1
|
Pan JZ, Wang Z, Sun W, Pan P, Li W, Sun Y, Chen S, Lin A, Tan W, He L, Greene J, Yao V, An L, Liang R, Li Q, Yu J, Zhang L, Kyritsis N, Fernandez XD, Moncivais S, Mendoza E, Fung P, Wang G, Niu X, Du Q, Xiao Z, Chang Y, Lv P, Huie JR, Torres‐Espin A, Ferguson AR, Hemmerle DD, Talbott JF, Weinstein PR, Pascual LU, Singh V, DiGiorgio AM, Saigal R, Whetstone WD, Manley GT, Dhall SS, Bresnahan JC, Maze M, Jiang X, Singhal NS, Beattie MS, Su H, Guan Z. ATF3 is a neuron-specific biomarker for spinal cord injury and ischaemic stroke. Clin Transl Med 2024; 14:e1650. [PMID: 38649772 PMCID: PMC11035380 DOI: 10.1002/ctm2.1650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Although many molecules have been investigated as biomarkers for spinal cord injury (SCI) or ischemic stroke, none of them are specifically induced in central nervous system (CNS) neurons following injuries with low baseline expression. However, neuronal injury constitutes a major pathology associated with SCI or stroke and strongly correlates with neurological outcomes. Biomarkers characterized by low baseline expression and specific induction in neurons post-injury are likely to better correlate with injury severity and recovery, demonstrating higher sensitivity and specificity for CNS injuries compared to non-neuronal markers or pan-neuronal markers with constitutive expressions. METHODS In animal studies, young adult wildtype and global Atf3 knockout mice underwent unilateral cervical 5 (C5) SCI or permanent distal middle cerebral artery occlusion (pMCAO). Gene expression was assessed using RNA-sequencing and qRT-PCR, while protein expression was detected through immunostaining. Serum ATF3 levels in animal models and clinical human samples were measured using commercially available enzyme-linked immune-sorbent assay (ELISA) kits. RESULTS Activating transcription factor 3 (ATF3), a molecular marker for injured dorsal root ganglion sensory neurons in the peripheral nervous system, was not expressed in spinal cord or cortex of naïve mice but was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Additionally, ATF3 protein levels in mouse blood significantly increased 1 day after SCI or ischemic stroke. Importantly, ATF3 protein levels in human serum were elevated in clinical patients within 24 hours after SCI or ischemic stroke. Moreover, Atf3 knockout mice, compared to the wildtype mice, exhibited worse neurological outcomes and larger damage regions after SCI or ischemic stroke, indicating that ATF3 has a neuroprotective function. CONCLUSIONS ATF3 is an easily measurable, neuron-specific biomarker for clinical SCI and ischemic stroke, with neuroprotective properties. HIGHLIGHTS ATF3 was induced specifically in neurons of the spinal cord or cortex within 1 day after SCI or ischemic stroke, respectively. Serum ATF3 protein levels are elevated in clinical patients within 24 hours after SCI or ischemic stroke. ATF3 exhibits neuroprotective properties, as evidenced by the worse neurological outcomes and larger damage regions observed in Atf3 knockout mice compared to wildtype mice following SCI or ischemic stroke.
Collapse
Affiliation(s)
- Jonathan Z. Pan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhanqiang Wang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurologyCangzhou People's HospitalCangzhouChina
| | - Wei Sun
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Peipei Pan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Wei Li
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Yongtao Sun
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyQianfoshan Hospital, Shandong UniversityJinanChina
| | - Shoulin Chen
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyThe Second Affiliated Hospital, Nanchang UniversityNanchangChina
| | - Amity Lin
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Wulin Tan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyGuangzhou Medical UniversityGuangzhouChina
| | - Liangliang He
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of Pain ManagementXuanwu Hospital, Capital Medical UniversityBeijingChina
| | - Jacob Greene
- Medical SchoolUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Virginia Yao
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lijun An
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyNo. 1 People's HospitalHuaianChina
| | - Rich Liang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Qifeng Li
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of NeurosurgeryTianjin Medical University General HospitalTianjinChina
| | - Jessica Yu
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lingyi Zhang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Nikolaos Kyritsis
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Xuan Duong Fernandez
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sara Moncivais
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Esmeralda Mendoza
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Pamela Fung
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Gongming Wang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Xinhuan Niu
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Qihang Du
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyShandong Provincial Hospital, Shandong UniversityJinanChina
| | - Zhaoyang Xiao
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Department of AnesthesiologyThe Second Affiliated Hospital, Dalian Medical UniversityDalianChina
| | - Yuwen Chang
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Peiyuan Lv
- Department of AnesthesiologyThe Second Affiliated Hospital, Dalian Medical UniversityDalianChina
- Department of NeurologyHebei Medical UniversityShijiazhuangChina
| | - J. Russell Huie
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Abel Torres‐Espin
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Adam R. Ferguson
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Debra D. Hemmerle
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Jason F. Talbott
- Department of RadiologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Philip R. Weinstein
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Lisa U. Pascual
- Department of Orthopedic SurgeryOrthopaedic Trauma InstituteUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Vineeta Singh
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Anthony M. DiGiorgio
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Rajiv Saigal
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - William D. Whetstone
- Department of Emergency MedicineUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Geoffrey T. Manley
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Sanjay S. Dhall
- Department of NeurosurgeryHarbor UCLA Medical CenterTorranceCaliforniaUSA
| | - Jacqueline C. Bresnahan
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Mervyn Maze
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Xiangning Jiang
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Neel S. Singhal
- Department of NeurologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Michael S. Beattie
- Department of Neurological SurgeryUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Brain and Spinal Injury CenterUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Hua Su
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- Center for Cerebrovascular ResearchUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| | - Zhonghui Guan
- Department of Anesthesia and Perioperative CareUniversity of California San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
2
|
Revisiting Traumatic Brain Injury: From Molecular Mechanisms to Therapeutic Interventions. Biomedicines 2020; 8:biomedicines8100389. [PMID: 33003373 PMCID: PMC7601301 DOI: 10.3390/biomedicines8100389] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/15/2022] Open
Abstract
Studying the complex molecular mechanisms involved in traumatic brain injury (TBI) is crucial for developing new therapies for TBI. Current treatments for TBI are primarily focused on patient stabilization and symptom mitigation. However, the field lacks defined therapies to prevent cell death, oxidative stress, and inflammatory cascades which lead to chronic pathology. Little can be done to treat the mechanical damage that occurs during the primary insult of a TBI; however, secondary injury mechanisms, such as inflammation, blood-brain barrier (BBB) breakdown, edema formation, excitotoxicity, oxidative stress, and cell death, can be targeted by therapeutic interventions. Elucidating the many mechanisms underlying secondary injury and studying targets of neuroprotective therapeutic agents is critical for developing new treatments. Therefore, we present a review on the molecular events following TBI from inflammation to programmed cell death and discuss current research and the latest therapeutic strategies to help understand TBI-mediated secondary injury.
