Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer’s disease

Authors: Minsook Ye, Hwan-Suck Chung, Chanju Lee, Moon Sik Yoon, A. Ram Yu, Jin Su Kim, Deok-Sang Hwang, Insop Shim, Hyunsu Bae

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Alzheimer’s disease (AD) is a severe neuroinflammatory disease. CD4+Foxp3+ regulatory T cells (Tregs) modulate various inflammatory diseases via suppressing Th cell activation. There are increasing evidences that Tregs have beneficial roles in neurodegenerative diseases. Previously, we found the population of Treg cells was significantly increased by bee venom phospholipase A2 (bvPLA2) treatment in vivo and in vitro.

Methods

To examine the effects of bvPLA2 on AD, bvPLA2 was administered to 3xTg-AD mice, mouse model of Alzheimer’s disease. The levels of amyloid beta (Aβ) deposits in the hippocampus, glucose metabolism in the brain, microglia activation, and CD4+ T cell infiltration were analyzed to evaluate the neuroprotective effect of bvPLA2.

Results

bvPLA2 treatment significantly enhanced the cognitive function of the 3xTg-AD mice and increased glucose metabolism, as assessed with 18F-2 fluoro-2-deoxy-D-glucose ([F-18] FDG) positron emission tomography (PET). The levels of Aβ deposits in the hippocampus were dramatically decreased by bvPLA2 treatment. This neuroprotective effect of bvPLA2 was associated with microglial deactivation and reduction in CD4+ T cell infiltration. Interestingly, the neuroprotective effects of bvPLA2 were abolished in Treg-depleted mice.

Conclusions

The present studies strongly suggest that the increase of Treg population by bvPLA2 treatment might inhibit progression of AD in the 3xTg AD mice.
Literature
1.
go back to reference Finder VH. Alzheimer's disease: a general introduction and pathomechanism. J Alzheimers Dis. 2010;22 Suppl 3:5–19.PubMed Finder VH. Alzheimer's disease: a general introduction and pathomechanism. J Alzheimers Dis. 2010;22 Suppl 3:5–19.PubMed
2.
go back to reference Honjo K, Black SE, Verhoeff NP. Alzheimer’s disease, cerebrovascular disease, and the beta-amyloid cascade. Can J Neurol Sci. 2012;39:712–28.PubMedCrossRef Honjo K, Black SE, Verhoeff NP. Alzheimer’s disease, cerebrovascular disease, and the beta-amyloid cascade. Can J Neurol Sci. 2012;39:712–28.PubMedCrossRef
3.
go back to reference Kim DKSJ, Park JD, Choi BS. Copper induces the accumulation of amyloid-beta in the brain. Mol Cell Toxicol. 2013;9:57–66.CrossRef Kim DKSJ, Park JD, Choi BS. Copper induces the accumulation of amyloid-beta in the brain. Mol Cell Toxicol. 2013;9:57–66.CrossRef
5.
go back to reference Weiner HL, Frenkel D. Immunology and immunotherapy of Alzheimer’s disease. Nat Rev Immunol. 2006;6:404–16.PubMedCrossRef Weiner HL, Frenkel D. Immunology and immunotherapy of Alzheimer’s disease. Nat Rev Immunol. 2006;6:404–16.PubMedCrossRef
6.
go back to reference McGeer EG, McGeer PL. Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:741–9.PubMedCrossRef McGeer EG, McGeer PL. Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27:741–9.PubMedCrossRef
7.
go back to reference Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, et al. In-vivo measurement of activated microglia in dementia. Lancet. 2001;358:461–7.PubMedCrossRef Cagnin A, Brooks DJ, Kennedy AM, Gunn RN, Myers R, Turkheimer FE, et al. In-vivo measurement of activated microglia in dementia. Lancet. 2001;358:461–7.PubMedCrossRef
8.
