1
|
Kim JE, Kim DS, Jin Ryu H, Il Kim W, Kim MJ, Won Kim D, Young Choi S, Kang TC. The effect of P2X7 receptor activation on nuclear factor-κB phosphorylation induced by status epilepticus in the rat hippocampus. Hippocampus 2013; 23:500-14. [PMID: 23564500 DOI: 10.1002/hipo.22109] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2013] [Indexed: 11/10/2022]
Abstract
Nuclear factor-kappa B (NFκB) signal is essential for neuronal survival and its activation may protect neuron against various stimuli. Since purinergic signals activate NFκB through the P2X7 receptor, we investigated the distinct pattern of NF-κB phosphorylation in neurons by P2X7 receptor activation following status epilepticus (SE) in an effort to understand the role of P2X7 receptor in epileptogenic insult. In non-SE animals, 2'(3')-O-(4-benzoyl)benzoyl adenosine 5'-triphosphate (BzATP, a P2X7R agonist) treatment increased only p52-Ser869 NF-κB phosphorylation in neuron. Following SE, p52-Ser865, p52-Ser869, p65-Ser276, p65-Ser311, p65-Ser468, and p65-Ser529 NF-κB phosphorylation was significantly decreased in CA1 and CA3 neurons. However, BzATP treatment prevented reductions in p65-Ser276, p65-Ser311, p65-Ser529, and p52-Ser869 NF-κB phosphorylations in CA1 and/or CA3 neurons induced by SE. Furthermore, BzATP treatment reduced SE-induced p65-Ser311, p65-Ser468, p65-Ser536, and p52-Ser869 NF-κB phosphorylations in astrocytes. These findings indicate that P2X7 functions may be involved in the regulation of SE-induced reactive astrocytes and neuronal degeneration via NF-κB phosphorylations in response to pilocarpine-induced SE in the rat hippocampus.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chunchon, Kangwon-Do, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Mannick EE, Mishra J, Marque J, Clavell M, Miller MJ, Oliver PD. Inhibitors of nuclear factor kappa B cause apoptosis in cultured macrophages. Mediators Inflamm 2012; 6:225-32. [PMID: 18472824 PMCID: PMC2365832 DOI: 10.1080/09629359791721] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
The precise role of the transcription factor nuclear factor kappa B (NF- κB) in the regulation of cell survival and cell death is still unresolved and may depend on cell type and position in the cell cycle. The aim of this study was to determine if three pharmacologic inhibitors of NF-κB, pyrrolidine dithiocarbamate, N-tosyl-L-lysl chloromethyl ketone and calpain I inhibitor, induce apoptosis in a murine macrophage cell line (RAW 264.7) at doses similar to those required for NF-κB inhibition. We found that each of the three inhibitors resulted in a dose- and time-dependent increase in morphologic indices of apoptosis in unstimulated, LPS-stimulated and TNF-stimulated cells. Lethal doses were consistent with those required for NF- κB inhibition. We conclude that nuclear NF-κB activation may represent an important survival mechanism in macrophages.
Collapse
Affiliation(s)
- E E Mannick
- Department of Pediatrics Louisiana State University New Orleans LA 70112 USA
| | | | | | | | | | | |
Collapse
|
3
|
Ryu H, Kim JE, Yeo SI, Kim MJ, Jo SM, Kang TC. RelA/p65-serine 536 nuclear factor-kappa B phosphorylation is related to vulnerability to status epilepticus in the rat hippocampus. Neuroscience 2011; 187:93-102. [DOI: 10.1016/j.neuroscience.2011.04.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2011] [Revised: 04/19/2011] [Accepted: 04/19/2011] [Indexed: 01/03/2023]
|
4
|
Kho Y, Kim S, Yoon BS, Moon JH, Kwak S, Park G, Woo J, Oh S, Hong K, Kim S, Kim H, You S, Choi Y. WDNM1 is associated with differentiation and apoptosis of mammary epithelial cells. Anim Biotechnol 2008; 19:89-103. [PMID: 18432400 DOI: 10.1080/10495390801887361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In this study, we show that expression of the Westmead DMBA8 nonmetastatic cDNA 1 (WDNM1) gene was increased upon SFM and/or TNFalpha treatment, with a corresponding increase in apoptotic cells, and gradually decreased following re-stimulation with serum in HC11 mammary epithelial cells. TNFalpha induced WDNM1 expression showed the NFkappaB-dependent mechanism since it's expression was abrogated in IkappaBalphaM (super-repressor of NFkappaB)-transfected cells, but not those transfected with control vector. Furthermore, overexpression of WDNM1 suppressed growth and differentiation, and accelerated apoptosis of HC11 cells. Thus, our results demonstrate that WDNM1 gene expression, regulated by the TNFalpha-NFkappaB signal pathway, is associated with HC11 cell apoptosis.
