Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2012

Open Access 01-12-2012 | Research

Hydrogen sulfide attenuates spatial memory impairment and hippocampal neuroinflammation in beta-amyloid rat model of Alzheimer’s disease

Authors: Aiguo Xuan, Dahong Long, Jianhua Li, Weidong Ji, Meng Zhang, Lepeng Hong, Jihong Liu

Published in: Journal of Neuroinflammation | Issue 1/2012

Login to get access

Abstract

Background

Endogenously produced hydrogen sulfide (H2S) may have multiple functions in brain. An increasing number of studies have demonstrated its anti-inflammatory effects. In the present study, we investigated the effect of sodium hydrosulfide (NaHS, a H2S donor) on cognitive impairment and neuroinflammatory changes induced by injections of Amyloid-β1-40 (Aβ1-40), and explored possible mechanisms of action.

Methods

We injected Aβ1-40 into the hippocampus of rats to mimic rat model of Alzheimer’s disease (AD). Morris water maze was used to detect the cognitive function. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to detect neuronal apoptosis. Immunohistochemistry analyzed the response of glia. The expression of interleukin (IL)-1β and tumor necrosis factor (TNF)-α was measured by enzyme-linked immunosorbent assay (ELISA) and quantitative real-time polymerase chain reaction (qRT-PCR). The expression of Aβ1-40, phospho-p38 mitogen-activated protein kinase (MAPK), phospho-p65 Nuclear factor (NF)-κB, and phospho-c-Jun N-terminal Kinase (JNK) was analyzed by western blot.

Results

We demonstrated that pretreatment with NaHS ameliorated learning and memory deficits in an Aβ1-40 rat model of AD. NaHS treatment suppressed Aβ1-40-induced apoptosis in the CA1 subfield of the hippocampus. Moreover, the over-expression in IL-1β and TNF-α as well as the extensive astrogliosis and microgliosis in the hippocampus induced by Aβ1-40 were significantly reduced following administration of NaHS. Concomitantly, treatment with NaHS alleviated the levels of p38 MAPK and p65 NF-κB phosphorylation but not JNK phosphorylation that occurred in the Aβ1-40-injected hippocampus.

