1
|
Choi YN, Seo TW, Lee YT, Jeong DH, Yoo SJ. Nuclear endonuclease G controls cell proliferation in ovarian cancer. FEBS Open Bio 2023; 13:655-669. [PMID: 36734593 PMCID: PMC10068316 DOI: 10.1002/2211-5463.13572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/22/2022] [Accepted: 02/02/2023] [Indexed: 02/04/2023] Open
Abstract
Ovarian cancer is characterized by a high degree of genetic heterogeneity. Platinum-based chemotherapy and some gene-targeted therapies have shown limited treatment efficacy due to toxicity and recurrence, and thus, it is essential to identify additional therapeutic targets based on an understanding of the pathological mechanism. Here, we report that endonuclease G, which exhibits altered expression in ovarian cancer, does not function as a cell death effector that digests chromosomal DNA in ovarian cancer. Endonuclease G is modulated by intracellular reactive oxygen species dynamics and plays a role in cell proliferation in ovarian cancer, suggesting that targeting endonuclease G alone or in combination with other antitumor agents may have the potential for development into a treatment for endonuclease G-overexpressing cancers, including ovarian cancer.
Collapse
Affiliation(s)
- Ye Na Choi
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Tae Woong Seo
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Yui Taek Lee
- Department of Biology, Kyung Hee University, Seoul, South Korea
| | - Dar Heum Jeong
- Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Soon Ji Yoo
- Department of Biology, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
2
|
Yesilot S, Bayram D, Özgöçmen M, Toğay VA. Apoptotic effects of Phlomis armeniaca mediated biosynthesized silver nanoparticles in monolayer (2D) and spheroid (3D) cultures of human breast cancer cell lines. 3 Biotech 2023; 13:4. [PMID: 36514484 PMCID: PMC9741690 DOI: 10.1007/s13205-022-03417-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 11/26/2022] [Indexed: 12/14/2022] Open
Abstract
The purpose of current research was to assess the apoptotic effects of biofabrication silver nanoparticles (AgNPs) mediated by the aqueous extract of Phlomis armeniaca on human breast cancer cells (MCF-7 and MDA-MB-231) in monolayer (2D) and spheroid (3D) cultures. The biosynthesized AgNPs were characterized by UV-Vis spectrophotometer (the peaks of resonances at 432 nm), scanning electron microscopy (SEM) and energy-dispersive X-ray spectrometry (EDS). 1-20 µM/mL AgNPs were applied to MCF-7 and MDA-MB-231 cell lines to determine IC50 values at 24, 48 and 72nd h and were found to be 10 µM/mL for both cell lines. Immunohistochemical staining results of BrdU, TUNEL, caspase-3 and Endo G in both 2D and 3D cultures and gene expression levels of caspases (caspase-3, -8 and -9) and Endo G were evaluated. Moreover, the total oxidant/antioxidant status (TOS-TAS) due to AgNPs application in both cell culture mediums was evaluated. AgNPs treatment results in both cell lines in both 2D and 3D cultures showed a significant decrease in the BrdU labeling index, while large amounts of cells were labelled with TUNEL and Endo G. In 2D culture, Endo G expression increased in MCF-7 cells at 48 and 72nd hours, while it increased significantly in MDA-MB-231 cells at all hours. OSI results show that ROS production is increased in cell medium treated with AgNPs. In conclusion, AgNPs mediated by Phlomis armeniaca, synthesized by a green method, successfully induced damage to mitochondria, resulting in cell cycle arrest and consequent cell proliferation blockade and death in both MCF-7 and MDA-MB-231 cells.
Collapse
Affiliation(s)
- Sukriye Yesilot
- Department of Health and Biomedical Sciences, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
- Department of Nursing, Bucak School of Health, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Dilek Bayram
- Department of Histology and Embryology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Meltem Özgöçmen
- Department of Histology and Embryology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Vehbi Atahan Toğay
- Department of Medical Biology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| |
Collapse
|
3
|
Jackson TC, Dezfulian C, Vagni VA, Stezoski J, Janesko-Feldman K, Kochanek PM. PHLPP Inhibitor NSC74429 Is Neuroprotective in Rodent Models of Cardiac Arrest and Traumatic Brain Injury. Biomolecules 2022; 12:1352. [PMID: 36291561 PMCID: PMC9599532 DOI: 10.3390/biom12101352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 09/07/2024] Open
Abstract
Pleckstrin homology domain and leucine rich repeat protein phosphatase (PHLPP) knockout mice have improved outcomes after a stroke, traumatic brain injury (TBI), and decreased maladaptive vascular remodeling following vascular injury. Thus, small-molecule PHLPP inhibitors have the potential to improve neurological outcomes in a variety of conditions. There is a paucity of data on the efficacy of the known experimental PHLPP inhibitors, and not all may be suited for targeting acute brain injury. Here, we assessed several PHLPP inhibitors not previously explored for neuroprotection (NSC13378, NSC25247, and NSC74429) that had favorable predicted chemistries for targeting the central nervous system (CNS). Neuronal culture studies in staurosporine (apoptosis), glutamate (excitotoxicity), and hydrogen peroxide (necrosis/oxidative stress) revealed that NSC74429 at micromolar concentrations was the most neuroprotective. Subsequent testing in a rat model of asphyxial cardiac arrest, and in a mouse model of severe TBI, showed that serial dosing of 1 mg/kg of NSC74429 over 3 days improved hippocampal survival in both models. Taken together, NSC74429 is neuroprotective across multiple insult mechanisms. Future pharmacokinetic and pharmacodynamic (PK/PD) studies are warranted to optimize dosing, and mechanistic studies are needed to determine the percentage of neuroprotection mediated by PHLPP1/2 inhibition, or potentially from the modulation of PHLPP-independent targets.
Collapse
Affiliation(s)
- Travis C. Jackson
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs BLDV, Tampa, FL 33612, USA
- USF Health Heart Institute, Morsani College of Medicine, University of South Florida, 560 Channelside Dr, Tampa, FL 33602, USA
| | - Cameron Dezfulian
- Safar Center for Resuscitation Research, UPMC Children’s Hospital of Pittsburgh, Rangos Research Center—6th Floor, Pittsburgh, PA 15224, USA
- Department of Pediatrics, Baylor College of Medicine, 6651 Main Street, Houston, TX 77030, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Vincent A. Vagni
- Safar Center for Resuscitation Research, UPMC Children’s Hospital of Pittsburgh, Rangos Research Center—6th Floor, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Jason Stezoski
- Safar Center for Resuscitation Research, UPMC Children’s Hospital of Pittsburgh, Rangos Research Center—6th Floor, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, UPMC Children’s Hospital of Pittsburgh, Rangos Research Center—6th Floor, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, UPMC Children’s Hospital of Pittsburgh, Rangos Research Center—6th Floor, Pittsburgh, PA 15224, USA
- Department of Critical Care Medicine, University of Pittsburgh School of Medicine, 4401 Penn Avenue, Pittsburgh, PA 15224, USA
| |
Collapse
|
4
|
VDAC1 regulates neuronal cell loss after retinal trauma injury by a mitochondria-independent pathway. Cell Death Dis 2022; 13:393. [PMID: 35449127 PMCID: PMC9023530 DOI: 10.1038/s41419-022-04755-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 12/23/2022]
Abstract
The voltage-dependent anion channel 1 (VDAC1) was first described as a mitochondrial porin that mediates the flux of metabolites and ions, thereby integrating both cell survival and death signals. In the nervous system, the functional roles of VDAC1 remain poorly understood. Herein, the rat retina was employed to study VDAC1. First, it was observed that even subtle changes in VDAC1 levels affect neuronal survival, inducing severe alterations in the retinal morphology. We next examined the regulation of VDAC1 after traumatic retinal injury. After mechanical trauma, SOD1 translocates towards the nucleus, which is insufficient to contain the consequences of oxidative stress, as determined by the evaluation of protein carbonylation. Using in vitro models of oxidative stress and mechanical injury in primary retinal cell cultures, it was possible to determine that inhibition of VDAC1 oligomerization by 4'-diisothiocyano-2,2'-disulfonic acid stilbene (DIDS) rescues cell viability, impacting microglial cell activation. We next focused on the regulation of VDAC1 after retinal mechanical injury. VDAC1 was promptly upregulated 2 h after lesion in the plasma membrane and endoplasmic reticulum rather than in the mitochondria, and multimers of VDAC1 were assembled after lesion. DIDS intraocular application decreased apoptosis and prevented microglial polarization, which confirmed in vitro observations. Considering the role of microglia in neuroinflammation, multiplex evaluation of cytokines showed that DIDS application disorganized the inflammatory response 2 h after the lesion, matching the fast regulation of VDAC1. Taken together, data disclosed that fine regulation of VDAC1 influences neuronal survival, and pharmacological inhibition after trauma injury has neuroprotective effects. This protection may be attributed to the effects on VDAC1 abnormal accumulation in the plasma membrane, thereby controlling the activation of microglial cells. We concluded that VDAC1 is a putative therapeutic target in neuronal disorders since it integrates both death and survival cellular signaling.