Collapse
Affiliation(s)
- Yoonjung Kho
- School of Agricultural and Biotechnology, College of Agriculture and Life Sciences Seoul National University, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Shaw MM, Gurr WK, McCrimmon RJ, Schorderet DF, Sherwin RS. 5'AMP-activated protein kinase alpha deficiency enhances stress-induced apoptosis in BHK and PC12 cells. J Cell Mol Med 2007; 11:286-98. [PMID: 17488477 PMCID: PMC3822827 DOI: 10.1111/j.1582-4934.2007.00023.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
5'AMP-activated protein kinase (AMPK) activation occurs under a variety of stress conditions but the role of this enzyme in the promotion or inhibition of stress-induced cell death is unclear. To address this issue, we transformed two different cell lines with shRNA-expressing plasmids, targeting the alpha subunit of AMPK, and verified AMPKalpha downregulation. The cell lines were then stressed by exposure to medium without glucose (PC12 cells) or with the viral thymidine kinase-specific DNA replication inhibitors: acyclovir, penciclovir and ganciclovir (herpes simplex virus thymidine kinase-expressing Baby Hamster Kidney cells). In non-AMPK-downregulated cells, these stress treatments induced AMPK upregulation and phosphorylation, leaving open the question whether the association of AMPK activation with stress-induced cell death reflects a successful death-promoting or an ineffective death-inhibiting activity. In AMPKalpha-deficient cells (expressing AMPKalpha-specific shRNAs or treated with Compound C) exposure to low glucose medium or DNA replication inhibitors led to an enhancement of cell death, indicating that, under the conditions examined, the role of activated AMPK is not to promote, but to protect from or delay stress-induced cell death.
Collapse
Affiliation(s)
- Margaret M Shaw
- Institut de Recherche en Ophtalmologie, Avenue de Grand-Champsec 64, 1950 Sion, Switzerland.
| | | | | | | | | |
Collapse
|
6
|
Kim SB, Kim JS, Lee JH, Yoon WJ, Lee DS, Ko MS, Kwon BS, Choi DH, Cho HR, Lee BJ, Chung DK, Lee HW, Park JW. NF-κB activation is required for cisplatin-induced apoptosis in head and neck squamous carcinoma cells. FEBS Lett 2005; 580:311-8. [PMID: 16376337 DOI: 10.1016/j.febslet.2005.12.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2005] [Revised: 11/07/2005] [Accepted: 12/02/2005] [Indexed: 02/07/2023]
Abstract
This study demonstrates a requirement for NF-kappaB activation in cis-diamminedichloroplatinum (cisplatin)-induced apoptosis in human head and neck squamous cell carcinoma (HNSCC) cell lines. This conclusion was supported by the following observations: cisplatin induced IkappaBalpha degradation and NF-kappaB-dependent transcriptional activation prior to cell death; pyrrolidine dithiocarbamate (PDTC), a chemical inhibitor of NF-kappaB activation, prevented apoptosis; lactacystin, an inhibitor of IkappaBalpha degradation, also prevented apoptosis; and finally, the expression of a super-repressor mutant IkappaBalpha blocked apoptosis. The expression of tumor necrosis factor alpha (TNFalpha) was promoted by cisplatin treatment and was suppressed by PDTC treatment. In addition, a neutralizing antibody against TNFalpha protected cells from cisplatin-induced apoptosis. These findings suggest that NF-kappaB activation is required for cisplatin-induced apoptosis and TNFalpha may play an important role in NF-kappaB-mediated apoptosis in cisplatin-treated HNSCC cell lines.
Collapse
Affiliation(s)
- Seong Bum Kim
- Department of Biological Sciences, University of Ulsan, Nam-Gu, Ulsan 680-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Su JH, Anderson AJ, Cribbs DH, Tu C, Tong L, Kesslack P, Cotman CW. Fas and Fas ligand are associated with neuritic degeneration in the AD brain and participate in beta-amyloid-induced neuronal death. Neurobiol Dis 2003; 12:182-93. [PMID: 12742739 DOI: 10.1016/s0969-9961(02)00019-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
It has recently been suggested that neuronal cell death in response to many brain insults may be mediated by the upregulation of tumor necrosis factor receptor (TNFR) family members and their ligands. In the present study, we investigated whether the expression of the TNFR family death domain receptor, Fas, and its ligand, FasL, is altered in association with neuropathology and activated caspase markers in Alzheimer disease (AD) brain, and Abeta-induced neuronal cell death in vitro. To evaluate this hypothesis, we examined Fas and FasL expression in AD and control brain, and Abeta-treated primary neurons, using immunocytochemistry and Western blots. Neurons in both AD brain and Abeta-treated cultures exhibited FasL upregulation and changes in immunoreactivity for Fas receptor. Further, FasL expression was remarkably elevated in senile plaques and neurofilament-positive dystrophic neurites, and in association with caspase activation and neuritic apoptosis in AD brain. Based on these and previous data regarding protection of primary neuronal cultures from Abeta(1-42)-induced apoptosis by blockade of Fas-associated death domain signaling, we also tested the hypothesis that dynamic regulation of Fas and FasL may contribute to Abeta-mediated neuronal cell death. Accordingly, neuronal cultures derived from mice carrying inactivating mutations in Fas (Faslpr) or FasL (Fasgld) exhibited protection from Abeta(1-42)-induced cell death. These findings suggest that Fas-FasL interactions may contribute to mechanisms of neuronal loss and neuritic degeneration in AD.