Conclusions

These results indicate that NaHS could significantly ameliorate Aβ1-40-induced spatial learning and memory impairment, apoptosis, and neuroinflammation at least in part via the inhibition of p38 MAPK and p65 NF-κB activity, suggesting that administration of NaHS could provide a therapeutic approach for AD.
Literature
1.
go back to reference Mildner A, Schlevogt B, Kierdorf K, Böttcher C, Erny D, Kummer MP, Quinn M, Brück W, Bechmann I, Heneka MT, Priller J, Prinz M: Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer’s disease. J Neurosci 2011, 31:11159–11171.CrossRefPubMed Mildner A, Schlevogt B, Kierdorf K, Böttcher C, Erny D, Kummer MP, Quinn M, Brück W, Bechmann I, Heneka MT, Priller J, Prinz M: Distinct and non-redundant roles of microglia and myeloid subsets in mouse models of Alzheimer’s disease. J Neurosci 2011, 31:11159–11171.CrossRefPubMed
2.
go back to reference Eikelenboom P, van Exel E, Hoozemans JJ, Veerhuis R, Rozemuller AJ, van Gool WA: Neuroinflammation-an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis 2010, 7:38–41.CrossRefPubMed Eikelenboom P, van Exel E, Hoozemans JJ, Veerhuis R, Rozemuller AJ, van Gool WA: Neuroinflammation-an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis 2010, 7:38–41.CrossRefPubMed
3.
go back to reference Seabrook TJ, Jiang L, Maier M, Lemere CA: Minocycline affects microglia activation, Abeta deposition, and behavior in APP-tg mice. Glia 2006, 53:776–782.CrossRefPubMed Seabrook TJ, Jiang L, Maier M, Lemere CA: Minocycline affects microglia activation, Abeta deposition, and behavior in APP-tg mice. Glia 2006, 53:776–782.CrossRefPubMed
4.
go back to reference Weldon DT, Rogers SD, Ghilard JR, Finke MP, Cleary JP, O’Hare E, Esler WP, Maggio JE, Mantyh PW: Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J Neurosci 1998, 18:2161–2173.PubMed Weldon DT, Rogers SD, Ghilard JR, Finke MP, Cleary JP, O’Hare E, Esler WP, Maggio JE, Mantyh PW: Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J Neurosci 1998, 18:2161–2173.PubMed
5.
go back to reference Potter PE: Investigational medications for treatment of patients with Alzheimer disease. J Am Osteopath Assoc 2010, Suppl 8:27–36. Potter PE: Investigational medications for treatment of patients with Alzheimer disease. J Am Osteopath Assoc 2010, Suppl 8:27–36.
6.
7.
go back to reference Shibuya N, Tanaka M, Yoshida M, Ogasawara Y, Togawa T, Ishii K, Kimura H: 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid Redox Signal 2009, 11:703–714.CrossRefPubMed Shibuya N, Tanaka M, Yoshida M, Ogasawara Y, Togawa T, Ishii K, Kimura H: 3-Mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid Redox Signal 2009, 11:703–714.CrossRefPubMed
8.
go back to reference Kimura Y, Kimura H: Hydrogen sulfide protects neurons from oxidative stress. FASEB J 2004, 18:1165–1167.PubMed Kimura Y, Kimura H: Hydrogen sulfide protects neurons from oxidative stress. FASEB J 2004, 18:1165–1167.PubMed
9.
go back to reference Yin WL, He JQ, Hu B, Jiang ZS, Tang XQ: Hydrogen sulfide inhibits MPP(+)-induced apoptosis in PC12 cells. Life Sci 2009, 85:269–275.CrossRefPubMed Yin WL, He JQ, Hu B, Jiang ZS, Tang XQ: Hydrogen sulfide inhibits MPP(+)-induced apoptosis in PC12 cells. Life Sci 2009, 85:269–275.CrossRefPubMed
10.
go back to reference Lee SW, Hu YS, Hu LF, Lu Q, Dawe GS, Moore PK, Wong PT, Bian JS: Hydrogen sulphide regulates calcium homeostasis in microglial cells. Glia 2006, 54:116–124.CrossRefPubMed Lee SW, Hu YS, Hu LF, Lu Q, Dawe GS, Moore PK, Wong PT, Bian JS: Hydrogen sulphide regulates calcium homeostasis in microglial cells. Glia 2006, 54:116–124.CrossRefPubMed