go back to reference Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, et al. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology. 2009;72:56–62.PubMedPubMedCentralCrossRef Okello A, Edison P, Archer HA, Turkheimer FE, Kennedy J, Bullock R, et al. Microglial activation and amyloid deposition in mild cognitive impairment: a PET study. Neurology. 2009;72:56–62.PubMedPubMedCentralCrossRef
9.
go back to reference Rogers J, Lue LF. Microglial chemotaxis, activation, and phagocytosis of amyloid beta-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int. 2001;39:333–40.PubMedCrossRef Rogers J, Lue LF. Microglial chemotaxis, activation, and phagocytosis of amyloid beta-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int. 2001;39:333–40.PubMedCrossRef
10.
go back to reference Cheng D, Spiro AS, Jenner AM, Garner B, Karl T. Long-term cannabidiol treatment prevents the development of social recognition memory deficits in Alzheimer’s disease transgenic mice. J Alzheimers Dis. 2014. Cheng D, Spiro AS, Jenner AM, Garner B, Karl T. Long-term cannabidiol treatment prevents the development of social recognition memory deficits in Alzheimer’s disease transgenic mice. J Alzheimers Dis. 2014.
12.
go back to reference Terry Jr AV, Buccafusco JJ. The cholinergic hypothesis of age and Alzheimer’s disease-related cognitive deficits: recent challenges and their implications for novel drug development. J Pharmacol Exp Ther. 2003;306:821–7.PubMedCrossRef Terry Jr AV, Buccafusco JJ. The cholinergic hypothesis of age and Alzheimer’s disease-related cognitive deficits: recent challenges and their implications for novel drug development. J Pharmacol Exp Ther. 2003;306:821–7.PubMedCrossRef
13.
go back to reference Rountree SD, Chan W, Pavlik VN, Darby EJ, Siddiqui S, Doody RS. Persistent treatment with cholinesterase inhibitors and/or memantine slows clinical progression of Alzheimer disease. Alzheimers Res Ther. 2009;1:7.PubMedPubMedCentralCrossRef Rountree SD, Chan W, Pavlik VN, Darby EJ, Siddiqui S, Doody RS. Persistent treatment with cholinesterase inhibitors and/or memantine slows clinical progression of Alzheimer disease. Alzheimers Res Ther. 2009;1:7.PubMedPubMedCentralCrossRef
14.
15.
go back to reference Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–21.PubMedCrossRef Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed R, et al. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron. 2003;39:409–21.PubMedCrossRef
16.
go back to reference Gimenez-Llort L, Blazquez G, Canete T, Johansson B, Oddo S, Tobena A, et al. Modeling behavioral and neuronal symptoms of Alzheimer’s disease in mice: a role for intraneuronal amyloid. Neurosci Biobehav Rev. 2007;31:125–47.PubMedCrossRef Gimenez-Llort L, Blazquez G, Canete T, Johansson B, Oddo S, Tobena A, et al. Modeling behavioral and neuronal symptoms of Alzheimer’s disease in mice: a role for intraneuronal amyloid. Neurosci Biobehav Rev. 2007;31:125–47.PubMedCrossRef
17.
go back to reference Oddo S, Caccamo A, Tran L, Lambert MP, Glabe CG, Klein WL, et al. Temporal profile of amyloid-beta (Abeta) oligomerization in an in vivo model of Alzheimer disease. A link between Abeta and tau pathology. J Biol Chem. 2006;281:1599–604.PubMedCrossRef Oddo S, Caccamo A, Tran L, Lambert MP, Glabe CG, Klein WL, et al. Temporal profile of amyloid-beta (Abeta) oligomerization in an in vivo model of Alzheimer disease. A link between Abeta and tau pathology. J Biol Chem. 2006;281:1599–604.PubMedCrossRef
19.
go back to reference Linderoth L, Peters GH, Jorgensen K, Madsen R, Andresen TL. Synthesis of sn-1 functionalized phospholipids as substrates for secretory phospholipase A2. Chem Phys Lipids. 2007;146:54–66.PubMedCrossRef Linderoth L, Peters GH, Jorgensen K, Madsen R, Andresen TL. Synthesis of sn-1 functionalized phospholipids as substrates for secretory phospholipase A2. Chem Phys Lipids. 2007;146:54–66.PubMedCrossRef
20.