Collapse
Affiliation(s)
- Joseph H Su
- Institute for Brain Aging and Dementia, 1113 Gillespie Neuroscience Research Facility, University of California Irvine, 92697-4540, USA
| | | | | | | | | | | | | |
Collapse
|
8
|
Liu Y, Song XD, Liu W, Zhang TY, Zuo J. Glucose deprivation induces mitochondrial dysfunction and oxidative stress in PC12 cell line. J Cell Mol Med 2003; 7:49-56. [PMID: 12767261 PMCID: PMC6740129 DOI: 10.1111/j.1582-4934.2003.tb00202.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Glucose metabolism plays a pivotal role in many physiological and pathological conditions. To investigate the effect of hypoglycemia (obtained by glucose deprivation) on PC12 cell line, we analyzed the cell viability, mitochondrial function (assessed by MTT reduction, cellular ATP level, mitochondrial transmembrane potential), and the level of reactive oxygen species (ROS) after glucose deprivation (GD). Upon exposure to GD, ROS level increased and MTT reduction decreased immediately, intracellular ATP level increased in the first 3 hours, followed by progressive decrease till the end of GD treatment, and the mitochondrial transmembrane potential (deltapsi(m)) dropped after 6 hours. Both necrosis and apoptosis occurred apparently after 24 hours which was determined by nuclei staining with propidium iodide(PI) and Hoechst 33342. These data suggested that cytotoxicity of GD is mainly due to ROS accumulation and ATP depletion in PC12 cells.
Collapse
Affiliation(s)
- Yan Liu
- Department of Cellular and Genetic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
9
|
Tabakman R, Lazarovici P, Kohen R. Neuroprotective effects of carnosine and homocarnosine on pheochromocytoma PC12 cells exposed to ischemia. J Neurosci Res 2002; 68:463-9. [PMID: 11992473 DOI: 10.1002/jnr.10228] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The development of neuroprotective drugs against ischemic insults is hampered by the lack of pharmacological in vitro models. We developed an ischemic model using PC12 cell cultures exposed to oxygen-glucose-deprivation (OGD) followed by reoxygenation (18 hr) under regular atmospheric oxygen level. The toxicity induced in this model, that is partially caused by generation of reactive oxygen species (ROS), was measured morphologically as well as by the release of lactate dehydrogenase (LDH) and the prostaglandin PGE(2) from the cells. Carnosine and homocarnosine, histidine dipeptides antioxidants, found in high concentration in the brain, have been suggested to provide neuroprotection. Using the OGD model we found that 5 mM carnosine and 1 mM homocarnosine provided maximal neuroprotection of about 50% against OGD insult. This neuroprotective effect was similar to that of a known antioxidant, 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl (tempol), and was not observed in a serum-deprivation toxicity model of PC12 cells, indicating that carnosine and homocarnosine may act as antioxidant-neuroprotective agents in the brain. Our ischemic model may provide a useful tool for investigating the mechanisms involved in the neuroprotection afforded by histidine dipeptides.
Collapse
Affiliation(s)
- Rinat Tabakman
- Department of Pharmacology and Experimental Therapeutics, Hebrew University of Jerusalem, Jerusalem, Israel
| | | | | |
Collapse
|
10
|
Shou Y, Li N, Li L, Borowitz JL, Isom GE. NF-kappaB-mediated up-regulation of Bcl-X(S) and Bax contributes to cytochrome c release in cyanide-induced apoptosis. J Neurochem 2002; 81:842-52. [PMID: 12065643 DOI: 10.1046/j.1471-4159.2002.00880.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cyanide induces apoptosis through cytochrome c activated caspase cascade in primary cultured cortical neurons. The underlying mechanism for cytochrome c release from mitochondria after cyanide treatment is still unclear. In this study, the roles of endogenous Bcl-2 proteins in cyanide-induced apoptosis were investigated. After cyanide (100-500 microm) treatment for 24 h, two pro-apoptotic Bcl-2 proteins, Bcl-X(S) and Bax were up-regulated as shown by western blot and RT-PCR analysis. The expression levels of two antiapoptotic Bcl-2 proteins, Bcl-2 and Bcl-X(L), remained unchanged after cyanide treatment, whereas the mRNA levels of Bcl-X(S) and Bax began to increase within 2 h and their protein levels increased 6 h after treatment. NF-kappaB, a redox-sensitive transcription factor activated after cyanide treatment, is responsible for the up-regulation of Bcl-X(S) and Bax. SN50, which is a synthetic peptide that blocks translocation of NF-kappaB from cytosol to nucleus, inhibited the up-regulation of Bcl-X(S) and Bax. Similar results were obtained using a specific kappaB decoy DNA. NMDA receptor activation and reactive oxygen species (ROS) generation are upstream events of NF-kappaB activation, as blockade of these two events by MK801, l-NAME or PBN inhibited cyanide-induced up-regulation of Bcl-X(S) and Bax. Up-regulation of pro-apoptotic Bcl-X(S) and Bax contributed to cyanide-induced cytochrome c release, because SN50 and a specific Bax antisense oligodeoxynucleotide significantly reduced release of cytochrome c from mitochondria as shown by western blot analysis. It was concluded that NF-kappaB-mediated up-regulation of Bcl-X(S) and Bax is involved in regulating cytochrome c release in cyanide-induced apoptosis.
