51
|
Switzer CH, Cheng RY, Vitek TM, Christensen DJ, Wink DA, Vitek MP. Targeting SET/I(2)PP2A oncoprotein functions as a multi-pathway strategy for cancer therapy. Oncogene 2011; 30:2504-13. [PMID: 21297667 PMCID: PMC3107340 DOI: 10.1038/onc.2010.622] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Revised: 12/16/2010] [Accepted: 12/17/2010] [Indexed: 12/14/2022]
Abstract
The SET oncoprotein participates in cancer progression by affecting multiple cellular processes, inhibiting the tumor suppressor protein phosphatase 2A (PP2A), and inhibiting the metastasis suppressor nm23-H1. On the basis of these multiple activities, we hypothesized that targeted inhibition of SET would have multiple discrete and measurable effects on cancer cells. Here, the effects of inhibiting SET oncoprotein function on intracellular signaling and proliferation of human cancer cell lines was investigated. We observed the effects of COG112, a novel SET interacting peptide, on PP2A activity, Akt signaling, nm23-H1 activity and cellular migration/invasion in human U87 glioblastoma and MDA-MB-231 breast adenocarcinoma cancer cell lines. We found that COG112 interacted with SET protein and inhibited the association between SET and PP2A catalytic subunit (PP2A-c) and nm23-H1. The interaction between COG112 and SET caused PP2A phosphatase and nm23-H1 exonuclease activities to increase. COG112-mediated increases in PP2A activity resulted in the inhibition of Akt signaling and cellular proliferation. Additionally, COG112 inhibited SET association with Ras-related C(3) botulinum toxin substrate 1 (Rac1), leading to decreased cellular migration and invasion. COG112 treatment releases the SET-mediated inhibition of the tumor suppressor PP2A, as well as the metastasis suppressor nm23-H1. These results establish SET as a novel molecular target and that the inhibition of SET may have beneficial effects in cancer chemotherapy.
Collapse
Affiliation(s)
- Christopher H. Switzer
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Robert Y.S. Cheng
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Timothy M. Vitek
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | - David A. Wink
- Radiation Biology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michael P. Vitek
- Cognosci, Inc., Research Triangle Park, NC 27709
- Division of Neurology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
52
|
Singh K, Chaturvedi R, Barry DP, Coburn LA, Asim M, Lewis ND, Piazuelo MB, Washington MK, Vitek MP, Wilson KT. The apolipoprotein E-mimetic peptide COG112 inhibits NF-kappaB signaling, proinflammatory cytokine expression, and disease activity in murine models of colitis. J Biol Chem 2011; 286:3839-50. [PMID: 21115487 PMCID: PMC3030385 DOI: 10.1074/jbc.m110.176719] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD), consisting of Crohn's disease and ulcerative colitis, is a source of substantial morbidity and remains difficult to treat. New strategies for beneficial anti-inflammatory therapies would be highly desirable. Apolipoprotein (apo) E has immunomodulatory effects and synthetically derived apoE-mimetic peptides are beneficial in models of sepsis and neuroinflammation. We have reported that the antennapedia-linked apoE-mimetic peptide COG112 inhibits the inflammatory response to the colitis-inducing pathogen Citrobacter rodentium in vitro by inhibiting NF-κB activation. We now determined the effect of COG112 in mouse models of colitis. Using C. rodentium as an infection model, and dextran sulfate sodium (DSS) as an injury model, mice were treated with COG112 by intraperitoneal injection. With C. rodentium, COG112 improved the clinical parameters of survival, body weight, colon weight, and histologic injury. With DSS, COG112 ameliorated the loss of body weight, reduction in colon length, and histologic injury, whether administered concurrently with induction of colitis, during induction plus recovery, or only during the recovery phase of disease. In both colitis models, COG112 inhibited colon tissue inducible nitric-oxide synthase (iNOS), KC, TNF-α, IFN-γ, and IL-17 mRNA expression, and reduced nuclear translocation of NF-κB, as determined by immunoblot and immunofluorescence confocal microscopy. IκB kinase (IKK) activity was also reduced, which is necessary for activation of the canonical NF-κB pathway. Isolated colonic epithelial cells exhibited marked attenuation of expression of iNOS and the CXC chemokines KC and MIP-2. These studies indicate that apoE-mimetic peptides such as COG112 are novel potential therapies for IBD.