Collapse
|
3
|
Wu J, Lipinski MM. Autophagy in Neurotrauma: Good, Bad, or Dysregulated. Cells 2019; 8:E693. [PMID: 31295858 PMCID: PMC6678153 DOI: 10.3390/cells8070693] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/06/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a physiological process that helps maintain a balance between the manufacture of cellular components and breakdown of damaged organelles and other toxic cellular constituents. Changes in autophagic markers are readily detectable in the spinal cord and brain following neurotrauma, including traumatic spinal cord and brain injury (SCI/TBI). However, the role of autophagy in neurotrauma remains less clear. Whether autophagy is good or bad is under debate, with strong support for both a beneficial and detrimental role for autophagy in experimental models of neurotrauma. Emerging data suggest that autophagic flux, a measure of autophagic degradation activity, is impaired in injured central nervous systems (CNS), and interventions that stimulate autophagic flux may provide neuroprotection in SCI/TBI models. Recent data demonstrating that neurotrauma can cause lysosomal membrane damage resulting in pathological autophagosome accumulation in the spinal cord and brain further supports the idea that the impairment of the autophagy-lysosome pathway may be a part of secondary injury processes of SCI/TBI. Here, we review experimental work on the complex and varied responses of autophagy in terms of both the beneficial and detrimental effects in SCI and TBI models. We also discuss the existing and developing therapeutic options aimed at reducing the disruption of autophagy to protect the CNS after injuries.
Collapse
Affiliation(s)
- Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| | - Marta M Lipinski
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
4
|
Singh VK, Singh HB, Upadhyay RS. Role of fusaric acid in the development of 'Fusarium wilt' symptoms in tomato: Physiological, biochemical and proteomic perspectives. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2017; 118:320-332. [PMID: 28683401 DOI: 10.1016/j.plaphy.2017.06.028] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/25/2017] [Accepted: 06/26/2017] [Indexed: 05/18/2023]
Abstract
Fusarium wilt is one of the most prevalent and damaging diseases of tomato. Among various toxins secreted by the Fusarium oxysporum f. sp. lycopersici (causal agent of Fusarium wilt of tomato), fusaric acid (FA) is suspected to be a potent pathogenicity factor in tomato wilt disease development. With this rationale the present study was carried out with physiological, biochemical and proteomic perspectives. Treatment of FA was given to the leaves of tomato directly through infiltration to show the characteristic features of Fusarium wilt of tomato. The phytotoxic effect of FA was assessed in the form of cell death in tomato leaves which was observed by increased uptake of Evans blue stain. The measurement of electrolyte leakage was used as an indicator of the extent of cell death. The influence of FA on the leaf photosynthesis of tomato plant was investigated and it was found that FA strongly reduced the photosynthetic pigment contents of tomato leaves resulting to heavy suppression of leaf photosynthesis processes, which therefore affected leaf physiology finally leading to leaf wilting and necrosis. This cell death inducer (FA) produced an enormous oxidative burst during which large quantities of reactive oxygen species (ROS) like H2O2 was generated in the treated leaf tissues of tomato plants which was evident from enhancement in lipid peroxidation. To assess the involvement of proteolysis in the cell death cascade induced by FA treatment, total protease activity was measured in the leaf tissues and it was found that the total protease activity increased with the treatment and leading to cell death. Furthermore, proteomic study was used as a powerful tool to understand the alterations in cellular protein expression in response to FA exposure. Differential expression in several proteins was observed in the present study. Proteomic analyses, thus, clearly indicate that proteins belonging to different functional classes are significantly affected in the plant leaf tissues after FA exposure leading to deterioration of structure and metabolism of cells. Thus, it is concluded that FA plays an important role in fungal pathogenicity by decreasing cell viability.
Collapse
Affiliation(s)
- Vivek Kumar Singh
- Laboratory of Mycopathology and Microbial Technology, Centre of Advanced Study in Botany, Banaras Hindu University, Varanasi, India.
| | - Harikesh Bahadur Singh
- Department of Mycology & Plant Pathology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Ram Sanmukh Upadhyay
- Laboratory of Mycopathology and Microbial Technology, Centre of Advanced Study in Botany, Banaras Hindu University, Varanasi, India
| |
Collapse
|
5
|
Reshi L, Wu JL, Wang HV, Hong JR. Aquatic viruses induce host cell death pathways and its application. Virus Res 2015; 211:133-44. [PMID: 26494167 DOI: 10.1016/j.virusres.2015.10.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 10/07/2015] [Accepted: 10/14/2015] [Indexed: 11/15/2022]
Abstract
Virus infections of mammalian and animal cells consist of a series of events. As intracellular parasites, viruses rely on the use of host cellular machinery. Through the use of cell culture and molecular approaches over the past decade, our knowledge of the biology of aquatic viruses has grown exponentially. The increase in aquaculture operations worldwide has provided new approaches for the transmission of aquatic viruses that include RNA and DNA viruses. Therefore, the struggle between the virus and the host for control of the cell's death machinery is crucial for survival. Viruses are obligatory intracellular parasites and, as such, must modulate apoptotic pathways to control the lifespan of their host to complete their replication cycle. This paper updates the discussion on the detailed mechanisms of action that various aquatic viruses use to induce cell death pathways in the host, such as Bad-mediated, mitochondria-mediated, ROS-mediated and Fas-mediated cell death circuits. Understanding how viruses exploit the apoptotic pathways of their hosts may provide great opportunities for the development of future potential therapeutic strategies and pathogenic insights into different aquatic viral diseases.
Collapse
Affiliation(s)
- Latif Reshi
- Laboratory of Molecular Virology and Biotechnology, College of Bioscience and Biotechnology, Institute of Biotechnology, National Cheng Kung University, No 1. University Road, Tainan City 701, Taiwan, ROC; Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, No. 1. University Road, Tainan City 701, Taiwan, ROC
| | - Jen-Leih Wu
- Laboratory of Marine Molecular Biology and Biotechnology, Institute of Cellular and Organismic Biology, Academia Sinica, Nankang, Taipei 115, Taiwan, ROC
| | - Hao-Ven Wang
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, No. 1. University Road, Tainan City 701, Taiwan, ROC
| | - Jiann-Ruey Hong
- Laboratory of Molecular Virology and Biotechnology, College of Bioscience and Biotechnology, Institute of Biotechnology, National Cheng Kung University, No 1. University Road, Tainan City 701, Taiwan, ROC.
| |
Collapse
|
6
|
Abstract
Traumatic brain injury (TBI) is a major cause of mortality and morbidity worldwide. Despite extensive preclinical research supporting the effectiveness of neuroprotective therapies for brain trauma, there have been no successful randomized controlled clinical trials to date. TBI results in delayed secondary tissue injury due to neurochemical, metabolic and cellular changes; modulating such effects has provided the basis for neuroprotective interventions. To establish more effective neuroprotective treatments for TBI it is essential to better understand the complex cellular and molecular events that contribute to secondary injury. Here we critically review relevant research related to causes and modulation of delayed tissue damage, with particular emphasis on cell death mechanisms and post-traumatic neuroinflammation. We discuss the concept of utilizing multipotential drugs that target multiple secondary injury pathways, rather than more specific "laser"-targeted strategies that have uniformly failed in clinical trials. Moreover, we assess data supporting use of neuroprotective drugs that are currently being evaluated in human clinical trials for TBI, as well as promising emerging experimental multipotential drug treatment strategies. Finally, we describe key challenges and provide suggestions to improve the likelihood of successful clinical translation.