Collapse
Affiliation(s)
- Yan Shou
- Neurotoxicology Laboratory, Department of Medicinal Chemistry and Molecular Pharmacology, Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907-1333, USA
| | | | | | | | | |
Collapse
|
11
|
Panet H, Barzilai A, Daily D, Melamed E, Offen D. Activation of nuclear transcription factor kappa B (NF-kappaB) is essential for dopamine-induced apoptosis in PC12 cells. J Neurochem 2001; 77:391-8. [PMID: 11299301 DOI: 10.1046/j.1471-4159.2001.00213.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The etiology of Parkinson's disease is still unknown, though current investigations support the notion of the pivotal involvement of oxidative stress in the process of neurodegeneration in the substantia nigra (SN). In the present study, we investigated the molecular mechanisms underlying cellular response to a challenge by dopamine, one of the local oxidative stressors in the SN. Based on studies showing that nuclear factor kappa B (NF-kappaB) is activated by oxidative stress, we studied the involvement of NF-kappaB in the toxicity of PC12 cells following dopamine exposure. We found that dopamine (0.1-0.5 m M) treatment increased the phosphorylation of the IkappaB protein, the inhibitory subunit of NF-kappaB in the cytoplasm. Immunoblot analysis demonstrated the presence of NF-kappaB-p65 protein in the nuclear fraction and its disappearance from the cytoplasmic fraction after 2 h of dopamine exposure. Dopamine-induced NF-kappaB activation was also evidenced by electromobility shift assay using radioactive labeled NF-kappaB consensus DNA sequence. Cell-permeable NF-kappaB inhibitor SN-50 rescued the cells from dopamine-induced apoptosis and showed the importance of NF-kappaB activation to the induction of apoptosis. Furthermore, flow cytometry assay demonstrated a higher level of translocated NF-kappaB-p65 in the apoptotic nuclei than in the unaffected nuclei. In conclusion, our findings suggest that NF-kappaB activation is essential to dopamine-induced apoptosis in PC12 cells and it may be involved in nigral neurodegeneration in patients with Parkinson's disease.
Collapse
Affiliation(s)
- H Panet
- Department of Neurology, Rabin Medical Center and the Laboratory for Neurosciences, Felsenstein Medical Research Center, Petah Tikva and Sackler School of Medicine, Tel Aviv, Israel
| | | | | | | | | |
Collapse
|
12
|
Uberti D, Grilli M, Memo M. Contribution of NF-kappaB and p53 in the glutamate-induced apoptosis. Int J Dev Neurosci 2000; 18:447-54. [PMID: 10817929 DOI: 10.1016/s0736-5748(00)00018-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Exposure of primary cultures of cerebellar granule cells for 15 min to micromolar concentrations of glutamate results in cell death of both necrotic and apoptotic types. Among the intracellular events triggered by glutamate, we identified two transcriptional factors: the p50 member of the NF-kappaB family and the tumor suppressor phosphoprotein p53. Pretreatment of the cultures with aspirin, which inhibits NF-kappaB activation, or with specific p53 antisense oligonucleotide, which inhibits p53 transcription, resulted in a complete prevention of glutamate-induced p53 induction and apoptosis. These findings suggest the existence of a transcriptional program activated by glutamate receptor stimulation in which p50 and p53 play a relevant role. Then, we studied the expression of two p53 downstream genes that could participate in the glutamate-induced pro-apoptotic pathway: p21, which codes for an inhibitor of different cyclin dependent kinases, and MSH2, which codes for a protein involved in the recognition and repair of DNA mismatches. We found that primary cerebellar neurons expressed p21 and MSH2 at very low levels in basal conditions. However, very soon after a brief exposure of the cells to glutamate, the expression of both proteins was dramatically enhanced.On these bases, we propose NF-kappaB, p53, p21 and MSH2 as relevant contributors of the glutamate-induced pro-apoptotic pathway. Understanding this cascade of nuclear events may unravel specific targets for pharmacological intervention for those neurological diseases in which excitatory amino acid-induced apoptosis plays a relevant role.
Collapse
Affiliation(s)
- D Uberti
- Division of Pharmacology, Department of Biomedical Sciences and Biotechnologies, School of Medicine, University of Brescia, Via Valsabbina 19, 25123, Brescia, Italy
| | | | | |
Collapse
|
13
|
Abstract
Expression of the Wnt-1 oncogene in PC12 cells induces morphological and biochemical changes, including up-regulation of cell adhesion and lack of differentiation in response to growth factors. The survival of PC12 cells is known to be mediated in part by phosphatidylinositol-3 kinase (PI-3 kinase)-dependent activation of the transcription factor nuclear factor-kappaB (NF-kappaB). We investigated the effect of Wnt-1 expression on cell survival and NF-kappaB activation using PC12 cells expressing Wnt-1 (PC12/Wnt1) and a reporter vector in which firefly luciferase expression is under the control of NF-kappaB consensus sequences. Serum deprivation caused apoptosis and decreased NF-kappaB activity in wild type PC12 cells. PC12/Wnt-1 cells showed less apoptosis in the absence of serum, and the levels of NF-kappaB activity were higher than in wild type PC12 cells. NF-kappaB activity was also increased by the transient expression of Wnt-1 in PC12 cells and it was completely inhibited in both PC12 and PC12/Wnt-1 cells by a dominant negative mutant IkappaB-alpha that has been shown to prevent NF-kappaB activation. Agents known to inhibit NF-kappaB-induced apoptosis in PC12 as well as in PC12/Wnt-1 cells, indicating a role of NF-kappaB activation in the anti-apoptotic effect of Wnt-1. Inhibition of PI-3 kinase with wortmannin, or with a dominant negative p85 regulatory subunit of the PI-3 kinase, blocked NF-kappaB activity in PC12 cells but caused only partial inhibition in PC12/Wnt-1 cells. The effect of Wnt-1 in activating NF-kappaB can be mimicked by inhibition of glycogen synthase kinase-3beta (GSK-3beta) with lithium or with a dominant negative GSK-3beta. Our results show that expression of Wnt-1 increases survival of PC12 cells in the absence of serum by activating the anti-apoptotic factor NF-kappaB. Wnt-1-induced activation of NF-kappaB is partially independent of PI-3 kinase and can be mimicked by inhibition of GSK-3beta.