11.
go back to reference Nagai Y, Tsugane M, Oka J, Kimura H: Hydrogen sulfide induces calcium waves in astrocytes. FASEB J 2004, 18:557–559.PubMed Nagai Y, Tsugane M, Oka J, Kimura H: Hydrogen sulfide induces calcium waves in astrocytes. FASEB J 2004, 18:557–559.PubMed
12.
go back to reference Hu LF, Wong PT, Moore PK, Bian JS: Hydrogen sulfide attenuates lipopolysaccharide induced inflammation by inhibition of p38 mitogen-activated protein kinase in microglia. J Neurochem 2007, 100:1121–1128.CrossRefPubMed Hu LF, Wong PT, Moore PK, Bian JS: Hydrogen sulfide attenuates lipopolysaccharide induced inflammation by inhibition of p38 mitogen-activated protein kinase in microglia. J Neurochem 2007, 100:1121–1128.CrossRefPubMed
13.
go back to reference Morrison LD, Smith DD, Kish SJ: Brain S-adenosylmethionine levels are severely decreased in Alzheimer’s disease. J Neurochem 1996, 67:1328–1331.CrossRefPubMed Morrison LD, Smith DD, Kish SJ: Brain S-adenosylmethionine levels are severely decreased in Alzheimer’s disease. J Neurochem 1996, 67:1328–1331.CrossRefPubMed
14.
go back to reference Lee M, Schwab C, Yu S, McGeer E, McGeer PL: Astrocytes produce the antiinflammatory and neuroprotective agent hydrogen sulfide. Neurobiol Aging 2009, 30:1523–1534.CrossRefPubMed Lee M, Schwab C, Yu S, McGeer E, McGeer PL: Astrocytes produce the antiinflammatory and neuroprotective agent hydrogen sulfide. Neurobiol Aging 2009, 30:1523–1534.CrossRefPubMed
15.
go back to reference Gong QH, Wang Q, Pan LL, Liu XH, Huang H, Zhu YZ: Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: A pro-inflammatory pathway in rats. Pharmacol Biochem BE 2010, 96:52–58.CrossRef Gong QH, Wang Q, Pan LL, Liu XH, Huang H, Zhu YZ: Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: A pro-inflammatory pathway in rats. Pharmacol Biochem BE 2010, 96:52–58.CrossRef
16.
go back to reference Qu K, Chen CP, Halliwell B, Moore PK, Wong PT: Hydrogen sulfide is a mediator of cerebral ischemic damage. Stroke 2006, 37:889–893.CrossRefPubMed Qu K, Chen CP, Halliwell B, Moore PK, Wong PT: Hydrogen sulfide is a mediator of cerebral ischemic damage. Stroke 2006, 37:889–893.CrossRefPubMed
17.
go back to reference Huang HJ, Liang KC, Chen CP, Chen CM, Hsieh-Li HM: Intrahippocampal administration of A beta(1–40) impairs spatial learning and memory in hyperglycemic mice. Neurobiol Learn Mem 2007, 87:483–494.CrossRefPubMed Huang HJ, Liang KC, Chen CP, Chen CM, Hsieh-Li HM: Intrahippocampal administration of A beta(1–40) impairs spatial learning and memory in hyperglycemic mice. Neurobiol Learn Mem 2007, 87:483–494.CrossRefPubMed
18.
go back to reference Morris R: Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 1984, 11:47–60.CrossRefPubMed Morris R: Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 1984, 11:47–60.CrossRefPubMed
19.
go back to reference Srivareerat M, Tran TT, Salim S, Aleisa AM, Alkadhi KA: Chronic nicotine restores normal Aβ levels and prevents short-term memory and E-LTP impairment in Aβ rat model of Alzheimer’s disease. Neurobiol Aging 2011, 32:834–844.CrossRefPubMed Srivareerat M, Tran TT, Salim S, Aleisa AM, Alkadhi KA: Chronic nicotine restores normal Aβ levels and prevents short-term memory and E-LTP impairment in Aβ rat model of Alzheimer’s disease. Neurobiol Aging 2011, 32:834–844.CrossRefPubMed
20.
