1
|
Kumar NN, Pizzo ME, Nehra G, Wilken-Resman B, Boroumand S, Thorne RG. Passive Immunotherapies for Central Nervous System Disorders: Current Delivery Challenges and New Approaches. Bioconjug Chem 2018; 29:3937-3966. [PMID: 30265523 PMCID: PMC7234797 DOI: 10.1021/acs.bioconjchem.8b00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Passive immunotherapy, i.e., the administration of exogenous antibodies that recognize a specific target antigen, has gained significant momentum as a potential treatment strategy for several central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and brain cancer, among others. Advances in antibody engineering to create therapeutic antibody fragments or antibody conjugates have introduced new strategies that may also be applied to treat CNS disorders. However, drug delivery to the CNS for antibodies and other macromolecules has thus far proven challenging, due in large part to the blood-brain barrier and blood-cerebrospinal fluid barriers that greatly restrict transport of peripherally administered molecules from the systemic circulation into the CNS. Here, we summarize the various passive immunotherapy approaches under study for the treatment of CNS disorders, with a primary focus on disease-specific and target site-specific challenges to drug delivery and new, cutting edge methods.
Collapse
Affiliation(s)
- Niyanta N. Kumar
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Michelle E. Pizzo
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Geetika Nehra
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Brynna Wilken-Resman
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Sam Boroumand
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Robert G. Thorne
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Neuroscience Training Program & Center for
Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705, United
States
- Cellular and Molecular Pathology Graduate Training Program,
University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
2
|
Cheng L, Zhang J, Li XY, Yuan L, Pan YF, Chen XR, Gao TM, Qiao JT, Qi JS. A novel antibody targeting sequence 31-35 in amyloid β protein attenuates Alzheimer's disease-related neuronal damage. Hippocampus 2016; 27:122-133. [PMID: 27784133 DOI: 10.1002/hipo.22676] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 11/10/2022]
Abstract
Amyloid β protein (Aβ) plays a critical role in pathogenesis of Alzheimer's disease (AD). Our previous studies indicated that the sequence 31-35 in Aβ molecule is an effective active center responsible for Aβ neurotoxicity in vivo and in vitro. In the present study, we prepared a novel antibody specifically targeting the sequence 31-35 of amyloid β protein, and investigated the neuroprotection of the anti-Aβ31-35 antibody against Aβ1-42 -induced impairments in neuronal viability, spatial memory, and hippocampal synaptic plasticity in rats. The results showed that the anti-Aβ31-35 antibody almost equally bound to both Aβ31-35 and Aβ1-42 , and pretreatment with the antibody dose-dependently prevented Aβ1-42 -induced cytotoxicity on cultured primary cortical neurons. In behavioral study, intracerebroventricular (i.c.v.) injection of anti-Aβ31-35 antibody efficiently attenuated Aβ1-42 -induced impairments in spatial learning and memory of rats. In vivo electrophysiological experiments further indicated that Aβ1-42 -induced suppression of hippocampal synaptic plasticity was effectively reversed by the antibody. These results demonstrated that the sequence 31-35 of Aβ may be a new therapeutic target, and the anti-Aβ31-35 antibody could be a novel immunotheraputic approach for the treatment of AD. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Li Cheng
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China.,The General Hospital of TISCO Affiliated to Shanxi Medical University, Taiyuan, 030003, China
| | - Jun Zhang
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xin-Yi Li
- Department of Neurology, Shanxi Dayi Hospital, Taiyuan, 030032, China
| | - Li Yuan
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Yan-Fang Pan
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao-Rong Chen
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Tian-Ming Gao
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China.,Department of Neurobiology, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Tian Qiao
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin-Shun Qi
- Department of Physiology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
3
|
Determining binding sites of polycyclic aromatic small molecule-based amyloid-beta peptide aggregation modulators using sequence-specific antibodies. Anal Biochem 2015; 470:61-70. [DOI: 10.1016/j.ab.2014.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/21/2014] [Accepted: 10/27/2014] [Indexed: 12/22/2022]
|
4
|
Guo W, Sha S, Jiang T, Xing X, Cao Y. A new DNA vaccine fused with the C3d-p28 induces a Th2 immune response against amyloid-beta. Neural Regen Res 2014; 8:2581-90. [PMID: 25206569 PMCID: PMC4145937 DOI: 10.3969/j.issn.1673-5374.2013.27.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 08/02/2013] [Indexed: 01/11/2023] Open
Abstract
To enhance anti-amyloid-beta (Aβ) antibody generation and induce a Th2 immune response, we constructed a new DNA vaccine p(Aβ3–10)10-C3d-p28.3 encoding ten repeats of Aβ3–10 and three copies of C3d-p28 as a molecular adjuvant. In this study, we administered this adjuvant cularly to female C57BL/6J mice at 8–10 weeks of age. Enzyme linked immunosorbent assay was used to detect the titer of serum anti-Aβ antibody, isotypes, and cytokines in splenic T cells. A 3-(4,5-cimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was used to detect the prolifera-tion rate of splenic T cells. Brain sections from a 12-month-old APP/PS1 transgenic mouse were used for detecting the binding capacities of anti-Aβ antibodies to Aβ plaques. The p(Aβ3–10)10-C3d-p28.3 vaccine induced high titers of anti-amyloid-β antibodies, which bound to Aβ plaques in APP/PS1 transgenic mouse brain tissue, demonstrating that the vaccine is effective against plaques in a mouse model of Alzheimer's disease. Moreover, the vaccine elicited a predo-minantly IgG1 humoral response and low levels of interferon-γ in ex vivo cultured splenocytes, dicating that the vaccine could shift the cellular immune response towards a Th2 phenotype. This indicated that the vaccine did not elicit a detrimental immune response and had a favorable safety profile. Our results indicate that the p(Aβ3–10)10-C3d-p28.3 vaccine is a promising immunothe-peutic option for Aβ vaccination in Alzheimer's disease.
Collapse
Affiliation(s)
- Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Sha Sha
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Tongzi Jiang
- Department of Neurology, First People's Hospital of Shenyang City, Shenyang 110041, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
5
|
Marciani DJ. New Th2 adjuvants for preventive and active immunotherapy of neurodegenerative proteinopathies. Drug Discov Today 2014; 19:912-20. [PMID: 24607730 DOI: 10.1016/j.drudis.2014.02.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 02/13/2014] [Accepted: 02/27/2014] [Indexed: 01/01/2023]
Abstract
Active immunotherapy of neurodegenerative proteinopathies, such as Alzheimer's disease, requires a Th2 antibody immune response with exclusion of damaging inflammatory Th1 immunity. Because these diseases affect the elderly whom, owing to immunosenescence, are frequently immune compromised, a successful therapy would require immune agonists capable of eliciting Th2 immunity solely while ameliorating the immune decline linked to aging; an objective hampered by the scarcity of Th2 immune agonists. The fact that some helminth-derived glycans and modified triterpene glycosides elicit Th2 immunity only, even when administered with antigens carrying T cell epitopes, presents new ways to improve the active immune therapy of proteinopathies. Of additional benefit is that these triterpene glycosides could amend some of the detrimental effects of the immunosenescence.