Collapse
|
5
|
Guo X, Ma L, Li H, Qi X, Wei Y, Duan Z, Xu J, Wang C, You C, Tian M. Brainstem iron overload and injury in a rat model of brainstem hemorrhage. J Stroke Cerebrovasc Dis 2020; 29:104956. [PMID: 32689646 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104956] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/27/2020] [Accepted: 05/11/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Brainstem hemorrhage (BSH) is the most devastating subtype of intracerebral hemorrhage (ICH) with the highest mortality ranging from 56 % to 61.2 %. However, there is no effective medical or surgical therapy to improve its outcomes in clinic to date due to lack of understanding of its injury mechanisms. Herein, we explored the brainstem iron overload and injury in a rat model of BSH. METHODS Neurological scores were examined on day 1, 3, and 7 after modeling, and mortality of the rats was recorded to draft a survival curve. Rats were monitored by MRI using T2 and susceptibility weighted imaging (SWI) before sacrifice for examination of histology and immunofluorescence on day 1, 3, and 7. RESULTS BSH rats had a high mortality of 56 % and demonstrated the severe neurological deficits mimicking the clinical conditions. SWI showed that the same increasing tendency in change of hypointense area with that in iron deposition by Perls staining from day 1 to 7. Expression of heme oxygenase 1 (HO-1) and generation of reactive oxygen species (ROS) had similar tendency and both peaked on day 3. Neuronal degeneration occurred and stayed elevated from day 1 to 7, while myelin sheath injury was initially observed on day 1 but without significant difference within 7 days. CONCLUSIONS The time courses of erythrocyte lysis, HO-1 expression, iron deposition and ROS generation are related to each other after BSH. Besides, brainstem injury including neuronal degeneration and myelin damage were observed and discussed.
Collapse
Affiliation(s)
- Xi Guo
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lu Ma
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hao Li
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Qi
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yang Wei
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zhongxin Duan
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jiake Xu
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chengwei Wang
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Integrated Traditional and Western Medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Chao You
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Meng Tian
- Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
6
|
Guo X, Qi X, Li H, Duan Z, Wei Y, Zhang F, Tian M, Ma L, You C. Deferoxamine Alleviates Iron Overload and Brain Injury in a Rat Model of Brainstem Hemorrhage. World Neurosurg 2019; 128:e895-e904. [PMID: 31082547 DOI: 10.1016/j.wneu.2019.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Brainstem hemorrhage (BSH) is the most dangerous and devastating subtype of intracerebral hemorrhage and is associated with high morbidity and mortality. However, to date, no effective prevention methods or specific therapies have been available to improve its clinical outcomes. We preliminarily explored the efficacy of deferoxamine (DFO), a clinical chelator known for its iron-scavenging activities, in a rat model of BSH induced with collagenase infusion. METHODS DFO or saline was administrated 6 hours after BSH induction and then every 12 hours for ≤7 days. The survival curve of the rats was created, and the neurological scores were examined on days 1, 3, and 7 after BSH. The rats were sacrificed after 1, 3, and 7 days of DFO treatment for histological examination and immunohistochemistry. RESULTS The results showed that administration of DFO delayed erythrocytes lysis, reduced iron deposition, reduced reactive oxygen species generation, reduced heme oxygenase-1 expression, and alleviated brain injury such as neuron degeneration and myelin sheath injury. However, DFO did not improve the survival rate and neurobehavioral outcomes in this model. CONCLUSIONS Administration of DFO had limited therapeutic effects on collagenase-induced brainstem hemorrhage in rats. Some potential explanations were proposed, and more preclinical work is required to clarify the controversial curative effect of DFO in ICH.
Collapse
Affiliation(s)
- Xi Guo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Qi
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongxin Duan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Wei
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meng Tian
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Shin YS, Ryall JG, Britto JM, Lau CL, Devenish RJ, Nagley P, Beart PM. Inhibition of bioenergetics provides novel insights into recruitment of PINK1-dependent neuronal mitophagy. J Neurochem 2019; 149:269-283. [PMID: 30664245 DOI: 10.1111/jnc.14667] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/11/2018] [Accepted: 01/08/2019] [Indexed: 01/01/2023]
Abstract
Contributions of damaged mitochondria to neuropathologies have stimulated interest in mitophagy. We investigated triggers of neuronal mitophagy by disruption of mitochondrial energy metabolism in primary neurons. Mitophagy was examined in cultured murine cerebellar granule cells after inhibition of mitochondrial respiratory chain by drugs rotenone, 3-nitropropionic acid, antimycin A, and potassium cyanide, targeting complexes I, II, III, and IV, respectively. Inhibitor concentrations producing slow cellular demise were determined from analyses of cellular viability, morphology of neuritic damage, plasma membrane permeability, and oxidative phosphorylation. Live cell imaging of dissipation of mitochondrial membrane potential (ΔΨm ) by drugs targeting mitochondrial complexes was referenced to complete depolarization by carbonyl cyanide m-chlorophenyl hydrazone. While inhibition of complexes I, III and IV effected rapid dissipation of ΔΨm , inhibition of complex II using 3-nitropropionic acid led to minimal depolarization of mitochondria. Nonetheless, all respiratory chain inhibitors triggered mitophagy as indicated by increased aggregation of mitochondrially localized PINK1. Mitophagy was further analyzed using a dual fluorescent protein biosensor reporting mitochondrial relocation to acidic lysosomal environment. Significant acidification of mitochondria was observed in neurons treated with rotenone or 3-nitropropionic acid, revealing mitophagy at distal processes. Neurons treated with antimycin A or cyanide failed to show mitochondrial acidification. Minor dissipation of ΔΨm by 3-nitropropionic acid coupled with vigorous triggering of mitophagy suggested depolarization of mitochondria is not a necessary condition to trigger mitophagy. Moreover, weak elicitation of mitophagy by antimycin A, subsequent to loss of ΔΨm , suggested that mitochondrial depolarization is not a sufficient condition for triggering robust neuronal mitophagy. Our findings provide new insight into complexities of mitophagic clearance of neuronal mitochondria.
Collapse
Affiliation(s)
- Yea Seul Shin
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | - James G Ryall
- Department of Physiology, University of Melbourne, Parkville, Vic., Australia
| | - Joanne M Britto
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia
| | - Chew L Lau
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia
| | - Rodney J Devenish
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Phillip Nagley
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Vic., Australia
| | - Philip M Beart
- Florey Institute of Neuroscience and Mental Health, Parkville, Vic., Australia.,Department of Pharmacology, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
8
|
Verma DK, Gupta S, Biswas J, Joshi N, Singh A, Gupta P, Tiwari S, Sivarama Raju K, Chaturvedi S, Wahajuddin M, Singh S. New therapeutic activity of metabolic enhancer piracetam in treatment of neurodegenerative disease: Participation of caspase independent death factors, oxidative stress, inflammatory responses and apoptosis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2078-2096. [DOI: 10.1016/j.bbadis.2018.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 02/26/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
|
9
|
Abstract
Mild traumatic brain injury (mTBI) represents a significant public healthcare concern, accounting for the majority of all head injuries. While symptoms are generally transient, some patients go on to experience long-term cognitive impairments and additional mild impacts can result in exacerbated and persisting negative outcomes. To date, studies using a range of experimental models have reported chronic behavioral deficits in the presence of axonal injury and inflammation following repeated mTBI; assessments of oxidative stress and myelin pathology have thus far been limited. However, some models employed induced acute focal damage more suggestive of moderate–severe brain injury and are therefore not relevant to repeated mTBI. Given that the nature of mechanical loading in TBI is implicated in downstream pathophysiological changes, the mechanisms of damage and chronic consequences of single and repeated closed-head mTBI remain to be fully elucidated. This review covers literature on potential mechanisms of damage following repeated mTBI, integrating known mechanisms of pathology underlying moderate–severe TBIs, with recent studies on adult rodent models relevant to direct impact injuries rather than blast-induced damage. Pathology associated with excitotoxicity and cerebral blood flow-metabolism uncoupling, oxidative stress, cell death, blood-brain barrier dysfunction, astrocyte reactivity, microglial activation, diffuse axonal injury, and dysmyelination is discussed, followed by a summary of functional deficits and preclinical assessments of therapeutic strategies. Comprehensive characterization of the pathology underlying delayed and persisting deficits following repeated mTBI is likely to facilitate further development of therapeutic strategies to limit long-term sequelae.