Collapse
Affiliation(s)
- Kshipra Singh
- From the Departments of Medicine, Division of Gastroenterology
- the Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, and
| | - Rupesh Chaturvedi
- From the Departments of Medicine, Division of Gastroenterology
- the Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, and
| | - Daniel P. Barry
- From the Departments of Medicine, Division of Gastroenterology
| | - Lori A. Coburn
- the Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, and
| | - Mohammad Asim
- From the Departments of Medicine, Division of Gastroenterology
- the Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, and
| | - Nuruddeen D. Lewis
- From the Departments of Medicine, Division of Gastroenterology
- Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | | | | | | | - Keith T. Wilson
- From the Departments of Medicine, Division of Gastroenterology
- Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232
- the Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee 37212, and
| |
Collapse
|
53
|
Laskowitz DT, Song P, Wang H, Mace B, Sullivan PM, Vitek MP, Dawson HN. Traumatic Brain Injury Exacerbates Neurodegenerative Pathology: Improvement with an Apolipoprotein E-Based Therapeutic. J Neurotrauma 2010; 27:1983-95. [DOI: 10.1089/neu.2010.1396] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Daniel T. Laskowitz
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
| | - Pingping Song
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Haichen Wang
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| | - Brian Mace
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Patrick M. Sullivan
- Department of Medicine (Geriatrics), Duke University Medical Center, Durham, North Carolina
| | - Michael P. Vitek
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina
- Cognosci, Inc., Research Triangle Park, North Carolina
| | - Hana N. Dawson
- Department of Medicine (Neurology), Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
54
|
Elliott DA, Weickert CS, Garner B. Apolipoproteins in the brain: implications for neurological and psychiatric disorders. CLINICAL LIPIDOLOGY 2010; 51:555-573. [PMID: 21423873 PMCID: PMC3058497 DOI: 10.2217/clp.10.37] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The brain is the most lipid-rich organ in the body and, owing to the impermeable nature of the blood-brain barrier, lipid and lipoprotein metabolism within this organ is distinct from the rest of the body. Apolipoproteins play a well-established role in the transport and metabolism of lipids within the CNS; however, evidence is emerging that they also fulfill a number of functions that extend beyond lipid transport and are critical for healthy brain function. The importance of apolipoproteins in brain physiology is highlighted by genetic studies, where apolipoprotein gene polymorphisms have been identified as risk factors for several neurological diseases. Furthermore, the expression of brain apolipoproteins is significantly altered in several brain disorders. The purpose of this article is to provide an up-to-date assessment of the major apolipoproteins found in the brain (ApoE, ApoJ, ApoD and ApoA-I), covering their proposed roles and the factors influencing their level of expression. Particular emphasis is placed on associations with neurological and psychiatric disorders.
Collapse
Affiliation(s)
- David A Elliott
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
| | - Cyndi Shannon Weickert
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
- Schizophrenia Research Institute, Sydney, NSW, Australia
- School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Brett Garner
- Prince of Wales Medical Research Institute, Randwick, Sydney, NSW 2031, Australia
- School of Biological Sciences, Faculty of Science, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
55
|
Li FQ, Fowler KA, Neil JE, Colton CA, Vitek MP. An apolipoprotein E-mimetic stimulates axonal regeneration and remyelination after peripheral nerve injury. J Pharmacol Exp Ther 2010; 334:106-15. [PMID: 20406857 PMCID: PMC2912037 DOI: 10.1124/jpet.110.167882] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Accepted: 04/19/2010] [Indexed: 12/30/2022] Open
Abstract
Elevated apolipoprotein E (apoE) synthesis within crushed sciatic nerves advocates that apoE could benefit axonal repair and reconstruction of axonal and myelin membranes. We created an apoE-mimetic peptide, COG112 (acetyl-RQIKIWFQNRRMKWKKCLRVRLASHLRKLRKRLL-amide), and found that postinjury treatment with COG112 significantly improved recovery of motor and sensory function following sciatic nerve crush in C57BL/6 mice. Morphometric analysis of injured sciatic nerves revealed that COG112 promoted axonal regrowth after 2 weeks of treatment. More strikingly, the thickness of myelin sheaths was increased by COG112 treatment. Consistent with these histological findings, COG112 potently elevated growth associated protein 43 (GAP-43) and peripheral myelin protein zero (P0), which are markers of axon regeneration and remyelination, respectively. Electron microscopic examination further suggested that the apoE-mimetic COG112 may increase clearance of myelin debris. Schwann cell uptake of cholesterol-containing low-density lipoprotein particles was selectively enhanced by COG112 treatment in a Schwann cell line S16. Moreover, COG112 significantly promoted axon elongation in primary dorsal root ganglion cultures from rat pups. Considering that cholesterol and lipids are needed for reconstructing myelin sheaths and axon extension, these data support a hypothesis where supplementation with exogenous apoE-mimetics such as COG112 may be a promising strategy for restoring lost functional and structural elements following nerve injury.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Apolipoproteins E/chemistry
- Apolipoproteins E/metabolism
- Axons/drug effects
- Axons/metabolism
- Blotting, Western
- Disease Models, Animal
- Female
- Lipoproteins, LDL/metabolism
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/metabolism
- Mice
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Molecular Sequence Data
- Myelin Sheath/metabolism
- Nerve Crush
- Nerve Fibers, Myelinated/drug effects
- Nerve Fibers, Myelinated/metabolism
- Nerve Regeneration/drug effects
- Peptides/administration & dosage
- Peptides/chemistry
- Peptides/pharmacology
- Peptides/therapeutic use
- Rats
- Schwann Cells
- Sciatic Nerve/drug effects
- Sciatic Nerve/injuries
- Sciatic Nerve/ultrastructure
- Sciatic Neuropathy/drug therapy
- Sciatic Neuropathy/metabolism
- Treatment Outcome
- Walking
Collapse
Affiliation(s)
- Feng-Qiao Li
- Cognosci, Inc., Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
56
|
Influence of apolipoprotein E plasma levels and tobacco smoking on the induction of neutralising antibodies to interferon-beta. J Neurol 2010; 257:1703-7. [DOI: 10.1007/s00415-010-5606-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Revised: 05/15/2010] [Accepted: 05/17/2010] [Indexed: 12/18/2022]
|
57
|
The immune-modulatory role of apolipoprotein E with emphasis on multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Dev Immunol 2010; 2010:186813. [PMID: 20613949 PMCID: PMC2896842 DOI: 10.1155/2010/186813] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 02/18/2010] [Indexed: 01/28/2023]
Abstract
Apolipoprotein E (apoE) is a 34.2 kDa glycoprotein characterized by its wide tissue distribution and multiple functions. The nonlipid-related properties of apoE include modulating inflammation and oxidation, suppressing T cell proliferation, regulating macrophage functions, and facilitating lipid antigen presentation by CD1 molecules to natural killer T (NKT) cells, and so forth. Increasing studies have revealed that APOE ε allele might be associated with multiple sclerosis (MS), although evidence is still not sufficient enough. In this review, we summarized the current progress of the immunomodulatory functions of apoE, with special focus on the association of APOE ε allele with the clinical features of MS and of its animal model experimental autoimmune encephalomyelitis (EAE).