Collapse
Affiliation(s)
- Dante J Marciani
- Qantu Therapeutics, LLC, 612 E Main Street, Lewisville, TX 75057, USA.
| |
Collapse
|
6
|
Abstract
Alzheimer's disease (AD) is a common and devastating neurodegenerative disease. The incidence of AD is increasing in Western societies. The current treatment of AD is mostly symptomatic and ineffective in stopping or reversing the cognitive impairment. One of the exciting and effective new treatments developed in experimental AD is immunization against amyloid-beta peptide. This article provides an overview of immunization therapy in AD and examines the future prospects of this therapeutic modality.
Collapse
Affiliation(s)
- Felix Mor
- Tel-Aviv University, Weizmann Institute of Science Department of Immunology, Rehovot, Israel.
| | | |
Collapse
|
7
|
Guo W, Sha S, Xing X, Jiang T, Cao Y. Reduction of cerebral Aβ burden and improvement in cognitive function in Tg-APPswe/PSEN1dE9 mice following vaccination with a multivalent Aβ3-10 DNA vaccine. Neurosci Lett 2013; 549:109-15. [PMID: 23800542 DOI: 10.1016/j.neulet.2013.06.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/28/2013] [Accepted: 06/13/2013] [Indexed: 12/22/2022]
Abstract
To develop a safe and efficient Aβ vaccine for Alzheimer's disease, we constructed a plasmid DNA vaccine encoding ten repeats of Aβ3-10 and three copies of C3d-p28 as a molecular adjuvant and administered it intramuscularly in 12-month-old female Tg-APPswe/PSEN1dE9 mice. Therapeutic immunization with p(Aβ3-10)10-C3d-p28.3 stimulated a Th2 immune response that elicited therapeutic levels of anti-Aβ antibodies and improved cognitive function. In addition, the vaccine reduced the cerebral Aβ burden and astrocytosis without increasing the incidence of microhemorrhage. Our results indicate that the p(Aβ3-10)10-C3d-p28.3 vaccine is a promising immunotherapeutic option for Aβ vaccination in Alzheimer's disease.
Collapse
Affiliation(s)
- Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang 110001, Liao Ning Province, China
| | | | | | | | | |
Collapse
|
8
|
Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, Sha S, Liu L, Cao YP. Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Aβ3-10 reduces AD-like pathology and cognitive impairment in Tg-APPswe/PSEN1dE9 mice. J Neuroimmunol 2012; 249:16-26. [DOI: 10.1016/j.jneuroim.2012.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 04/09/2012] [Accepted: 04/15/2012] [Indexed: 10/28/2022]
|
9
|
Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Aβ3–10 induces Th2 immune response against amyloid-β in wild-type mouse. Neurosci Lett 2011; 505:128-33. [DOI: 10.1016/j.neulet.2011.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 09/22/2011] [Accepted: 10/02/2011] [Indexed: 10/16/2022]
|
10
|
Chauhan NB, Davis F, Xiao C. Wheat germ agglutinin enhanced cerebral uptake of anti-Aβ antibody after intranasal administration in 5XFAD mice. Vaccine 2011; 29:7631-7. [PMID: 21840361 DOI: 10.1016/j.vaccine.2011.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 07/22/2011] [Accepted: 08/01/2011] [Indexed: 12/28/2022]
Abstract
Alzheimer's disease (AD) is the 6th leading cause of death in United States afflicting >5 million Americans. This number is estimated to triple by the middle of the century if effective treatments are not discovered. Current therapy for AD is mainly symptomatic. Effective disease-modifying treatments are needed that would eliminate the cause rather than the symptoms of the disease. Polymerization of monomeric beta-amyloid peptide (Aβ) into dimers, soluble oligomers and insoluble fibrils is considered the prime causative factor in triggering AD pathogenesis. Based on these facts, removal/reduction of Aβ has gained importance as a primary therapeutic target in treating the cause of the disease. In that regard, passive immunotherapy with direct delivery of anti-Aβ antibodies to the brain has shown great promise, but awaits the challenge of overcoming greater influx of anti-Aβ antibody into the brain. This investigation was undertaken to maximize direct delivery of immunotherapeutics to the brain by using wheat germ agglutinin (WGA) as a novel axonal transporter-carrier to be conjugated with anti-Aβ antibody (6E10) raised against EFRHDS 3-8 amino acid (aa) epitopes of Aβ known to react with 1-16 aa residues of mono-/di-/oligomeric Aβ. This is the first report showing the use of WGA as an efficient axonal transporter carrier that not only enhanced the influx of anti-Aβ antibody directly into the brain but also resulted in greater reduction of cerebral Aβ compared to the unconjugated anti-Aβ antibody delivered intranasally in Alzheimer's 5XFAD model.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Neuroscience Research, Jesse Brown VA Medical Center, Chicago, IL 60612, USA.
| | | | | |
Collapse
|
11
|
Chauhan NB, Gatto R. Synergistic benefits of erythropoietin and simvastatin after traumatic brain injury. Brain Res 2010; 1360:177-92. [PMID: 20833152 DOI: 10.1016/j.brainres.2010.09.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 08/30/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
Abstract
Simvastatin and recombinant human erythropoietin (rhEpo) are implicated as potential therapeutic candidates for traumatic brain injury (TBI). Prominent effects of simvastatin include its anti-inflammatory, neurotrophic and neuroregenerative actions studied in various models of neuronal injury. On the other hand, rhEpo has been shown to promote cell survival mechanisms by producing anti-apoptotic and cell proliferative actions. Beneficial effects of rhEpo and statin monotherapies have been well studied. However, there are no reports showing combined use of rhEpo and statins after TBI. This investigation examined if combined efficacy of cell proliferative ability of rhEpo along with the neuroregenerative ability of simvastatin will render maximum recovery in a controlled cortical impact (CCI) mouse model of TBI. Results showed that compared to baseline TBI, rhEpo was more effective than simvastatin in promoting cell proliferation while simvastatin was more effective than rhEpo in restoring axonal damage following TBI. Combined treatment with simvastatin and rhEpo maximally restored axonal integrity while simultaneously inducing greater proliferation of newly formed cells resulting in better functional recovery after TBI than either alone. This is the first study showing the efficacy of erythropoietin-simvastatin combinational therapeutic approach in achieving greater structural and cognitive recovery after TBI.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Jesse Brown VA Medical Center, University of Illinois, Chicago, USA.