Collapse
Affiliation(s)
- David J Loane
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bogdan A Stoica
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Li J, Cui G, Sun L, Wang SJ, Tian S, Guan Z, Fan WS, Yan ZF, Yang YZ, You YQ, Fu XY, Li LA, Huang K, Li YL, Meng YG. ARHI overexpression induces epithelial ovarian cancer cell apoptosis and excessive autophagy. Int J Gynecol Cancer 2014; 24:437-43. [PMID: 24476894 DOI: 10.1097/igc.0000000000000065] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE ARHI is a maternally imprinted tumor suppressor gene that is responsible for initiating programmed cell death and inhibiting cancer cell growth. However, the influence of ARHI on epithelial ovarian cancer cell death and the underlying mechanisms behind how ARHI regulates cancer cells still require further studies. METHODS Epithelial ovarian cancer cells TOV112D and ES-2 were used in this in vitro study. Cell proliferation, apoptosis, and autophagy activities were compared in TOV112D and ES-2 cells transfected with ARHI vectors or control vectors. Bcl-2 siRNA was transfected into TOV112D cells to investigate the roles of Bcl-2 played in regulating apoptosis and autophagy. RESULTS ARHI expression was reduced in TOV112D and ES-2 cells compared with normal epithelial ovarian cells (NOE095 and HOSEpiC). Overexpressed ARHI inhibited cancer cell proliferation, whereas induced forced cell apoptosis and excessive formation of autophagosomes inhibited promoted cell death. Furthermore, we found that Bcl-2 expression moderately declined in response to ARHI overexpressing in ES-2 and TOV112D cells; meanwhile, more apoptotic cells and higher LC3 level presented after silence of Bcl-2 in TOV112D cells. Reduced Bcl-2-Beclin 1 complex were observed in ARHI overexpressing cells. Moreover, modulation of ARHI to Bcl-2 expression could be ascribed partially to the activation of PI3k/AKT pathway. The addition of LY294002 enabled to suppress Bcl-2 expression and cell proliferation. CONCLUSIONS The silence of ARHI expression in vitro seems to accelerate the malignant transformation of healthy ovarian cells by restraining apoptosis and autophagy. The overexpressed ARHI in TOV112D cancer cells suppresses the activation of PI3K/AKT and reduces the expression of Bcl-2, leading to enhanced cell apoptosis and autophagic cancer cell death.
Collapse
Affiliation(s)
- Jie Li
- Departments of *Gynecology and Obstetrics, †Orthopedics, and ‡Pathology, General Hospital of PLA, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
It is well known that the death of dopaminergic neurons of the substantia nigra pars compacta (SNc) is the pathological hallmark of Parkinson's disease (PD), the second most common and disabling condition in the expanding elderly population. Nevertheless, the intracellular cascade of events leading to dopamine cell death is still unknown and, consequently, treatment is largely symptomatic rather than preventive. Moreover, the mechanisms whereby nigral dopaminergic neurons may degenerate still remain controversial. Hitherto, several data have shown that the earlier cellular disturbances occurring in dopaminergic neurons include oxidative stress, excitotoxicity, inflammation, mitochondrial dysfunction and altered proteolysis. These alterations, rather than killing neurons, trigger subsequent death-related molecular pathways, including elements of apoptosis. In rare incidences, PD may be inherited; this evidence has opened a new and exciting area of research, attempting to shed light on the nature of the more common idiopathic PD form. In this review, the characteristics of the SNc dopaminergic neurons and their lifecycle from birth to death are reviewed. In addition, of the mechanisms by which the aforementioned alterations cause neuronal dopaminergic death, particular emphasis will be given to the role played by inflammation, and the relevance of the possible use of anti-inflammatory drugs in the treatment of PD. Finally, new evidence of a possible de novo neurogenesis in the SNc of adult animals and in PD patients will also be examined.
Collapse
Affiliation(s)
- Ennio Esposito
- Istituto di Ricerche Farmacologiche Mario Negri, Consorzio Mario Negri Sud, Via Nazionale 8, 66030 Santa Maria Imbaro (Chieti), Italy.
| | | | | |
Collapse
|
9
|
Proteomic analysis of apoptotic and oncotic pancreatic acinar AR42J cells treated with caerulein. Mol Cell Biochem 2013; 382:1-17. [PMID: 23884867 DOI: 10.1007/s11010-013-1603-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/23/2013] [Indexed: 01/08/2023]
Abstract
This study aims to determine the differentially expressed proteins in the pancreatic acinar cells undergoing apoptosis and oncosis stimulated with caerulein to explore different cell death process of the acinar cell. AR42J cells were treated with caerulein to induce cell model of acute pancreatitis. Cells that were undergoing apoptosis and oncosis were separated by flow cytometry. Then differentially expressed proteins in the two groups of separated cells were detected by shotgun liquid chromatography-tandem mass spectrometry. The results showed that 11 proteins were detected in both apoptosis group and oncosis group, 17 proteins were detected only in apoptosis group and 29 proteins were detected only in oncosis group. KEGG analysis showed that proteins detected only in apoptosis group were significantly enriched in 10 pathways, including ECM-receptor interaction, cell adhesion molecules, and proteins detected only in oncosis group were significantly enriched in three pathways, including endocytosis, base excision repair, and RNA degradation. These proteins we detected are helpful for us to understand the process of cell death in acute pancreatitis and may be useful for changing the death mode of pancreatic acinar cells, thus attenuating the severity of pancreatitis.
Collapse
|
10
|
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
|
11
|
Puyal J, Ginet V, Clarke PGH. Multiple interacting cell death mechanisms in the mediation of excitotoxicity and ischemic brain damage: a challenge for neuroprotection. Prog Neurobiol 2013; 105:24-48. [PMID: 23567504 DOI: 10.1016/j.pneurobio.2013.03.002] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 03/05/2013] [Accepted: 03/13/2013] [Indexed: 02/09/2023]
Abstract
There is currently no approved neuroprotective pharmacotherapy for acute conditions such as stroke and cerebral asphyxia. One of the reasons for this may be the multiplicity of cell death mechanisms, because inhibition of a particular mechanism leaves the brain vulnerable to alternative ones. It is therefore essential to understand the different cell death mechanisms and their interactions. We here review the multiple signaling pathways underlying each of the three main morphological types of cell death--apoptosis, autophagic cell death and necrosis--emphasizing their importance in the neuronal death that occurs during cerebral ischemia and hypoxia-ischemia, and we analyze the interactions between the different mechanisms. Finally, we discuss the implications of the multiplicity of cell death mechanisms for the design of neuroprotective strategies.
Collapse
Affiliation(s)
- Julien Puyal
- Département des Neurosciences Fondamentales, Université de Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland.
| | | | | |
Collapse
|
12
|
Bouzas-Rodríguez J, Zárraga-Granados G, Sánchez-Carbente MDR, Rodríguez-Valentín R, Gracida X, Anell-Rendón D, Covarrubias L, Castro-Obregón S. The nuclear receptor NR4A1 induces a form of cell death dependent on autophagy in mammalian cells. PLoS One 2012; 7:e46422. [PMID: 23071566 PMCID: PMC3465341 DOI: 10.1371/journal.pone.0046422] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/29/2012] [Indexed: 11/18/2022] Open
Abstract
The control of cell death is a biological process essential for proper development, and for preventing devastating pathologies like cancer and neurodegeneration. On the other hand, autophagy regulation is essential for protein and organelle degradation, and its dysfunction is associated with overlapping pathologies like cancer and neurodegeneration, but also for microbial infection and aging. In the present report we show that two evolutionarily unrelated receptors--Neurokinin 1 Receptor (NK(1)R,) a G-protein coupled receptor, and Insulin-like Growth Factor 1 Receptor (IGF1R), a tyrosine kinase receptor--both induce non-apoptotic cell death with autophagic features and requiring the activity of the autophagic core machinery proteins PI3K-III, Beclin-1 and Atg7. Remarkably, this form of cell death occurs in apoptosis-competent cells. The signal transduction pathways engaged by these receptors both converged on the activation of the nuclear receptor NR4A1, which has previously been shown to play a critical role in some paradigms of apoptosis and in NK(1)R-induced cell death. The activity of NR4A1 was necessary for IGF1R-induced cell death, as well as for a canonical model of cell death by autophagy induced by the presence of a pan-caspase inhibitor, suggesting that NR4A1 is a general modulator of this kind of cell death. During cell death by autophagy, NR4A1 was transcriptionally competent, even though a fraction of it was present in the cytoplasm. Interestingly, NR4A1 interacts with the tumor suppressor p53 but not with Beclin-1 complex. Therefore the mechanism to promote cell death by autophagy might involve regulation of gene expression, as well as protein interactions. Understanding the molecular basis of autophagy and cell death mediation by NR4A1, should provide novel insights and targets for therapeutic intervention.