Collapse
|
58
|
Minami SS, Cordova A, Cirrito JR, Tesoriero JA, Babus LW, Davis GC, Dakshanamurthy S, Turner RS, Pak DTS, Rebeck GW, Paige M, Hoe HS. ApoE mimetic peptide decreases Abeta production in vitro and in vivo. Mol Neurodegener 2010; 5:16. [PMID: 20406479 PMCID: PMC2890633 DOI: 10.1186/1750-1326-5-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/20/2010] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Apolipoprotein E (apoE) is postulated to affect brain Abeta levels through multiple mechanisms--by altering amyloid precursor protein (APP) processing, Abeta degradation, and Abeta clearance. We previously showed that an apoE-derived peptide containing a double repeat of the receptor-binding region was similarly effective in increasing APP processing in vivo. Here, we further examined whether peptides containing tandem repeats of the apoE receptor-binding region (amino acids 141-149) affected APP trafficking, APP processing, and Abeta production. RESULTS We found that peptides containing a double or triple tandem repeat of the apoE receptor-binding region, LRKLRKRLL, increased cell surface APP and decreased Abeta levels in PS1-overexpressing PS70 cells and in primary neurons. This effect was potentiated by a sequential increase in the number of apoE receptor-binding domain repeats (trimer > dimer > monomer). We previously showed that the apoE dimer increased APP CTF in vivo; to determine whether the dimer also affected secreted APP or Abeta levels, we performed a single hippocampal injection of the apoE dimer in wild-type mice and analyzed its effect on APP processing. We found increased sAPPalpha and decreased Abeta levels at 24 hrs after treatment, suggesting that the apoE dimer may increase alpha-secretase cleavage. CONCLUSIONS These data suggest that small peptides consisting of tandem repeats of the apoE receptor-binding region are sufficient to alter APP trafficking and processing. The potency of these peptides increased with increasing repeats of the receptor binding domain of apoE. In addition, in vivo administration of the apoE peptide (dimer) increased sAPPalpha and decreased Abeta levels in wild-type mice. Overall, these findings contribute to our understanding of the effects of apoE on APP processing and Abeta production both in vitro and in vivo.
Collapse
Affiliation(s)
- S Sakura Minami
- Department of Neuroscience, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Antoinette Cordova
- Drug Discovery and Drug Therapeutics, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - John R Cirrito
- Department of Neurology, Washington University, St. Louis, MO 63110, USA
| | - Joseph A Tesoriero
- Department of Neurology, Georgetown University, 4000 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Lenard W Babus
- Department of Neurology, Georgetown University, 4000 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Gary C Davis
- Drug Discovery and Drug Therapeutics, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Sivanesan Dakshanamurthy
- Drug Discovery and Drug Therapeutics, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - R Scott Turner
- Department of Neurology, Georgetown University, 4000 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Daniel TS Pak
- Department of Pharmacology, Georgetown University Medical Center, 3900 Reservoir Rd. NW, Washington, DC 20057, USA
| | - G William Rebeck
- Department of Neuroscience, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Mikell Paige
- Drug Discovery and Drug Therapeutics, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
| | - Hyang-Sook Hoe
- Department of Neuroscience, Georgetown University, 3970 Reservoir Rd. NW, Washington, DC 20057, USA
- Department of Neurology, Georgetown University, 4000 Reservoir Rd. NW, Washington, DC 20057, USA
| |
Collapse
|
59
|
Yancey PG, Blakemore J, Ding L, Fan D, Overton CD, Zhang Y, Linton MF, Fazio S. Macrophage LRP-1 controls plaque cellularity by regulating efferocytosis and Akt activation. Arterioscler Thromb Vasc Biol 2010; 30:787-95. [PMID: 20150557 PMCID: PMC2845445 DOI: 10.1161/atvbaha.109.202051] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The balance between apoptosis susceptibility and efferocytosis of macrophages is central to plaque remodeling and inflammation. LRP-1 and its ligand, apolipoprotein E, have been implicated in efferocytosis and apoptosis in some cell types. We investigated the involvement of the macrophage LRP-1/apolipoprotein E axis in controlling plaque apoptosis and efferocytosis. Method and Results- LRP-1(-/-) macrophages displayed nearly 2-fold more TUNEL positivity compared to wild-type cells in the presence of DMEM alone or with either lipopolysaccharide or oxidized low-density lipoprotein. The survival kinase, phosphorylated Akt, was barely detectable in LRP-1(-/-) cells, causing decreased phosphorylated Bad and increased cleaved caspase-3. Regardless of the apoptotic stimulation and degree of cell death, LRP-1(-/-) macrophages displayed enhanced inflammation with increased IL-1 beta, IL-6, and tumor necrosis factor-alpha expression. Efferocytosis of apoptotic macrophages was reduced by 60% in LRP-1(-/-) vs wild-type macrophages despite increased apolipoprotein E expression by both LRP-1(-/-) phagocytes and wild-type apoptotic cells. Compared to wild-type macrophage lesions, LRP-1(-/-) lesions had 5.7-fold more necrotic core with more dead cells not associated with macrophages. CONCLUSIONS Macrophage LRP-1 deficiency increases cell death and inflammation by impairing phosphorylated Akt activation and efferocytosis. Increased apolipoprotein E expression in LRP-1(-/-) macrophages suggests that the LRP-1/apolipoprotein E axis regulates the balance between apoptosis and efferocytosis, thereby preventing necrotic core formation.