| | | |
Collapse
|
12
|
Huang X, Wang J, Cui L, Zou X, Zhang Y. Recombinant GST-I-Aβ28-induced efficient serum antibody against Aβ42. J Neurosci Methods 2010; 186:52-9. [DOI: 10.1016/j.jneumeth.2009.10.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Revised: 08/31/2009] [Accepted: 10/30/2009] [Indexed: 10/20/2022]
|
13
|
Lord A, Gumucio A, Englund H, Sehlin D, Sundquist VS, Söderberg L, Möller C, Gellerfors P, Lannfelt L, Pettersson FE, Nilsson LNG. An amyloid-beta protofibril-selective antibody prevents amyloid formation in a mouse model of Alzheimer's disease. Neurobiol Dis 2009; 36:425-34. [PMID: 19703562 DOI: 10.1016/j.nbd.2009.08.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2009] [Revised: 08/11/2009] [Accepted: 08/14/2009] [Indexed: 01/08/2023] Open
Abstract
Human genetics link Alzheimer's disease pathogenesis to excessive accumulation of amyloid-beta (Abeta) in brain, but the symptoms do not correlate with senile plaque burden. Since soluble Abeta aggregates can cause synaptic dysfunctions and memory deficits, these species could contribute to neuronal dysfunction and dementia. Here we explored selective targeting of large soluble aggregates, Abeta protofibrils, as a new immunotherapeutic strategy. The highly protofibril-selective monoclonal antibody mAb158 inhibited in vitro fibril formation and protected cells from Abeta protofibril-induced toxicity. When the mAb158 antibody was administered for 4 months to plaque-bearing transgenic mice with both the Arctic and Swedish mutations (tg-ArcSwe), Abeta protofibril levels were lowered while measures of insoluble Abeta were unaffected. In contrast, when treatment began before the appearance of senile plaques, amyloid deposition was prevented and Abeta protofibril levels diminished. Therapeutic intervention with mAb158 was however not proven functionally beneficial, since place learning depended neither on treatment nor transgenicity. Our findings suggest that Abeta protofibrils can be selectively cleared with immunotherapy in an animal model that display highly insoluble Abeta deposits, similar to those of Alzheimer's disease brain.
Collapse
Affiliation(s)
- Anna Lord
- Department of Public Health and Caring Sciences/Molecular Geriatrics, Uppsala University, Rudbeck Laboratory, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Rajkannan R, Arul V, Malar EJP, Jayakumar R. Preparation, physiochemical characterization, and oral immunogenicity of Abeta(1-12), Abeta(29-40), and Abeta(1-42) loaded PLG microparticles formulations. J Pharm Sci 2009; 98:2027-39. [PMID: 18980172 DOI: 10.1002/jps.21600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Alzheimer's disease (AD) is caused by the deposition of beta-amyloid (Abeta) protein in brain. The current AD immunotherapy aims to prevent Abeta plaque deposition and enhance its degradation in the brain. In this work, the peptides B-cell epitope Abeta(1-12), T-cell epitope Abeta(29-40) and full-length Abeta(1-42) were loaded separately to the poly (D,L-lactide co-glycolide) (PLG) microparticles by using W/O/W double emulsion solvent evaporation method with entrapment efficacy of 70.46%, 60.93%, and 65.98%, respectively. The prepared Abeta PLG microparticles were smooth, spherical, individual, and nonporous in nature with diameters ranging from 2 to 12 microm. The cumulative in vitro release profiles of Abeta(1-12), Abeta(29-40), and Abeta(1-42) from PLG microparticles sustained for long periods and progressively reached to 73.89%, 69.29%, and 70.08% by week 15. In vitro degradation studies showed that the PLG microparticles maintained the surface integrity up to week 8 and eroded completely by week 16. Oral immunization of Abeta peptides loaded microparticles in mice elicited stronger immune response by inducing anti-Abeta antibodies for prolonged time (24 weeks). The physicochemical characterization and immunogenic potency of Abeta peptides incorporated PLG microparticles suggest that the microparticles formulation of Abeta can be a potential oral AD vaccine.
Collapse
Affiliation(s)
- R Rajkannan
- Bioorganic and Neurochemistry Laboratory, Central Leather Research Institute, Adyar, Chennai 600 020, Tamil Nadu, India
| | | | | | | |
Collapse
|
15
|
Zameer A, Kasturirangan S, Emadi S, Nimmagadda SV, Sierks MR. Anti-oligomeric Abeta single-chain variable domain antibody blocks Abeta-induced toxicity against human neuroblastoma cells. J Mol Biol 2008; 384:917-28. [PMID: 18929576 DOI: 10.1016/j.jmb.2008.09.068] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 09/19/2008] [Accepted: 09/23/2008] [Indexed: 12/29/2022]
Abstract
The Amyloid-beta (Abeta) peptide is a major component of the amyloid plaques associated with Alzheimer's disease (AD). Recent studies suggest that the most toxic forms of Abeta are small, soluble oligomeric aggregates. Here, we report the isolation and characterization of a single-chain variable domain (scFv) antibody isolated against oligomeric Abeta using a protocol developed in our laboratory that combines phage display technology and atomic force microscopy (AFM). Starting with a randomized, single framework phage display library, after three rounds of selection against oligomeric Abeta, we identified an scFv that bound oligomeric Abeta specifically, but not monomeric or fibrillar forms. The anti-oligomeric scFv inhibits Abeta aggregation and toxicity, and reduces the toxicity of preformed oligomeric Abeta towards human neuroblastoma cells. When used to probe samples of human brain tissue, the scFv reacted with AD tissue but not a healthy control or Parkinson's disease brain samples. The anti-oligomeric Abeta scFv therefore has potential therapeutic and diagnostic applications in specifically targeting or identifying the toxic morphologies of Abeta in AD brains.