Collapse
Affiliation(s)
- Brooke Fehily
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Melinda Fitzgerald
- 1 Experimental and Regenerative Neurosciences, School of Biological sciences, The University of Western Australia, Perth, Western Australia, Australia.,2 Curtin Health Innovation Research Institute, Curtin University, Perth, Western Australia, Australia.,3 Perron Institute for Neurological and Translational Science, Sarich Neuroscience Research Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
10
|
Picroside II Exerts a Neuroprotective Effect by Inhibiting mPTP Permeability and EndoG Release after Cerebral Ischemia/Reperfusion Injury in Rats. J Mol Neurosci 2017; 64:144-155. [PMID: 29256102 DOI: 10.1007/s12031-017-1012-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 12/01/2017] [Indexed: 11/27/2022]
Abstract
Mitochondrial membrane permeability is closely related to cerebral ischemia/reperfusion (I/R) injury. This paper explored the neuroprotective effect of picroside II (Picr), which inhibits the permeability of mitochondrial permeability transition pore (mPTP) and endonuclease G (EndoG) release from mitochondria into cytoplasm after cerebral I/R in rats. After 2 h of cerebral ischemia and 24 h of reperfusion in rats with different intervention measures, the neurobehavioral function, infarction volume, and reactive oxygen species (ROS) content in brain tissues were observed by modified neurological severity scale (mNSS), triphenyl tetrazolium chloride (TTC) staining, and enzyme-linked immunosorbent assay, respectively. The permeability of mPTP was assayed using spectrophotometry. The morphology and apoptotic cells of brain tissues were observed by hematoxylin-eosin staining and terminal deoxynucleotidyl transferase dUTP nick end labeling assay, respectively. The expressions of EndoG and voltage-dependent anion channel 1 (VDAC1) were determined by immunohistochemical assay and western blot. The Picr group exhibited clear decreases in mNSS scores, ROS content, number of apoptotic cells, mPTP permeability and expression of VDAC1, and EndoG in cytoplasm and nuclei, and the morphology of brain tissue was improved as compared with the model group (P < 0.05). Picr could attenuate cerebral I/R injury by downregulating the expression of VDAC1 and decreasing the permeability of mPTP, thereby inhibiting EndoG release from mitochondria into cytoplasm.
Collapse
|
11
|
Li Z, Fan EK, Liu J, Scott MJ, Li Y, Li S, Xie W, Billiar TR, Wilson MA, Jiang Y, Wang P, Fan J. Cold-inducible RNA-binding protein through TLR4 signaling induces mitochondrial DNA fragmentation and regulates macrophage cell death after trauma. Cell Death Dis 2017; 8:e2775. [PMID: 28492546 PMCID: PMC5584526 DOI: 10.1038/cddis.2017.187] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 03/24/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023]
Abstract
Trauma is a major cause of systemic inflammatory response syndrome and multiple organ dysfunction syndrome. Macrophages (Mϕ) direct trauma-induced inflammation, and Mϕ death critically influences the progression of the inflammatory response. In the current study, we explored an important role of trauma in inducing mitochondrial DNA (mtDNA) damage in Mϕ and the subsequent regulation of Mϕ death. Using an animal pseudo-fracture trauma model, we demonstrated that tissue damage induced NADPH oxidase activation and increased the release of reactive oxygen species via cold-inducible RNA-binding protein (CIRP)–TLR4–MyD88 signaling. This in turn, activates endonuclease G, which serves as an executor for the fragmentation of mtDNA in Mϕ. We further showed that fragmented mtDNA triggered both p62-related autophagy and necroptosis in Mϕ. However, autophagy activation also suppressed Mϕ necroptosis and pro-inflammatory responses. This study demonstrates a previously unidentified intracellular regulation of Mϕ homeostasis in response to trauma.
Collapse
Affiliation(s)
- Zhigang Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Erica K Fan
- University of Pittsburgh School of Arts and Science, Pittsburgh, PA 15213, USA
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| | - Melanie J Scott
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Song Li
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Wen Xie
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Mark A Wilson
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| | - Ping Wang
- The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
12
|
|
13
|
Misic V, El-Mogy M, Haj-Ahmad Y. Role of Endonuclease G in Exogenous DNA Stability in HeLa Cells. BIOCHEMISTRY (MOSCOW) 2016; 81:163-75. [PMID: 27260396 DOI: 10.1134/s0006297916020103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Endonuclease G (EndoG) is a well-conserved mitochondrial-nuclear nuclease with dual lethal and vital roles in the cell. The aim of our study was to examine whether EndoG exerts its nuclease activity on exogenous DNA substrates such as plasmid DNA (pDNA), considering their importance in gene therapy applications. The effects of EndoG knockdown on pDNA stability and levels of encoded reporter gene expression were evaluated in the cervical carcinoma HeLa cells. Transfection of pDNA vectors encoding short-hairpin RNAs (shRNAs) reduced levels of EndoG mRNA in HeLa cells. In physiological circumstances, EndoG knockdown did not have an effect on the stability of pDNA or the levels of encoded transgene expression as measured over a four-day time course. However, when endogenous expression of EndoG was induced by an extrinsic stimulus, targeting of EndoG by shRNA improved the perceived stability and transgene expression of pDNA vectors. Therefore, EndoG is not a mediator of exogenous DNA clearance, but in non-physiological circumstances, it may nonspecifically cleave intracellular DNA regardless of its origin. These findings make it unlikely that targeting of EndoG is a viable strategy for improving the duration and level of transgene expression from nonviral DNA vectors in gene therapy efforts.
Collapse
Affiliation(s)
- V Misic
- Brock University, Department of Biological Sciences, St. Catharines, ON, L2S 3A1, Canada.
| | | | | |
Collapse
|
14
|
Intracerebral Hemorrhage, Oxidative Stress, and Antioxidant Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:1203285. [PMID: 27190572 PMCID: PMC4848452 DOI: 10.1155/2016/1203285] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/20/2015] [Accepted: 03/28/2016] [Indexed: 12/20/2022]
Abstract
Hemorrhagic stroke is a common and severe neurological disorder and is associated with high rates of mortality and morbidity, especially for intracerebral hemorrhage (ICH). Increasing evidence demonstrates that oxidative stress responses participate in the pathophysiological processes of secondary brain injury (SBI) following ICH. The mechanisms involved in interoperable systems include endoplasmic reticulum (ER) stress, neuronal apoptosis and necrosis, inflammation, and autophagy. In this review, we summarized some promising advances in the field of oxidative stress and ICH, including contained animal and human investigations. We also discussed the role of oxidative stress, systemic oxidative stress responses, and some research of potential therapeutic options aimed at reducing oxidative stress to protect the neuronal function after ICH, focusing on the challenges of translation between preclinical and clinical studies, and potential post-ICH antioxidative therapeutic approaches.