Collapse
Affiliation(s)
- Patricia G. Yancey
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - John Blakemore
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - Lei Ding
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - Daping Fan
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - Cheryl D. Overton
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
- Department of Pathology, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - Youmin Zhang
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - MacRae F. Linton
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville TN 37232-6300
| | - Sergio Fazio
- Atherosclerosis Research Unit, Division of Cardiology, Department of Medicine, Vanderbilt University Medical Center, Nashville TN 37232-6300
- Department of Pathology, Vanderbilt University Medical Center, Nashville TN 37232-6300
| |
Collapse
|
60
|
Sarantseva S, Timoshenko S, Bolshakova O, Karaseva E, Rodin D, Schwarzman AL, Vitek MP. Apolipoprotein E-mimetics inhibit neurodegeneration and restore cognitive functions in a transgenic Drosophila model of Alzheimer's disease. PLoS One 2009; 4:e8191. [PMID: 19997607 PMCID: PMC2782140 DOI: 10.1371/journal.pone.0008191] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Accepted: 10/28/2009] [Indexed: 11/21/2022] Open
Abstract
Background Mutations of the amyloid precursor protein gene (APP) are found in familial forms of Alzheimer's disease (AD) and some lead to the elevated production of amyloid-β-protein (Aβ). While Aβ has been implicated in the causation of AD, the exact role played by Aβ and its APP precursor are still unclear. Principal Findings In our study, Drosophila melanogaster transgenics were established as a model to analyze AD-like pathology caused by APP overexpression. We demonstrated that age related changes in the levels and pattern of synaptic proteins accompanied progressive neurodegeneration and impairment of cognitive functions in APP transgenic flies, but that these changes may be independent from the generation of Aβ. Using novel peptide mimetics of Apolipoprotein-E, COG112 or COG133 proved to be neuroprotective and significantly improved the learning and memory of APP transgenic flies. Conclusions The development of neurodegeneration and cognitive deficits was corrected by injections of COG112 or COG133, novel mimetics of apolipoprotein-E (apoE) with neuroprotective activities.
Collapse
Affiliation(s)
- Svetlana Sarantseva
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
| | - Svetlana Timoshenko
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
| | - Olga Bolshakova
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
| | - Eugenia Karaseva
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
| | - Dmitry Rodin
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
| | - Alexander L. Schwarzman
- Petersburg Institute of Nuclear Physics, Russian Academy of Sciences, Gatchina, Russia
- Institute for Experimental Medicine, Russian Academy of Medical Sciences, St. Petersburg, Russia
| | - Michael P. Vitek
- Division of Neurology, Duke University Medical Center, Durham, North Carolina, United States of America
- Cognosci, Inc., Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
61
|
Crutcher KA, Lilley HN, Anthony SR, Zhou W, Narayanaswami V. Full-length apolipoprotein E protects against the neurotoxicity of an apoE-related peptide. Brain Res 2009; 1306:106-15. [PMID: 19836363 DOI: 10.1016/j.brainres.2009.10.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 10/06/2009] [Accepted: 10/08/2009] [Indexed: 11/26/2022]
Abstract
Apolipoprotein E was found to protect against the neurotoxic effects of a dimeric peptide derived from the receptor-binding region of this protein (residues 141-149). Both apoE3 and apoE4 conferred protection but the major N-terminal fragment of each isoform did not. Nor was significant protection provided by bovine serum albumin or apoA-I. Full-length apoE3 and apoE4 also inhibited the uptake of a fluorescent-labeled derivative of the peptide, suggesting that the mechanism of inhibition might involve competition for cell surface receptors/proteoglycans that mediate endocytosis and/or signaling pathways. These results might bear on the question of the role of apoE in neuronal degeneration, such as occurs in Alzheimer's disease where apoE4 confers a significantly greater risk of pathology.
Collapse
Affiliation(s)
- K A Crutcher
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | | | | | | | | |
Collapse
|
62
|
Fissolo N, Haag S, de Graaf KL, Drews O, Stevanovic S, Rammensee HG, Weissert R. Naturally presented peptides on major histocompatibility complex I and II molecules eluted from central nervous system of multiple sclerosis patients. Mol Cell Proteomics 2009; 8:2090-101. [PMID: 19531498 DOI: 10.1074/mcp.m900001-mcp200] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Tandem mass spectrometry was used to identify naturally processed peptides bound to major histocompatibility complex (MHC) I and MHC II molecules in central nervous system (CNS) of eight patients with multiple sclerosis (MS). MHC molecules were purified from autopsy CNS material by immunoaffinity chromatography with monoclonal antibody directed against HLA-A, -B, -C, and -DR. Subsequently peptides were separated by reversed-phase HPLC and analyzed by mass spectrometry. Database searches revealed 118 amino acid sequences from self-proteins eluted from MHC I molecules and 191 from MHC II molecules, corresponding to 174 identified source proteins. These sequences define previously known and potentially novel autoantigens in MS possibly involved in disease induction and antigen spreading. Taken together, we have initiated the characterization of the CNS-expressed MHC ligandome in CNS diseases and were able to demonstrate the presentation of naturally processed myelin basic protein peptides in the brain of MS patients.