Collapse
Affiliation(s)
- J C Bournat
- The Lankenau Medical Research Center, Wynnewood, Pennsylvania 19096, USA
| | | | | |
Collapse
|
14
|
Gu Z, Cain L, Werrbach-Perez K, Perez-Polo JR. Differential alterations of NF-kappaB to oxidative stress in primary basal forebrain cultures. Int J Dev Neurosci 2000; 18:185-92. [PMID: 10715573 DOI: 10.1016/s0736-5748(99)00087-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress has been linked to neuronal cell death resulting from either acute insults due to ischemia, trauma, excitotoxicity, or chronic neurodegenerative diseases. Cholinergic basal forebrain neurons (CBFNs) compete for nerve growth factor (NGF) synthesized in the hippocampus and cortex via retrograde transport. NGF affects CBFN survival and cholinergic function via activation of the NF-kappaB transcription factor and this signaling pathway appears to be impaired in aged rats. Here, we demonstrate that activation of NF-kappaB in basal forebrain primary culture via treatment with hydrogen peroxide or TNF-alpha is predominantly restricted to CBFNs, and that NF-kappaB activation appears to mostly affect p65 translocation to the nucleus, but not the p50 subunit. These results are consistent with NF-kappaB activation being a part of recovery processes after acute oxidative stress. Since p50 or p49 (also called p52) binding to promoter sites does not stimulate transcription - both p50 and p49 lack an activating domain - and p65 does contain an activating domain and thus can act as a transcription enhancer, differential translocation of different NF-kappaB dimers can act as repressors of constitutive activity or enhancers. These results are in agreement with the hypothesis that p50/p65 is the active trans-activating species of NF-kappaB, as compared to p50/p50 homodimers which bind to NF-kappaB binding sites but do not trans-activate promoters. Our results also suggest that selective activation of different NF-kappaB dimer species may have regulatory significance in neuronal responses to acute or chronic insults to CNS. Thus, increased p65 translocation could have enhancing effects while increased p50 translocation could have a repressor role. Manipulation of the types of NF-kappaB species being translocated could provide a basis for therapeutic strategies.
Collapse
Affiliation(s)
- Z Gu
- Department of Human Biological Chemistry, The University of Texas Medical Branch at Galveston, Room 436, Gail Borden Bldg, 301 University Blvd, Galveston, TX 77555-0652, USA
| | | | | | | |
Collapse
|
15
|
Mastrangelo AJ, Hardwick JM, Zou S, Betenbaugh MJ. Part II. Overexpression of bcl-2 family members enhances survival of mammalian cells in response to various culture insults. Biotechnol Bioeng 2000; 67:555-64. [PMID: 10649230 DOI: 10.1002/(sici)1097-0290(20000305)67:5<555::aid-bit6>3.0.co;2-t] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A number of bioreactor configurations have been developed for the manufacture of products from mammalian cell hosts. Even in the most efficient of these, however, problems such as nutrient exhaustion, growth factor deprivation, and toxin accumulations may arise. Consequently, the current effort focused on the feasibility of overexpressing anti-apoptosis genes in baby hamster kidney (BHK) and Chinese hamster ovary (CHO) cells as a means of limiting cell death upon exposure to three such insults. Extended periods of glucose deprivation, serum withdrawal, and treatment with ammonium chloride each caused significant damage, often apoptotic in nature, to BHK and CHO cells, typically rendering cultures completely nonviable. The overexpression of bcl-2 and bcl-x(L), however, was able to abrogate the cell death in BHK cultures, though to varying degrees. For instance, the presence of Bcl-2, which did little to suppress apoptosis upon glucose deprivation, significantly improved the viabilities of these cells during serum withdrawal. In contrast, bcl-x(L) overexpression provided BHK cells with enhanced protection in the absence of glucose, allowing cultures to remain viable throughout the entire three week study. CHO cultures, on the other hand, displayed similar trends in survival in response to both glucose and serum deprivation. During these studies, Bcl-x(L) was consistently able to afford cells the highest degree of protection, though Bcl-2 also enhanced culture viabilities and viable numbers. Death suppression following exposure to 50 mM ammonium chloride was observed to a limited extent in both BHK and CHO cells overexpressing bcl-2 and bcl-x(L). However, even during such harsh treatment, Bcl-x(L) was able to enhance the survival of both cultures, providing CHO cells with viable numbers that were nearly 20-fold that of the controls after five days of exposure. Furthermore, the extensions in cell survival provided by the anti-apoptosis gene products enabled the recovery of many of the cultures during rescue attempts in which the death-inducing stimulus was removed. Clearly, engineering cells to better withstand and recover from the insults common during the large scale cultivation of mammalian cells has a number of potential applications in the biopharmaceutical industries where cell death can limit culture productivities.