Collapse
Affiliation(s)
- Jimena Bouzas-Rodríguez
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Gabriela Zárraga-Granados
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Maria del Rayo Sánchez-Carbente
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Rocío Rodríguez-Valentín
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Xicotencatl Gracida
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Dámaris Anell-Rendón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Luis Covarrubias
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Susana Castro-Obregón
- Developmental Genetics and Molecular Physiology Department, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
- * E-mail:
| |
Collapse
|
13
|
Azab KS, Osman SA, El-Fatih NM. Implication of caspase-3 and granzyme B expression and activity in spleenocytes of ehrlich ascites carcinoma mice subjected to immunotherapy. NORTH AMERICAN JOURNAL OF MEDICAL SCIENCES 2011; 3:371-7. [PMID: 22171245 PMCID: PMC3234140 DOI: 10.4297/najms.2011.3371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Caspase-3 and granzyme B were claimed as apoptotic manipulative enzymes. Aims The present study was to determine the enzymes expression and activity in cancer and cancer immune therapeutic status and the possible association to cancer common pathological signs targeting the improvement of therapeutic conditions. Material and Methods Mice were immunized with cell lyaste or cell lysate + CKI in the right thigh and challenged with live cells of ehrlich ascites carcinoma (EAC) in the left thigh. The expression and activity of both enzymes in the spleenocytes derived from different subjects (normal, EAC and cell lysate or cell lysate + CKI immunized mice) after cultured with EAC viable cells were determined by colorimetric assay and western blot analysis. In addition, the subjects DNA ladder and serum metalloproteases (MMP 2 and 9) zymography were observed. Results The experimental data revealed over expression of caspase3 and granzyme B in the groups of cell lysate or cell lysate + CKI immunized mice compared to control while down expression were recorded in the EAC subject. The over expression of the 2 enzymes were accompanied with increases in the activities of caspase3 and granzyme B, changes in DNA fragmentation and inhibition of metalloproteases. Conclusion It could be suggested that, the parameter estimation within the present experimental framework could identify the efficiency of therapeutic vaccine protocols and elucidate the impact of CKI adjuvant with vaccines therapy.
Collapse
Affiliation(s)
- Khaled Sh Azab
- Radiation Biology Department, National Center for Radiation Research and Technology Atomic Energy Authority, Cairo, Egypt
| | | | | |
Collapse
|
14
|
Huang X, Huang Y, Ouyang Z, Xu L, Yan Y, Cui H, Han X, Qin Q. Singapore grouper iridovirus, a large DNA virus, induces nonapoptotic cell death by a cell type dependent fashion and evokes ERK signaling. Apoptosis 2011; 16:831-45. [PMID: 21656148 DOI: 10.1007/s10495-011-0616-y] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Virus induced cell death, including apoptosis and nonapoptotic cell death, plays a critical role in the pathogenesis of viral diseases. Singapore grouper iridovirus (SGIV), a novel iridovirus of genus Ranavirus, causes high mortality and heavy economic losses in grouper aquaculture. Here, using fluorescence microscopy, electron microscopy and biochemical assays, we found that SGIV infection in host (grouper spleen, EAGS) cells evoked nonapoptotic programmed cell death (PCD), characterized by appearance of cytoplasmic vacuoles and distended endoplasmic reticulum, in the absence of DNA fragmentation, apoptotic bodies and caspase activation. In contrast, SGIV induced typical apoptosis in non-host (fathead minnow, FHM) cells, as evidenced by caspase activation and DNA fragmentation, suggesting that SGIV infection induced nonapoptotic cell death by a cell type dependent fashion. Furthermore, viral replication was essential for SGIV induced nonapoptotic cell death, but not for apoptosis. Notably, the disruption of mitochondrial transmembrane potential (ΔΨm) and externalization of phosphatidylserine (PS) were not detected in EAGS cells but in FHM cells after SGIV infection. Moreover, the extracellular signal-regulated kinase (ERK) signaling was involved in SGIV infection induced nonapoptotic cell death and viral replication. This is a first demonstration of ERK-mediated nonapoptotic cell death induced by a DNA virus. These findings contribute to understanding the mechanisms of iridovirus pathogenesis.
Collapse
Affiliation(s)
- Xiaohong Huang
- Key Laboratory of Marine Bio-Resources Sustainable Utilization, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Modulatory profiling identifies mechanisms of small molecule-induced cell death. Proc Natl Acad Sci U S A 2011; 108:E771-80. [PMID: 21896738 DOI: 10.1073/pnas.1106149108] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cell death is a complex process that plays a vital role in development, homeostasis, and disease. Our understanding of and ability to control cell death is impeded by an incomplete characterization of the full range of cell death processes that occur in mammalian systems, especially in response to exogenous perturbations. We present here a general approach to address this problem, which we call modulatory profiling. Modulatory profiles are composed of the changes in potency and efficacy of lethal compounds produced by a second cell death-modulating agent in human cell lines. We show that compounds with the same characterized mechanism of action have similar modulatory profiles. Furthermore, clustering of modulatory profiles revealed relationships not evident when clustering lethal compounds based on gene expression profiles alone. Finally, modulatory profiling of compounds correctly predicted three previously uncharacterized compounds to be microtubule-destabilizing agents, classified numerous compounds that act nonspecifically, and identified compounds that cause cell death through a mechanism that is morphologically and biochemically distinct from previously established ones.
Collapse
|
16
|
Adaptation to moderate hypoxia protects cortical neurons against ischemia-reperfusion injury and excitotoxicity independently of HIF-1α. Exp Neurol 2011; 230:302-10. [PMID: 21619879 DOI: 10.1016/j.expneurol.2011.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 04/19/2011] [Accepted: 05/10/2011] [Indexed: 11/20/2022]
Abstract
Continuous exposure of cultured cortical neurons to moderate hypoxia (1% O(2)) elevates cellular accumulation of hypoxia-inducible factor-1α (HIF-1α) and improves basal survival of cultured cortical neurons. We examined the effects of adaptation to moderate hypoxia on the vulnerability of cultured neurons to the acute injury of simulated ischemia-reperfusion. Cortical neurons cultured continuously in 1% O(2) were markedly protected against simulated ischemia-reperfusion, with protection persisting through 72h after ischemia. Neurons from 1% O(2) conditions were also highly resistant to glutamate-induced NMDA receptor-dependent excitotoxic injury, despite expression of NMDA receptors at levels not significantly changed from controls. Inhibition of prolyl hydroxylase, mimicking cellular signaling effects of hypoxia including HIF-1α stabilization, also protected neurons against simulated ischemia-reperfusion injury. Nevertheless, genetic deletion of HIF-1α expression did not diminish the protection of neurons adapted to 1% O(2) from excitotoxicity or ischemia-reperfusion injury, nor did it prevent the protective effect of prolyl hydroxylase inhibition. We conclude that chronic exposure to moderate hypoxia, through HIF-1α-independent mechanisms, produces strong protective effects against excitotoxic and ischemia-reperfusion related injury.