Collapse
|
15
|
Effects of helium on inflammatory and oxidative stress-induced endothelial cell damage. Exp Cell Res 2015; 337:37-43. [PMID: 26096659 DOI: 10.1016/j.yexcr.2015.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 11/22/2022]
Abstract
Helium induces preconditioning in human endothelium protecting against postischemic endothelial dysfunction. Circulating endothelial microparticles are markers of endothelial dysfunction derived in response to injury. Another noble gas, xenon, protected human umbilical vein endothelial cells (HUVEC) against inflammatory stress in vitro. We hypothesised that helium protects the endothelium in vitro against inflammatory and oxidative stress. HUVEC were isolated from fresh umbilical cords and grown upon confluence. Cells were subjected to starving medium for 12h before the experiment and treated for either 3 × 5 min or 1 × 30 min with helium (5% CO2, 25% O2, 70% He) or control gas (5% CO2, 25% O2, 70% N2) in a specialised gas chamber. Subsequently, cells were stimulated with TNF-α (40 ng/ml for 24h or 10 ng/ml for 2h) or H2O2 (500 μM for 2h) or left untreated. Adhesion molecule expression was analysed using real-time quantitative polymerase chain reaction. Caspase-3 expression and viability of the cells was measured by flowcytometry. Microparticles were investigated by nanoparticle tracking analysis. Helium had no effect on adhesion molecule expression after TNF-α stimulation but in combination with oxidative stress decreased cell viability (68.9 ± 1.3% and 58 ± 1.9%) compared to control. Helium further increased TNF-α induced release of caspase-3 containing particles compared to TNF-α alone (6.4 × 10(6) ± 1.1 × 10(6) and 2.9 × 10(6) ± 0.7 × 10(6), respectively). Prolonged exposure of helium increased microparticle formation (2.4 × 10(9) ± 0.5 × 10(9)) compared to control (1.7 × 10(9) ± 0.2 × 10(9)). Summarized, helium increases inflammatory and oxidative stress-induced endothelial damage and is thus not biologically inert. A possible noxious effects on the cellular level causing alterations in microparticle formation both in number and content should be acknowledged.
Collapse
|
16
|
Seo TW, Lee JS, Yoo SJ. Cellular inhibitor of apoptosis protein 1 ubiquitinates endonuclease G but does not affect endonuclease G-mediated cell death. Biochem Biophys Res Commun 2014; 451:644-9. [PMID: 25139236 DOI: 10.1016/j.bbrc.2014.08.047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 08/09/2014] [Indexed: 11/30/2022]
Abstract
Inhibitors of Apoptosis Proteins (IAPs) are evolutionarily well conserved and have been recognized as the key negative regulators of apoptosis. Recently, the role of IAPs as E3 ligases through the Ring domain was revealed. Using proteomic analysis to explore potential target proteins of DIAP1, we identified Drosophila Endonuclease G (dEndoG), which is known as an effector of caspase-independent cell death. In this study, we demonstrate that human EndoG interacts with IAPs, including human cellular Inhibitor of Apoptosis Protein 1 (cIAP1). EndoG was ubiquitinated by IAPs in vitro and in human cell lines. Interestingly, cIAP1 was capable of ubiquitinating EndoG in the presence of wild-type and mutant Ubiquitin, in which all lysines except K63 were mutated to arginine. cIAP1 expression did not change the half-life of EndoG and cIAP1 depletion did not alter its levels. Expression of dEndoG 54310, in which the mitochondrial localization sequence was deleted, led to cell death that could not be suppressed by DIAP1 in S2 cells. Moreover, EndoG-mediated cell death induced by oxidative stress in HeLa cells was not affected by cIAP1. Therefore, these results indicate that IAPs interact and ubiquitinate EndoG via K63-mediated isopeptide linkages without affecting EndoG levels and EndoG-mediated cell death, suggesting that EndoG ubiquitination by IAPs may serve as a regulatory signal independent of proteasomal degradation.
Collapse
Affiliation(s)
- Tae Woong Seo
- Department of Biology, Research Institute for Basic Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ji Sun Lee
- Department of Nanopharmaceutical Life Sciences, Research Institute for Basic Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Soon Ji Yoo
- Department of Biology, Research Institute for Basic Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea; Department of Nanopharmaceutical Life Sciences, Research Institute for Basic Sciences, Kyung Hee University, Seoul 130-701, Republic of Korea.
| |
Collapse
|
17
|
Cheng YH, Lai SW, Chen PY, Chang JH, Chang NW. PPARα activation attenuates amyloid-β-dependent neurodegeneration by modulating Endo G and AIF translocation. Neurotox Res 2014; 27:55-68. [PMID: 25048111 DOI: 10.1007/s12640-014-9485-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 06/19/2014] [Accepted: 07/04/2014] [Indexed: 12/31/2022]
Abstract
The accumulation of a large amount of amyloid-β (Aβ42) in brain neurons is one of the debilitating characteristics of Alzheimer's disease. In this study, we determined the effects of peroxisome proliferator-activated receptor alpha (PPARα) activation on neuronal degeneration using a model of Aβ42-induced cytotoxicity. We found that 0.5 μM Aβ42 induced DNA damage and apoptosis in NT2N cells after 6 h of treatment. Co-treatment of Aβ42-treated cells with Wy14643, a PPARα ligand, significantly increased cell viability after 24 h compared with cells treated with Aβ42 alone. There were no differences in the protein levels of caspase-3, Bcl-2/Bax or p53 between cells treated with Aβ42 alone and those treated with both Aβ42 and Wy14643. However, the addition of Wy14643 significantly suppressed the Aβ42-induced upregulation of Endo G and AIF protein levels. Immunohistochemical analyses further demonstrated that Wy14643 reduced the expression of Endo G and AIF translocated from the cytoplasm into the nucleus, which occurred concomitantly with the decrease in DNA damage in Aβ42-treated cells. Our data clearly show that PPARα activation prevents DNA damage and neuronal cell apoptosis by decreasing the expression and translocation of AIF/Endo G to the nucleus in a caspase-3- and p53-independent pathway in the NT2N cell model. This role of PPARα in promoting neuron survival suggests a possible clinical application in treating Aβ42-associated neurotoxicity in Alzheimer's disease.
Collapse
Affiliation(s)
- Ya-Hsin Cheng
- Department of Physiology, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | | | | | | | | |
Collapse
|
18
|
O'Hare Doig RL, Bartlett CA, Maghzal GJ, Lam M, Archer M, Stocker R, Fitzgerald M. Reactive species and oxidative stress in optic nerve vulnerable to secondary degeneration. Exp Neurol 2014; 261:136-46. [PMID: 24931225 DOI: 10.1016/j.expneurol.2014.06.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/24/2014] [Accepted: 06/05/2014] [Indexed: 12/20/2022]
Abstract
Secondary degeneration contributes substantially to structural and functional deficits following traumatic injury to the CNS. While it has been proposed that oxidative stress is a feature of secondary degeneration, contributing reactive species and resultant oxidized products have not been clearly identified in vivo. The study is designed to identify contributors to, and consequences of, oxidative stress in a white matter tract vulnerable to secondary degeneration. Partial dorsal transection of the optic nerve (ON) was used to model secondary degeneration in ventral nerve unaffected by the primary injury. Reactive species were assessed using fluorescent labelling and liquid chromatography/tandem mass spectroscopy (LC/MS/MS). Antioxidant enzymes and oxidized products were semi-quantified immunohistochemically. Mitophagy was assessed by electron microscopy. Fluorescent indicators of reactive oxygen and/or nitrogen species increased at 1, 3 and 7days after injury, in ventral ON. LC/MS/MS confirmed increases in reactive species linked to infiltrating microglia/macrophages in dorsal ON. Similarly, immunoreactivity for glutathione peroxidase and haem oxygenase-1 increased in ventral ON at 3 and 7days after injury, respectively. Despite increased antioxidant immunoreactivity, DNA oxidation was evident from 1day, lipid oxidation at 3days, and protein nitration at 7days after injury. Nitrosative and oxidative damage was particularly evident in CC1-positive oligodendrocytes, at times after injury at which structural abnormalities of the Node of Ranvier/paranode complex have been reported. The incidence of mitochondrial autophagic profiles was also significantly increased from 3days. Despite modest increases in antioxidant enzymes, increased reactive species are accompanied by oxidative and nitrosative damage to DNA, lipid and protein, associated with increasing abnormal mitochondria, which together may contribute to the deficits of secondary degeneration.
