1
|
Chen Y, Wang K, Di J, Guan C, Wang S, Li Q, Qu Y. Mutation of the BAG-1 domain decreases its protective effect against hypoxia/reoxygenation by regulating HSP70 and the PI3K/AKT signalling pathway in SY-SH5Y cells. Brain Res 2020; 1751:147192. [PMID: 33152339 DOI: 10.1016/j.brainres.2020.147192] [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: 06/06/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 10/23/2022]
Abstract
BCL-2-associated athanogene-1 (BAG-1) is a multifunctional protein that was first identified as a binding partner of BCL-2. Our previous results indicated that BAG-1 large (BAG-1L) overexpression significantly increases cell viability and decreases apoptosis by upregulating HSP70 and p-AKT in response to hypoxia/reoxygenation in SY-SH5Y cells. However, the functional domain of BAG-1L that exerts these protective effects against hypoxic damage has not been identified. In this study, we examined changes in HSP70 and p-AKT protein levels in SH-SY5Y cells with or without BAG-1L domain mutation after six hours of hypoxia/reoxygenation treatment. The BAG-1 domain mutant (BAG-1MUT) attenuated neuronal viability and proliferation while enhancing apoptosis after hypoxia/reoxygenation, which was achieved in part by inhibiting the HSP70 and p-AKT signalling pathways. This evidence illustrates that the BAG-1 domain plays a key role in protecting cells from hypoxia/reoxygenation injury.
Collapse
Affiliation(s)
- Ying Chen
- School of Nursing, Medical College of Qingdao University, Qingdao 26600, Shandong, China; Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Keke Wang
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Jie Di
- School of Nursing, Medical College of Qingdao University, Qingdao 26600, Shandong, China; Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Chun Guan
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Sumei Wang
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Qingshu Li
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China.
| | - Yan Qu
- Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
2
|
Abstract
Since ancient times, opioids have been used clinically and abused recreationally. In the early stages (about 1,000 AD) of opium history, an Arab physician, Avicenna, administered opioids to control diarrhea and eye diseases. 1 Opioids have very strong pain relieving properties and they also regulate numerous cellular responses. Opioid receptors are expressed throughout the body, including the nervous system, heart, lungs, liver, gastrointestinal tract, and retina. 2-6 Delta opioid receptors (DORs) are a very attractive target from the perspective of both receptor function and their therapeutic potential. Due to a rapid progress in mouse mutagenesis and development of small molecules as DOR agonist, novel functions and roles of DORs have emerged in recent years. This review article focuses on the recent advances in the neuroprotective roles of DOR agonists in general and retina neuroprotection in particular. Rather than being exhaustive, this review highlights the selected studies of DOR function in neuroprotection. We also highlight our preclinical studies using rodent models to demonstrate the potentials of DOR agonists for retinal neuroprotection. Based on existing literature and our recently published data on the eye, DOR agonists possess therapeutic abilities that protect the retina and optic nerve injury against glaucoma and perhaps other retinopathies as well. This review also highlights the signaling events associated with DOR for neuroprotection in the eye. There is a need for translational research on DORs to recognize their potential for clinical application such as in glaucoma.