Collapse
Affiliation(s)
- Andleeb Zameer
- Department of Chemical Engineering, Arizona State University, Tempe, AZ 85287, USA
| | | | | | | | | |
Collapse
|
16
|
Petrushina I, Ghochikyan A, Mkrtichyan M, Mamikonyan G, Movsesyan N, Ajdari R, Vasilevko V, Karapetyan A, Lees A, Agadjanyan MG, Cribbs DH. Mannan-Abeta28 conjugate prevents Abeta-plaque deposition, but increases microhemorrhages in the brains of vaccinated Tg2576 (APPsw) mice. J Neuroinflammation 2008; 5:42. [PMID: 18823564 PMCID: PMC2567310 DOI: 10.1186/1742-2094-5-42] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2008] [Accepted: 09/29/2008] [Indexed: 12/30/2022] Open
Abstract
Background New pre-clinical trials in AD mouse models may help to develop novel immunogen-adjuvant configurations with the potential to avoid the adverse responses that occurred during the clinical trials with AN-1792 vaccine formulation. Recently, we have pursued an alternative immunization strategy that replaces QS21 the Th1 type adjuvant used in the AN-1792 clinical trial with a molecular adjuvant, mannan that can promote a Th2-polarized immune response through interactions with mannose-binding and CD35/CD21 receptors of the innate immune system. Previously we established that immunization of wild-type mice with mannan-Aβ28 conjugate promoted Th2-mediated humoral and cellular immune responses. In the current study, we tested the efficacy of this vaccine configuration in amyloid precursor protein (APP) transgenic mice (Tg2576). Methods Mannan was purified, activated and chemically conjugated to Aβ28 peptide. Humoral immune responses induced by the immunization of mice with mannan-Aβ28 conjugate were analyzed using a standard ELISA. Aβ42 and Aβ40 amyloid burden, cerebral amyloid angiopathy (CAA), astrocytosis, and microgliosis in the brain of immunized and control mice were detected using immunohistochemistry. Additionally, cored plaques and cerebral vascular microhemorrhages in the brains of vaccinated mice were detected by standard histochemistry. Results Immunizations with low doses of mannan-Aβ28 induced potent and long-lasting anti-Aβ humoral responses in Tg2576 mice. Even 11 months after the last injection, the immunized mice were still producing low levels of anti-Aβ antibodies, predominantly of the IgG1 isotype, indicative of a Th2 immune response. Vaccination with mannan-Aβ28 prevented Aβ plaque deposition, but unexpectedly increased the level of microhemorrhages in the brains of aged immunized mice compared to two groups of control animals of the same age either injected with molecular adjuvant fused with an irrelevant antigen, BSA (mannan-BSA) or non-immunized mice. Of note, mice immunized with mannan-Aβ28 showed a trend toward elevated levels of CAA in the neocortex and in the leptomeninges compared to that in mice of both control groups. Conclusion Mannan conjugated to Aβ28 provided sufficient adjuvant activity to induce potent anti-Aβ antibodies in APP transgenic mice, which have been shown to be hyporesponsive to immunization with Aβ self-antigen. However, in old Tg2576 mice there were increased levels of cerebral microhemorrhages in mannan-Aβ28 immunized mice. This effect was likely unrelated to the anti-mannan antibodies induced by the immunoconjugate, because control mice immunized with mannan-BSA also induced antibodies specific to mannan, but did not have increased levels of cerebral microhemorrhages compared with non-immunized mice. Whether these anti-mannan antibodies increased the permeability of the blood brain barrier thus allowing elevated levels of anti-Aβ antibodies entry into cerebral perivascular or brain parenchymal spaces and contributed to the increased incidence of microhemorrhages remains to be investigated in the future studies.
Collapse
Affiliation(s)
- Irina Petrushina
- The Institute for Brain Aging and Dementia, University of California Irvine, Irvine, CA 92697-4540, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Damante CA, Osz K, Nagy Z, Pappalardo G, Grasso G, Impellizzeri G, Rizzarelli E, Sóvágó I. The metal loading ability of beta-amyloid N-terminus: a combined potentiometric and spectroscopic study of copper(II) complexes with beta-amyloid(1-16), its short or mutated peptide fragments, and its polyethylene glycol (PEG)-ylated analogue. Inorg Chem 2008; 47:9669-83. [PMID: 18808108 DOI: 10.1021/ic8006052] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is becoming a rapidly growing health problem, as it is one of the main causes of dementia in the elderly. Interestingly, copper(II) (together with zinc and iron) ions are accumulated in amyloid deposits, suggesting that metal binding to Abeta could be involved in AD pathogenesis. In Abeta, the metal binding is believed to occur within the N-terminal region encompassing the amino acid residues 1-16. In this work, potentiometric, spectroscopic (UV-vis, circular dichroism, and electron paramagnetic resonance), and electrospray ionization mass spectrometry (ESI-MS) approaches were used to investigate the copper(II) coordination features of a new polyethylene glycol (PEG)-conjugated Abeta peptide fragment encompassing the 1-16 amino acid residues of the N-terminal region (Abeta(1-16)PEG). The high water solubility of the resulting metal complexes allowed us to obtain a complete complex speciation at different metal-to-ligand ratios ranging from 1:1 to 4:1. Potentiometric and ESI-MS data indicate that Abeta(1-16)PEG is able to bind up to four copper(II) ions. Furthermore, in order to establish the coordination environment at each metal binding site, a series of shorter peptide fragments of Abeta, namely, Abeta(1-4), Abeta(1-6), AcAbeta(1-6), and AcAbeta(8-16)Y10A, were synthesized, each encompassing a potential copper(II) binding site. The complexation properties of these shorter peptides were also comparatively investigated by using the same experimental approach.
Collapse
Affiliation(s)
- Chiara A Damante
- Department of Chemical Sciences, University of Catania, V.le A. Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Sun X, Becker M, Pankow K, Krause E, Ringling M, Beyermann M, Maul B, Walther T, Siems WE. Catabolic attacks of membrane-bound angiotensin-converting enzyme on the N-terminal part of species-specific amyloid-beta peptides. Eur J Pharmacol 2008; 588:18-25. [PMID: 18495113 DOI: 10.1016/j.ejphar.2008.03.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Revised: 03/14/2008] [Accepted: 03/31/2008] [Indexed: 10/22/2022]
Abstract
Catabolic processes play a crucial role in the steady state of the amyloid-beta peptide (Abeta). Neprilysin (NEP) and angiotensin-converting enzyme (ACE), two transmembranal enzymes with greatest importance in peptide pharmacology, are known to play a role in Abeta catabolism. This paper focuses on the N-terminal part of Abeta. This region contains the three amino acid residues that determine the differences between human (hAbeta) and murine Abeta (mAbeta). Moreover, the N-terminal part of Abeta contains the zinc-binding site of the molecule. Consequently, all hydrolytic attacks on this part of the Alzheimer peptide should be of exceptional interest. We investigated domain-selective forms of ACE in HPLC-monitored peptide degradation studies and used mass spectrometry for product analyses. We found that ACE-evoked a hydrolysis of the N-terminal part of m- and hAbeta. The hAbeta sequence hAbeta (4-15) was found to be a better substrate for ACE compared to the corresponding murine form. Moreover, we localized the corresponding cleavage sites in the N-terminal part of Abeta as well as in the full-length molecule and identified new sites of endopeptidolytic attack by ACE. Finally, we demonstrate that both catalytic domains of mACE have similar hydrolytic activity on the N-terminal part of Abeta. Our results show that ACE besides its typical function as a dipeptidyl-carboxypeptidase has also unequivocal endopeptidolytic activities.