Collapse
Affiliation(s)
- Ryan L O'Hare Doig
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Carole A Bartlett
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Ghassan J Maghzal
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; University of New South Wales, NSW, Australia
| | - Magdalena Lam
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Michael Archer
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; University of New South Wales, NSW, Australia
| | - Melinda Fitzgerald
- Experimental and Regenerative Neurosciences, School of Animal Biology, The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
19
|
Hamada M, Wakabayashi K, Masui A, Iwai S, Imai T, Yura Y. Involvement of hydrogen peroxide in safingol-induced endonuclease G-mediated apoptosis of squamous cell carcinoma cells. Int J Mol Sci 2014; 15:2660-71. [PMID: 24549171 PMCID: PMC3958874 DOI: 10.3390/ijms15022660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 01/03/2014] [Accepted: 02/13/2014] [Indexed: 01/09/2023] Open
Abstract
Safingol, a L-threo-dihydrosphingosine, induced the nuclear translocation of a mitochondrial apoptogenic mediator—endonuclease G (endo G)—and apoptosis of human oral squamous cell carcinoma (SCC) cells. Upstream mediators remain largely unknown. The levels of hydrogen peroxide (H2O2) in cultured oral SCC cells were measured. Treatment with safingol increased intracellular H2O2 levels but not extracellular H2O2 levels, indicating the production of H2O2. The cell killing effect of safingol and H2O2 was diminished in the presence of reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Dual staining of cells with annexin V and propidium iodide (PI) revealed that apoptotic cell death occurred by treatment with H2O2 and safingol. The number of apoptotic cells was reduced in the presence of NAC. In untreated cells, endo G distributed in the cytoplasm and an association of endo G with mitochondria was observed. After treatment with H2O2 and safingol, endo G was distributed to the nucleus and cytoplasm, indicating the nuclear translocation of the mitochondrial factor. NAC prevented the increase of apoptotic cells and the translocation of endo G. Knock down of endo G diminished the cell killing effect of H2O2 and safingol. These results suggest that H2O2 is involved in the endo G-mediated apoptosis of oral SCC cells by safingol.
Collapse
Affiliation(s)
- Masakazu Hamada
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Ken Wakabayashi
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Atsushi Masui
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Soichi Iwai
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Tomoaki Imai
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Yoshiaki Yura
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
20
|
Hogg MC, Prehn JHM. Endonuclease-G and the pathways to dopaminergic neurodegeneration: a question of location? EMBO J 2013; 32:3014-6. [PMID: 24162725 PMCID: PMC3844957 DOI: 10.1038/emboj.2013.238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
EMBO J (2013 ) 32 23 , 3041 –3054 ; DOI: 10.1038/emboj.2013.228 ; published online 10 15 2013 Parkinson’s disease (PD) is characterised by the loss of dopaminergic neurons in the substantia nigra and accumulation of pathological inclusions containing α -synuclein termed Lewy bodies. However, to date the precise function of α -synuclein has remained elusive along with the mechanism of neuronal death. A paper published in this issue of EMBO Journal provides insight into the mechanism of α -synuclein-induced neuronal death and thereby reveals potential new targets for therapeutic intervention. Büttner et al (2013) highlight Endonuclease-G as a key executioner in α -synuclein toxicity and show that inhibiting mitochondrial release of Endonuclease-G can protect against α -synuclein toxicity.
Collapse
Affiliation(s)
- Marion C Hogg
- Centre for the Study of Neurological Disorders, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jochen H M Prehn
- Centre for the Study of Neurological Disorders, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
21
|
Don Paul C, Kiss C, Traore DAK, Gong L, Wilce MCJ, Devenish RJ, Bradbury A, Prescott M. Phanta: a non-fluorescent photochromic acceptor for pcFRET. PLoS One 2013; 8:e75835. [PMID: 24098733 PMCID: PMC3786930 DOI: 10.1371/journal.pone.0075835] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/22/2013] [Indexed: 11/24/2022] Open
Abstract
We have developed an orange non-fluorescent photochromic protein (quantum yield, 0.003) we call Phanta that is useful as an acceptor in pcFRET applications. Phanta can be repeatedly inter-converted between the two absorbing states by alternate exposure to cyan and violet light. The absorption spectra of Phanta in one absorbing state shows excellent overlap with the emission spectra of a number of donor green fluorescent proteins including the commonly used EGFP. We show that the Phanta-EGFP FRET pair is suitable for monitoring the activation of caspase 3 in live cells using readily available instrumentation and a simple protocol that requires the acquisition of two donor emission images corresponding to Phanta in each of its photoswitched states. This the first report of a genetically encoded non-fluorescent acceptor for pcFRET.
Collapse
Affiliation(s)
- Craig Don Paul
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
| | - Csaba Kiss
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Daouda A. K. Traore
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Lan Gong
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Matthew C. J. Wilce
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
| | - Rodney J. Devenish
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
| | - Andrew Bradbury
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Mark Prescott
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
22
|
Aleksandrushkina NI, Vanyushin BF. Endonucleases and apoptosis in animals. BIOCHEMISTRY (MOSCOW) 2013; 77:1436-51. [PMID: 23379520 DOI: 10.1134/s0006297912130032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Endonucleases are the main instruments of obligatory DNA degradation in apoptosis. Many endonucleases have marked processive action; initially they split DNA in chromatin into very large domains, and then they perform in it internucleosomal fragmentation of DNA followed by its hydrolysis to small fragments (oligonucleotides). During apoptosis, DNA of chromatin is attacked by many nucleases that are different in activity, specificity, and order of action. The activity of every endonuclease is regulated in the cell through its own regulatory mechanism (metal ions and other effectors, possibly also S-adenosylmethionine). Apoptosis is impossible without endonucleases as far as it leads to accumulation of unnecessary (defective) DNA, disorders in cell differentiation, embryogenesis, the organism's development, and is accompanied by various severe diseases. The interpretation of the structure and functions of endonucleases and of the nature and action of their modulating effectors is important not only for elucidation of mechanisms of apoptosis, but also for regulation and control of programmed cell death, cell differentiation, and development of organisms.
Collapse
Affiliation(s)
- N I Aleksandrushkina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | | |
Collapse
|
23
|
CHIP has a protective role against oxidative stress-induced cell death through specific regulation of endonuclease G. Cell Death Dis 2013; 4:e666. [PMID: 23764847 PMCID: PMC3698548 DOI: 10.1038/cddis.2013.181] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Oxidative stress is implicated in carcinogenesis, aging, and neurodegenerative diseases. The E3 ligase C terminus of Hsc-70 interacting protein (CHIP) has a protective role against various stresses by targeting damaged proteins for proteasomal degradation, and thus maintains protein quality control. However, the detailed mechanism by which CHIP protects cells from oxidative stress has not been demonstrated. Here, we show that depletion of CHIP led to elevated Endonuclease G (EndoG) levels and enhanced cell death upon oxidative stress. In contrast, CHIP overexpression reduced EndoG levels, and resulted in reduced or no oxidative stress-induced cell death in cancer cells and primary rat cortical neurons. Under normal conditions Hsp70 mediated the interaction between EndoG and CHIP, downregulating EndoG levels in a Hsp70/proteasome-dependent manner. However, under oxidative stress Hsp70 no longer interacted with EndoG, and the stabilized EndoG translocated to the nucleus and degraded chromosomal DNA. Our data suggest that regulation of the level of EndoG by CHIP in normal conditions may determine the sensitivity to cell death upon oxidative stress. Indeed, injection of H2O2 into the rat brain markedly increased cell death in aged mice compared with young mice, which correlated with elevated levels of EndoG and concurrent downregulation of CHIP in aged mice. Taken together, our findings demonstrate a novel protective mechanism of CHIP against oxidative stress through regulation of EndoG, and provide an opportunity to modulate oxidative stress-induced cell death in cancer and aging.
Collapse
|
24
|
Tichy ED, Stephan ZA, Osterburg A, Noel G, Stambrook PJ. Mouse embryonic stem cells undergo charontosis, a novel programmed cell death pathway dependent upon cathepsins, p53, and EndoG, in response to etoposide treatment. Stem Cell Res 2013; 10:428-41. [PMID: 23500643 DOI: 10.1016/j.scr.2013.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/18/2013] [Accepted: 01/29/2013] [Indexed: 02/09/2023] Open
Abstract
Embryonic stem cells (ESCs) are hypersensitive to many DNA damaging agents and can rapidly undergo cell death or cell differentiation following exposure. Treatment of mouse ESCs (mESCs) with etoposide (ETO), a topoisomerase II poison, followed by a recovery period resulted in massive cell death with characteristics of a programmed cell death pathway (PCD). While cell death was both caspase- and necroptosis-independent, it was partially dependent on the activity of lysosomal proteases. A role for autophagy in the cell death process was eliminated, suggesting that ETO induces a novel PCD pathway in mESCs. Inhibition of p53 either as a transcription factor by pifithrin α or in its mitochondrial role by pifithrin μ significantly reduced ESC death levels. Finally, EndoG was newly identified as a protease participating in the DNA fragmentation observed during ETO-induced PCD. We coined the term charontosis after Charon, the ferryman of the dead in Greek mythology, to refer to the PCD signaling events induced by ETO in mESCs.