Collapse
Affiliation(s)
- Shahid Husain
- Hewitt Laboratory of the Ola B. Williams Glaucoma Center, Department of Ophthalmology, Medical University of South Carolina , Charleston, South Carolina
| |
Collapse
|
3
|
Song Y, Li Z, Wang Y, Qu Y, Li Q, Man X, Wang F, Hu D. Inhibition of BAG‐1 induced SH‐SY5Y cell apoptosis without affecting Hsp70 expression. J Cell Biochem 2019; 121:1728-1735. [PMID: 31609014 DOI: 10.1002/jcb.29408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 08/28/2019] [Indexed: 01/10/2023]
Affiliation(s)
- Yan‐Kun Song
- Department of Pediatric Emergency The Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Zhi Li
- Department of Intensive Care Unit Qingdao Municipal Hospital Affiliated to Qingdao University Qingdao Shandong China
| | - Yun Wang
- Department of Pediatric Emergency The Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Yan Qu
- Department of Intensive Care Unit Qingdao Municipal Hospital Affiliated to Qingdao University Qingdao Shandong China
| | - Qing‐Shu Li
- Department of Intensive Care Unit Qingdao Municipal Hospital Affiliated to Qingdao University Qingdao Shandong China
| | - Xiao‐Yun Man
- Department of Intensive Care Unit Qingdao Municipal Hospital Affiliated to Qingdao University Qingdao Shandong China
| | - Feng‐Tao Wang
- Department of Pediatric Emergency The Affiliated Hospital of Qingdao University Qingdao Shandong China
| | - Dan Hu
- Department of Intensive Care Unit Qingdao Municipal Hospital Affiliated to Qingdao University Qingdao Shandong China
| |
Collapse
|
4
|
BAG-1L Protects SH-SY5Y Neuroblastoma Cells Against Hypoxia/Re-oxygenation Through Up-Regulating HSP70 and Activating PI3K/AKT Signaling Pathway. Neurochem Res 2017; 42:2861-2868. [PMID: 28523530 DOI: 10.1007/s11064-017-2304-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 05/07/2017] [Accepted: 05/11/2017] [Indexed: 01/01/2023]
Abstract
BCL-2-associated athanogene-1(BAG-1) is a multifunctional and anti-apoptotic protein that was first identified as a binding partner of BCL-2. But the effects and mechanisms for BAG-1 against hypoxic damage is unclear up to now. Whether BAG-1 could protect the human brain against hypoxic damage through up-regulating 70 kDa heat shock proteins (HSP70) and PI3K/AKT pathway activation? In present study, we examined the changes of HSP70 and AKT and p-AKT protein level in SH-SY5Y cells with BAG-1L gene over-expression subjected to hypoxia/re-oxygenation injury. BAG-1L over-expression increased neuronal viability, and it reduced apoptosis of neurons after hypoxia/re-oxygenation for 8 h. BAG-1L over-expression enhanced the HSP70 protein levels and increased p-AKT/total AKT ratio after hypoxia/re-oxygenation for 8 h. These results suggest that BAG-1L over-expression protects against hypoxia/re-oxygenation injury, at least in part, by interacting with HSP70, and by accelerating the activation of PI3K/AKT pathways.
Collapse
|
5
|
Staples M, Acosta S, Tajiri N, Pabon M, Kaneko Y, Borlongan CV. Delta opioid receptor and its peptide: a receptor-ligand neuroprotection. Int J Mol Sci 2013; 14:17410-9. [PMID: 23979422 PMCID: PMC3794733 DOI: 10.3390/ijms140917410] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 08/15/2013] [Accepted: 08/19/2013] [Indexed: 11/29/2022] Open
Abstract
In pursuit of neurological therapies, the opioid system, specifically delta opioid receptors and delta opioid peptides, demonstrates promising therapeutic potential for stroke, Parkinson’s disease, and other degenerative neurological conditions. Recent studies offer strong evidence in support of the therapeutic use of delta opioid receptors, and provide insights into the underlying mechanisms of action. Delta opioid receptors have been shown to confer protective effects by mediating ionic homeostasis and activating endogenous neuroprotective pathways. Additionally, delta opioid agonists such as (D-Ala 2, D-Leu 5) enkephalin (DADLE) have been shown to decrease apoptosis and promote neuronal survival. In its entirety, the delta opioid system represents a promising target for neural therapies.