go back to reference Bagheri M, Roghani M, Joghataei MT, Mohseni S: Genistein inhibits aggregation of exogenous amyloid-beta1–40 and alleviates astrogliosis in the hippocampus of rats. Brain Res 2012, 1429:145–154.CrossRefPubMed Bagheri M, Roghani M, Joghataei MT, Mohseni S: Genistein inhibits aggregation of exogenous amyloid-beta1–40 and alleviates astrogliosis in the hippocampus of rats. Brain Res 2012, 1429:145–154.CrossRefPubMed
21.
go back to reference Miguel-Hidalgo JJ, Cacabelos R: Beta-amyloid(1–40)-induced neurodegeneration in the rat hippocampal neurons of the CA1 subfield. Acta Neuropathol 1998, 95:455–465.CrossRefPubMed Miguel-Hidalgo JJ, Cacabelos R: Beta-amyloid(1–40)-induced neurodegeneration in the rat hippocampal neurons of the CA1 subfield. Acta Neuropathol 1998, 95:455–465.CrossRefPubMed
22.
go back to reference Walsh DM, Selkoe DJ: Deciphering the molecular basis of memory failure in Alzheimer’s disease. Neuron 2004, 44:181–193.CrossRefPubMed Walsh DM, Selkoe DJ: Deciphering the molecular basis of memory failure in Alzheimer’s disease. Neuron 2004, 44:181–193.CrossRefPubMed
23.
go back to reference Hashimoto T, Adams KW, Fan Z, McLean PJ, Hyman BT: Characterization of oligomer formation of amyloid-beta peptide using a split-luciferase complementation assay. J Biol Chem 2011, 286:27081–27091.CrossRefPubMedPubMedCentral Hashimoto T, Adams KW, Fan Z, McLean PJ, Hyman BT: Characterization of oligomer formation of amyloid-beta peptide using a split-luciferase complementation assay. J Biol Chem 2011, 286:27081–27091.CrossRefPubMedPubMedCentral
24.
go back to reference Shin RW, Ogino K, Kondo A, Saido TC, Trojanowski JQ, Kitamoto T, Tateishi J: Amyloid beta-protein (Abeta) 1–40 but not Abeta1–42 contributes to the experimental formation of Alzheimer disease amyloid fibrils in rat brain. J Neurosci 1997, 17:8187–8193.PubMed Shin RW, Ogino K, Kondo A, Saido TC, Trojanowski JQ, Kitamoto T, Tateishi J: Amyloid beta-protein (Abeta) 1–40 but not Abeta1–42 contributes to the experimental formation of Alzheimer disease amyloid fibrils in rat brain. J Neurosci 1997, 17:8187–8193.PubMed
25.
go back to reference Yamaguchi Y, Miyashita H, Tsunekawa H, Mouri A, Kim HC, Saito K, Matsuno T, Kawashima S, Nabeshima T: Effects of a novel cognitive enhancer, spiro[imidazo-[1,2-a] pyridine −3,2- indan]-2(3H)-one (ZSET1446), on learning impairments induced by amyloid-beta1–40 in the rat. J Pharmacol Exp Ther 2006, 317:1079–1087.CrossRefPubMed Yamaguchi Y, Miyashita H, Tsunekawa H, Mouri A, Kim HC, Saito K, Matsuno T, Kawashima S, Nabeshima T: Effects of a novel cognitive enhancer, spiro[imidazo-[1,2-a] pyridine −3,2- indan]-2(3H)-one (ZSET1446), on learning impairments induced by amyloid-beta1–40 in the rat. J Pharmacol Exp Ther 2006, 317:1079–1087.CrossRefPubMed
26.
go back to reference Zou K, Kim D, Kakio A, Byun K, Gong JS, Kim J, Kim M, Sawamura N, Nishimoto S, Matsuzaki K, Lee B, Yanagisawa K, Michikawa M: Amyloid beta-protein (Abeta)1–40 protects neurons from damage induced by Abeta1–42 in culture and in rat brain. J Neurochem 2003, 87:609–619.CrossRefPubMed Zou K, Kim D, Kakio A, Byun K, Gong JS, Kim J, Kim M, Sawamura N, Nishimoto S, Matsuzaki K, Lee B, Yanagisawa K, Michikawa M: Amyloid beta-protein (Abeta)1–40 protects neurons from damage induced by Abeta1–42 in culture and in rat brain. J Neurochem 2003, 87:609–619.CrossRefPubMed
27.