Collapse
Affiliation(s)
- Nicolas Fissolo
- Hertie Institute for Clinical Brain Research, Experimental Neuroimmunology, University of Tuebingen, 72076 Tuebingen, Germany
| | | | | | | | | | | | | |
Collapse
|
63
|
Elliott DA, Tsoi K, Holinkova S, Chan SL, Kim WS, Halliday GM, Rye KA, Garner B. Isoform-specific proteolysis of apolipoprotein-E in the brain. Neurobiol Aging 2009; 32:257-71. [PMID: 19278755 DOI: 10.1016/j.neurobiolaging.2009.02.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2008] [Revised: 02/01/2009] [Accepted: 02/03/2009] [Indexed: 11/19/2022]
Abstract
Apolipoprotein-E (apoE) plays important roles in neurobiology and the apoE4 isoform increases risk for Alzheimer's disease (AD). ApoE peptides are biologically active and may be produced in the brain. It is unclear if apoE proteolysis is dependent on isoform or AD status and this was addressed here. Hippocampus, frontal cortex, occipital lobe and cerebellum samples were homogenized into fractions that were soluble in Tris-buffered saline (TBS), Triton X-100 or guanidine hydrochloride and analysed for apoE fragmentation by Western blotting. Approximately 20% of apoE3 was detected as fragments and this was predominantly as a 25 kDa peptide in TBS-soluble fractions. The concentration of TBS-soluble apoE fragments was two- to three-fold higher in apoE3 compared to apoE4 subjects. This difference was observed in all areas of the brain examined and was not related to AD status. Cathepsin-D treatment generated apoE fragments that were very similar to those detected in brain, however, no apoE isoform-specific differences in susceptibility to cathepsin-D proteolysis were detected. This indicates that proteolytic processing of apoE to form soluble fragments in the human brain is dependent on apoE isoform but not AD status.
Collapse
Affiliation(s)
- David A Elliott
- Prince of Wales Medical Research Institute, Randwick, NSW 2031, Australia
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Hoane MR, Kaufman N, Vitek MP, McKenna SE. COG1410 improves cognitive performance and reduces cortical neuronal loss in the traumatically injured brain. J Neurotrauma 2009; 26:121-9. [PMID: 19119914 PMCID: PMC2749004 DOI: 10.1089/neu.2008.0565] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
We have previously shown that a single dose of COG1410, a small molecule ApoE-mimetic peptide derived from the apolipoprotein E (ApoE) receptor binding region, improves sensorimotor and motor outcome following cortical contusion injury (CCI). The present study evaluated a regimen of COG1410 following frontal CCI in order to examine its preclinical efficacy on cognitive recovery. Animals were prepared with a bilateral CCI of the frontal cortex. A regimen of COG1410 (0.8mg/kg intravenously [IV]) was administered twice, at 30min and again at 24h post-CCI. Starting on day 11, the animals were tested for their acquisition of a reference memory task in the Morris water maze (MWM), followed by a working memory task in the MWM on day 15. Following CCI, the animals were also tested on the bilateral tactile adhesive removal test to measure sensorimotor dysfunction. On all of the behavioral tests the COG1410 group was no different from the uninjured sham group. Administration of the regimen of COG1410 significantly improved recovery on the reference and working memory tests, as well as on the sensorimotor test. Lesion analysis revealed that COG1410 significantly reduced the size of the injury cavity. Administration of COG1410 also reduced the number of degenerating neurons, as measured by Fluoro-Jade C staining, in the frontal cortex at 48h post-CCI. These results suggest that a regimen of COG1410 appeared to block the development of significant behavioral deficits and reduced tissue loss. These combined findings suggest that COG1410 appears to have strong preclinical efficacy when administered following traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Michael R Hoane
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology,Southern Illinois University, Carbondale, IL 62901, USA.
| | | | | | | |
Collapse
|
65
|
Gay EA, Klein RC, Melton MA, Blackshear PJ, Yakel JL. Inhibition of native and recombinant nicotinic acetylcholine receptors by the myristoylated alanine-rich C kinase substrate peptide. J Pharmacol Exp Ther 2008; 327:884-90. [PMID: 18812491 PMCID: PMC2709273 DOI: 10.1124/jpet.108.144758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A variety of peptide ligands are known to inhibit the function of neuronal nicotinic acetylcholine receptors (nAChRs), including small toxins and brain-derived peptides such as beta-amyloid(1-42) and synthetic apolipoproteinE peptides. The myristoylated alanine-rich C kinase substrate (MARCKS) protein is a major substrate of protein kinase C and is highly expressed in the developing and adult brain. The ability of a 25-amino acid synthetic MARCKS peptide, derived from the effector domain (ED), to modulate nAChR activity was tested. To determine the effects of the MARCKS ED peptide on nAChR function, receptors were expressed in Xenopus laevis oocytes, and two-electrode voltage-clamp experiments were performed. The MARCKS ED peptide completely inhibited acetylcholine (ACh)-evoked responses from alpha7 nAChRs in a dose-dependent manner, yielding an IC(50) value of 16 nM. Inhibition of ACh-induced responses was both activity- and voltage-independent. The MARCKS ED peptide was unable to block alpha-bungarotoxin binding. A MARCKS ED peptide in which four serine residues were replaced with aspartate residues was unable to inhibit alpha7 nAChR-mediated currents. The MARCKS ED peptide inhibited ACh-induced alpha4beta2 and alpha2beta2 responses, although with decreased potency. The effects of the MARCKS ED peptide on native nAChRs were tested using acutely isolated rat hippocampal slices. In hippocampal interneurons, the MARCKS ED peptide was able to block native alpha7 nAChRs in a dose-dependent manner. The MARCKS ED peptide represents a novel antagonist of neuronal nAChRs that has considerable utility as a research tool.