Collapse
Affiliation(s)
- A J Mastrangelo
- Department of Chemical Engineering, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, USA
| | | | | | | |
Collapse
|
16
|
|
17
|
Camandola S, Poli G, Mattson MP. The lipid peroxidation product 4-hydroxy-2,3-nonenal increases AP-1-binding activity through caspase activation in neurons. J Neurochem 2000; 74:159-68. [PMID: 10617117 DOI: 10.1046/j.1471-4159.2000.0740159.x] [Citation(s) in RCA: 319] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The transcription factor activator protein-1 (AP-1) is activated in response to physiological activity in neuronal circuits and in response to neuronal injury associated with various acute and chronic neurodegenerative conditions. The membrane lipid peroxidation product 4-hydroxy-2,3-nonenal (HNE) is increasingly implicated in the disruption of neuronal calcium homeostasis that occurs in various paradigms of neuronal excitotoxicity and apoptosis. The possible mechanistic links between lipid peroxidation and alterations in gene transcription during neuronal apoptosis have not previously been examined. We now report that exposure of cultured rat cortical neurons to an apoptotic concentration of HNE results in a large increase in AP-1 DNA-binding activity. The protein synthesis inhibitor cycloheximide blocked the induction of AP-1, consistent with a requirement for induction of expression of AP-1 family members. The broad-spectrum caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone and the caspase-3 inhibitor N-acetyl-Asp-Glu-Val-Asp-aldehyde blocked HNE-induced increases in AP-1 DNA-binding activity, demonstrating a requirement for caspase activation in the activation of AP-1. HNE induced phosphorylation of c-Jun N-terminal kinase (JNK), which was prevented by caspase inhibitors, indicating that HNE was acting at or upstream of JNK phosphorylation. The intracellular calcium chelator BAPTA-acetoxymethyl ester completely prevented stimulation of AP-1 DNA-binding by HNE, indicating a requirement for calcium. Moreover, agents that suppress mitochondrial calcium uptake (ruthenium red) and membrane permeability transition (cyclosporin A) attenuated AP-1 activation by HNE, suggesting a contribution of mitochondrial alterations to AP-1 activation. Collectively, our data suggest a scenario in which HNE disrupts neuronal calcium homeostasis and perturbs mitochondrial function, resulting in caspase activation. Activated caspases, in turn, induce activation of JNK, resulting in stimulation of AP-1 DNA-binding protein production. This transcriptional pathway induced by HNE may modulate the cell death process.
Collapse
Affiliation(s)
- S Camandola
- Sanders-Brown Center on Aging and Department of Anatomy and Neurobiology, University of Kentucky, Lexington, USA
| | | | | |
Collapse
|
18
|
Macdonald NJ, Perez-Polo JR, Bennett AD, Taglialatela G. NGF-resistant PC12 cell death induced by arachidonic acid is accompanied by a decrease of active PKC zeta and nuclear factor kappa B. J Neurosci Res 1999; 57:219-26. [PMID: 10398299 DOI: 10.1002/(sici)1097-4547(19990715)57:2<219::aid-jnr7>3.0.co;2-c] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Inflammation and the associated release of inflammatory cytokines such as tumor necrosis factor alpha (TNFalpha) may be a component of neurodegenerative diseases associated with aging or chronic HIV-1 infection. Most of the neurons that are affected under these conditions require a constant supply of trophic factors such as nerve growth factor (NGF) for survival. NGF acts via binding to a specific tyrosine kinase receptor (TrkA). NGF also binds to the common neurotrophin receptor (p75(NTR)), a member of the TNFalpha receptor (TNFR-I) superfamily, whose function may be to modulate apoptosis via the release of ceramide and the activation of the transcription factor nuclear factor kappa B (NFkappaB). The similarity between p75(NTR) and TNFR-I signal transduction pathways suggests that one of the mechanisms by which TNFalpha affects neuronal survival is by impacting upon these pathways that normally promote NGF support of neurons. Here we show that arachidonic acid (AA), a signaling lipid potentially associated with TNFR-I signal cascade, induces apoptosis in PC12 cells through inhibition of both protein kinase C zeta (PKCzeta) and NFkappaB activity. We also show that apoptosis induced by AA cannot be prevented by NGF. These data support the idea that PKCzeta and NFkappaB are both essential signaling elements for mediating NGF-promoted rescue from apoptosis. Our results also suggest that AA, an inflammatory signal lipid induced by TNFalpha via binding to TNFR-I, may reduce neuronal survival by inhibiting elements of the signal cascade induced by NGF.
Collapse
Affiliation(s)
- N J Macdonald
- Department of Human Biological Chemistry and Genetics, The University of Texas Medical Branch at Galveston, USA
| | | | | | | |
Collapse
|
19
|
Ramesh GT, Manna SK, Aggarwal BB, Jadhav AL. Lead activates nuclear transcription factor-kappaB, activator protein-1, and amino-terminal c-Jun kinase in pheochromocytoma cells. Toxicol Appl Pharmacol 1999; 155:280-6. [PMID: 10079214 DOI: 10.1006/taap.1999.8624] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Lead (Pb) is a ubiquitous environmental contaminant that produces variety of effects on the central and peripheral nervous system, induces inflammatory response, and modulates immune functions. Though increase in lipid peroxidation and reactive oxygen intermediates (ROI) have been observed in Pb-induced toxicity, the molecular mechanism underlying these effects is largely unknown. Since nuclear factor kappa B (NF-kappaB) and activator protein (AP-1) are known to be activated by oxidative stress, we hypothesized that Pb-induced effects may be modulated via these transcription factors. The effects of Pb on NF-kappaB, AP-1, and related kinases were studied in pheochromocytoma cells (PC-12). Our results showed that treatment of murine PC-12 cells with Pb resulted in activation of NF-kappaB and degradation of IkappaBalpha (the inhibitory subunit of NF-kappaB). Pb-induced NF-kappaB dependent gene expression was also enhanced. The binding of Pb-induced NF-kappaB to DNA was blocked by antibodies for p65 and p50 but not by c-Rel or nonspecific antibodies such as cyclin D-1 and preimmune serum, suggesting that NF-kappaB consisted of p65 and p50 subunits. Similar to its effects on NF-kappaB, Pb also activated AP-1 in a time- and dose-dependent manner. Besides activating these transcription factors, Pb was also found to upregulate the related kinases such as mitogen activated protein kinase kinase (MEK) and c-Jun N-terminal kinase (JNK) (also known as stress-activated protein kinase) in a dose- and time-dependent manner. Thus, these results suggest that NF-kappaB, AP-1, MEK, and JNK may be important mediators of Pb-induced signaling in gene expression mediating inflammatory response and immunomodulation.