Collapse
Affiliation(s)
- Xiaoou Sun
- Leibniz-Institut für Molekulare Pharmakologie (FMP), D-13125 Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Miles LA, Wun KS, Crespi GAN, Fodero-Tavoletti MT, Galatis D, Bagley CJ, Beyreuther K, Masters CL, Cappai R, McKinstry WJ, Barnham KJ, Parker MW. Amyloid-beta-anti-amyloid-beta complex structure reveals an extended conformation in the immunodominant B-cell epitope. J Mol Biol 2008; 377:181-92. [PMID: 18237744 DOI: 10.1016/j.jmb.2007.12.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 12/15/2007] [Accepted: 12/17/2007] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Amyloid-beta (A beta) peptide, generated by proteolytic cleavage of the amyloid precursor protein, is central to AD pathogenesis. Most pharmaceutical activity in AD research has focused on A beta, its generation and clearance from the brain. In particular, there is much interest in immunotherapy approaches with a number of anti-A beta antibodies in clinical trials. We have developed a monoclonal antibody, called WO2, which recognises the A beta peptide. To this end, we have determined the three-dimensional structure, to near atomic resolution, of both the antibody and the complex with its antigen, the A beta peptide. The structures reveal the molecular basis for WO2 recognition and binding of A beta. The A beta peptide adopts an extended, coil-like conformation across its major immunodominant B-cell epitope between residues 2 and 8. We have also studied the antibody-bound A beta peptide in the presence of metals known to affect its aggregation state and show that WO2 inhibits these interactions. Thus, antibodies that target the N-terminal region of A beta, such as WO2, hold promise for therapeutic development.
Collapse
Affiliation(s)
- Luke A Miles
- Biota Structural Biology Laboratory, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Victoria 3065, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that is characterized pathologically by the deposition of beta-amyloid (A beta)-containing extracellular neuritic plaques, intracellular neurofibrillary tangles and neuronal loss. Much evidence supports the hypothesis that A beta peptide aggregation contributes to AD pathogenesis, however, currently approved therapeutic treatments do nothing to stop or reverse A beta deposition. The success of active and passive anti-A beta immunotherapies in both preventing and clearing parenchymal amyloid in transgenic mouse models led to the initiation of an active anti-A beta vaccination (AN1792) trial in human patients with mild-to-moderate AD, but was prematurely halted when 6% of inoculated patients developed aseptic meningoencephalitis. Autopsy results from the brains of four individuals treated with AN1792 revealed decreased plaque burden in select brain areas, as well as T-cell lymphocytes in three of the patients. Furthermore, antibody responders showed some improvement in memory task measures. These findings indicated that anti-A beta therapy might still be a viable option for the treatment of AD, if potentially harmful proinflammatory processes can be avoided. Over the past 6 years, this target has led to the development of novel experimental immunization strategies, including selective A beta epitope targeting, antibody and adjuvant modifications, as well as alternative routes and mechanisms of vaccine delivery, to generate anti-A beta antibodies that selectively target and remove specific A beta species without evoking autoimmunity. Results from the passive vaccination AD clinical trials that are currently underway will provide invaluable information about both the effectiveness of newly improved anti-A beta vaccines in clinical treatment, as well as the role of the A beta peptide in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Cheryl A Hawkes
- Center for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.
| | | |
Collapse
|
21
|
Rak M, Del Bigio MR, Mai S, Westaway D, Gough K. Dense-core and diffuse Abeta plaques in TgCRND8 mice studied with synchrotron FTIR microspectroscopy. Biopolymers 2007; 87:207-17. [PMID: 17680701 DOI: 10.1002/bip.20820] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Plaques composed of the Abeta peptide are the main pathological feature of Alzheimer's disease. Dense-core plaques are fibrillar deposits of Abeta, showing all the classical properties of amyloid including beta-sheet secondary structure, while diffuse plaques are amorphous deposits. We studied both plaque types, using synchrotron infrared (IR) microspectroscopy, a technique that allows the chemical composition and average protein secondary structure to be investigated in situ. We examined plaques in hippocampal, cortical and caudal tissue from 5- to 21-month-old TgCRND8 mice, a transgenic model expressing doubly mutant amyloid precursor protein, and displaying impaired hippocampal function and robust pathology from an early age. Spectral analysis confirmed that the congophilic plaque cores were composed of protein in a beta-sheet conformation. The amide I maximum of plaque cores was at 1623 cm(-1), and unlike for in vitro Abeta fibrils, the high-frequency (1680-1690 cm(-1)) component attributed to antiparallel beta-sheet was not observed. A significant elevation in phospholipids was found around dense-core plaques in TgCRND8 mice ranging in age from 5 to 21 months. In contrast, diffuse plaques were not associated with IR detectable changes in protein secondary structure or relative concentrations of any other tissue components.