go back to reference Yu Y, Zhou L, Sun M, Zhou T, Zhong K, Wang H, Liu Y, Liu X, Xiao R, Ge J, Tu P, Fan DS, Lan Y, Hui C, Chui D: Xylocoside G reduces amyloid-β induced neurotoxicity by inhibiting NF-κB signaling pathway in neuronal cells. J Alzheimers Dis 2012, 30:263–275.PubMed Yu Y, Zhou L, Sun M, Zhou T, Zhong K, Wang H, Liu Y, Liu X, Xiao R, Ge J, Tu P, Fan DS, Lan Y, Hui C, Chui D: Xylocoside G reduces amyloid-β induced neurotoxicity by inhibiting NF-κB signaling pathway in neuronal cells. J Alzheimers Dis 2012, 30:263–275.PubMed
28.
go back to reference Hwang DY, Chae KR, Kang TS, Hwang JH, Lim CH, Kang HK, Goo JS, Lee MR, Lim HJ, Min SH, Cho JY, Hong JT, Song CW, Paik SG, Cho JS, Kim YK: Alterations in behavior, amyloid beta-42, caspase-3, and Cox-2 in mutant PS2 transgenic mouse model of Alzheimer’s disease. FASEB J 2002, 6:805–813.CrossRef Hwang DY, Chae KR, Kang TS, Hwang JH, Lim CH, Kang HK, Goo JS, Lee MR, Lim HJ, Min SH, Cho JY, Hong JT, Song CW, Paik SG, Cho JS, Kim YK: Alterations in behavior, amyloid beta-42, caspase-3, and Cox-2 in mutant PS2 transgenic mouse model of Alzheimer’s disease. FASEB J 2002, 6:805–813.CrossRef
29.
go back to reference Dragunow M, Faull RL, Lawlor P, Beilharz EJ, Singleton K, Walker EB, Mee E: In situ evidence for DNA fragmentation in Huntington’s disease striatum and Alzheimer’s disease temporal lobes. Neuroreport 1995, 6:1053–1057.CrossRefPubMed Dragunow M, Faull RL, Lawlor P, Beilharz EJ, Singleton K, Walker EB, Mee E: In situ evidence for DNA fragmentation in Huntington’s disease striatum and Alzheimer’s disease temporal lobes. Neuroreport 1995, 6:1053–1057.CrossRefPubMed
30.
go back to reference Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, Luo W, Li Y, Caracciolo L, Russo I, Barlati S, Ray B, Lahiri DK, Bosetti F, Greig NH, Rosi S: Tumor necrosis factor-α synthesis inhibitor 3,6′-dithiothalidomide attenuates markers of inflammation. Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer’s disease. J Neuroinflammation 2012, 9:106.CrossRefPubMedPubMedCentral Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, Luo W, Li Y, Caracciolo L, Russo I, Barlati S, Ray B, Lahiri DK, Bosetti F, Greig NH, Rosi S: Tumor necrosis factor-α synthesis inhibitor 3,6′-dithiothalidomide attenuates markers of inflammation. Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer’s disease. J Neuroinflammation 2012, 9:106.CrossRefPubMedPubMedCentral
31.
go back to reference Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rübe CE, Walter J, Heneka MT, Hartmann T, Menger MD, Fassbender K: TLR2 is a primary receptor for Alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J Immunol 2012, 188:1098–1107.CrossRefPubMed Liu S, Liu Y, Hao W, Wolf L, Kiliaan AJ, Penke B, Rübe CE, Walter J, Heneka MT, Hartmann T, Menger MD, Fassbender K: TLR2 is a primary receptor for Alzheimer’s amyloid β peptide to trigger neuroinflammatory activation. J Immunol 2012, 188:1098–1107.CrossRefPubMed
32.
go back to reference Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T, Staufenbiel M, Iwata N, Ono M, Saido TC, Suzuki K, Mori H, Higuchi M, Suhara T: Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of Alzheimer’s disease enabled by positron emission tomography. J Neurosci 2007, 27:10957–10968.CrossRefPubMed Maeda J, Ji B, Irie T, Tomiyama T, Maruyama M, Okauchi T, Staufenbiel M, Iwata N, Ono M, Saido TC, Suzuki K, Mori H, Higuchi M, Suhara T: Longitudinal, quantitative assessment of amyloid, neuroinflammation, and anti-amyloid treatment in a living mouse model of Alzheimer’s disease enabled by positron emission tomography. J Neurosci 2007, 27:10957–10968.CrossRefPubMed
33.