go back to reference Dennis EA. Diversity of group types, regulation, and function of phospholipase A2. J Biol Chem. 1994;269:13057–60.PubMed Dennis EA. Diversity of group types, regulation, and function of phospholipase A2. J Biol Chem. 1994;269:13057–60.PubMed
21.
go back to reference Mukherjee AB, Miele L, Pattabiraman N. Phospholipase A2 enzymes: regulation and physiological role. Biochem Pharmacol. 1994;48:1–10.PubMedCrossRef Mukherjee AB, Miele L, Pattabiraman N. Phospholipase A2 enzymes: regulation and physiological role. Biochem Pharmacol. 1994;48:1–10.PubMedCrossRef
22.
go back to reference Dennis EA, Rhee SG, Billah MM, Hannun YA. Role of phospholipase in generating lipid second messengers in signal transduction. FASEB J. 1991;5:2068–77.PubMed Dennis EA, Rhee SG, Billah MM, Hannun YA. Role of phospholipase in generating lipid second messengers in signal transduction. FASEB J. 1991;5:2068–77.PubMed
23.
go back to reference Granata F, Frattini A, Loffredo S, Del Prete A, Sozzani S, Marone G, et al. Signaling events involved in cytokine and chemokine production induced by secretory phospholipase A2 in human lung macrophages. Eur J Immunol. 2006;36:1938–50.PubMedCrossRef Granata F, Frattini A, Loffredo S, Del Prete A, Sozzani S, Marone G, et al. Signaling events involved in cytokine and chemokine production induced by secretory phospholipase A2 in human lung macrophages. Eur J Immunol. 2006;36:1938–50.PubMedCrossRef
24.
go back to reference Kim H, Keum DJ, Kwak J, Chung HS, Bae H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS One. 2014;9:e114726.PubMedPubMedCentralCrossRef Kim H, Keum DJ, Kwak J, Chung HS, Bae H. Bee venom phospholipase A2 protects against acetaminophen-induced acute liver injury by modulating regulatory T cells and IL-10 in mice. PLoS One. 2014;9:e114726.PubMedPubMedCentralCrossRef
25.
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.PubMedCrossRef Morris R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods. 1984;11:47–60.PubMedCrossRef
26.
go back to reference Ye M, Lee SG, Chung ES, Lim SJ, Kim WS, Yoon H, et al. Neuroprotective effects of cuscutae semen in a mouse model of Parkinson's disease. Evid Based Complement Alternat Med. 2014;2014:150153.PubMedPubMedCentral Ye M, Lee SG, Chung ES, Lim SJ, Kim WS, Yoon H, et al. Neuroprotective effects of cuscutae semen in a mouse model of Parkinson's disease. Evid Based Complement Alternat Med. 2014;2014:150153.PubMedPubMedCentral
27.
go back to reference Sala C, Rudolph-Correia S, Sheng M. Developmentally regulated NMDA receptor-dependent dephosphorylation of cAMP response element-binding protein (CREB) in hippocampal neurons. J Neurosci. 2000;20:3529–36.PubMed Sala C, Rudolph-Correia S, Sheng M. Developmentally regulated NMDA receptor-dependent dephosphorylation of cAMP response element-binding protein (CREB) in hippocampal neurons. J Neurosci. 2000;20:3529–36.PubMed
28.
go back to reference Paxinos G, Watson C, Pennisi M, Topple A. Bregma, lambda and the interaural midpoint in stereotaxic surgery with rats of different sex, strain and weight. J Neurosci Methods. 1985;13:139–43.PubMedCrossRef Paxinos G, Watson C, Pennisi M, Topple A. Bregma, lambda and the interaural midpoint in stereotaxic surgery with rats of different sex, strain and weight. J Neurosci Methods. 1985;13:139–43.PubMedCrossRef
29.