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| | | | | | | | | |
Collapse
|
25
|
Polster BM. AIF, reactive oxygen species, and neurodegeneration: a "complex" problem. Neurochem Int 2012; 62:695-702. [PMID: 23246553 DOI: 10.1016/j.neuint.2012.12.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 11/26/2012] [Accepted: 12/05/2012] [Indexed: 12/21/2022]
Abstract
Apoptosis-inducing factor (AIF) is a flavin-binding mitochondrial intermembrane space protein that is implicated in diverse but intertwined processes that include maintenance of electron transport chain function, reactive oxygen species regulation, cell death, and neurodegeneration. In acute brain injury, AIF acquires a pro-death role upon translocation from the mitochondria to the nucleus, where it initiates chromatin condensation and large-scale DNA fragmentation. Although harlequin mice exhibiting an 80-90% global reduction in AIF protein are resistant to numerous forms of acute brain injury, they paradoxically undergo slow, progressive neurodegeneration beginning at three months of age. Brain deterioration, accompanied by markers of oxidative stress, is most pronounced in the cerebellum and retina, although it also occurs in the cortex, striatum, and thalamus. Loss of an AIF pro-survival function linked to assembly or stabilization of electron transport chain complex I underlies chronic neurodegeneration. To date, most studies of neurodegeneration have failed to adequately separate the relative importance of the mitochondrial and nuclear functions of AIF in determining the extent of injury, or whether oxidative stress plays a causative role. This review explores the complicated relationship among AIF, complex I, and the regulation of mitochondrial reactive oxygen species levels. It also discusses the controversial role of complex I deficiency in Parkinson's disease, and what can be learned from the AIF- and complex I-depleted harlequin mouse.
Collapse
Affiliation(s)
- Brian M Polster
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore St., MSTF 5-34, Baltimore, MD 21201, USA.
| |
Collapse
|
26
|
Datta K, Hyduke DR, Suman S, Moon BH, Johnson MD, Fornace AJ. Exposure to ionizing radiation induced persistent gene expression changes in mouse mammary gland. Radiat Oncol 2012; 7:205. [PMID: 23216862 PMCID: PMC3551737 DOI: 10.1186/1748-717x-7-205] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 11/16/2012] [Indexed: 12/02/2022] Open
Abstract
Background Breast tissue is among the most sensitive tissues to the carcinogenic actions of ionizing radiation and epidemiological studies have linked radiation exposure to breast cancer. Currently, molecular understanding of radiation carcinogenesis in mammary gland is hindered due to the scarcity of in vivo long-term follow up data. We undertook this study to delineate radiation-induced persistent alterations in gene expression in mouse mammary glands 2-month after radiation exposure. Methods Six to eight week old female C57BL/6J mice were exposed to 2 Gy of whole body γ radiation and mammary glands were surgically removed 2-month after radiation. RNA was isolated and microarray hybridization performed for gene expression analysis. Ingenuity Pathway Analysis (IPA) was used for biological interpretation of microarray data. Real time quantitative PCR was performed on selected genes to confirm the microarray data. Results Compared to untreated controls, the mRNA levels of a total of 737 genes were significantly (p<0.05) perturbed above 2-fold of control. More genes (493 genes; 67%) were upregulated than the number of downregulated genes (244 genes; 33%). Functional analysis of the upregulated genes mapped to cell proliferation and cancer related canonical pathways such as ‘ERK/MAPK signaling’, ‘CDK5 signaling’, and ‘14-3-3-mediated signaling’. We also observed upregulation of breast cancer related canonical pathways such as ‘breast cancer regulation by Stathmin1’, and ‘HER-2 signaling in breast cancer’ in IPA. Interestingly, the downregulated genes mapped to fewer canonical pathways involved in cell proliferation. We also observed that a number of genes with tumor suppressor function (GPRC5A, ELF1, NAB2, Sema4D, ACPP, MAP2, RUNX1) persistently remained downregulated in response to radiation exposure. Results from qRT-PCR on five selected differentially expressed genes confirmed microarray data. The PCR data on PPP4c, ELF1, MAPK12, PLCG1, and E2F6 showed similar trend in up and downregulation as has been observed with the microarray. Conclusions Exposure to a clinically relevant radiation dose led to long-term activation of mammary gland genes involved in proliferative and metabolic pathways, which are known to have roles in carcinogenesis. When considered along with downregulation of a number of tumor suppressor genes, our study has implications for breast cancer initiation and progression after therapeutic radiation exposure.
Collapse
Affiliation(s)
- Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, 3970 Reservoir Rd, Washington, DC, NW 20057-1468, USA.
| | | | | | | | | | | |
Collapse
|
27
|
Higgins GC, Devenish RJ, Beart PM, Nagley P. Transitory phases of autophagic death and programmed necrosis during superoxide-induced neuronal cell death. Free Radic Biol Med 2012; 53:1960-7. [PMID: 22982049 DOI: 10.1016/j.freeradbiomed.2012.08.586] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 08/20/2012] [Accepted: 08/24/2012] [Indexed: 01/08/2023]
Abstract
Neurons can undergo a diverse range of death responses under oxidative stress, encompassing apoptosis (caspase-dependent, programmed cell death) to various forms of caspase-independent death, including necrosis. We recently showed that primary murine cortical neurons exposed acutely to hydrogen peroxide undergo caspase-independent death, both autophagic cell death and programmed necrosis. To determine how oxidative stress induced by superoxide affects the route to cellular demise, we exposed primary cortical neurons to extended superoxide insult (provided by exogenous xanthine and xanthine oxidase in the presence of catalase). Under these conditions, over 24h, the nitroblue tetrazolium-reducing activity (indicative of superoxide) rose significantly during the first 4 to 8h and then declined to background levels. As with hydrogen peroxide, this superoxide insult failed to activate downstream caspases (-3, -7, and -9). Substantial depolarization of mitochondria occurred after 1h, and nuclear morphology changes characteristic of oxidative stress became maximal after 2h. However, death indicated by plasma membrane permeabilization (cellular uptake of propidium iodide) approached maximal levels only after 4h, at which time substantial redistribution to the cytosol of death-associated mitochondrial intermembrane space proteins, notably endonuclease G, had occurred. Applying established criteria for autophagic death (knockdown of Atg7) or programmed necrosis (knockdown of endonuclease G), cells treated with the relevant siRNA showed significant blockade of each type of cell death, 4h after onset of the superoxide flux. Yet at later times, siRNA-mediated knockdown failed to prevent death, monitored by cellular uptake of propidium iodide. We conclude that superoxide initially invokes a diverse programmed caspase-independent death response, involving transient manifestation in parallel of autophagic death and programmed necrosis. Ultimately most neurons become overwhelmed by the consequences of severe oxidative stress and die. This study reveals the multiple phases of neuronal cell death modalities under extended oxidative stress.
Collapse
Affiliation(s)
- Gavin C Higgins
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | |
Collapse
|
28
|
Liu W, Fan Z, Han Y, Zhang D, Li J, Wang H. Intranuclear localization of apoptosis-inducing factor and endonuclease G involves in peroxynitrite-induced apoptosis of spiral ganglion neurons. Neurol Res 2012; 34:915-22. [PMID: 23006905 DOI: 10.1179/1743132812y.0000000098] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The present study was designed to determine whether or not the caspase-independent apoptotic pathway participated in the cellular death of spiral ganglion neurons (SGNs) after exposure to peroxynitrite (ONOO(-)), with particular attention given to the intranuclear translocation of mitochondrial apoptosis-inducing factor (AIF) and endonuclease G (Endo G) in this process. METHODS The rat SGNs were isolated and primary cultured in vitro and were exposed to ONOO(-) with pre-treatment of pan-caspase inhibitor. Morphological changes of SGNs were observed by acridine orange cytochemistry staining, and apoptosis was examined by flow cytometry. The translocation of mitochondrial AIF and Endo G was detected by immunocytochemistry and Western blot. The protein expressions of Bcl-2 family in SGNs exposed to ONOO(-) were determined by Western blot. RESULTS Treatment of SGNs with ONOO(-) resulted in the occurrence of caspase-independent apoptosis as evidenced by acridine orange staining and flow cytometry analysis. The immunocytochemical analysis showed that AIF and Endo G labeling were marked in neuronal nuclei, while the Western blot demonstrated the intranuclear localization of AIF and Endo G in SGNs treated with ONOO(-). Western blot analysis demonstrated that ONOO(-) increased the Bax expression while reducing Bcl-2 expression, which was not prevented by pre-treatment with caspase inhibitor. CONCLUSION These data indicate that ONOO(-) can trigger caspase-independent apoptosis in SGNs associated with mitochondrial AIF and Endo G intranuclear localization.