Collapse
Affiliation(s)
- G T Ramesh
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, 77004, USA
| | | | | | | |
Collapse
|
20
|
Abstract
Oxidative insults, whether over-excitation, excessive release of glutamate or ATP caused by stroke, ischemia or inflammation, exposure to ionizing radiation, heavy-metal ions or oxidized lipoproteins may initiate various signaling cascades leading to apoptotic cell death and neurodegenerative disorders. Among the various reactive oxygen species (ROS) generated in the living organism, hydroxyl and peroxynitrite are the most potent and can damage proteins, lipids and nucleic acids. It appears that some natural antioxidants (tocopherol, ascorbic acid and glutathione) and defense enzyme systems (superoxide dismutase, catalase and glutathione peroxidase) may provide some protection against oxidative damage. Recent findings indicate several polyphenols and antioxidant drugs (probucol, seligilline) are effective in protecting the cells from ROS attack. Further development of these antioxidant molecules may be of value in preventing the development of neurodegenerative diseases.
Collapse
Affiliation(s)
- A Y Sun
- Department of Pharmacology, University of Missouri, Columbia, MO 65212, USA.
| | | |
Collapse
|
21
|
Abstract
We have previously shown that extracellular ATP caused cell death in PC12 cells through activation of its receptors. Oxidative stress has been implicated as a mechanism of cell death caused by extracellular ATP. In the present study we examined the possible signal transduction cascades leading to cell death by extracellular ATP. We found, using the electrophoretic mobility shift assay, that transcription factor AP-1 DNA binding activity was stimulated by extracellular ATP. Northern blot analysis showed that mRNA levels of c-fos, c-jun were elevated after treatment with ATP. The stimulation was receptor mediated, since it was blocked by the ATP receptor antagonist, suramin. The stimulated AP-1 binding was also blocked by the antioxidant N-acetyl-L-cysteine, indicating that reactive oxygen species generated following ATP stimulation were involved in the induction of AP-1 activity. It appears that both translational and posttranslational events contributed to the increased AP-1 DNA binding since cyclohexamide (a protein synthesis inhibitor), genistein (tyrosine kinase inhibitor) and staurosporine (PKC inhibitor) each partially blocked the AP-1 activation. Changes in AP-1 DNA binding activity may modulate expression of target genes involved in cell death pathways.
Collapse
Affiliation(s)
- Y Chen
- Dept. of Pharmacology, University of Missouri, Columbia 65212, USA
| | | |
Collapse
|
22
|
Qin ZH, Wang Y, Nakai M, Chase TN. Nuclear factor-kappa B contributes to excitotoxin-induced apoptosis in rat striatum. Mol Pharmacol 1998; 53:33-42. [PMID: 9443930 DOI: 10.1124/mol.53.1.33] [Citation(s) in RCA: 140] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Excitotoxin-induced destruction of striatal neurons, proposed as a model of Huntington's disease, involves a process having the biochemical stigmata of apoptosis. Recent studies suggested that transcription factor nuclear factor (NF)-kappa B may be involved in excitotoxicity. To further analyze the contribution of NF kappa B to excitotoxic neuronal death in vivo, changes in binding activities of NF kappa B and other transcription factors as well as the consequences of inhibiting NF kappa B nuclear translocation were measured after the infusion of quinolinic acid (120 nmol) into rat striatum. Internucleosomal DNA fragmentation and terminal transferase-mediated dUTP digoxigenin nick end labeling-positive nuclei appeared 12 hr later and intensified over the next 12 hr. NF kappa B binding activity increased several-fold from 2 to 12 hr, then gradually declined during the next 12 hr. Other transcription factor changes included AP-1, whose binding peaked about 6 hr after quinolinic acid administration, and E2F-1, which was only modestly and transiently elevated. In contrast, quinolinic acid lead to a reduction in OCT-1, beginning after 12 hr, and briefly in SP-1 binding. The NF kappa B, AP-1, and OCT-1 changes were attenuated both by the N-methyl-D-aspartate receptor antagonist MK-801 and the protein synthesis inhibitor cycloheximide. Moreover, quinolinic acid-induced internucleosomal DNA fragmentation and striatal cell death were significantly reduced by the intrastriatal administration of NF kappa B SN50, a cell-permeable recombinant peptide that blocks NF kappa B nuclear translocation. These results illustrate the complex temporal pattern of transcription factor change attending the apoptotic destruction produced in rat striatum by quinolinic acid. They further suggest that NF kappa B activation contributes to the excitotoxin-induced death of striatal neurons.
Collapse
Affiliation(s)
- Z H Qin
- Experimental Therapeutics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|
23
|
Pan Z, Sampath D, Jackson G, Werrbach-Perez K, Perez-Polo R. Nerve growth factor and oxidative stress in the nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1997; 429:173-93. [PMID: 9413574 DOI: 10.1007/978-1-4757-9551-6_13] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Z Pan
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch at Galveston 77555-0652, USA
| | | | | | | | | |
Collapse
|
24
|
Sikora E, Bielak-Zmijewska A, Piwocka K, Skierski J, Radziszewska E. Inhibition of proliferation and apoptosis of human and rat T lymphocytes by curcumin, a curry pigment. Biochem Pharmacol 1997; 54:899-907. [PMID: 9354590 DOI: 10.1016/s0006-2952(97)00251-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Curcumin (diferuoylmethane), the yellow pigment in the rhizome of tumeric (Curcuma longa), an ingredient of curry spice, is known to exhibit a variety of pharmacological effects including antitumor, antiinflammatory, and antiinfectious activities. Although its precise mode of action remains elusive, curcumin has been shown to suppress the activity of the AP-1 transcription factor in cells stimulated to proliferate. In this study, we observed that curcumin (50 microM) inhibited proliferation of rat thymocytes stimulated with concanavalin A (Con A) as well as that of human Jurkat lymphoblastoid cells in the logarithmic growth phase. The pigment also inhibited apoptosis in dexamethasone-treated rat thymocytes and in UV-irradiated Jurkat cells as judged by DNA ladder formation, cellular morphological changes, and flow cytometry analysis. The inhibition of apoptosis by curcumin in rat thymocytes was accompanied by partial suppression of AP-1 activity. Complete suppression of AP-1 activity was observed in Con A-treated, proliferating thymocytes. The capacity of curcumin to inhibit both cell growth and death strongly implies that these two biological processes share a common pathway at some point and that curcumin affects a common step, presumably involving a modulation of the AP-1 transcription factor.
Collapse
Affiliation(s)
- E Sikora
- Department of Cellular Biochemistry, Nencki Institute of Experimental Biology, Warsaw, Poland.
| | | | | | | | | |
Collapse
|
25
|
Matthews CC, Odeh HM, Feldman EL. Insulin-like growth factor-I is an osmoprotectant in human neuroblastoma cells. Neuroscience 1997; 79:525-34. [PMID: 9200735 DOI: 10.1016/s0306-4522(96)00611-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A role in neuronal homeostasis is suggested by the persistent expression of the insulin-like growth factors in the adult nervous system. SH-SY5Y human neuroblastoma cells, a well-characterized in vitro model of human neurons, were used to investigate the effects of hyperosmotic stress on neurons. Neuronal DNA fragmentation was detected within 1 h and pyknotic nuclei were apparent in attached cells after 12 h of hyperosmotic stress. In parallel, flow cytometry measurements revealed a sudden increase in the rate of cells irreversibly undergoing programmed cell death after 12 h of hyperosmotic exposure. Insulin-like growth factor-I delayed the onset of a laddered DNA fragmentation pattern for 24 h and provided continuing protection against hyperosmotic exposure for 72 h. Amino acid uptake was decreased in hyperosmotic medium even in the presence of insulin-like growth factor-I; the protein synthesis inhibitor cycloheximide neither prevented the induction of programmed cell death nor interfered with the ability of insulin-like growth factor-I to act as an osmoprotectant in hyperosmotic medium. Cysteine and serine protease inhibitors each prevented DNA fragmentation under hyperosmotic conditions, suggesting that the osmoprotectant activity of insulin-like growth factor-I involves the suppression of protease activity. Collectively, these results indicate that insulin-like growth factor-I limits the death of neurons under stressful environmental conditions, suggesting that it may provide a candidate therapy in the treatment of hyperosmolar coupled neurological injury.
Collapse
Affiliation(s)
- C C Matthews
- Department of Neurology, University of Michigan, Ann Arbor 48109-0588, U.S.A
| | | | | |
Collapse
|
26
|
Taglialatela G, Robinson R, Perez-Polo JR. Inhibition of nuclear factor kappa B (NF?B) activity induces nerve growth factor-resistant apoptosis in PC12 cells. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970115)47:2<155::aid-jnr4>3.0.co;2-e] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
27
|
Sheng P, Ladenheim B, Moran TH, Wang XB, Cadet JL. Methamphetamine-induced neurotoxicity is associated with increased striatal AP-1 DNA-binding activity in mice. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 1996; 42:171-4. [PMID: 8915598 DOI: 10.1016/s0169-328x(96)00192-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Multiple injections of methamphetamine (METH) produce long-lasting neurotoxic effects on the nigrostriatal dopamine (DA) system. The drug also causes increases in AP-1 DNA-binding activity in mice. In the present study, we tested the idea that toxic doses of METH might cause long-term increases in AP-1 DNA-binding. Mice were given 10 mg/kg of METH 2, 3 or 4 times at a 2 h interval in 1 day. Striatal DA levels were markedly decreased at 3 h and 24 h in all injection groups. After 1 week, striatal DA level recovered to near control in the METH x2 group, but were still significantly decreased in the METH x3 and x4 groups. Similar drug administration schedules caused increases in AP-1 DNA-binding activity at the 3 h time point in all groups. The AP-1-binding activity almost returned back to control level in the x2 and x3 injection groups at the 24 h and 1 week time point, but there were still increased levels of AP-1-binding activity in the METH x4 group. These findings raise the possibility that METH-induced neurotoxicity might involve prolonged activation of AP-1 transcription factor. This might be related to the report that c-fos or c-jun activation may be important in some models of neurodegeneration.
Collapse
Affiliation(s)
- P Sheng
- Molecular Neuropsychiatry Section, NIH/NIDA IRP, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
28
|
Perez-Polo JR. Genotoxic elements in glial degeneration. Neurobiol Aging 1996; 17:487-8; discussion 488-90. [PMID: 8725913 DOI: 10.1016/0197-4580(96)00015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- J R Perez-Polo
- University of Texas Medical Branch, Galveston 77555-0652, USA
| |
Collapse
|