go back to reference Fueger BJ, Czernin J, Hildebrandt I, Tran C, Halpern BS, Stout D, et al. Impact of animal handling on the results of 18F-FDG PET studies in mice. J Nucl Med. 2006;47:999–1006.PubMed Fueger BJ, Czernin J, Hildebrandt I, Tran C, Halpern BS, Stout D, et al. Impact of animal handling on the results of 18F-FDG PET studies in mice. J Nucl Med. 2006;47:999–1006.PubMed
30.
go back to reference Bao Q, Newport D, Chen M, Stout DB, Chatziioannou AF. Performance evaluation of the inveon dedicated PET preclinical tomograph based on the NEMA NU-4 standards. J Nucl Med. 2009;50:401–8.PubMedPubMedCentralCrossRef Bao Q, Newport D, Chen M, Stout DB, Chatziioannou AF. Performance evaluation of the inveon dedicated PET preclinical tomograph based on the NEMA NU-4 standards. J Nucl Med. 2009;50:401–8.PubMedPubMedCentralCrossRef
31.
go back to reference Swartzwelder HS, Dyer RS, Holahan W, Myers RD. Activity changes in rats following acute trimethyltin exposure. Neurotoxicology. 1981;2:589–93.PubMed Swartzwelder HS, Dyer RS, Holahan W, Myers RD. Activity changes in rats following acute trimethyltin exposure. Neurotoxicology. 1981;2:589–93.PubMed
32.
go back to reference Setiady YY, Coccia JA, Park PU. In vivo depletion of CD4+FOXP3+ Treg cells by the PC61 anti-CD25 monoclonal antibody is mediated by FcgammaRIII+ phagocytes. Eur J Immunol. 2010;40:780–786.PubMedCrossRef Setiady YY, Coccia JA, Park PU. In vivo depletion of CD4+FOXP3+ Treg cells by the PC61 anti-CD25 monoclonal antibody is mediated by FcgammaRIII+ phagocytes. Eur J Immunol. 2010;40:780–786.PubMedCrossRef
33.
go back to reference Lee H, Kwon Y, Lee JH, Kim J, Shin MK, Kim SH, Bae H: Methyl gallate exhibits potent antitumor activities by inhibiting tumor infiltration of CD4+CD25+ regulatory T cells. J Immunol. 2010;185:6698–705.PubMedCrossRef Lee H, Kwon Y, Lee JH, Kim J, Shin MK, Kim SH, Bae H: Methyl gallate exhibits potent antitumor activities by inhibiting tumor infiltration of CD4+CD25+ regulatory T cells. J Immunol. 2010;185:6698–705.PubMedCrossRef
34.
35.
go back to reference Schmole AC, Lundt R, Ternes S, Albayram O, Ulas T, Schultze JL, et al. Cannabinoid receptor 2 deficiency results in reduced neuroinflammation in an Alzheimer’s disease mouse model. Neurobiol Aging. 2015;36:710–9.PubMedCrossRef Schmole AC, Lundt R, Ternes S, Albayram O, Ulas T, Schultze JL, et al. Cannabinoid receptor 2 deficiency results in reduced neuroinflammation in an Alzheimer’s disease mouse model. Neurobiol Aging. 2015;36:710–9.PubMedCrossRef
37.
go back to reference Kadiu I, Glanzer JG, Kipnis J, Gendelman HE, Thomas MP. Mononuclear phagocytes in the pathogenesis of neurodegenerative diseases. Neurotox Res. 2005;8:25–50.PubMedCrossRef Kadiu I, Glanzer JG, Kipnis J, Gendelman HE, Thomas MP. Mononuclear phagocytes in the pathogenesis of neurodegenerative diseases. Neurotox Res. 2005;8:25–50.PubMedCrossRef
38.
go back to reference McGeer PL, McGeer EG. The inflammatory response system of brain: implications for therapy of Alzheimer and other neurodegenerative diseases. Brain Res Brain Res Rev. 1995;21:195–218.PubMedCrossRef McGeer PL, McGeer EG. The inflammatory response system of brain: implications for therapy of Alzheimer and other neurodegenerative diseases. Brain Res Brain Res Rev. 1995;21:195–218.PubMedCrossRef
40.