Collapse
Affiliation(s)
- Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | | | | | | | | | | |
Collapse
|
29
|
Oh JM, Choi EK, Carp RI, Kim YS. Oxidative stress impairs autophagic flux in prion protein-deficient hippocampal cells. Autophagy 2012; 8:1448-61. [PMID: 22889724 DOI: 10.4161/auto.21164] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We previously reported that autophagy is upregulated in Prnp-deficient (Prnp ( 0/0) ) hippocampal neuronal cells in comparison to cellular prion protein (PrP (C) )-expressing (Prnp (+/+) ) control cells under conditions of serum deprivation. In this study, we determined whether a protective mechanism of PrP (C) is associated with autophagy using Prnp ( 0/0) hippocampal neuronal cells under hydrogen peroxide (H 2O 2)-induced oxidative stress. We found that Prnp ( 0/0) cells were more susceptible to oxidative stress than Prnp (+/+) cells in a dose- and time-dependent manner. In addition, we observed enhanced autophagy by immunoblotting, which detected the conversion of microtubule-associated protein 1 light chain 3 β (LC3B)-I to LC3B-II, and we observed increased punctate LC3B immunostaining in H 2O 2-treated Prnp ( 0/0) cells compared with H 2O 2-treated control cells. Interestingly, this enhanced autophagy was due to impaired autophagic flux in the H 2O 2-treated Prnp ( 0/0) cells, while the H 2O 2-treated Prnp (+/+) cells showed enhanced autophagic flux. Furthermore, caspase-dependent and independent apoptosis was observed when both cell lines were exposed to H 2O 2. Moreover, the inhibition of autophagosome formation by Atg7 siRNA revealed that increased autophagic flux in Prnp (+/+) cells contributes to the prosurvival effect of autophagy against H 2O 2 cytotoxicity. Taken together, our results provide the first experimental evidence that the deficiency of PrP (C) may impair autophagic flux via H 2O 2-induced oxidative stress.
Collapse
Affiliation(s)
- Jae-Min Oh
- Ilsong Institute of Life Science, Hallym University, Anyang, Korea
| | | | | | | |
Collapse
|
30
|
Pettikiriarachchi A, Gong L, Perugini MA, Devenish RJ, Prescott M. Ultramarine, a chromoprotein acceptor for Förster resonance energy transfer. PLoS One 2012; 7:e41028. [PMID: 22815901 PMCID: PMC3397996 DOI: 10.1371/journal.pone.0041028] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 06/17/2012] [Indexed: 11/29/2022] Open
Abstract
We have engineered a monomeric blue non-fluorescent chromoprotein called Ultramarine (fluorescence quantum yield, 0.001; ε 585nm, 64,000 M−1. cm−1) for use as a Förster resonance energy transfer acceptor for a number of different donor fluorescent proteins. We show its use for monitoring activation of caspase 3 in live cells using fluorescence lifetime imaging. Ultramarine has the potential to increase the number of cellular parameters that can be imaged simultaneously.
Collapse
Affiliation(s)
- Anne Pettikiriarachchi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Victoria, Australia
| | - Lan Gong
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Victoria, Australia
- ARC Centre of Excellence for Structural and Functional Microbial Genomics, Monash University, Clayton Campus, Victoria, Australia
| | - Matthew A. Perugini
- Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Rodney J. Devenish
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Victoria, Australia
- ARC Centre of Excellence for Structural and Functional Microbial Genomics, Monash University, Clayton Campus, Victoria, Australia
| | - Mark Prescott
- Department of Biochemistry and Molecular Biology, Monash University, Clayton Campus, Victoria, Australia
- * E-mail:
| |
Collapse
|
31
|
Higgins GC, Devenish RJ, Beart PM, Nagley P. Autophagic activity in cortical neurons under acute oxidative stress directly contributes to cell death. Cell Mol Life Sci 2011; 68:3725-40. [PMID: 21437645 PMCID: PMC11115140 DOI: 10.1007/s00018-011-0667-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Revised: 02/04/2011] [Accepted: 03/08/2011] [Indexed: 10/18/2022]
Abstract
Primary neurons undergo insult-dependent programmed cell death. We examined autophagy as a process contributing to cell death in cortical neurons after treatment with either hydrogen peroxide (H(2)O(2)) or staurosporine. Although caspase-9 activation and cleavage of procaspase-3 were significant following staurosporine treatment, neither was observed following H(2)O(2) treatment, indicating a non-apoptotic death. Autophagic activity increased rapidly with H(2)O(2), but slowly with staurosporine, as quantified by processing of endogenous LC3. Autophagic induction by both stressors increased the abundance of fluorescent puncta formed by GFP-LC3, which could be blocked by 3-methyladenine. Significantly, such inhibition of autophagy blocked cell death induced by H(2)O(2) but not staurosporine. Suppression of Atg7 inhibited cell death by H(2)O(2), but not staurosporine, whereas suppression of Beclin 1 prevented cell death by both treatments, suggesting it has a complex role regulating both apoptosis and autophagy. We conclude that autophagic mechanisms are activated in an insult-dependent manner and that H(2)O(2) induces autophagic cell death.
Collapse
Affiliation(s)
- Gavin C. Higgins
- Department of Biochemistry and Molecular Biology, Monash University, Building 13D, Clayton Campus, Clayton, VIC 3800 Australia
| | - Rodney J. Devenish
- Department of Biochemistry and Molecular Biology, Monash University, Building 13D, Clayton Campus, Clayton, VIC 3800 Australia
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Monash University, Clayton Campus, Clayton, VIC 3800 Australia
| | - Philip M. Beart
- Florey Neuroscience Institutes, University of Melbourne, Parkville, VIC 3010 Australia
- Department of Pharmacology, University of Melbourne, Parkville, VIC 3010 Australia
| | - Phillip Nagley
- Department of Biochemistry and Molecular Biology, Monash University, Building 13D, Clayton Campus, Clayton, VIC 3800 Australia
- Australian Research Council Centre of Excellence in Structural and Functional Microbial Genomics, Monash University, Clayton Campus, Clayton, VIC 3800 Australia
| |
Collapse
|
32
|
Wells J, Kilburn MR, Shaw JA, Bartlett CA, Harvey AR, Dunlop SA, Fitzgerald M. Early in vivo changes in calcium ions, oxidative stress markers, and ion channel immunoreactivity following partial injury to the optic nerve. J Neurosci Res 2011; 90:606-18. [DOI: 10.1002/jnr.22784] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 08/12/2011] [Indexed: 01/19/2023]
|
33
|
Guttmann RP, Ghoshal S. Thiol-protease oxidation in age-related neuropathology. Free Radic Biol Med 2011; 51:282-8. [PMID: 21565267 DOI: 10.1016/j.freeradbiomed.2011.04.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Revised: 04/07/2011] [Accepted: 04/07/2011] [Indexed: 11/27/2022]
Abstract
Increased oxidative stress is a hallmark of every major neurodegenerative disease that has been studied. Numerous biomarkers of oxidative stress have been found, indicating that waves of oxidation had, at one time or another, overwhelmed antioxidant defenses, leaving behind a host of oxidized DNA, lipids, and proteins in their path. Although some level of oxidation may be beneficial, perhaps mediated by a hormetic response, the extent and types of oxidation detected in neuropathological states would suggest that oxidative stress contributes to a loss of homeostasis and cellular dysfunction. Although there are many targets of oxidants, this review emphasizes protein oxidation with a focus on an important group of redox-sensitive enzymes, the thiol-proteases. Both the direct and the indirect effects of oxidation and their potential importance in neurodegeneration are considered.