Collapse
Affiliation(s)
- Margaret Rak
- Department of Chemistry, University of Manitoba, Winnipeg, MB, Canada R3T 2N2
| | | | | | | | | |
Collapse
|
22
|
Chauhan NB. Intracerebroventricular passive immunization with anti-oligoAbeta antibody in TgCRND8. J Neurosci Res 2007; 85:451-63. [PMID: 17086547 DOI: 10.1002/jnr.21110] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Based on the central dogma of beta-amyloid (Abeta) as a key seeding event in the pathogenesis of Alzheimer disease (AD), immunoneutralization strategies have been actively pursued both in AD and in models of AD as a potential means for treating AD. Both active and passive immunizations targeted at fibrillar Abeta successfully remove cerebral plaque load and attenuate Abeta-induced toxicity. Consistently with this, intracerebroventricular (ICV) passive immunization established in our laboratory using antibody against fibrillar Abeta (anti-fAbeta) reduced cerebral plaque load and reversed early synaptic deficits at pre/early plaque stage when there is an abundance of soluble dimeric/oligomeric Abeta but sparse fibrillar Abeta, indicating that anti-fAbeta-mediated partial neutralization of toxic oligomeric Abeta species might have reduced early synaptotoxicity. In the previous investigation, we found that immunoneutralization with anti-fAbeta transiently reduced cerebral Abeta and associated toxicity. The current investigation tested whether ICV im munization using antibody to conformationally changed oligomeric Abeta (anti-oligoAbeta) will overcome the transient restorative nature of anti-fAbeta and produce persistent, long-lasting preventive effects. Because oligomeric Abeta is strongly correlated with synaptotoxicity, we investigated whether immunoneutralization of oligomeric Abeta will reverse synaptic deficits by analyzing presynaptic molecular marker (SNAP-25) profile within hippocampal dendritic fields, where SNAP-25 is abundantly expressed. Results show that, in contrast to ICV anti-fAbeta antibody, ICV anti-oligoAbeta antibody significantly prevented cerebral Abeta build and almost completely restored SNAP-25 immunoreaction up to 8 weeks postinjection in TgCRND8 brain. Results show that ICV passive immunization with anti-oligoAbeta antibody might be an improved ICV immunization strategy for preventing permanent structural damage in AD.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Research and Development (151), Jesse Brown VA Medical Center Chicago, Chicago, Illinois 60612, USA.
| |
Collapse
|
23
|
Prada CM, Garcia-Alloza M, Betensky RA, Zhang-Nunes SX, Greenberg SM, Bacskai BJ, Frosch MP. Antibody-mediated clearance of amyloid-beta peptide from cerebral amyloid angiopathy revealed by quantitative in vivo imaging. J Neurosci 2007; 27:1973-80. [PMID: 17314293 PMCID: PMC6673561 DOI: 10.1523/jneurosci.5426-06.2007] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is the accumulation of amyloid-beta peptide (Abeta) in the vessel wall of arteries in the brain. Because CAA is commonly associated with Alzheimer's disease (AD), characterized by parenchymal deposition of the same peptide in the form of senile plaques, there is considerable interest in the relationship of the two deposits in generating human disease. The study of CAA is of particular importance for immunotherapeutic approaches to AD, because reports of anti-Abeta immunotherapy in mice and humans have suggested that, whereas CAA appeared resistant to clearance, its response to this treatment promoted potential adverse effects, including meningoencephalitis. We used multiphoton microscopy and longitudinal imaging to monitor CAA in a mouse model of amyloid deposition to evaluate the effects of anti-Abeta passive immunotherapy. We found detectable clearance of CAA deposits within 1 week after a single administration of antibody directly to the brain, an effect that was short-lived. Chronic administration of antibody over 2 weeks led to more robust clearance without evidence of hemorrhage or other destructive changes. We found that the progressive clearance of Abeta from vessels follows distinct kinetics from what has been previously reported for clearance of plaques (parenchymal deposits of Abeta). This quantitative in vivo imaging approach directly demonstrates that CAA in a transgenic mouse model can be cleared with an optimized immunotherapy.
Collapse
Affiliation(s)
- Claudia M. Prada
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
| | - Monica Garcia-Alloza
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
| | - Rebecca A. Betensky
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts 02115, and
| | - Sandy X. Zhang-Nunes
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
| | - Steven M. Greenberg
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
| | - Brian J. Bacskai
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
| | - Matthew P. Frosch
- Department of Neurology/Alzheimer Research Unit, Massachusetts General Hospital, Charlestown, Massachusetts 02129
- C. S. Kubik Laboratory for Neuropathology, Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114
| |
Collapse
|
24
|
Kim HD, Tahara K, Maxwell JA, Lalonde R, Fukuiwa T, Fujihashi K, Van Kampen KR, Kong FK, Tang DCC, Fukuchi KI. Nasal inoculation of an adenovirus vector encoding 11 tandem repeats of Abeta1-6 upregulates IL-10 expression and reduces amyloid load in a Mo/Hu APPswe PS1dE9 mouse model of Alzheimer's disease. J Gene Med 2007; 9:88-98. [PMID: 17219449 PMCID: PMC2446608 DOI: 10.1002/jgm.993] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND One of the pathological hallmarks of Alzheimer's disease (AD) is deposits of amyloid beta-peptide (Abeta) in neuritic plaques and cerebral vessels. Immunization of AD mouse models with Abeta reduces Abeta deposits and improves memory and learning deficits. Because recent clinical trials of immunization with Abeta were halted due to brain inflammation that was presumably induced by a T-cell-mediated autoimmune response, vaccination modalities that elicit predominantly humoral immune responses are currently being developed. METHODS We have nasally immunized a young AD mouse model with an adenovirus vector encoding 11 tandem repeats of Abeta1-6 fused to the receptor-binding domain (Ia) of Pseudomonas exotoxin A (PEDI), AdPEDI-(Abeta1-6)(11), in order to evaluate the efficacy of the vector in preventing Abeta deposits in the brain. We also have investigated immune responses of mice to AdPEDI-(Abeta1-6)(11). RESULTS Nasal immunization of an AD mouse model with AdPEDI-(Abeta1-6)(11) elicited a predominant IgG1 response and reduced Abeta load in the brain. The plasma IL-10 level in the AD mouse model was upregulated after immunization and, upon the stimulation with PEDI-(Abeta1-6)(11), marked IL-10 responses were found in splenic CD4(+) T cells from C57BL/6 mice that had been immunized with AdPEDI-(Abeta1-6)(11). CONCLUSIONS These results suggest that the induction of Th2-biased responses with AdPEDI-(Abeta1-6)(11) in mice is mediated in part through the upregulation of IL-10, which inhibits activation of dendritic cells that dictate the induction of Th1 cells.
Collapse
Affiliation(s)
- Hong-Duck Kim
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, PO Box 1649, Peoria, IL 61656, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The pathological hallmarks of Alzheimer's disease (AD) include beta-amyloid (Abeta) plaques, dystrophic neurites and neurofibrillary pathology, which eventually result in the degeneration of neurons and subsequent dementia. In 1999, international interest in a new therapeutic approach to the treatment of AD was ignited following transgenic mouse studies that indicated that it might be possible to immunise against the pathological alterations in Abeta that lead to aggregation of this protein in the brain. A subsequent phase I human trial for safety, tolerability and immunogenicity using an active immunisation strategy against Abeta had a positive outcome. However, phase IIA human trials involving active immunisation were halted following the diagnosis of aseptic meningoencephalitis in 6% of immunised subjects. Research into immunisation strategies involving transgenic AD mouse models has subsequently been refocused to determine the mechanisms by which plaque clearance and reduced memory deficits are attained, and to establish safer therapeutic approaches that may reduce potentially harmful brain inflammation. The vigour of international research on immunotherapy for AD provides significant hope for a strong therapeutic lead for the escalating number of individuals who will develop this otherwise incurable condition.
