal mouse muscles. Furthermore, myotubes formed in culture from human laminin-alpha2-deficient patient myoblasts produced high levels of activated caspase-3 when grown on poly-L-lysine, but not when grown on a laminin-alpha2-containing substrate or when treated with BIPs. Finally, cytoplasmic Ku70 in human laminin-alpha2-deficient myotubes was both reduced in amount and more highly acetylated than in normal myotubes. Increased susceptibility to cell death thus appears to be an intrinsic property of human laminin-alpha2-deficient myotubes. These results identify Ku70 as a regulator of Bax-mediated pathogenesis and a therapeutic target in laminin-alpha2-deficiency.
Collapse
Affiliation(s)
- Vivek K Vishnudas
- Neuromuscular Biology & Disease Group, Boston Biomedical Research Institute, 64 Grove Street, Watertown, MA 02478, USA
| | | |
Collapse
|
21
|
Robertson CL, Scafidi S, McKenna MC, Fiskum G. Mitochondrial mechanisms of cell death and neuroprotection in pediatric ischemic and traumatic brain injury. Exp Neurol 2009; 218:371-80. [PMID: 19427308 PMCID: PMC3096876 DOI: 10.1016/j.expneurol.2009.04.030] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Revised: 04/23/2009] [Accepted: 04/28/2009] [Indexed: 11/30/2022]
Abstract
There are several forms of acute pediatric brain injury, including neonatal asphyxia, pediatric cardiac arrest with global ischemia, and head trauma, that result in devastating, lifelong neurologic impairment. The only clinical intervention that appears neuroprotective is hypothermia initiated soon after the initial injury. Evidence indicates that oxidative stress, mitochondrial dysfunction, and impaired cerebral energy metabolism contribute to the brain cell death that is responsible for much of the poor neurologic outcome from these events. Recent results obtained from both in vitro and animal models of neuronal death in the immature brain point toward several molecular mechanisms that are either induced or promoted by oxidative modification of macromolecules, including consumption of cytosolic and mitochondrial NAD(+) by poly-ADP ribose polymerase, opening of the mitochondrial inner membrane permeability transition pore, and inactivation of key, rate-limiting metabolic enzymes, e.g., the pyruvate dehydrogenase complex. In addition, the relative abundance of pro-apoptotic proteins in immature brains and neurons, and particularly within their mitochondria, predisposes these cells to the intrinsic, mitochondrial pathway of apoptosis, mediated by Bax- or Bak-triggered release of proteins into the cytosol through the mitochondrial outer membrane. Based on these pathways of cell dysfunction and death, several approaches toward neuroprotection are being investigated that show promise toward clinical translation. These strategies include minimizing oxidative stress by avoiding unnecessary hyperoxia, promoting aerobic energy metabolism by repletion of NAD(+) and by providing alternative oxidative fuels, e.g., ketone bodies, directly interfering with apoptotic pathways at the mitochondrial level, and pharmacologic induction of antioxidant and anti-inflammatory gene expression.
Collapse
Affiliation(s)
- Courtney L. Robertson
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore MD, USA
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore MD, USA
| | - Susanna Scafidi
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore MD, USA
| | - Mary C. McKenna
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore MD, USA
| | - Gary Fiskum
- Department of Anesthesiology, University of Maryland School of Medicine, Baltimore MD, USA
- Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore MD, USA
| |
Collapse
|