go back to reference Kuniyasu Y, Takahashi T, Itoh M, Shimizu J, Toda G, Sakaguchi S. Naturally anergic and suppressive CD25(+)CD4(+) T cells as a functionally and phenotypically distinct immunoregulatory T cell subpopulation. Int Immunol. 2000;12:1145–55.PubMedCrossRef Kuniyasu Y, Takahashi T, Itoh M, Shimizu J, Toda G, Sakaguchi S. Naturally anergic and suppressive CD25(+)CD4(+) T cells as a functionally and phenotypically distinct immunoregulatory T cell subpopulation. Int Immunol. 2000;12:1145–55.PubMedCrossRef
41.
go back to reference Wang L, Xie Y, Zhu LJ, Chang TT, Mao YQ, Li J. An association between immunosenescence and CD4(+)CD25(+) regulatory T cells: a systematic review. Biomed Environ Sci. 2010;23:327–32.PubMedCrossRef Wang L, Xie Y, Zhu LJ, Chang TT, Mao YQ, Li J. An association between immunosenescence and CD4(+)CD25(+) regulatory T cells: a systematic review. Biomed Environ Sci. 2010;23:327–32.PubMedCrossRef
42.
go back to reference Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005;76:77–98.PubMedCrossRef Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol. 2005;76:77–98.PubMedCrossRef
43.
go back to reference Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer’s disease. J Immunol. 2008;181:7254–62.PubMedPubMedCentralCrossRef Jana M, Palencia CA, Pahan K. Fibrillar amyloid-beta peptides activate microglia via TLR2: implications for Alzheimer’s disease. J Immunol. 2008;181:7254–62.PubMedPubMedCentralCrossRef
44.
go back to reference Muehlhauser F, Liebl U, Kuehl S, Walter S, Bertsch T, Fassbender K. Aggregation-dependent interaction of the Alzheimer’s beta-amyloid and microglia. Clin Chem Lab Med. 2001;39:313–6.PubMedCrossRef Muehlhauser F, Liebl U, Kuehl S, Walter S, Bertsch T, Fassbender K. Aggregation-dependent interaction of the Alzheimer’s beta-amyloid and microglia. Clin Chem Lab Med. 2001;39:313–6.PubMedCrossRef
45.
go back to reference Delgado S, Sheremata WA. The role of CD4+ T-cells in the development of MS. Neurol Res. 2006;28:245–9.PubMedCrossRef Delgado S, Sheremata WA. The role of CD4+ T-cells in the development of MS. Neurol Res. 2006;28:245–9.PubMedCrossRef
46.
go back to reference Town T, Tan J, Flavell RA, Mullan M. T-cells in Alzheimer’s disease. Neuromolecular Med. 2005;7:255–64.PubMedCrossRef Town T, Tan J, Flavell RA, Mullan M. T-cells in Alzheimer’s disease. Neuromolecular Med. 2005;7:255–64.PubMedCrossRef
47.
go back to reference Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62.PubMedCrossRef Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annu Rev Immunol. 2004;22:531–62.PubMedCrossRef
48.
go back to reference Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–87.PubMedCrossRef Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133:775–87.PubMedCrossRef
49.
go back to reference Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009;15:192–9.PubMedCrossRef Liesz A, Suri-Payer E, Veltkamp C, Doerr H, Sommer C, Rivest S, et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat Med. 2009;15:192–9.PubMedCrossRef
50.
go back to reference Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82:1083–94.PubMedCrossRef Reynolds AD, Banerjee R, Liu J, Gendelman HE, Mosley RL. Neuroprotective activities of CD4+CD25+ regulatory T cells in an animal model of Parkinson’s disease. J Leukoc Biol. 2007;82:1083–94.PubMedCrossRef
51.