Collapse
Affiliation(s)
- Elaine A Gay
- Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709, USA
| | | | | | | | | |
Collapse
|
66
|
Toma HS, Murina AT, Areaux RG, Neumann DM, Bhattacharjee PS, Foster TP, Kaufman HE, Hill JM. Ocular HSV-1 latency, reactivation and recurrent disease. Semin Ophthalmol 2008; 23:249-73. [PMID: 18584563 DOI: 10.1080/08820530802111085] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ocular infection with HSV-1 continues to be a serious clinical problem despite the availability of effective antivirals. Primary infection with HSV-1 can involve ocular and adenaxial sites and can manifest as blepharitis, conjunctivitis, or corneal epithelial keratitis. After initial ocular infection, HSV-1 can establish latent infection in the trigeminal ganglia for the lifetime of the host. During latency, the viral genome is retained in the neuron without producing viral proteins. However, abundant transcription occurs at the region encoding the latency-associated transcript, which may play significant roles in the maintenance of latency as well as neuronal reactivation. Many host and viral factors are involved in HSV-1 reactivation from latency. HSV-1 DNA is shed into tears and saliva of most adults, but in most cases this does not result in lesions. Recurrent disease occurs as HSV-1 is carried by anterograde transport to the original site of infection, or any other site innervated by the latently infected ganglia, and can reinfect the ocular tissues. Recurrent corneal disease can lead to corneal scarring, thinning, stromal opacity and neovascularization and, eventually, blindness. In spite of intensive antiviral and anti-inflammatory therapy, a significant percentage of patients do not respond to chemotherapy for herpetic necrotizing stromal keratitis. Therefore, the development of therapies that would reduce asymptomatic viral shedding and lower the risks of recurrent disease and transmission of the virus is key to decreasing the morbidity of ocular herpetic disease. This review will highlight basic HSV-1 virology, and will compare the animal models of latency, reactivation, and recurrent ocular disease to the current clinical data.
Collapse
Affiliation(s)
- Hassanain S Toma
- Louisiana State University Health Sciences Center, New Orleans, LA 70112-2234, USA
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Cedazo-Mínguez A. Apolipoprotein E and Alzheimer's disease: molecular mechanisms and therapeutic opportunities. J Cell Mol Med 2008; 11:1227-38. [PMID: 18205697 PMCID: PMC4401287 DOI: 10.1111/j.1582-4934.2007.00130.x] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Multiple genetic and environmental factors are likely to contribute to the development of Alzheimer's disease (AD). The most important known risk factor for AD is presence of the E4 isoform of apolipoprotein E (apoE). Epidemiological studies demonstrated that apoE4 carriers have a higher risk and develop the disease and an early onset. Moreover, apoE4 is the only molecule that has been associated with all the biochemical disturbances characteristic of the disease: amyloid-beta (Abeta) deposition, tangle formation, oxidative stress, lipid homeostasis deregulation, synaptic plasticity loss and cholinergic dysfunction. This large body of evidence suggest that apoE is a key player in the pathogenesis of AD. This short review examines the current facts and hypotheses of the association between apoE4 and AD, as well as the therapeutic possibilities that apoE might offer for the treatment of this disease.
Collapse
Affiliation(s)
- Angel Cedazo-Mínguez
- Karolinska Institutet, Department of Neurobiology, Care Sciences and Society, KI-Alzheimer's Disease Research Center, NOVUM, Stockholm, Sweden.
| |
Collapse
|
68
|
Singh K, Chaturvedi R, Asim M, Barry DP, Lewis ND, Vitek MP, Wilson KT. The apolipoprotein E-mimetic peptide COG112 inhibits the inflammatory response to Citrobacter rodentium in colonic epithelial cells by preventing NF-kappaB activation. J Biol Chem 2008; 283:16752-61. [PMID: 18417477 DOI: 10.1074/jbc.m710530200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Inflammatory bowel disease arises from the interplay between luminal bacteria and the colonic mucosa. Targeted inhibition of pro-inflammatory pathways without global immunosuppression is highly desirable. Apolipoprotein (apo) E has immunomodulatory effects and synthetically derived apoE-mimetic peptides are beneficial in models of sepsis and neuroinflammation. Citrobacter rodentium is the rodent equivalent of enteropathogenic Escherichia coli, and it causes colitis in mice by colonizing the surface of colonic epithelial cells and inducing signaling events. We have reported that mice deficient in inducible nitric-oxide (NO) synthase (iNOS) have attenuated C. rodentium-induced colitis. We used young adult mouse colon (YAMC) cells that mimic primary colonic epithelial cells to study effects of an antennapedia-linked apoE-mimetic peptide, COG112, on C. rodentium-activated cells. COG112 significantly attenuated induction of NO production, and iNOS mRNA and protein expression, in a concentration-dependent manner. COG112 inhibited the C. rodentium-stimulated induction of iNOS and the CXC chemokines KC and MIP-2 to the same degree as the NF-kappaB inhibitors MG132 or BAY 11-7082, and there was no additive effect when COG112 and these inhibitors were combined. COG112 significantly reduced nuclear translocation of NF-kappaB, when assessed by electromobility shift assay, immunoblotting, and immunofluorescence for p65. This correlated with inhibition of both C. rodentium-stimulated IkappaB-alpha phosphorylation and degradation, and IkappaB kinase activity, which occurred by inhibition of IkappaB kinase complex formation rather than by a direct effect on the enzyme itself. These studies indicate that apoE-mimetic peptides may have novel therapeutic potential by inhibiting NF-kappaB-driven proinflammatory epithelial responses to pathogenic colonic bacteria.