Collapse
Affiliation(s)
- Meaghan Staples
- Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL 33612, USA.
| | | | | | | | | | | |
Collapse
|
6
|
Increased expression of BAG-1 in rat brain cortex after traumatic brain injury. J Mol Histol 2012; 43:335-42. [DOI: 10.1007/s10735-012-9408-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/19/2012] [Indexed: 12/14/2022]
|
7
|
Borlongan CV, Hayashi T, Oeltgen PR, Su TP, Wang Y. Hibernation-like state induced by an opioid peptide protects against experimental stroke. BMC Biol 2009; 7:31. [PMID: 19534760 PMCID: PMC2708132 DOI: 10.1186/1741-7007-7-31] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 06/17/2009] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Delta opioid peptide [D-ala2,D-leU5]enkephalin (DADLE) induces hibernation in summer ground squirrels, and enhances preservation and survival of isolated or transplanted lungs and hearts. In the present study, we investigated the protective effect of DADLE in the central nervous system. RESULTS Adult Sprague-Dawley rats were pretreated with DADLE (4 mg/kg every 2 h x 4 injections, i.p.) or saline prior to unilateral occlusion of the middle cerebral artery (MCA). Daily behavioral tests revealed that ischemic animals treated with DADLE did not show any significant behavioral dysfunctions compared with saline-treated ischemic animals. Opioid antagonists only transiently inhibited the protective effect of DADLE, indicating the participation of non-opioid mechanisms in DADLE neuroprotection. Histological examination using triphenyltetrazolium chloride (TTC) revealed that brains from ischemic animals treated with DADLE, either alone or with adjuvant opioid blockers, exhibited almost completely intact striata. In contrast, brains from ischemic animals that received saline showed significant infarction in the lateral striatum. Analyses of apoptotic cell death revealed a significant increase in the p-53 mRNA expression in the striatum of ischemic animals that received saline, while those that received DADLE exhibited near normal striatal p-53 expression. This protective effect was accompanied by significant increments in protein levels of glial cell line-derived neurotrophic factor in the striatum of DADLE-treated ischemic animals. CONCLUSION These results indicate that DADLE protected against necrotic and apoptotic cell death processes associated with ischemia-reperfusion injury. The present study demonstrates that delta opioids are crucially involved in stroke, suggesting that the opioid system is important in the study of brain injury and protection.
Collapse
Affiliation(s)
- Cesar V Borlongan
- National Institutes of Health, National Institute on Drug Abuse Intramural Research Program, Cellular Neurobiology Branch, Baltimore, MD, USA.
| | | | | | | | | |
Collapse
|
8
|
Curcio C, Asheld JJ, Chabla JM, Ayubcha D, Hallas BH, Horowitz JM, Torres G. Expression profile of Bag 1 in the postmortem brain. J Chem Neuroanat 2006; 32:191-5. [PMID: 17046197 PMCID: PMC1769523 DOI: 10.1016/j.jchemneu.2006.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 09/08/2006] [Accepted: 09/08/2006] [Indexed: 12/15/2022]
Abstract
Bag 1 is a protein intimately involved in signaling pathways that regulate cell survival. Here we examined the expression profile of Bag 1 in the brain to consider issues associated with the sampling of anti-apoptotic proteins in a rat model of the human postmortem process. Following a 4h postmortem interval, we analyzed the hippocampus of rats maintained at 24 or 4 degrees C storage temperatures using immunocytochemical and Western blotting techniques. Remarkably, postmortem tissue (up to 4h) showed a significant and prominent up-regulation of Bag 1 in CA1 and CA3 subfields of the hippocampal formation. Over-expression of Bag 1, however, could only be traced down to a storage temperature of 24 degrees C. These data suggest that storage temperatures, but not postmortem intervals, significantly affect the expression profile and cellular stability of Bag 1 proteins.