go back to reference Zhu LH, Bi W, Qi RB, Wang HD, Wang ZG, Zeng Q, Zhao YR, Lu DX: Luteolin reduces primary hippocampal neurons death induced by neuroinflammation. Neurol Res 2011, 33:927–934.CrossRefPubMed Zhu LH, Bi W, Qi RB, Wang HD, Wang ZG, Zeng Q, Zhao YR, Lu DX: Luteolin reduces primary hippocampal neurons death induced by neuroinflammation. Neurol Res 2011, 33:927–934.CrossRefPubMed
34.
go back to reference Lee JY, Cho E, Ko YE, Kim I, Lee KJ, Kwon SU, Kang DW, Kim JS: Ibudilast, a phosphodiesterase inhibitor with anti-inflammatory activity, protects against ischemic brain injury in rats. Brain Res 2012, 1431:97–106.CrossRefPubMed Lee JY, Cho E, Ko YE, Kim I, Lee KJ, Kwon SU, Kang DW, Kim JS: Ibudilast, a phosphodiesterase inhibitor with anti-inflammatory activity, protects against ischemic brain injury in rats. Brain Res 2012, 1431:97–106.CrossRefPubMed
35.
go back to reference Liu T, Jin H, Sun QR, Xu JH, Hu HT: The neuroprotective effects of tanshinone IIA on β-amyloid-induced toxicity in rat cortical neurons. Neuropharmacology 2010, 59:595–604.CrossRefPubMed Liu T, Jin H, Sun QR, Xu JH, Hu HT: The neuroprotective effects of tanshinone IIA on β-amyloid-induced toxicity in rat cortical neurons. Neuropharmacology 2010, 59:595–604.CrossRefPubMed
36.
go back to reference Franciosi S, Ryu JK, Choi HB, Radov L, Kim SU, McLarnon JG: Broad-spectrum effects of 4-aminopyridine to modulate amyloid beta1–42-induced cell signaling and functional responses in human microglia. J Neurosci 2006, 26:11652–11664.CrossRefPubMed Franciosi S, Ryu JK, Choi HB, Radov L, Kim SU, McLarnon JG: Broad-spectrum effects of 4-aminopyridine to modulate amyloid beta1–42-induced cell signaling and functional responses in human microglia. J Neurosci 2006, 26:11652–11664.CrossRefPubMed
37.
go back to reference Peila R, Launer LJ: Inflammation and dementia: epidemiologic evidence. Acta Neurol Scand Suppl 2006, 185:102–106.CrossRefPubMed Peila R, Launer LJ: Inflammation and dementia: epidemiologic evidence. Acta Neurol Scand Suppl 2006, 185:102–106.CrossRefPubMed
38.
go back to reference Morales I, Farías G, Maccioni RB: Neuroimmunomodulation in the pathogenesis of Alzheimer’s disease. Neuroimmunomodulation 2010, 17:202–204.CrossRefPubMed Morales I, Farías G, Maccioni RB: Neuroimmunomodulation in the pathogenesis of Alzheimer’s disease. Neuroimmunomodulation 2010, 17:202–204.CrossRefPubMed
39.
go back to reference Angelopoulos P, Agouridaki H, Vaiopoulos H, Siskou E, Doutsou K, Costa V, Baloyiannis SI: Cytokines in Alzheimer’s disease and vascular dementia. Int J Neurosci 2008, 118:1659–1672.CrossRefPubMed Angelopoulos P, Agouridaki H, Vaiopoulos H, Siskou E, Doutsou K, Costa V, Baloyiannis SI: Cytokines in Alzheimer’s disease and vascular dementia. Int J Neurosci 2008, 118:1659–1672.CrossRefPubMed
40.
go back to reference Forlenza OV, Diniz BS, Talib LL, Mendonça VA, Ojopi EB, Gattaz WF, Teixeira AL: Increased serum IL-1beta level in Alzheimer’s disease and mild cognitive impairment. Dement Geriatr Cogn Disord 2009, 28:507–512.CrossRefPubMed Forlenza OV, Diniz BS, Talib LL, Mendonça VA, Ojopi EB, Gattaz WF, Teixeira AL: Increased serum IL-1beta level in Alzheimer’s disease and mild cognitive impairment. Dement Geriatr Cogn Disord 2009, 28:507–512.CrossRefPubMed
41.
go back to reference Alkam T, Nitta A, Mizoguchi H, Saito K, Seshima M, Itoh A, Yamada K, Nabeshima T: Restraining tumor necrosis factor-alpha by thalidomide prevents the amyloid beta-induced impairment of recognition memory in mice. Behav Brain Res 2008, 189:100–106.CrossRefPubMed Alkam T, Nitta A, Mizoguchi H, Saito K, Seshima M, Itoh A, Yamada K, Nabeshima T: Restraining tumor necrosis factor-alpha by thalidomide prevents the amyloid beta-induced impairment of recognition memory in mice. Behav Brain Res 2008, 189:100–106.CrossRefPubMed