Collapse
Affiliation(s)
- Adele Woodhouse
- School of Medicine, NeuroRepair Group, University of Tasmania, Hobart, Tasmania, Australia.
| | | | | |
Collapse
|
26
|
Chauhan NB. Effect of aged garlic extract on APP processing and tau phosphorylation in Alzheimer's transgenic model Tg2576. JOURNAL OF ETHNOPHARMACOLOGY 2006; 108:385-94. [PMID: 16842945 DOI: 10.1016/j.jep.2006.05.030] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2005] [Revised: 05/21/2006] [Accepted: 05/25/2006] [Indexed: 05/10/2023]
Abstract
Multiple components present in garlic and various garlic preparations are known to exert pleiotropic protective effects as demonstrated in various in vitro and in vivo model systems. However, garlic pleiotropy in relation to Alzheimer's pathophysiology has not been explored extensively. Current study investigated anti-amyloidogenic, anti-inflammatory and anti-tangle effects of dietary aged garlic extract (AGE) (2%) and compared with its prominent constituents, i.e. S-allyl-cysteine (SAC) (20 mg/kg) and di-allyl-disulfide (DADS) (20 mg/kg) in Alzheimer's Swedish double mutant mouse model (Tg2576). Possible cholesterol-dependent and cholesterol-independent mechanisms of actions of AGE, SAC and DADS in exerting anti-amyloidogenic, anti-inflammatory and anti-tangle effects are discussed. Finally, ameliorative effects of dietary interventions were found to be in the order of AGE>SAC>DADS. If validated pre-clinically, dietary intervention with herbal alternative such as AGE having pleiotropic useful properties and least adverse effects may provide greater therapeutic benefit over a single-ingredient synthetic pharmaceutical drug having serious side effects in treating Alzheimer's disease.
Collapse
Affiliation(s)
- Neelima B Chauhan
- Department of Anesthesiology, University of Illinois at Chicago, USA.
| |
Collapse
|
27
|
Virus and virus-like particle-based immunogens for Alzheimer's disease induce antibody responses against amyloid-beta without concomitant T cell responses. Vaccine 2006; 24:6321-31. [PMID: 16806604 DOI: 10.1016/j.vaccine.2006.05.059] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2005] [Revised: 05/15/2006] [Accepted: 05/22/2006] [Indexed: 11/17/2022]
Abstract
A vaccine targeting the amyloid-beta (Abeta) peptide is a promising potential immunotherapy for Alzheimer's disease patients. However, experience from a recent clinical trial of a candidate Abeta vaccine has suggested that it is important to develop techniques to induce high titer antibodies against Abeta associated with vaccine efficacy while reducing the T cell responses against Abeta that were potentially responsible for serious side effects. We have previously demonstrated that immunization with self- and foreign antigens arrayed in a repetitive fashion on the surface of virus-like particles (VLPs) induces high titer antibody responses at low doses and in the absence of potentially inflammatory adjuvants. In this study, we examined the antibody and T cell responses upon immunization with human papillomavirus VLP- and Qbeta bacteriophage-based Abeta vaccines. Immunization with Abeta conjugated to VLPs or Qbeta elicited anti-Abeta antibody responses at low doses and without the use of adjuvants. The flexibility of these virus-based display systems allowed us to link and induce antibodies against short Abeta-derived peptides from the amino- and carboxyl-termini of the peptide. Immunization of mice with Abeta peptide in combination with Freund's adjuvant elicited predominantly IgG2c antibodies and strong T cell proliferative responses against Abeta. In contrast, VLP-conjugated Abeta peptides elicited more balanced isotype responses, dominated by IgG1. Both VLP and Qbeta-based Abeta vaccines induced weak or negligible T cell responses against Abeta. T cell responses were largely directed against linked viral epitopes. Taken together, virus-based vaccines that allow the presentation of Abeta in a repetitive dense array are new and potentially more effective vaccine candidates for Alzheimer's disease.
Collapse
|
28
|
Carty NC, Wilcock DM, Rosenthal A, Grimm J, Pons J, Ronan V, Gottschall PE, Gordon MN, Morgan D. Intracranial administration of deglycosylated C-terminal-specific anti-Abeta antibody efficiently clears amyloid plaques without activating microglia in amyloid-depositing transgenic mice. J Neuroinflammation 2006; 3:11. [PMID: 16686956 PMCID: PMC1479322 DOI: 10.1186/1742-2094-3-11] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2006] [Accepted: 05/10/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Antibodies against the Ass peptide clear Ass deposits when injected intracranially. Deglycosylated antibodies have reduced effector functions compared to their intact counterparts, potentially avoiding immune activation. METHODS Deglycosylated or intact C-terminal specific high affinity anti-Abeta antibody (2H6) were intracranially injected into the right frontal cortex and hippocampus of amyloid precursor protein (APP) transgenic mice. The untreated left hemisphere was used to normalize for the extent of amyloid deposition present in each mouse. Control transgenic mice were injected with an antibody against a drosophila-specific protein (amnesiac). Tissues were examined for brain amyloid deposition and microglial responses 3 days after the injection. RESULTS The deglycosylated 2H6 antibody had lower affinity for several murine Fcgamma receptors and human complement than intact 2H6 without a change in affinity for Ass. Immunohistochemistry for Abeta and thioflavine-S staining revealed that both diffuse and compact deposits were reduced by both antibodies. In animals treated with the intact 2H6 antibody, a significant increase in Fcgamma-receptor II/III immunostaining was observed compared to animals treated with the control IgG antibody. No increase in Fcgamma-receptor II/III was found with the deglycosylated 2H6 antibody. Immunostaining for the microglial activation marker CD45 demonstrated a similar trend. CONCLUSION These findings suggest that the deglycosylated 2H6 is capable of removing both compact and diffuse plaques without activating microglia. Thus, antibodies with reduced effector functions may clear amyloid without concomitant immune activation when tested as immunotherapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Niki C Carty
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Donna M Wilcock
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Arnon Rosenthal
- Rinat Neuroscience Corp. 3155 Porter Drive, Palo Alto, California, 94304, USA
| | - Jan Grimm
- Rinat Neuroscience Corp. 3155 Porter Drive, Palo Alto, California, 94304, USA
| | - Jaume Pons