Collapse
Affiliation(s)
- Kshipra Singh
- Department of Medicine, Division of Gastroenterology, Vandergilt University School of Medicine, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | |
Collapse
|
69
|
Laskowitz DT, Vitek MP. Apolipoprotein E and neurological disease: therapeutic potential and pharmacogenomic interactions. Pharmacogenomics 2008; 8:959-69. [PMID: 17716229 DOI: 10.2217/14622416.8.8.959] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The apolipoprotein E (apoE) polymorphism is emerging as a uniquely important genetic modifier that affects functional outcome from both acute and chronic neurological injuries. Recent attention has focused on common denominator mechanisms by which apoE might affect brain injury and/or brain repair responses in clinically diverse diseases. Although endogenous apoE likely serves several adaptive functions in the injured CNS, there is growing evidence that its effect on modifying brain inflammatory responses and providing protection from excitotoxic injury may be central to its protective properties. A more complete understanding of the role that apoE plays in the injured brain has led to novel therapeutic strategies for both acute and chronic neurological disease.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Duke University Medical Center, Department of Medicine (Neurology), Box 2900, Durham, NC 27710, USA.
| | | |
Collapse
|
70
|
Laskowitz DT, McKenna SE, Song P, Wang H, Durham L, Yeung N, Christensen D, Vitek MP. COG1410, a novel apolipoprotein E-based peptide, improves functional recovery in a murine model of traumatic brain injury. J Neurotrauma 2007; 24:1093-107. [PMID: 17610350 DOI: 10.1089/neu.2006.0192] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is a silent epidemic affecting approximately 1.4 million Americans annually, at an estimated annual cost of $60 billion in the United States alone. Despite an increased understanding of the pathophysiology of closed head injury, there remains no pharmacological intervention proven to improve functional outcomes in this setting. Currently, the existing standard of care for TBI consists primarily of supportive measures. Apolipoprotein E (apoE) is the primary apolipoprotein synthesized in the brain in response to injury, where it modulates several components of the neuroinflammatory cascade associated with TBI. We have previously demonstrated that COG133, an apoE mimetic peptide, improved functional outcomes and attenuated neuronal death when administered as a single intravenous injection at 30 min post-TBI in mice. Using the principles of rational drug design, we developed a more potent analog, COG1410, which expands the therapeutic window for the treatment of TBI by a factor of four, from 30 min to 2 h. Mice that received a single intravenous injection of COG1410 at 120 min post-TBI exhibited significant improvement on a short term test of vestibulomotor function and on a long term test of spatial learning and memory. This was associated with a significant attenuation of microglial activation and neuronal death in the hippocampus, the neuroanatomical substrate for learning and memory. Rationally derived apoE mimetic peptides have been demonstrated to exert neuroprotective and anti-inflammatory effects in vitro and in clinically relevant models of brain injury. This represents a novel therapeutic strategy in the treatment of TBI.
Collapse
Affiliation(s)
- Daniel T Laskowitz
- Division of Neurology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | |
Collapse
|
71
|
Hoane MR, Pierce JL, Holland MA, Birky ND, Dang T, Vitek MP, McKenna SE. The novel apolipoprotein E-based peptide COG1410 improves sensorimotor performance and reduces injury magnitude following cortical contusion injury. J Neurotrauma 2007; 24:1108-18. [PMID: 17610351 DOI: 10.1089/neu.2006.0254] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
It has previously been shown that small peptide molecules derived from the apolipoprotein E (ApoE) receptor binding region are anti-inflammatory in nature and can improve outcome following head injury. The present study evaluated the preclinical efficacy of COG1410, a small molecule ApoE-mimetic peptide (1410 daltons), following cortical contusion injury (CCI). Animals were prepared with a unilateral CCI of the sensorimotor cortex (SMC) or sham procedure. Thirty mins post-CCI the animals received i.v. infusions of 0.8 mg/kg COG1410, 0.4 mg/kg COG1410, or vehicle. Starting on day 2, the animals were tested on a battery of behavioral measures to assess sensorimotor (vibrissae-forelimb placing and forelimb use-asymmetry), and motor (tapered balance beam) performance. Administration of the 0.8 mg/kg dose of COG1410 significantly improved recovery on the vibrissae-forelimb and limb asymmetry tests. However, no facilitation was observed on the tapered beam. The low dose (0.4 mg/kg) of COG1410 did not show any significant differences compared to vehicle. Lesion analysis revealed that the 0.8 mg/kg dose of COG1410 significantly reduced the size of the injury cavity compared to the 0.4 mg/kg dose and vehicle. The 0.8 mg/kg dose also reduced the number of glial fibrillary acid protein (GFAP+) reactive cells in the injured cortex. These results suggest that a single dose of COG1410 facilitates behavioral recovery and provides neuroprotection in a dose and task-dependent manner. Thus, the continued clinical development of ApoE based therapeutics is warranted and could represent a novel strategy for the treatment of traumatic brain injuries.