42.
go back to reference Koenigsknecht-Talboo J, Landreth GE: Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci 2005, 25:8240–8249.CrossRefPubMed Koenigsknecht-Talboo J, Landreth GE: Microglial phagocytosis induced by fibrillar beta-amyloid and IgGs are differentially regulated by proinflammatory cytokines. J Neurosci 2005, 25:8240–8249.CrossRefPubMed
43.
go back to reference Abe K, Kimura H: The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci 1996, 16:1066–1071.PubMed Abe K, Kimura H: The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci 1996, 16:1066–1071.PubMed
44.
go back to reference Li L, Bhatia M, Zhu YZ, Zhu YC, Ramnath RD, Wang ZJ, Anuar FB, Whiteman M, Salto-Tellez M, Moore PK: Hydrogen sulfide is a novel mediator of lipopolysaccharide-induced inflammation in the mouse. FASEB J 2005, 19:1196–1198.PubMed Li L, Bhatia M, Zhu YZ, Zhu YC, Ramnath RD, Wang ZJ, Anuar FB, Whiteman M, Salto-Tellez M, Moore PK: Hydrogen sulfide is a novel mediator of lipopolysaccharide-induced inflammation in the mouse. FASEB J 2005, 19:1196–1198.PubMed
45.
go back to reference Lee M, Sparatore A, Del Soldato P, McGeer E, McGeer PL: Hydrogen sulfide-releasing NSAIDs attenuate neuroinflammation induced by microglial and astrocytic activation. Glia 2010, 58:103–113.CrossRefPubMed Lee M, Sparatore A, Del Soldato P, McGeer E, McGeer PL: Hydrogen sulfide-releasing NSAIDs attenuate neuroinflammation induced by microglial and astrocytic activation. Glia 2010, 58:103–113.CrossRefPubMed
46.
go back to reference Gong QH, Wang Q, Pan LL, Liu XH, Huang H, Zhu YZ: Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: a pro-inflammatory pathway in rats. Pharmacol Biochem Behav 2010, 96:52–58.CrossRefPubMed Gong QH, Wang Q, Pan LL, Liu XH, Huang H, Zhu YZ: Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: a pro-inflammatory pathway in rats. Pharmacol Biochem Behav 2010, 96:52–58.CrossRefPubMed
47.
go back to reference Giovannini MG, Scal C, Prosperi C, Bellucci A, Vannucchi MG, Rosi S, Pepeu G, Casamenti F: Beta-amyloid-induced inflammation and cholinergic hypofunction in the rat brain in vivo: involvement of the p38MAPK pathway. Neurobiol Dis 2002, 11:257–274.CrossRefPubMed Giovannini MG, Scal C, Prosperi C, Bellucci A, Vannucchi MG, Rosi S, Pepeu G, Casamenti F: Beta-amyloid-induced inflammation and cholinergic hypofunction in the rat brain in vivo: involvement of the p38MAPK pathway. Neurobiol Dis 2002, 11:257–274.CrossRefPubMed
48.
go back to reference Ji C, Aisa HA, Yang N, Li Q, Wang T, Zhang L, Qu K, Zhu HB, Zuo PP: Gossypium herbaceam extracts inhibited NF-kappaB activation to attenuate spatial memory impairment and hippocampal neurodegeneration induced by amyloid-beta in rats. J Alzheimers Dis 2008, 14:271–283.PubMed Ji C, Aisa HA, Yang N, Li Q, Wang T, Zhang L, Qu K, Zhu HB, Zuo PP: Gossypium herbaceam extracts inhibited NF-kappaB activation to attenuate spatial memory impairment and hippocampal neurodegeneration induced by amyloid-beta in rats. J Alzheimers Dis 2008, 14:271–283.PubMed
49.
go back to reference Minogue AM, Lynch AM, Loane DJ, Herron CE, Lynch MA: Modulation of amyloid-beta-induced and age-associated changes in rat hippocampus by eicosapentaenoic acid. J Neurochem 2007, 103:914–926.CrossRefPubMed Minogue AM, Lynch AM, Loane DJ, Herron CE, Lynch MA: Modulation of amyloid-beta-induced and age-associated changes in rat hippocampus by eicosapentaenoic acid. J Neurochem 2007, 103:914–926.CrossRefPubMed
50.
go back to reference Wang C, Li J, Liu Q, Yang R, Zhang JH, Cao YP, Sun XJ: Hydrogen-rich saline reduces oxidative stress and inflammation by inhibit of JNK and NF-κB activation in a rat model of amyloid-beta-induced Alzheimer’s disease. Neurosci Lett 2011, 491:127–132.CrossRefPubMed Wang C, Li J, Liu Q, Yang R, Zhang JH, Cao YP, Sun XJ: Hydrogen-rich saline reduces oxidative stress and inflammation by inhibit of JNK and NF-κB activation in a rat model of amyloid-beta-induced Alzheimer’s disease. Neurosci Lett 2011, 491:127–132.CrossRefPubMed
51.
go back to reference Zhu X, Rottkamp CA, Hartzler A, Sun Z, Takeda A, Boux H, Shimohama S, Perry G, Smith MA: Activation of MKK6, an upstream activator of p38, in Alzheimer’s disease. J Neurochem 2001, 79:311–318.CrossRefPubMed Zhu X, Rottkamp CA, Hartzler A, Sun Z, Takeda A, Boux H, Shimohama S, Perry G, Smith MA: Activation of MKK6, an upstream activator of p38, in Alzheimer’s disease. J Neurochem 2001, 79:311–318.CrossRefPubMed
52.
go back to reference Kaltschmidt B, Uherek M, Volk B, Baeuerle PA, Kaltschmidt C: Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proc Natl Acad Sci U S A 1997, 94:2642–2647.CrossRefPubMedPubMedCentral Kaltschmidt B, Uherek M, Volk B, Baeuerle PA, Kaltschmidt C: Transcription factor NF-kappaB is activated in primary neurons by amyloid beta peptides and in neurons surrounding early plaques from patients with Alzheimer disease. Proc Natl Acad Sci U S A 1997, 94:2642–2647.CrossRefPubMedPubMedCentral
53.
go back to reference Kitamura Y, Shimohama S, Ota T, Matsuoka Y, Nomura Y, Taniguchi T: Alteration of transcription factors NF-kappaB and STAT1 in Alzheimer’s disease brains. Neurosci Lett 1997, 237:17–20.CrossRefPubMed Kitamura Y, Shimohama S, Ota T, Matsuoka Y, Nomura Y, Taniguchi T: Alteration of transcription factors NF-kappaB and STAT1 in Alzheimer’s disease brains. Neurosci Lett 1997, 237:17–20.CrossRefPubMed
54.
go back to reference Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi S, Mucke L, Gan L: SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem 2005, 280:40364–40374.CrossRefPubMed Chen J, Zhou Y, Mueller-Steiner S, Chen LF, Kwon H, Yi S, Mucke L, Gan L: SIRT1 protects against microglia-dependent amyloid-beta toxicity through inhibiting NF-kappaB signaling. J Biol Chem 2005, 280:40364–40374.CrossRefPubMed
55.
go back to reference Ebenezer PJ, Weidner AM, LeVine H, Markesbery WR, Murphy MP, Zhang L, Dasuri K, Fernandez-Kim SO, Bruce-Keller AJ, Gavilán E, Keller JN: Neuron specific toxicity of oligomeric amyloid-β: role for JUN-kinase and oxidative stress. J Alzheimers Dis 2010, 22:839–848.PubMedPubMedCentral Ebenezer PJ, Weidner AM, LeVine H, Markesbery WR, Murphy MP, Zhang L, Dasuri K, Fernandez-Kim SO, Bruce-Keller AJ, Gavilán E, Keller JN: Neuron specific toxicity of oligomeric amyloid-β: role for JUN-kinase and oxidative stress. J Alzheimers Dis 2010, 22:839–848.PubMedPubMedCentral
Metadata
Title
Hydrogen sulfide attenuates spatial memory impairment and hippocampal neuroinflammation in beta-amyloid rat model of Alzheimer’s disease
Authors
Aiguo Xuan
Dahong Long
Jianhua Li
Weidong Ji
Meng Zhang
Lepeng Hong
Jihong Liu
Publication date
01-12-2012
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2012
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/1742-2094-9-202

Other articles of this Issue 1/2012

Journal of Neuroinflammation 1/2012 Go to the issue