Collapse
Affiliation(s)
- Christine Curcio
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| | - John J. Asheld
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| | - Janet M. Chabla
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| | - Diana Ayubcha
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| | - Brian H. Hallas
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| | - Judith M. Horowitz
- Clinical Neuroscience Laboratory, Medaille College, Buffalo, New York 14214 USA
| | - German Torres
- Department of Neuroscience, New York College of Osteopathic Medicine of New York Institute of Technology, Old Westbury New York, 11568 USA
| |
Collapse
|
9
|
Zhou R, Gray NA, Yuan P, Li X, Chen J, Chen G, Damschroder-Williams P, Du J, Zhang L, Manji HK. The anti-apoptotic, glucocorticoid receptor cochaperone protein BAG-1 is a long-term target for the actions of mood stabilizers. J Neurosci 2006; 25:4493-502. [PMID: 15872096 PMCID: PMC6725025 DOI: 10.1523/jneurosci.4530-04.2005] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increasing data suggest that impairments of cellular plasticity/resilience underlie the pathophysiology of bipolar disorder. A series of microarray studies with validating criteria have recently revealed a common, novel target for the long-term actions of the structurally highly dissimilar mood stabilizers lithium and valproate: BAG-1 [BCL-2 (B-cell CLL/lymphoma 2)-associated athanogene]. Because BAG-1 attenuates glucocorticoid receptor (GR) nuclear translocation, activates ERK (extracellular signal-regulated kinase) MAP (mitogen-activated protein) kinases, and potentiates anti-apoptotic functions of BCL-2, extensive additional studies were undertaken. Chronic administration of both agents at therapeutic doses increased the expression of BAG-1 in rat hippocampus. Furthermore, these findings were validated at the protein level, and the effects were seen in a time frame consistent with therapeutic effects and were specific for mood stabilizers. Functional studies showed that either lithium or valproate, at therapeutically relevant levels, inhibited dexamethasone-induced GR nuclear translocation and inhibited GR transcriptional activity. Furthermore, small interfering RNA studies showed that these inhibitory effects on GR activity were mediated, at least in part, through BAG-1. The observation that BAG-1 inhibits glucocorticoid activation suggests that mood stabilizers may counteract the deleterious effects of hypercortisolemia seen in bipolar disorder by upregulating BAG-1. Additionally, these studies suggest that regulation of GR-mediated plasticity may play a role in the treatment of bipolar disorder and raise the possibility that agents affecting BAG-1 more directly may represent novel therapies for this devastating illness.
Collapse
Affiliation(s)
- Rulun Zhou
- Laboratory of Molecular Pathophysiology, National Institute of Mental Health, Bethesda, Maryland 20852, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kermer P, Digicaylioglu MH, Kaul M, Zapata JM, Krajewska M, Stenner-Liewen F, Takayama S, Krajewski S, Lipton SA, Reed JC. BAG1 over-expression in brain protects against stroke. Brain Pathol 2004; 13:495-506. [PMID: 14655755 PMCID: PMC8096046 DOI: 10.1111/j.1750-3639.2003.tb00480.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The co-chaperone BAG1 binds and regulates 70 kDa heat shock proteins (Hsp70/Hsc70) and exhibits cytoprotective activity in cell culture models. Recently, we observed that BAG1 expression is induced during neuronal differentiation in the developing brain. However, the in vivo effects of BAG1 during development and after maturation of the central nervous system have never been examined. We generated transgenic mice over-expressing BAG1 in neurons. While brain development was essentially normal, cultured cortical neurons from transgenic animals exhibited resistance to glutamate-induced, apoptotic neuronal death. Moreover, in an in vivo stroke model involving transient middle cerebral artery occlusion, BAG1 transgenic mice demonstrated decreased mortality and substantially reduced infarct volumes compared to wild-type littermates. Interestingly, brain tissue from BAG1 transgenic mice contained higher levels of neuroprotective Hsp70/Hsc70 protein but not mRNA, suggesting a potential mechanism whereby BAG1 exerts its anti-apoptotic effects. In summary, BAG1 displays potent neuroprotective activity in vivo against stroke, and therefore represents an interesting target for developing new therapeutic strategies including gene therapy and small-molecule drugs for reducing brain injury during cerebral ischemia and neurodegenerative diseases.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Blotting, Northern