Collapse
Affiliation(s)
- Rodney P Guttmann
- Department of Gerontology, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.
| | | |
Collapse
|
34
|
Akdag MZ, Dasdag S, Ulukaya E, Uzunlar AK, Kurt MA, Taşkin A. Effects of extremely low-frequency magnetic field on caspase activities and oxidative stress values in rat brain. Biol Trace Elem Res 2010; 138:238-49. [PMID: 20177816 DOI: 10.1007/s12011-010-8615-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 01/15/2010] [Indexed: 10/19/2022]
Abstract
This study was aimed to investigate the effect of extremely low-frequency magnetic field (ELF-MF) on apoptosis and oxidative stress values in the brain of rat. Rats were exposed to 100 and 500 µT ELF-MF, which are the safety standards of public and occupational exposure for 2 h/day for 10 months. Brain tissues were immunohistochemically stained for the active (cleaved) caspase-3 in order to measure the apoptotic index by a semi-quantitative scoring system. In addition, the levels of catalase (CAT), malondialdehyde (MDA), myeloperoxidase (MPO), total antioxidative capacity (TAC), total oxidant status (TOS), and oxidative stress index (OSI) were measured in rat brain. Final score of apoptosis and MPO activity were not significantly different between the groups. CAT activity decreased in both exposure groups (p < 0.05), while TAC was found to be lower in ELF 500 group than those in ELF-100 and sham groups (p < 0.05). MDA, TOS, and OSI values were found to be higher in ELF-500 group than those in ELF-100 and sham groups (p < 0.05). In conclusion, apoptosis was not changed by long-term ELF-MF exposure, while both 100 and 500 µT ELF-MF exposure induced toxic effect in the rat brain by increasing oxidative stress and diminishing antioxidant defense system.
Collapse
Affiliation(s)
- Mehmet Zulkuf Akdag
- Department of Biophysics, Medical School of Dicle University, 21280 Diyarbakir, Turkey
| | | | | | | | | | | |
Collapse
|
35
|
Chen L, Park SM, Turner JR, Peter ME. Cell death in the colonic epithelium during inflammatory bowel diseases: CD95/Fas and beyond. Inflamm Bowel Dis 2010; 16:1071-6. [PMID: 20049945 DOI: 10.1002/ibd.21191] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
CD95 is a member of the death receptor family. It is a prototypical inducer of apoptosis that, upon binding of its cognate ligand (CD95L), forms a death-inducing signaling complex composed of adaptor molecules and initiator caspases that transmit the apoptosis signal. The CD95/CD95L system was implicated in the etiology of inflammatory bowel disease (IBD) based, primarily, on the finding that CD95 is highly expressed in the intestinal epithelial cells and that epithelial apoptosis is increased in IBD. In recent years it has been recognized that CD95, while playing an important role as an apoptosis-inducing receptor in the immune system, also has multiple nonapoptotic functions on nonimmune cells. This review critically discusses the data on the possible function of CD95 as an apoptosis-inducing receptor in IBD and discusses alternative mechanisms for epithelial cell loss in IBD.
Collapse
Affiliation(s)
- Lina Chen
- Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois 60637, USA
| | | | | | | |
Collapse
|
36
|
Xu Z, Zhang J, David KK, Yang ZJ, Li X, Dawson TM, Dawson VL, Koehler RC. Endonuclease G does not play an obligatory role in poly(ADP-ribose) polymerase-dependent cell death after transient focal cerebral ischemia. Am J Physiol Regul Integr Comp Physiol 2010; 299:R215-21. [PMID: 20427721 DOI: 10.1152/ajpregu.00747.2009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Activation of poly(ADP-ribose) polymerase (PARP) and subsequent translocation of apoptosis-inducing factor contribute to caspase-independent neuronal injury from N-methyl-d-aspartate, oxygen-glucose deprivation, and ischemic stroke. Some studies have implicated endonuclease G in the DNA fragmentation associated with caspase-independent cell death. Here, we compared wild-type and endonuclease G null mice to investigate whether endonuclease G plays a role in the PARP-dependent injury that results from transient focal cerebral ischemia. Latex casts did not reveal differences in the cerebral arterial distribution territory or posterior communicating arterial diameter, and the decrease in laser-Doppler flux during middle cerebral artery occlusion was similar in wild-type and endonuclease G null mice. After 90 min of occlusion and 1 day of reperfusion, similar degrees of nuclear translocation of apoptosis-inducing factor and DNA degradation were evident in male wild-type and null mice. At 3 days of reperfusion, infarct volume and neurological deficit scores were not different between male wild-type and endonuclease G null mice or between female wild-type and endonuclease G null mice. These data demonstrate that endonuclease G is not required for the pathogenesis of transient focal ischemia in either male or female mice. Treatment with a PARP inhibitor decreased infarct volume and deficit scores equivalently in male wild-type and endonuclease G null mice, indicating that the injury in endonuclease G null mice remains dependent on PARP. Thus endonuclease G is not obligatory for executing PARP-dependent injury during ischemic stroke.
Collapse
Affiliation(s)
- Zhenfeng Xu
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Signaling mechanisms of apoptosis-like programmed cell death in unicellular eukaryotes. Comp Biochem Physiol B Biochem Mol Biol 2010; 155:341-53. [DOI: 10.1016/j.cbpb.2010.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 01/19/2010] [Accepted: 01/23/2010] [Indexed: 11/18/2022]
|
38
|
Abstract
Neurodegenerative diseases are characterized by progressive dysfunction of specific populations of neurons, determining clinical presentation. Neuronal loss is associated with extra and intracellular accumulation of misfolded proteins, the hallmarks of many neurodegenerative proteinopathies. Major basic processes include abnormal protein dynamics due to deficiency of the ubiquitin-proteosome-autophagy system, oxidative stress and free radical formation, mitochondrial dysfunction, impaired bioenergetics, dysfunction of neurotrophins, 'neuroinflammatory' processes and (secondary) disruptions of neuronal Golgi apparatus and axonal transport. These interrelated mechanisms lead to programmed cell death is a long run over many years. Neurodegenerative disorders are classified according to known genetic mechanisms or to major components of protein deposits, but recent studies showed both overlap and intraindividual diversities between different phenotypes. Synergistic mechanisms between pathological proteins suggest common pathogenic mechanisms. Animal models and other studies have provided insight into the basic neurodegeneration and cell death programs, offering new ways for future prevention/treatment strategies.
Collapse
Affiliation(s)
- Kurt A Jellinger
- Institute of Clinical Neurobiology, Kenyongasse, Vienna, Austria.
| |
Collapse
|
39
|
Multifaceted deaths orchestrated by mitochondria in neurones. Biochim Biophys Acta Mol Basis Dis 2010; 1802:167-85. [DOI: 10.1016/j.bbadis.2009.09.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Revised: 09/07/2009] [Accepted: 09/08/2009] [Indexed: 12/16/2022]
|
40
|
Yang Y, Xia X, Zhang Y, Wang Q, Li L, Luo G, Xia Y. delta-Opioid receptor activation attenuates oxidative injury in the ischemic rat brain. BMC Biol 2009; 7:55. [PMID: 19709398 PMCID: PMC2754429 DOI: 10.1186/1741-7007-7-55] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 08/26/2009] [Indexed: 02/06/2023] Open
Abstract
Background We have recently shown that δ-opioid receptors (DORs) play an important role in neuroprotection from hypoxic injury via the regulation of extracellular signaling-regulated kinase (ERK) and cytochrome c release. Since ERK and cytochrome c are differentially involved in caspase signaling of oxidative injury that significantly contributes to neuronal damage in ischemia/reperfusion, we considered if DOR activation protects the ischemic brain by attenuating oxidative injury. Results We observed that, in a model of cerebral ischemia with middle cerebral artery occlusion, DOR activation increased the activity of major antioxidant enzymes, glutathione peroxidase and superoxide dismutase, and decreased malondialdehyde and nitric oxide levels in the cortex exposed to cerebral ischemia/reperfusion. In addition, DOR activation reduced caspase 3 expression, though it did not significantly affect the increase in interleukin (IL)1β and tumor necrosis factor (TNF)α expression at the same timepoint. PD98059, an inhibitor of mitogen-activated protein kinase (MAPK) extracellular signaling-regulated kinase kinase, accelerated animal death during ischemia/reperfusion. Conclusion DOR activation attenuates oxidative injury in the brain exposed to ischemia/reperfusion by enhancing antioxidant ability and inhibiting caspase activity, which provides novel insights into the mechanism of DOR neuroprotection.
Collapse
Affiliation(s)
- Yilin Yang
- Third Clinical College of Schoow University, Changzhou, Jiangsu, PR China.
| | | | | | | | | | | | | |
Collapse
|