Collapse
|
17
|
Ashpole NM, Hudmon A. Excitotoxic neuroprotection and vulnerability with CaMKII inhibition. Mol Cell Neurosci 2011; 46:720-30. [PMID: 21316454 DOI: 10.1016/j.mcn.2011.02.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 12/15/2022] Open
Abstract
Aberrant calcium signaling is a common feature of ischemia and multiple neurodegenerative diseases. While activation of calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is a key event in calcium signaling, its role in excitotoxicity is controversial. Our findings demonstrate neuroprotection in neuronal cultures treated with the small molecule (KN-93) and peptide (tat-AIP and tat-CN21) inhibitors of CaMKII immediately prior to excitotoxic glutamate/glycine insult. Unlike KN-93 which blocks CaMKII activation, but not constitutively active forms of CaMKII, tat-CN21 and tat-AIP significantly reduced excitotoxicity in cultured neurons when applied post-insult. We observed that the neuroprotective effects of tat-CN21 are greatest when applied before the toxic glutamate challenge and diminish with time, with the neuroprotection associated with CaMKII inhibition diminishing back to control 3h post glutamate insult. Mechanistically, tat-CN21 inhibition of CaMKII resulted in an increase in CaMKII activity and the percentage of soluble αCaMKII observed in neuronal lysates 24h following glutamate stimulation. To address the impact of prolonged CaMKII inhibition prior to excitotoxic insult, neuronal cultures were treated with CaMKII inhibitors overnight and then subjected to a sub-maximal excitotoxic insult. In this model, CaMKII inhibition prior to insult exacerbated neuronal death, suggesting that a loss of CaMKII enhances neuronal vulnerability to glutamate. Although changes in αCaMKII or NR2B protein levels are not responsible for this enhanced glutamate vulnerability, this process is blocked by the protein translation inhibitor cycloheximide. In total, the neuroprotection afforded by CaMKII inhibition can be seen as neuroprotective immediately surrounding the excitotoxic insult, whereas sustained CaMKII inhibition produced by excitotoxicity leads to neuronal death by enhancing neuronal vulnerability to glutamate.
Collapse
Affiliation(s)
- Nicole M Ashpole
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
18
|
Torriglia A, Valamanesh F, Behar-Cohen F. On the retinal toxicity of intraocular glucocorticoids. Biochem Pharmacol 2010; 80:1878-86. [DOI: 10.1016/j.bcp.2010.07.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Revised: 07/06/2010] [Accepted: 07/08/2010] [Indexed: 10/24/2022]
|
19
|
Hoffman-Goetz L, Pervaiz N, Packer N, Guan J. Freewheel training decreases pro- and increases anti-inflammatory cytokine expression in mouse intestinal lymphocytes. Brain Behav Immun 2010; 24:1105-15. [PMID: 20510350 DOI: 10.1016/j.bbi.2010.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 05/10/2010] [Accepted: 05/10/2010] [Indexed: 01/25/2023] Open
Abstract
Intestinal inflammation and inflammatory bowel diseases (IBD) may occur due to imbalances in pro- and anti-inflammatory cytokines. Long-term exercise reduces the risk for IBD. The purpose of this study was to determine the effect of long-term wheel running in healthy mice on intestinal lymphocyte (IL) expression of pro- and anti-inflammatory cytokine proteins. In addition, pro- and anti-apoptotic proteins and the percentage of early apoptotic, late apoptotic, and dead IL were measured with wheel running and following acute aerobic exercise. Female C57BL/6 mice were given 16 weeks of wheel running (WR) or a control condition (No WR) and at the end of training were assigned to a single acute treadmill exercise session with sacrifice immediately, 2h after, or 24h after completion of exercise, or were not run (sedentary) with respect to the acute treadmill exercise. Intestinal lymphocytes were assessed for pro-(TNF-α, IL-17) and anti-(IL-10) inflammatory, and pleiotropic (IL-6) cytokines, and pro-(caspase 3 and 7, AIF) and anti-(Bcl-2) apoptotic protein expression. The percent of early (Annexin(+)) and late (Annexin(+)PI(+)) apoptotic, and dead (PI(+)) IL was determined. WR mice had lower TNF-α and caspase 7, and higher IL-10 and IL-6 expression in IL than No WR mice. A single exposure to intense aerobic treadmill exercise increased pro-(TNF-α) and anti-(IL-10) inflammatory cytokine and pro-apoptotic protein (caspase 3) expression in IL. The percent of early and late apoptotic, and dead IL were higher after acute exercise. Although long-term voluntary wheel running did not protect against acute exercise-induced changes in IL cytokine expression or apoptosis, there was an overall 'anti-inflammatory' effect observed as a result of wheel running in healthy mice.
Collapse
Affiliation(s)
- L Hoffman-Goetz
- Department of Health Studies and Gerontology, University of Waterloo, Waterloo, Ontario, Canada.
| | | | | | | |
Collapse
|
20
|
Halterman MW, Gill M, DeJesus C, Ogihara M, Schor NF, Federoff HJ. The endoplasmic reticulum stress response factor CHOP-10 protects against hypoxia-induced neuronal death. J Biol Chem 2010; 285:21329-40. [PMID: 20448044 DOI: 10.1074/jbc.m109.095299] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hypoxia-induced gene expression is a critical determinant of neuron survival after stroke. Understanding the cell autonomous genetic program controlling adaptive and pathological transcription could have important therapeutic implications. To identify the factors that modulate delayed neuronal apoptosis after hypoxic injury, we developed an in vitro culture model that recapitulates these divergent responses and characterized the sequence of gene expression changes using microarrays. Hypoxia induced a disproportionate number of bZIP transcription factors and related targets involved in the endoplasmic reticulum stress response. Although the temporal and spatial aspects of ATF4 expression correlated with neuron loss, our results did not support the anticipated pathological role for delayed CHOP expression. Rather, CHOP deletion enhanced neuronal susceptibility to both hypoxic and thapsigargin-mediated injury and attenuated brain-derived neurotrophic factor-induced neuroprotection. Also, enforced expression of CHOP prior to the onset of hypoxia protected wild-type cultures against subsequent injury. Collectively, these findings indicate CHOP serves a more complex role in the neuronal response to hypoxic stress with involvement in both ischemic preconditioning and delayed neuroprotection.