go back to reference Subramanian S, Ayala P, Wadsworth TL, Harris CJ, Vandenbark AA, Quinn JF, Offner H. CCR6: a biomarker for Alzheimer’s-like disease in a triple transgenic mouse model. J Alzheimers Dis. 2010;22:619–29.PubMedPubMedCentral Subramanian S, Ayala P, Wadsworth TL, Harris CJ, Vandenbark AA, Quinn JF, Offner H. CCR6: a biomarker for Alzheimer’s-like disease in a triple transgenic mouse model. J Alzheimers Dis. 2010;22:619–29.PubMedPubMedCentral
52.
go back to reference Larbi A, Pawelec G, Witkowski JM, Schipper HM, Derhovanessian E, Goldeck D, et al. Dramatic shifts in circulating CD4 but not CD8 T cell subsets in mild Alzheimer’s disease. J Alzheimers Dis. 2009;17:91–103.PubMed Larbi A, Pawelec G, Witkowski JM, Schipper HM, Derhovanessian E, Goldeck D, et al. Dramatic shifts in circulating CD4 but not CD8 T cell subsets in mild Alzheimer’s disease. J Alzheimers Dis. 2009;17:91–103.PubMed
53.
go back to reference Kim H, Lee H, Lee G, Jang H, Kim SS, Yoon H, et al. Phospholipase A2 inhibits cisplatin-induced acute kidney injury by modulating regulatory T cells by the CD206 mannose receptor. Kidney Int. 2015;88:550–9.PubMedCrossRef Kim H, Lee H, Lee G, Jang H, Kim SS, Yoon H, et al. Phospholipase A2 inhibits cisplatin-induced acute kidney injury by modulating regulatory T cells by the CD206 mannose receptor. Kidney Int. 2015;88:550–9.PubMedCrossRef
54.
go back to reference Klingenberg R, Gerdes N, Badeau RM, Gistera A, Strodthoff D, Ketelhuth DF, et al. Depletion of FOXP3+ regulatory T cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest. 2013;123:1323–34.PubMedPubMedCentralCrossRef Klingenberg R, Gerdes N, Badeau RM, Gistera A, Strodthoff D, Ketelhuth DF, et al. Depletion of FOXP3+ regulatory T cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest. 2013;123:1323–34.PubMedPubMedCentralCrossRef
55.
go back to reference Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204:57–63.PubMedPubMedCentralCrossRef Lahl K, Loddenkemper C, Drouin C, Freyer J, Arnason J, Eberl G, et al. Selective depletion of Foxp3+ regulatory T cells induces a scurfy-like disease. J Exp Med. 2007;204:57–63.PubMedPubMedCentralCrossRef
56.
go back to reference Taguchi O, Takahashi T. Administration of anti-interleukin-2 receptor alpha antibody in vivo induces localized autoimmune disease. Eur J Immunol. 1996;26:1608–12.PubMedCrossRef Taguchi O, Takahashi T. Administration of anti-interleukin-2 receptor alpha antibody in vivo induces localized autoimmune disease. Eur J Immunol. 1996;26:1608–12.PubMedCrossRef
57.
go back to reference McHugh RS, Shevach EM. Cutting edge: depletion of CD4+CD25+ regulatory T cells is necessary, but not sufficient, for induction of organ-specific autoimmune disease. J Immunol. 2002;168:5979–83.PubMedCrossRef McHugh RS, Shevach EM. Cutting edge: depletion of CD4+CD25+ regulatory T cells is necessary, but not sufficient, for induction of organ-specific autoimmune disease. J Immunol. 2002;168:5979–83.PubMedCrossRef
58.
go back to reference Weitz TM, Gate D, Rezai-Zadeh K, Town T. MyD88 is dispensable for cerebral amyloidosis and neuroinflammation in APP/PS1 transgenic mice. Am J Pathol. 2014;184:2855–61.PubMedPubMedCentralCrossRef Weitz TM, Gate D, Rezai-Zadeh K, Town T. MyD88 is dispensable for cerebral amyloidosis and neuroinflammation in APP/PS1 transgenic mice. Am J Pathol. 2014;184:2855–61.PubMedPubMedCentralCrossRef
59.