- Blotting, Western
- Brain/anatomy & histology
- Brain/metabolism
- Brain/pathology
- Brain Chemistry
- Cell Death
- Cells, Cultured
- DNA-Binding Proteins
- Disease Models, Animal
- Heat-Shock Proteins/metabolism
- Immunohistochemistry
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Male
- Membrane Proteins
- Mice
- Mice, Transgenic
- Microtubule-Associated Proteins/metabolism
- Neurons/metabolism
- Polymerase Chain Reaction/methods
- Proteins/metabolism
- RNA, Messenger/biosynthesis
- Regional Blood Flow
- Staining and Labeling
- Stroke/metabolism
- Stroke/pathology
- Time Factors
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Transfection
Collapse
Affiliation(s)
- Pawel Kermer
- Department of Neurology, University of Goettingen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Townsend PA, Cutress RI, Sharp A, Brimmell M, Packham G. BAG-1: a multifunctional regulator of cell growth and survival. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1603:83-98. [PMID: 12618309 DOI: 10.1016/s0304-419x(03)00002-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BAG-1 is multifunctional protein which interacts with a wide range of cellular targets to regulate growth control pathways important for normal and malignant cells, including apoptosis, signaling, proliferation, transcription and cell motility. Of particular relevance to tumour cells, BAG-1 interacts with the anti-apoptotic BCL-2 protein, various nuclear hormone receptors and the 70 kDa heat shock proteins, Hsc70 and Hsp70. Interaction with chaperones may account for many of the pleiotropic effects associated with BAG-1 overexpression. Recent studies have shown that BAG-1 expression is frequently altered in malignant cells, and BAG-1 expression may have clinical value as a prognostic/predictive marker. This review summarises current understanding of molecular mechanisms of BAG-1 expression and function.
Collapse
Affiliation(s)
- Paul A Townsend
- Cancer Research UK Oncology Unit, Cancer Sciences Division, School of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | | | | | | | | |
Collapse
|
12
|
Perkins D, Pereira EFR, Aurelian L. The herpes simplex virus type 2 R1 protein kinase (ICP10 PK) functions as a dominant regulator of apoptosis in hippocampal neurons involving activation of the ERK survival pathway and upregulation of the antiapoptotic protein Bag-1. J Virol 2003; 77:1292-305. [PMID: 12502846 PMCID: PMC140789 DOI: 10.1128/jvi.77.2.1292-1305.2003] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Herpes simplex virus types 1 and 2 (HSV-1 and HSV-2) can trigger or block apoptosis in a cell type-dependent manner. We have recently shown that the protein kinase activity of the large subunit of the HSV-2 ribonucleotide reductase (R1) protein (ICP10 PK) blocks apoptosis in cultured hippocampal neurons by activating the extracellular signal-regulated kinase (ERK) survival pathway (Perkins et al., J. Virol. 76:1435-1449, 2002). The present studies were designed to better elucidate the mechanism of ICP10 PK-induced neuroprotection and determine whether HSV-1 has similar activity. The data indicate that apoptosis inhibition by ICP10 PK involves a c-Raf-1-dependent mechanism and induction of the antiapoptotic protein Bag-1 by the activated ERK survival pathway. Also associated with neuroprotection by ICP10 PK are increased activation/stability of the transcription factor CREB and stabilization of the antiapoptotic protein Bcl-2. HSV-1 and the ICP10 PK-deleted HSV-2 mutant ICP10DeltaPK activate JNK, c-Jun, and ATF-2, induce the proapoptotic protein BAD, and trigger apoptosis in hippocampal neurons. c-Jun activation and apoptosis are inhibited in hippocampal cultures infected with HSV-1 in the presence of the JNK inhibitor SP600125, suggesting that JNK/c-Jun activation is required for HSV-1-induced apoptosis. Ectopically delivered ICP10 PK (but not its PK-negative mutant p139) inhibits apoptosis triggered by HSV-1 or ICP10DeltaPK. Collectively, the data indicate that ICP10 PK-induced activation of the ERK survival pathway results in Bag-1 upregulation and overrides the proapoptotic JNK/c-Jun signal induced by other viral proteins.
Collapse
Affiliation(s)
- D Perkins
- Departments of Pharmacology and Experimental Therapeutics, University of Maryland School of Medicine, Baltimore 21201, USA
| | | | | |
Collapse
|
13
|
Abstract
Herpes Simplex Viruses type 1 (HSV-1) and 2 (HSV-2) cause central nervous system (CNS) disease ranging from benign aseptic meningitis to fatal encephalitis. In adults, CNS infection with HSV-2 is most often associated with aseptic meningitis while HSV-1 frequently produces severe, focal encephalitis associated with high mortality and morbidity. Recent studies suggested that the distinct neurological outcome of CNS infection with the two viruses may be due to their distinct modulation of apoptotic cell death: HSV-1 triggers neuronal apoptosis, while HSV-2 is neuroprotective. Apoptosis also occurs in the etiology of neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease and Down's syndrome, and determines the loss of specific neuronal populations and the decline in cognitive functions. Notwithstanding, the therapy of these disorders may rely on the use of replication-defective HSV-1 vectors to deliver anti-apoptotic transgenes to the CNS. However, the recent discovery of a neuroprotective activity innate to the HSV-2 genome (the ICP10 PK gene) suggests that: i) ICP10 PK may constitute a novel therapeutic approach by targeting both the apoptotic cell death and the cognitive decline, and ii) HSV-2 may be more suitable than HSV-1 as a vector for targeting neuronal disease.
Collapse
|
14
|
Kermer P, Krajewska M, Zapata JM, Takayama S, Mai J, Krajewski S, Reed JC. Bag1 is a regulator and marker of neuronal differentiation. Cell Death Differ 2002; 9:405-13. [PMID: 11965493 DOI: 10.1038/sj.cdd.4400972] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2001] [Accepted: 10/10/2001] [Indexed: 11/08/2022] Open
Abstract
Bag 1 acts as a co-chaperone for Hsp70/Hsc70. We report here that stable over-expression of Bag1 in immortalized neuronal CSM14.1 cells prevents death following serum deprivation. Bag1 over-expression slowed the proliferative rate of CSM14.1 cells, resulted in increased levels of phospo-MAP kinases and accelerated neuronal differentiation. Immunocytochemistry revealed mostly nuclear localization of Bag1 protein in these cells. However, during differentiation in vitro, Bag1 protein shifted from predominantly nuclear to mostly cytosolic in CSM14.1 cells. To explore in vivo parallels of these findings, we investigated Bag1 expression in the developing mouse nervous system using immunohistochemical methods. Early in brain development, Bag1 was found in nuclei of neuronal precursor cells, whereas cytosolic Bag1 staining was observed mainly after completion of neuronal precursor migration and differentiation. Taken together, these findings raise the possibility that the Bag1 protein is expressed early in neurogenesis in vivo and is capable of modulating neuronal cell survival and differentiation at least in part from a nuclear location.
Collapse
Affiliation(s)
- P Kermer
- The Burnham Institute, Program on Apoptosis and Cell Death Research, 10901 N. Torrey Pines Road, La Jolla, California, CA 92037, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Lee HZ, Hsu SL, Liu MC, Wu CH. Effects and mechanisms of aloe-emodin on cell death in human lung squamous cell carcinoma. Eur J Pharmacol 2001; 431:287-95. [PMID: 11730720 DOI: 10.1016/s0014-2999(01)01467-4] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aloe-emodin (1,8-dihydroxy-3-(hydroxymethyl)-anthraquinone) is an active component from the root and rhizome of Rheum palmatum. The study investigated the effects and mechanisms of aloe-emodin-induced cell death in human lung squamous cell carcinoma cell line CH27. Aloe-emodin (40 microM)-induced CH27 cell apoptosis was confirmed by DNA fragmentation (DNA ladders and sub-G(1) formation). Aloe-emodin-induced apoptosis of CH27 cells involved modulation of the expression of Bcl-2 family proteins, such as BclX(L), Bag-1, and Bak, and was associated with the translocation of Bak and Bax from cytosolic to particulate fractions. Aloe-emodin-treated CH27 cells had an increased relative abundance of cytochrome c in the cytosolic fraction. Results demonstrated that the activation of caspase-3, caspase-8, and caspase-9 is an important determinant of apoptotic death induced by aloe-emodin. These results suggest that aloe-emodin induces CH27 cell death by the Bax and Fas death pathway.
Collapse
Affiliation(s)
- H Z Lee
- School of Pharmacy, China Medical College, 91, Hsueh-Shih Road, Taichung, 404, Taiwan.
| | | | | | | |
Collapse
|
16
|
Hayashi T, Sasaki C, Iwai M, Sato K, Zhang WR, Warita H, Abe K. Induction of PML immunoreactivity in rat brain neurons after transient middle cerebral artery occlusion. Neurol Res 2001; 23:772-6. [PMID: 11680520 DOI: 10.1179/016164101101199162] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Promyelocytic leukemia (PML) protein is involved in apoptotic death of cultured neuronal cells, but its role in ischemic brain damage remains uncertain. In this study, we investigated change of immunoreactivity for PML protein in rat brain after transient middle cerebral artery occlusion, and compared the results with that of terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end labeling (TUNEL). Western blotting analysis revealed that PML immunoreactivity was only scant in the sham-control brain, but it increased at 1 h and 1 day after reperfusion, and decreased in density thereafter. Immunohistochemical analysis revealed that nuclei of neurons were most densely stained. TUNEL positive cells appeared at 1 day and peaked at 3 days of reperfusion, indicating that PML protein induction preceded DNA fragmentation in neurons. The present results suggest that PML protein may be one of the key molecules in ischemic neuronal cell death.
Collapse
Affiliation(s)
- T Hayashi
- Department of Neurology, Okayama University School of Medicine, Japan.
| | | | | | | | | | | | | |
Collapse
|
17
|
Jin K, Mao XO, Eshoo MW, Nagayama T, Minami M, Simon RP, Greenberg DA. Microarray analysis of hippocampal gene expression in global cerebral ischemia. Ann Neurol 2001; 50:93-103. [PMID: 11456315 DOI: 10.1002/ana.1073] [Citation(s) in RCA: 139] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The brain's response to ischemia, which helps determine clinical outcome after stroke, is regulated partly by competing genetic programs that respectively promote cell survival and delayed cell death. Many genes involved in this response have been identified individually or systematically, providing insights into the molecular basis of ischemic injury and potential targets for therapy. The development of microarray systems for gene expression profiling permits screening of large numbers of genes for possible involvement in biological or pathological processes. Therefore, we used an oligodeoxynucleotide-based microarray consisting of 374 human genes, most implicated previously in apoptosis or related events, to detect alterations in gene expression in the hippocampus of rats subjected to 15 minutes of global cerebral ischemia followed by up to 72 hours of reperfusion. We found 1.7-fold or greater increases in the expression of 57 genes and 1.7-fold or greater decreases in the expression of 34 genes at 4, 24, or 72 hours after ischemia. The number of induced genes increased from 4 to 72 hours, whereas the number of repressed genes decreased. The induced genes included genes involved in protein synthesis, genes mutated in hereditary human diseases, proapoptotic genes, antiapoptotic genes, injury-response genes, receptors, ion channels, and enzymes. We detected transcriptional induction of several genes implicated previously in cerebral ischemia, including ALG2, APP, CASP3, CLU, ERCC3, GADD34, GADD153, IGFBP2, TIAR, VEGF, and VIM, as well as other genes not so implicated. We also found coinduction of several groups of related genes that might represent functional modules within the ischemic neuronal transcriptome, including VEGF and its receptor, NRP1; the IGF1 receptor and the IGF1-binding protein IGFBP2; Rb, the Rb-binding protein E2F1, and the E2F-related transcription factor, TFDP1; the CACNB3 and CACNB4 beta-subunits of the voltage-gated calcium channel; and caspase-3 and its substrates, ACINUS, FEM1, and GSN. To test the hypothesis that genes identified through this approach might have roles in the pathophysiology of cerebral ischemia, we measured expression of the products of two induced genes not heretofore implicated in cerebral ischemia-GRB2, an adapter protein involved in growth-factor signaling pathways, and SMN1, which participates in RNA processing and is deleted in most cases of spinal muscular atrophy. Western analysis showed enhanced expression of both proteins in hippocampus at 24 to 72 hours after ischemia, and SMN1 was localized by immunohistochemistry to hippocampal neurons. These results suggest that microarray analysis of gene expression may be useful for elucidating novel molecular mediators of cell death and survival in the ischemic brain.