Collapse
Affiliation(s)
- Marc W Halterman
- Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, USA.
| | | | | | | | | | | |
Collapse
|
21
|
Stoica BA, Faden AI. Cell death mechanisms and modulation in traumatic brain injury. Neurotherapeutics 2010; 7:3-12. [PMID: 20129492 PMCID: PMC2841970 DOI: 10.1016/j.nurt.2009.10.023] [Citation(s) in RCA: 217] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Accepted: 10/20/2009] [Indexed: 12/17/2022] Open
Abstract
Cell death after traumatic brain injury (TBI) is a major cause of neurological deficits and mortality. Understanding the mechanisms of delayed post-traumatic cell loss may lead to new therapies that improve outcome. Although TBI induces changes in multiple cell types, mechanisms of neuronal cell death have been the predominant focus. Recent work has emphasized the diversity of neuronal death phenotypes, which have generally been defined by either morphological or molecular changes. This diversity has led to confusing and at times contradictory nomenclature. Here we review the historical basis of proposed definitions of neuronal cell death, with the goal of clarifying critical research questions and implications for therapy in TBI. We believe that both morphological and molecular features must be used to clarify post-traumatic cell death and related therapeutic targets. Further, we underscore that the most effective neuroprotective strategies will need to target multiple pathways to reflect the regional and temporal changes underlying diverse neuronal cell death phenotypes.
Collapse
Affiliation(s)
- Bogdan A Stoica
- Shock, Trauma and Anesthesiology Research Center, National Study Center for Trauma and EMS, University of Maryland, School of Medicine, Baltimore, Maryland 21201, USA.
| | | |
Collapse
|
22
|
Liu J, Naegele JR, Lin SL. The DNA-PK catalytic subunit regulates Bax-mediated excitotoxic cell death by Ku70 phosphorylation. Brain Res 2009; 1296:164-75. [PMID: 19664609 DOI: 10.1016/j.brainres.2009.07.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 07/27/2009] [Accepted: 07/30/2009] [Indexed: 10/20/2022]
Abstract
DNA repair deficiency results in neurodegenerative disease and increased susceptibility to excitotoxic cell death, suggesting a critical but undefined role for DNA damage in neurodegeneration. We compared DNA damage, Ku70-Bax interaction, and Bax-dependent excitotoxic cell death in kainic acid-treated primary cortical neurons derived from both wild-type mice and mice deficient in the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) encoded by the Prkdc gene. In both wild-type and Prkdc(-/-) neurons, kainic acid treatment resulted in rapid induction of DNA damage (53BP1 foci formation) followed by nuclear pyknosis. Bax deficiency, by either Bax shRNA-mediated knockdown or gene deletion, protected wild-type and heterozygous but not Prkdc(-/-) neurons from kainate-induced excitotoxicity. Cotransfection of DNA-PKcs with Bax shRNA restored Bax shRNA-mediated neuroprotection in Prkdc(-/-) neurons, suggesting that DNA-PKcs is required for kainate-induced activation of the pro-apoptotic Bax pathway. Immunoprecipitation studies revealed that the DNA-PKcs-nonphosphorylatable Ku70 (S6A/S51A) bound 3- to 4-fold greater Bax than wild-type Ku70, suggesting that DNA-PKcs-mediated Ku70 phosphorylation causes release of Bax from Ku70. In support of this, kainic acid induced translocation of a Bax-EGFP fusion protein to the mitochondria in the presence of a cotransfected wild-type, but not mutant Ku70 (S6A/S51A) gene when examined at 4 and 8 h following kainate addition. We conclude that DNA-PKcs links DNA damage to Bax-dependent excitotoxic cell death, by phosphorylating Ku70 on serines 6 and/or 51, to initiate Bax translocation to the mitochondria and directly activate a pro-apoptotic Bax-dependent death cascade.
Collapse
Affiliation(s)
- Jia Liu
- Program in Neuroscience and Behavior, Department of Biology, Wesleyan University, Middletown, CT 06459-0170, USA
| | | | | |
Collapse
|
23
|
Watson CJ, Kreuzaler PA. The role of cathepsins in involution and breast cancer. J Mammary Gland Biol Neoplasia 2009; 14:171-9. [PMID: 19437107 DOI: 10.1007/s10911-009-9126-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 04/23/2009] [Indexed: 12/13/2022] Open
Abstract
Cysteine cathepsins are proteolytic enzymes that reside in endolysosomal vesicles. Some are expressed constitutively while others are transcriptionally regulated. However, the expression and subcellular localization of cathepsins changes during cancer progression and cathepsins have been shown to be causally involved in various aspects of tumorigenesis including metastasis. The use of mouse models of breast cancer genetically ablated for cathepsin B has shown that both the growth of the primary tumor and the extend of lung metastasis is reduced by the loss of cathepsin B. The role of cathepsins in involution of the mammary gland has received little attention although it is clear that cathepsins are involved in tissue remodeling in the second phase of involution. We discuss here the roles of cathepsins and their endogenous inhibitors in breast tumorigenesis and post-lactational involution.
Collapse
|
24
|
Paris I, Perez-Pastene C, Couve E, Caviedes P, LeDoux S, Segura-Aguilar J. Copper dopamine complex induces mitochondrial autophagy preceding caspase-independent apoptotic cell death. J Biol Chem 2009; 284:13306-13315. [PMID: 19265190 PMCID: PMC2679430 DOI: 10.1074/jbc.m900323200] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 02/25/2009] [Indexed: 12/19/2022] Open
Abstract
Parkinsonism is one of the major neurological symptoms in Wilson disease, and young workers who worked in the copper smelting industry also developed Parkinsonism. We have reported the specific neurotoxic action of copper dopamine complex in neurons with dopamine uptake. Copper dopamine complex (100 microm) induces cell death in RCSN-3 cells by disrupting the cellular redox state, as demonstrated by a 1.9-fold increase in oxidized glutathione levels and a 56% cell death inhibition in the presence of 500 microm ascorbic acid; disruption of mitochondrial membrane potential with a spherical shape and well preserved morphology determined by transmission electron microscopy; inhibition (72%, p < 0.001) of phosphatidylserine externalization with 5 microm cyclosporine A; lack of caspase-3 activation; formation of autophagic vacuoles containing mitochondria after 2 h; transfection of cells with green fluorescent protein-light chain 3 plasmid showing that 68% of cells presented autophagosome vacuoles; colocalization of positive staining for green fluorescent protein-light chain 3 and Rhod-2AM, a selective indicator of mitochondrial calcium; and DNA laddering after 12-h incubation. These results suggest that the copper dopamine complex induces mitochondrial autophagy followed by caspase-3-independent apoptotic cell death. However, a different cell death mechanism was observed when 100 microm copper dopamine complex was incubated in the presence of 100 microm dicoumarol, an inhibitor of NAD(P)H quinone:oxidoreductase (EC 1.6.99.2, also known as DT-diaphorase and NQ01), because a more extensive and rapid cell death was observed. In addition, cyclosporine A had no effect on phosphatidylserine externalization, significant portions of compact chromatin were observed within a vacuolated nuclear membrane, DNA laddering was less pronounced, the mitochondria morphology was more affected, and the number of cells with autophagic vacuoles was a near 4-fold less.
Collapse
Affiliation(s)
- Irmgard Paris
- Programme of Molecular and Clinical Pharmacology, Faculty of Medicine, Casilla 70000, Santiago 7, Chile; Department of Basic Sciences, Universidad Santo Tomás, 2561780 Viña del Mar, Chile; Department of Biological Sciences, Universidad Andrés Bello, 2561156 Viña del Mar, Chile
| | - Carolina Perez-Pastene
- Programme of Molecular and Clinical Pharmacology, Faculty of Medicine, Casilla 70000, Santiago 7, Chile
| | - Eduardo Couve
- Department of Biology, University of Valparaiso, Casilla 5030, Valparaiso, Chile
| | - Pablo Caviedes
- Programme of Molecular and Clinical Pharmacology, Faculty of Medicine, Casilla 70000, Santiago 7, Chile
| | - Susan LeDoux
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama 36608
| | - Juan Segura-Aguilar
- Programme of Molecular and Clinical Pharmacology, Faculty of Medicine, Casilla 70000, Santiago 7, Chile.
| |
Collapse
|
25
|
Jiménez C, Capasso JM, Edelstein CL, Rivard CJ, Lucia S, Breusegem S, Berl T, Segovia M. Different ways to die: cell death modes of the unicellular chlorophyte Dunaliella viridis exposed to various environmental stresses are mediated by the caspase-like activity DEVDase. JOURNAL OF EXPERIMENTAL BOTANY 2009; 60:815-28. [PMID: 19251986 PMCID: PMC2652065 DOI: 10.1093/jxb/ern330] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 11/20/2008] [Accepted: 11/24/2008] [Indexed: 05/23/2023]
Abstract
Programmed cell death is necessary for homeostasis in multicellular organisms and it is also widely recognized to occur in unicellular organisms. However, the mechanisms through which it occurs in unicells, and the enzymes involved within the final response is still the subject of heated debate. It is shown here that exposure of the unicellular microalga Dunaliella viridis to several environmental stresses, induced different cell death morphotypes, depending on the stimulus received. Senescent cells demonstrated classical and unambiguous apoptotic-like characteristics such as chromatin condensation, DNA fragmentation, intact organelles, and blebbing of the cell membrane. Acute heat shock caused general swelling and altered plasma membrane, but the presence of chromatin clusters and DNA strand breaks suggested a necrotic-like event. UV irradiated cells presented changes typical for necrosis, together with apoptotic characteristics resembling an intermediate cell-death phenotype termed aponecrosis-like. Cells subjected to hyperosmotic shock revealed chromatin spotting without DNA fragmentation, and extensive cytoplasmic swelling and vacuolization, comparable to a paraptotic-like cell death phenotype. Nitrogen-starved cells showed pyknosis, blebbing, and cytoplasmic consumption, indicating a similarity to autophagic/vacuolar-like cell death. The caspase-like activity DEVDase was measured by using the fluorescent substrate Ac-DEVD-AMC and antibodies against the human caspase-3 active enzyme cross-reacted with bands, the intensity of which paralleled the activity. All the environmental stresses tested produced a substantial increase in both DEVDase activity and protein levels. The irreversible caspase-3 inhibitor Z-DEVD-FMK completely inhibited the enzymatic activity whereas serine and aspartyl proteases inhibitors did not. These results show that cell death in D. viridis does not conform to a single pattern and that environmental stimuli may produce different types of cell death depending on the type and intensity of the stimulus, all of which help to understand the cell death-dependent and cell death-independent functions of caspase-like proteins. Hence, these data support the theory that alternative, non-apoptotic programmed cell death (PCDs), exist either in parallel or in an independent manner with apoptosis and were already present in single-celled organisms that evolved some 1.2-1.6 billion years ago.
Collapse
Affiliation(s)
- Carlos Jiménez
- Department of Ecology, Faculty of Sciences, University of Málaga, Bvd. Louis Pasteur s/n, E-29071 Málaga, Spain
| | - Juan M. Capasso
- Department of Renal Diseases and Hypertension, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - Charles L. Edelstein
- Department of Renal Diseases and Hypertension, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - Christopher J. Rivard
- Department of Renal Diseases and Hypertension, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - Scott Lucia
- Department of Pathology, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - Sophia Breusegem
- Department of Renal Diseases and Hypertension, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - Tomás Berl
- Department of Renal Diseases and Hypertension, School of Medicine, University of Colorado Health Sciences Center, 4200 E. 9th Av. Denver, CO 80262, USA
| | - María Segovia
- Department of Ecology, Faculty of Sciences, University of Málaga, Bvd. Louis Pasteur s/n, E-29071 Málaga, Spain
| |
Collapse
|
26
|
Abstract
High-content analysis (HCA) combines automated microscopy and automated image analysis to quantify complex cellular anatomy and biochemistry objectively, accurately and quickly. High-content assays that are applicable to neuroscience include those that can quantify various aspects of dendritic trees, protein aggregation, transcription factor translocation, neurotransmitter receptor internalization, neuron and synapse number, cell migration, proliferation and apoptosis. The data that are generated by HCA are rich and multiplexed. HCA thus provides a powerful high-throughput tool for neuroscientists.
Collapse
|
27
|
Van Vickle GD, Esh CL, Kokjohn TA, Patton RL, Kalback WM, Luehrs DC, Beach TG, Newel AJ, Lopera F, Ghetti B, Vidal R, Castaño EM, Roher AE. Presenilin-1 280Glu-->Ala mutation alters C-terminal APP processing yielding longer abeta peptides: implications for Alzheimer's disease. Mol Med 2008; 14:184-94. [PMID: 18317569 DOI: 10.2119/2007-00094.vanvickle] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 01/14/2008] [Indexed: 01/17/2023] Open
Abstract
Presenilin (PS) mutations enhance the production of the Abeta42 peptide that is derived from the amyloid precursor protein (APP). The pathway(s) by which the Abeta42 species is preferentially produced has not been elucidated, nor is the mechanism by which PS mutations produce early-onset dementia established. Using a combination of histological, immunohistochemical, biochemical, and mass spectrometric methods, we examined the structural and morphological nature of the amyloid species produced in a patient expressing the PS1 280Glu-->Ala familial Alzheimer's disease mutation. Abundant diffuse plaques were observed that exhibited a staining pattern and morphology distinct from previously described PS cases, as well as discreet amyloid plaques within the white matter. In addition to finding increased amounts of CT99 and Abeta42 peptides, our investigation revealed the presence of a complex array of Abeta peptides substantially longer than 42/43 amino acid residue species. The increased hydrophobic nature of longer Abeta species retained within the membrane walls could impact the structure and function of plasma membrane and organelles. These C-terminally longer peptides may, through steric effects, dampen the rate of turnover by critical amyloid degrading enzymes such as neprilysin and insulin degrading enzyme. A complete understanding of the deleterious side effects of membrane bound Abeta as a consequence of gamma-secretase alterations is needed to understand Alzheimer's disease pathophysiology and will aid in the design of therapeutic interventions.
Collapse
Affiliation(s)
- Gregory D Van Vickle
- The Longtine Center for Molecular Biology and Genetics, Sun Health Research Institute, Sun City, Arizona 85351, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Anthrax lethal toxin and Salmonella elicit the common cell death pathway of caspase-1-dependent pyroptosis via distinct mechanisms. Proc Natl Acad Sci U S A 2008; 105:4312-7. [PMID: 18337499 DOI: 10.1073/pnas.0707370105] [Citation(s) in RCA: 325] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Caspase-1 cleaves the inactive IL-1beta and IL-18 precursors into active inflammatory cytokines. In Salmonella-infected macrophages, caspase-1 also mediates a pathway of proinflammatory programmed cell death termed "pyroptosis." We demonstrate active caspase-1 diffusely distributed in the cytoplasm and localized in discrete foci within macrophages responding to either Salmonella infection or intoxication by Bacillus anthracis lethal toxin (LT). Both stimuli triggered caspase-1-dependent lysis in macrophages and dendritic cells. Activation of caspase-1 by LT required binding, uptake, and endosome acidification to mediate translocation of lethal factor (LF) into the host cell cytosol. Catalytically active LF cleaved cytosolic substrates and activated caspase-1 by a mechanism involving proteasome activity and potassium efflux. LT activation of caspase-1 is known to require the inflammasome adapter Nalp1. In contrast, Salmonella infection activated caspase-1 through an independent pathway requiring the inflammasome adapter Ipaf. These distinct mechanisms of caspase-1 activation converged on a common pathway of caspase-1-dependent cell death featuring DNA cleavage, cytokine activation, and, ultimately, cell lysis resulting from the formation of membrane pores between 1.1 and 2.4 nm in diameter and pathological ion fluxes that can be blocked by glycine. These findings demonstrate that distinct activation pathways elicit the conserved cell death effector mechanism of caspase-1-mediated pyroptosis and support the notion that this pathway of proinflammatory programmed cell death is broadly relevant to cell death and inflammation invoked by diverse stimuli.