- Rinat Neuroscience Corp. 3155 Porter Drive, Palo Alto, California, 94304, USA
| | - Victoria Ronan
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Paul E Gottschall
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Marcia N Gordon
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Dave Morgan
- Alzheimer's Research Laboratory, University of South Florida, Department of Molecular Pharmacology and Physiology, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA
| |
Collapse
|
29
|
Levites Y, Das P, Price RW, Rochette MJ, Kostura LA, McGowan EM, Murphy MP, Golde TE. Anti-Abeta42- and anti-Abeta40-specific mAbs attenuate amyloid deposition in an Alzheimer disease mouse model. J Clin Invest 2005; 116:193-201. [PMID: 16341263 PMCID: PMC1307561 DOI: 10.1172/jci25410] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2005] [Accepted: 10/18/2005] [Indexed: 11/17/2022] Open
Abstract
Accumulation and aggregation of amyloid beta peptide 1-42 (Abeta42) in the brain has been hypothesized as triggering a pathological cascade that causes Alzheimer disease (AD). To determine whether selective targeting of Abeta42 versus Abeta40 or total Abeta is an effective way to prevent or treat AD, we compared the effects of passive immunization with an anti-Abeta42 mAb, an anti-Abeta40 mAb, and multiple Abeta(1-16) mAbs. We established in vivo binding selectivity of the anti-Abeta42 and anti-Abeta40 mAbs using novel TgBRI-Abeta mice. We then conducted a prevention study in which the anti-Abeta mAbs were administered to young Tg2576 mice, which have no significant Abeta deposition, and therapeutic studies in which mAbs were administered to Tg2576 or CRND8 mice with modest levels of preexisting Abeta deposits. Anti-Abeta42, anti-Abeta40, and anti-Abeta(1-16) mAbs attenuated plaque deposition in the prevention study. In contrast, anti-Abeta42 and anti-Abeta40 mAbs were less effective in attenuating Abeta deposition in the therapeutic studies and were not effective in clearing diffuse plaques following direct injection into the cortex. These data suggest that selective targeting of Abeta42 or Abeta40 may be an effective strategy to prevent amyloid deposition, but may have limited benefit in a therapeutic setting.
Collapse
Affiliation(s)
- Yona Levites
- Department of Neuroscience, Mayo Clinic, Mayo Clinic College of Medicine, Jacksonville, Florida 32224, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zirah S, Kozin SA, Mazur AK, Blond A, Cheminant M, Ségalas-Milazzo I, Debey P, Rebuffat S. Structural changes of region 1-16 of the Alzheimer disease amyloid beta-peptide upon zinc binding and in vitro aging. J Biol Chem 2005; 281:2151-61. [PMID: 16301322 DOI: 10.1074/jbc.m504454200] [Citation(s) in RCA: 248] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Amyloid deposits within the cerebral tissue constitute a characteristic lesion associated with Alzheimer disease. They mainly consist of the amyloid peptide Abeta and display an abnormal content in Zn(2+) ions, together with many truncated, isomerized, and racemized forms of Abeta. The region 1-16 of Abeta can be considered the minimal zinc-binding domain and contains two aspartates subject to protein aging. The influence of zinc binding and protein aging related modifications on the conformation of this region of Abeta is of importance given the potentiality of this domain to constitute a therapeutic target, especially for immunization approaches. In this study, we determined from NMR data the solution structure of the Abeta-(1-16)-Zn(2+) complex in aqueous solution at pH 6.5. The residues His(6), His(13), and His(14) and the Glu(11) carboxylate were identified as ligands that tetrahedrally coordinate the Zn(II) cation. In vitro aging experiments on Abeta-(1-16) led to the formation of truncated and isomerized species. The major isomer generated, Abeta-(1-16)-l-iso-Asp(7), displayed a local conformational change in the His(6)-Ser(8) region but kept a zinc binding propensity via a coordination mode involving l-iso-Asp(7). These results are discussed here with regard to Abeta fibrillogenesis and the potentiality of the region 1-16 of Abeta to be used as a therapeutic target.
Collapse
Affiliation(s)
- Séverine Zirah
- Laboratoire de Chimie et Biochimie des Substances Naturelles, UMR 5154 CNRS-MNHN, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, 63 Rue Buffon, 75005 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Johanson CE, Duncan JA, Stopa EG, Baird A. Enhanced Prospects for Drug Delivery and Brain Targeting by the Choroid Plexus–CSF Route. Pharm Res 2005; 22:1011-37. [PMID: 16028003 DOI: 10.1007/s11095-005-6039-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Accepted: 04/12/2005] [Indexed: 02/07/2023]
Abstract
The choroid plexus (CP), i.e., the blood-cerebrospinal fluid barrier (BCSFB) interface, is an epithelial boundary exploitable for drug delivery to brain. Agents transported from blood to lateral ventricles are convected by CSF volume transmission (bulk flow) to many periventricular targets. These include the caudate, hippocampus, specialized circumventricular organs, hypothalamus, and the downstream pia-glia and arachnoid membranes. The CSF circulatory system normally provides micronutrients, neurotrophins, hormones, neuropeptides, and growth factors extensively to neuronal networks. Therefore, drugs directed to CSF can modulate a variety of endocrine, immunologic, and behavioral phenomema; and can help to restore brain interstitial and cellular homeostasis disrupted by disease and trauma. This review integrates information from animal models that demonstrates marked physiologic effects of substances introduced into the ventricular system. It also recapitulates how pharmacologic agents administered into the CSF system prevent disease or enhance the brain's ability to recover from chemical and physical insults. In regard to drug distribution in the CNS, the BCSFB interaction with the blood-brain barrier is discussed. With a view toward translational CSF pharmacotherapy, there are several promising innovations in progress: bone marrow cell infusions, CP encapsulation and transplants, neural stem cell augmentation, phage display of peptide ligands for CP epithelium, CSF gene transfer, regulation of leukocyte and cytokine trafficking at the BCSFB, and the purification of neurotoxic CSF in degenerative states. The progressively increasing pharmacological significance of the CP-CSF nexus is analyzed in light of treating AIDS, multiple sclerosis, stroke, hydrocephalus, and Alzheimer's disease.
Collapse
Affiliation(s)
- Conrad E Johanson
- Department of Clinical Neurosciences, Rhode Island Hospital, Brown Medical School, Providence, Rhode Island 02912, USA.
| | | | | | | |
Collapse
|