Collapse
Affiliation(s)
- K Jin
- Buck Institute for Age Research, Novato, CA 94945, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Niyaz Y, Zeiner M, Gehring U. Transcriptional activation by the human Hsp70-associating protein Hap50. J Cell Sci 2001; 114:1839-45. [PMID: 11329370 DOI: 10.1242/jcs.114.10.1839] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated human Hap50, the large isoform of the previously characterized Hsp70/Hsc70-associating protein Hap46, also called BAG-1, for effects on transcriptional activities. Overproduction by transient transfection led to enhanced expression of reporter gene constructs in various cell types using different promoters, suggesting independence of promoter type. Similarly, overexpression of Hap50 resulted in increased levels of poly(A)(+)mRNAs in HeLa, COS-7, 3T3 and HTC cells. Concomitantly, the expression of some selected endogenous genes, such as those coding for c-Jun and the glucocorticoid receptor, was enhanced significantly relative to actin. Nuclear runoff transcription assays using HeLa cells showed that the effect is caused by increased transcription rates rather than mRNA stabilization. Activation of transcription by Hap50 occurred at 37 degrees C and did not require prior thermal stress, as is the case for Hap46. In accordance with these biological effects, Hap50 is localized exclusively in the nuclear compartment of different cell types, whereas Hap46 is mostly cytoplasmic in unstressed cells, as revealed by use of fusion constructs with green fluorescent protein. High cellular levels of Hap50 were found to make cells less susceptible to adverse environmental effects such as heat stress. Our data suggest that Hap50 is a nuclear protein that acts in cells to increase the transcription of various genes.
Collapse
Affiliation(s)
- Y Niyaz
- Ruprecht-Karls-Universität Heidelberg, Biochemie-Zentrum Heidelberg, Biologische Chemie, Im Neuenheimer Feld 501, D-69120 Heidelberg, Germany
| | | | | |
Collapse
|
19
|
Sourisseau T, Desbois C, Debure L, Bowtell DD, Cato AC, Schneikert J, Moyse E, Michel D. Alteration of the stability of Bag-1 protein in the control of olfactory neuronal apoptosis. J Cell Sci 2001; 114:1409-16. [PMID: 11257006 DOI: 10.1242/jcs.114.7.1409] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Normal apoptosis occurs continuously in the olfactory neuroepithelium of adult vertebrates, making it a useful model for studying neuronal apoptosis. Here we demonstrate that overexpression of the anti-apoptotic Bag-1 gene in olfactory neuronal cells confers a strong resistance to apoptosis. Conversely decreased levels of Bag-1 were found to precede a massive wave of olfactory neuronal apoptosis triggered by synaptic target ablation. We show that the decrease is brought about by ubiquitination and subsequent degradation of the Bag-1 protein. The ring finger protein Siah-2 is a likely candidate for the ubiquitination reaction since Siah-2 mRNA accumulated in lesioned olfactory neuroepithelium and overexpression of Siah-2 stimulated Bag-1 ubiquitination and degradation in transient expression assays. These results together identify destabilization of Bag-1 as a necessary step in olfactory neuronal apoptosis.
Collapse
Affiliation(s)
- T Sourisseau
- Information et Programmation Cellulaire, UMR 6026 CNRS-Université de Rennes1, Campus de Beaulieu, Bat.13. 35042 Rennes cedex, France
| | | | | | | | | | | | | | | |
Collapse
|