Collapse
|
29
|
Abstract
DNA damage can, but does not always, induce cell death. While several pathways linking DNA damage signals to mitochondria-dependent and -independent death machineries have been elucidated, the connectivity of these pathways is subject to regulation by multiple other factors that are not well understood. We have proposed two conceptual models to explain the delayed and variable cell death response to DNA damage: integrative surveillance versus autonomous pathways. In this review, we discuss how these two models may explain the in vivo regulation of cell death induced by ionizing radiation (IR) in the developing central nervous system, where the death response is regulated by radiation dose, cell cycle status and neuronal development.
Collapse
|
30
|
Chu CT. Eaten alive: autophagy and neuronal cell death after hypoxia-ischemia. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 172:284-7. [PMID: 18202199 DOI: 10.2353/ajpath.2008.071064] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Charleen T Chu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
31
|
Mur LAJ, Kenton P, Lloyd AJ, Ougham H, Prats E. The hypersensitive response; the centenary is upon us but how much do we know? JOURNAL OF EXPERIMENTAL BOTANY 2008; 59:501-20. [PMID: 18079135 DOI: 10.1093/jxb/erm239] [Citation(s) in RCA: 408] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
With the centenary of the first descriptions of 'hypersensitiveness' following pathogenic challenge upon us, it is appropriate to assess our current understanding of the hypersensitive response (HR) form of cell death. In recent decades our understanding of the initiation, associated signalling, and some important proteolytic events linked to the HR has dramatically increased. Genetic approaches are increasingly elucidating the function of the HR initiating resistance genes and there have been extensive analyses of death-associated signals, calcium, reactive oxygen species (ROS), nitric oxide, salicylic acid, and now sphingolipids. At the same time, attempts to draw parallels between mammalian apoptosis and the HR have been largely unsuccessful and it may be better to consider the HR to be a distinctive form of plant cell death. We will consider if the HR form of cell death may occur through metabolic dysfunction in which malfunctioning organelles may play a major role. This review will highlight that although our knowledge of parts of the HR is excellent, a comprehensive molecular model is still to be attained.
Collapse
Affiliation(s)
- Luis A J Mur
- University of Wales Aberystwyth, Institute of Biological Sciences, Aberystwyth, Ceredigion SY23 2DA, UK.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Stroke is a devastating disorder that significantly contributes to death, disability, and healthcare costs. New therapeutic strategies have been recently focusing on the development of neuroprotective agents that could halt the underlying mechanisms of neuronal death leading to brain damage. Accumulating evidence implicates proteins that are normally involved in the regulation of the cell cycle to neuronal death following ischemic insult, suggesting that these proteins could be suitable targets for stroke therapy. In this brief review, we present in vitro and in vivo arguments linking cell cycle molecules, i.e., cyclins, mitotic cyclin-dependent kinases (Cdks), as well as non-mitotic Cdk5, to ischemic neuronal death. We also report the evaluation of the potential of Cdk inhibitors as neuroprotective strategy for ischemic injury.
Collapse
Affiliation(s)
- Serge Timsit
- Faculté de Médecine et des Sciences de la Santé de Brest, Brest, France.
| | | |
Collapse
|
33
|
Golstein P, Kroemer G. A multiplicity of cell death pathways. Symposium on apoptotic and non-apoptotic cell death pathways. EMBO Rep 2007; 8:829-33. [PMID: 17721445 PMCID: PMC1973949 DOI: 10.1038/sj.embor.7401042] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2007] [Accepted: 06/15/2007] [Indexed: 01/08/2023] Open
Affiliation(s)
- Pierre Golstein
- Centre d'Immunologie de Marseille–Luminy, Case 906, Avenue de Luminy, 13288 Marseille Cedex 9, France
- Centre d'Immunologie de Marseille–Luminy, Case 906, Avenue de Luminy, 13288 Marseille Cedex 9, France
- Centre d'Immunologie de Marseille–Luminy, Case 906, Avenue de Luminy, 13288 Marseille Cedex 9, France
| | - Guido Kroemer
- INSERM Unit ‘Apoptosis, Cancer and Immunity', Institut Gustave Roussy, PR1, 39 rue Camille Desmoulins, F-94805 Villejuif, France
- INSERM Unit ‘Apoptosis, Cancer and Immunity', Institut Gustave Roussy, PR1, 39 rue Camille Desmoulins, F-94805 Villejuif, France
- INSERM Unit ‘Apoptosis, Cancer and Immunity', Institut Gustave Roussy, PR1, 39 rue Camille Desmoulins, F-94805 Villejuif, France
- Tel: +33 1 42 11 60 46; Fax: +33 1 42 11 60 47;
| |
Collapse
|
34
|
Li D, Shao Z, Hoek TLV, Brorson JR. Reperfusion accelerates acute neuronal death induced by simulated ischemia. Exp Neurol 2007; 206:280-7. [PMID: 17599834 PMCID: PMC3648807 DOI: 10.1016/j.expneurol.2007.05.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2007] [Revised: 04/12/2007] [Accepted: 05/11/2007] [Indexed: 10/23/2022]
Abstract
Observations in real time can provide insights into the timing of injury and the mechanisms of damage in neural ischemia-reperfusion. Continuous digital imaging of morphology and cell viability was applied in a novel model of simulated ischemia-reperfusion in cultured cortical neurons, consisting of exposure to severe hypoxia combined with glucose deprivation, mild acidosis, hypercapnia, and elevated potassium, followed by return of oxygenated, glucose-containing physiological saline. Substantial acute injury resulted following 1 h of simulated ischemia, with 36+/-8% neurons dying within 2 h of reperfusion. Inclusion of moderate glutamate elevation (30 microM) in the simulation of ischemia increased the acute neuronal death to 51+/-6% at 2 h of reperfusion. While some swelling and neuritic breakdown occurred during ischemia, particularly with inclusion of glutamate, neuronal death, as marked by loss of somatic membrane integrity, was entirely restricted to the reperfusion phase. Morphological and cytoskeletal changes suggested a predominance of necrotic death in the acute phase of reperfusion, with more complete delayed death accompanied by some apoptotic features occurring over subsequent days. Prolonged simulated ischemia, without reperfusion, did not induce significant acute neuronal death even when extended to 3 h. We conclude that while morphological changes suggesting initiation of neuronal injury appear during severe simulated ischemia, the irreversible injury signaled by membrane breakdown is accelerated by the events of reperfusion itself.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Neurology, The University of Chicago
| | - Zuohui Shao
- Department of Medicine, The University of Chicago
| | | | | |
Collapse
|