Collapse
Affiliation(s)
- Michael R Hoane
- Restorative Neuroscience Laboratory, Center for Integrative Research in Cognitive and Neural Sciences, Department of Psychology, Southern Illinois University, Carbondale, Illinois 62901, USA.
| | | | | | | | | | | | | |
Collapse
|
72
|
Gay EA, Bienstock RJ, Lamb PW, Yakel JL. Structural determinates for apolipoprotein E-derived peptide interaction with the alpha7 nicotinic acetylcholine receptor. Mol Pharmacol 2007; 72:838-49. [PMID: 17609418 PMCID: PMC2742887 DOI: 10.1124/mol.107.035527] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neuronal nicotinic acetylcholine receptor (nAChR) signaling has been implicated in a variety of normal central nervous system (CNS) functions as well as an array of neuropathologies. Previous studies have demonstrated both neurotoxic and neuroprotective actions of peptides derived from apolipoprotein E (apoE). It has been discovered that apoE-derived peptides inhibit native and recombinant alpha7-containing nAChRs, indicating a direct interaction between apoE peptides and nAChRs. To probe the structure/function interaction between alpha7 nAChRs and the apoE peptide apoE(141-148), experiments were conducted in Xenopus laevis oocytes expressing wild-type and mutated nAChRs. Mutation of Trp55 to alanine blocks apoE peptide-induced inhibition of acetylcholine (ACh)-mediated alpha7 nAChR responses. Additional mutations at Trp55 suggest that hydrophobic interactions between the receptor and apoE(141-148) are essential for inhibition of alpha7 nAChR function. A mutated apoE peptide also demonstrated decreased inhibition at alpha7-W55A nAChRs as well as activity-dependent inhibition of both wild-type alpha7 nAChRs and alpha7-W55A receptors. Finally, a three-dimensional model of the alpha7 nAChR was developed based on the recently refined Torpedo marmorata nACh receptor. A structural model is proposed for the binding of apoE(141-148) to the alpha7 nAChR where the peptide binds at the interface between two subunits, near the ACh binding site. Similar to the functional data, the computational docking suggests the importance of hydrophobic interactions between the alpha7 nAChR and the apoE peptide for inhibition of receptor function. The current data suggest a mode for apoE peptide binding that directly blocks alpha7 nAChR activity and consequently may disrupt nAChR signaling.
Collapse
Affiliation(s)
- Elaine A Gay
- NIEHS, F2-08, P.O. Box 12233, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | | | | | | |
Collapse
|
73
|
Liu T, Donahue KC, Hu J, Kurnellas MP, Grant JE, Li H, Elkabes S. Identification of differentially expressed proteins in experimental autoimmune encephalomyelitis (EAE) by proteomic analysis of the spinal cord. J Proteome Res 2007; 6:2565-75. [PMID: 17571869 PMCID: PMC2430926 DOI: 10.1021/pr070012k] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The present study used isobaric tags for relative and absolute quantitation (iTRAQ) to identify novel targets in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. The expression of 41 proteins was significantly altered in the inflamed spinal cord. Twenty of these are implicated in EAE for the first time and many have previously been shown to play a role in antigen processing, inflammation, neuroprotection, or neurodegeneration.
Collapse
Affiliation(s)
- Tong Liu
- Center for Advanced Proteomics Research and Department of Biochemistry and Molecular Biology, UMDNJ-New Jersey Medical School Cancer Center, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | |
Collapse
|
74
|
Maezawa I, Zaja-Milatovic S, Milatovic D, Stephen C, Sokal I, Maeda N, Montine TJ, Montine KS. Apolipoprotein E isoform-dependent dendritic recovery of hippocampal neurons following activation of innate immunity. J Neuroinflammation 2006; 3:21. [PMID: 16934151 PMCID: PMC1584222 DOI: 10.1186/1742-2094-3-21] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2006] [Accepted: 08/25/2006] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Innate immune activation, including a role for cluster of differentiation 14/toll-like receptor 4 co-receptors (CD14/TLR-4) co-receptors, has been implicated in paracrine damage to neurons in several neurodegenerative diseases that also display stratification of risk or clinical outcome with the common alleles of the apolipoprotein E gene (APOE): APOE2, APOE3, and APOE4. Previously, we have shown that specific stimulation of CD14/TLR-4 with lipopolysaccharide (LPS) leads to greatest innate immune response by primary microglial cultures from targeted replacement (TR) APOE4 mice and greatest p38MAPK-dependent paracrine damage to neurons in mixed primary cultures and hippocampal slice cultures derived from TR APOE4 mice. In contrast, TR APOE2 astrocytes had the highest NF-kappaB activity and no neurotoxicity. Here we tested the hypothesis that direct activation of CD14/TLR-4 in vivo would yield different amounts of paracrine damage to hippocampal sector CA1 pyramidal neurons in TR APOE mice. METHODS We measured in vivo changes in dendrite length in hippocampal CA1 neurons using Golgi staining and determined hippocampal apoE levels by Western blot. Neurite outgrowth of cultured primary neurons in response to astrocyte conditioned medium was assessed by measuring neuron length and branch number. RESULTS Our results showed that TR APOE4 mice had slightly but significantly shorter dendrites at 6 weeks of age. Following exposure to intracerebroventricular LPS, there was comparable loss of dendrite length at 24 hr among the three TR APOE mice. Recovery of dendrite length over the next 48 hr was greater in TR APOE2 than TR APOE3 mice, while TR APOE4 mice had failure of dendrite regeneration. Cell culture experiments indicated that the enhanced neurotrophic effect of TR APOE2 was LDL related protein-dependent. CONCLUSION The data indicate that the environment within TR APOE2 mouse hippocampus was most supportive of dendrite regeneration while that within TR APOE4 hippocampus failed to support dendrite regeneration in this model of reversible paracrine damage to neurons from innate immune activation, and suggest an explanation for the stratification of clinical outcome with APOE seen in several degenerative diseases or brain that are associated with activated innate immune response.
Collapse
Affiliation(s)
- Izumi Maezawa
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Dejan Milatovic
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Izabela Sokal
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Nobuyo Maeda
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Thomas J Montine
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Kathleen S Montine
- Department of Pathology, University of Washington, Seattle, WA, USA
- Department of Pathology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|