go back to reference Goni F, Herline K, Peyser D, Wong K, Ji Y, Sun Y, et al. Immunomodulation targeting of both Abeta and tau pathological conformers ameliorates Alzheimer’s disease pathology in TgSwDI and 3xTg mouse models. J Neuroinflammation. 2013;10:150.PubMedPubMedCentralCrossRef Goni F, Herline K, Peyser D, Wong K, Ji Y, Sun Y, et al. Immunomodulation targeting of both Abeta and tau pathological conformers ameliorates Alzheimer’s disease pathology in TgSwDI and 3xTg mouse models. J Neuroinflammation. 2013;10:150.PubMedPubMedCentralCrossRef
60.
go back to reference Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM. Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol Aging. 2003;24:1063–70.PubMedCrossRef Oddo S, Caccamo A, Kitazawa M, Tseng BP, LaFerla FM. Amyloid deposition precedes tangle formation in a triple transgenic model of Alzheimer’s disease. Neurobiol Aging. 2003;24:1063–70.PubMedCrossRef
61.
go back to reference Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM. Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45:675–88.PubMedCrossRef Billings LM, Oddo S, Green KN, McGaugh JL, LaFerla FM. Intraneuronal Abeta causes the onset of early Alzheimer’s disease-related cognitive deficits in transgenic mice. Neuron. 2005;45:675–88.PubMedCrossRef
62.
go back to reference Sperling RA, Dickerson BC, Pihlajamaki M, Vannini P, LaViolette PS, Vitolo OV, et al. Functional alterations in memory networks in early Alzheimer’s disease. Neuromolecular Med. 2010;12:27–43.PubMedPubMedCentralCrossRef Sperling RA, Dickerson BC, Pihlajamaki M, Vannini P, LaViolette PS, Vitolo OV, et al. Functional alterations in memory networks in early Alzheimer’s disease. Neuromolecular Med. 2010;12:27–43.PubMedPubMedCentralCrossRef
63.
go back to reference Mirrione MM, Schiffer WK, Fowler JS, Alexoff DL, Dewey SL, Tsirka SE. A novel approach for imaging brain-behavior relationships in mice reveals unexpected metabolic patterns during seizures in the absence of tissue plasminogen activator. Neuroimage. 2007;38:34–42.PubMedPubMedCentralCrossRef Mirrione MM, Schiffer WK, Fowler JS, Alexoff DL, Dewey SL, Tsirka SE. A novel approach for imaging brain-behavior relationships in mice reveals unexpected metabolic patterns during seizures in the absence of tissue plasminogen activator. Neuroimage. 2007;38:34–42.PubMedPubMedCentralCrossRef
64.
go back to reference Mirrione MM, Schiffer WK, Siddiq M, Dewey SL, Tsirka SE. PET imaging of glucose metabolism in a mouse model of temporal lobe epilepsy. Synapse. 2006;59:119–21.PubMedCrossRef Mirrione MM, Schiffer WK, Siddiq M, Dewey SL, Tsirka SE. PET imaging of glucose metabolism in a mouse model of temporal lobe epilepsy. Synapse. 2006;59:119–21.PubMedCrossRef
65.
go back to reference Nordberg A, Svensson AL. Cholinesterase inhibitors in the treatment of Alzheimer’s disease: a comparison of tolerability and pharmacology. Drug Saf. 1998;19:465–80.PubMedCrossRef Nordberg A, Svensson AL. Cholinesterase inhibitors in the treatment of Alzheimer’s disease: a comparison of tolerability and pharmacology. Drug Saf. 1998;19:465–80.PubMedCrossRef
Metadata
Title
Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer’s disease
Authors
Minsook Ye
Hwan-Suck Chung
Chanju Lee
Moon Sik Yoon
A. Ram Yu
Jin Su Kim
Deok-Sang Hwang
Insop Shim
Hyunsu Bae
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0476-z

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue