1
|
Serag I, Abouzid M, Moawad MHED, Jaradat JH, Hendawy M, Hendi NI, Alkhawaldeh IM, Abdullah JA, Elsakka MM, Muneer MA, Elnagar MA, Fakher MA, Elkenani AJ, Abbas A. Vaccines for Alzheimer's disease: a brief scoping review. Neurol Sci 2025:10.1007/s10072-025-08073-2. [PMID: 40111670 DOI: 10.1007/s10072-025-08073-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder and the most common cause of dementia among older adults. Existing treatments-such as cholinesterase inhibitors, N-methyl-D-aspartate receptor antagonists, and monoclonal antibodies targeting amyloid beta-can improve functional and neuropsychiatric outcomes but fail to prevent disease onset, halt progression, or adequately reduce amyloid-beta burden. Consequently, research efforts have shifted to primary prevention through immunization, although the efficacy of these strategies remains uncertain. This review explores the efficacy, safety, and adverse events of current immunotherapies for AD and discusses future research and clinical implications. METHODS A scoping review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses for Scoping Reviews (PRISMA-SR) checklist. A systematic search was carried out using PubMed, Scopus, and Web of Science. RESULTS A total of 145 studies were included. Preclinical research often employed transgenic mouse models to investigate AD pathology and vaccine benefits, while Phase I and II clinical trials centered on safety and preliminary efficacy in humans. Most studies were conducted in the USA, China, and Japan, highlighting these countries' strong clinical trial infrastructure. Vaccination frequently reduced amyloid-beta or tau pathology in preclinical settings, although cognitive outcomes were inconsistent. Clinical trials primarily focused on safety and immune response, with newer vaccines such as ABvac40 demonstrating encouraging results and minimal adverse events. CONCLUSION Although AD vaccines show promise in preclinical settings, longer and more comprehensive clinical trials are necessary to determine their long-term efficacy and safety. Standardized protocols and efforts to reduce regional disparities in research would facilitate better comparability and generalizability of findings, thereby guiding the future development of effective immunotherapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Ibrahim Serag
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abouzid
- Department of Physical Pharmacy and Pharmacokinetics, Faculty of Pharmacy, Poznan University of Medical Sciences, Rokietnicka 3 St., 60-806, Poznan, Poland.
- Doctoral School, Poznan University of Medical Sciences, 60-812, Poznan, Poland.
| | - Mostafa Hossam El Din Moawad
- Alexandria Main University Hospital, Alexandria, Egypt
- Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Mohamed Hendawy
- Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | | | | | | | | | | | | | - Aya J Elkenani
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Abdallah Abbas
- Faculty of Medicine, Al-Azhar University, Damietta, Egypt
| |
Collapse
|
2
|
Rajendran K, Krishnan UM. Mechanistic insights and emerging therapeutic stratagems for Alzheimer's disease. Ageing Res Rev 2024; 97:102309. [PMID: 38615895 DOI: 10.1016/j.arr.2024.102309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
Alzheimer's disease (AD), a multi-factorial neurodegenerative disorder has affected over 30 million individuals globally and these numbers are expected to increase in the coming decades. Current therapeutic interventions are largely ineffective as they focus on a single target. Development of an effective drug therapy requires a deep understanding of the various factors influencing the onset and progression of the disease. Aging and genetic factors exert a major influence on the development of AD. Other factors like post-viral infections, iron overload, gut dysbiosis, and vascular dysfunction also exacerbate the onset and progression of AD. Further, post-translational modifications in tau, DRP1, CREB, and p65 proteins increase the disease severity through triggering mitochondrial dysfunction, synaptic loss, and differential interaction of amyloid beta with different receptors leading to impaired intracellular signalling. With advancements in neuroscience tools, new inter-relations that aggravate AD are being discovered including pre-existing diseases and exposure to other pathogens. Simultaneously, new therapeutic strategies involving modulation of gene expression through targeted delivery or modulation with light, harnessing the immune response to promote clearance of amyloid deposits, introduction of stem cells and extracellular vesicles to replace the destroyed neurons, exploring new therapeutic molecules from plant, marine and biological sources delivered in the free state or through nanoparticles and use of non-pharmacological interventions like music, transcranial stimulation and yoga. Polypharmacology approaches involving combination of therapeutic agents are also under active investigation for superior therapeutic outcomes. This review elaborates on various disease-causing factors, their underlying mechanisms, the inter-play between different disease-causing players, and emerging therapeutic options including those under clinical trials, for treatment of AD. The challenges involved in AD therapy and the way forward have also been discussed.
Collapse
Affiliation(s)
- Kayalvizhi Rajendran
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India
| | - Uma Maheswari Krishnan
- School of Chemical & Biotechnology, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; Centre for Nanotechnology & Advanced Biomaterials, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India; School of Arts, Sciences, Humanities & Education, SASTRA Deemed University, Thanjavur, Tamilnadu 613401, India.
| |
Collapse
|
3
|
Tondo G, De Marchi F, Bonardi F, Menegon F, Verrini G, Aprile D, Anselmi M, Mazzini L, Comi C. Novel Therapeutic Strategies in Alzheimer's Disease: Pitfalls and Challenges of Anti-Amyloid Therapies and Beyond. J Clin Med 2024; 13:3098. [PMID: 38892809 PMCID: PMC11172489 DOI: 10.3390/jcm13113098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) causes a significant challenge to global healthcare systems, with limited effective treatments available. This review examines the landscape of novel therapeutic strategies for AD, focusing on the shortcomings of traditional therapies against amyloid-beta (Aβ) and exploring emerging alternatives. Despite decades of research emphasizing the role of Aβ accumulation in AD pathogenesis, clinical trials targeting Aβ have obtained disappointing results, highlighting the complexity of AD pathophysiology and the need for investigating other therapeutic approaches. In this manuscript, we first discuss the challenges associated with anti-Aβ therapies, including limited efficacy and potential adverse effects, underscoring the necessity of exploring alternative mechanisms and targets. Thereafter, we review promising non-Aβ-based strategies, such as tau-targeted therapies, neuroinflammation modulation, and gene and stem cell therapy. These approaches offer new avenues for AD treatment by addressing additional pathological hallmarks and downstream effects beyond Aβ deposition.
Collapse
Affiliation(s)
- Giacomo Tondo
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Fabiola De Marchi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Francesca Bonardi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Federico Menegon
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Gaia Verrini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Davide Aprile
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Matteo Anselmi
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Letizia Mazzini
- Neurology Unit, Department of Translational Medicine, Maggiore della Carità Hospital, University of Piemonte Orientale, 28100 Novara, Italy; (G.T.); (F.B.); (F.M.); (G.V.); (D.A.); (M.A.); (L.M.)
| | - Cristoforo Comi
- Neurology Unit, Department of Translational Medicine, Sant’Andrea Hospital, University of Piemonte Orientale, Corso Abbiate 21, 13100 Vercelli, Italy;
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
4
|
Suliman M, Al-Hawary SIS, Al-Dolaimy F, Hjazi A, Almalki SG, Alkhafaji AT, Alawadi AH, Alsaalamy A, Bijlwan S, Mustafa YF. Inflammatory diseases: Function of LncRNAs in their emergence and the role of mesenchymal stem cell secretome in their treatment. Pathol Res Pract 2023; 249:154758. [PMID: 37660657 DOI: 10.1016/j.prp.2023.154758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/05/2023]
Abstract
One of the best treatments for inflammatory diseases such as COVID-19, respiratory diseases and brain diseases is treatment with stem cells. Here we investigate the effect of stem cell therapy in the treatment of brain diseases.Preclinical studies have shown promising results, including improved functional recovery and tissue repair in animal models of neurodegenerative diseases, strokes,and traumatic brain injuries. However,ethical implications, safety concerns, and regulatory frameworks necessitate thorough evaluation before transitioning to clinical applications. Additionally, the complex nature of the brain and its intricate cellular environment present unique obstacles that must be overcome to ensure the successful integration and functionality of genetically engineered MSCs. The careful navigation of this path will determine whether the application of genetically engineered MSCs in brain tissue regeneration ultimately lives up to the hype surrounding it.
Collapse
Affiliation(s)
- Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| | | | - Ahmed Hussien Alawadi
- College of technical engineering, the Islamic University, Najaf, Iraq; College of technical engineering, the Islamic University of Al Diwaniyah, Iraq; College of technical engineering, the Islamic University of Babylon, Iraq
| | - Ali Alsaalamy
- College of technical engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq
| | - Sheela Bijlwan
- Uttaranchal School of Computing Sciences, Uttaranchal University, Dehradun, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
5
|
Gadhave K, Kumar D, Uversky VN, Giri R. A multitude of signaling pathways associated with Alzheimer's disease and their roles in AD pathogenesis and therapy. Med Res Rev 2021; 41:2689-2745. [PMID: 32783388 PMCID: PMC7876169 DOI: 10.1002/med.21719] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/13/2020] [Accepted: 07/29/2020] [Indexed: 02/06/2023]
Abstract
The exact molecular mechanisms associated with Alzheimer's disease (AD) pathology continue to represent a mystery. In the past decades, comprehensive data were generated on the involvement of different signaling pathways in the AD pathogenesis. However, the utilization of signaling pathways as potential targets for the development of drugs against AD is rather limited due to the immense complexity of the brain and intricate molecular links between these pathways. Therefore, finding a correlation and cross-talk between these signaling pathways and establishing different therapeutic targets within and between those pathways are needed for better understanding of the biological events responsible for the AD-related neurodegeneration. For example, autophagy is a conservative cellular process that shows link with many other AD-related pathways and is crucial for maintenance of the correct cellular balance by degrading AD-associated pathogenic proteins. Considering the central role of autophagy in AD and its interplay with many other pathways, the finest therapeutic strategy to fight against AD is the use of autophagy as a target. As an essential step in this direction, this comprehensive review represents recent findings on the individual AD-related signaling pathways, describes key features of these pathways and their cross-talk with autophagy, represents current drug development, and introduces some of the multitarget beneficial approaches and strategies for the therapeutic intervention of AD.
Collapse
Affiliation(s)
- Kundlik Gadhave
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Deepak Kumar
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| | - Vladimir N. Uversky
- Department of Molecular Medicine and Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States of America
- Laboratory of New Methods in Biology, Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Rajanish Giri
- School of Basic Sciences, Indian Institute of Technology Mandi, Kamand, Himachal Pradesh, 175005, India
| |
Collapse
|
6
|
Kabir MT, Uddin MS, Mathew B, Das PK, Perveen A, Ashraf GM. Emerging Promise of Immunotherapy for Alzheimer's Disease: A New Hope for the Development of Alzheimer's Vaccine. Curr Top Med Chem 2021; 20:1214-1234. [PMID: 32321405 DOI: 10.2174/1568026620666200422105156] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the characteristics of this devastating disorder include the progressive and disabling deficits in the cognitive functions including reasoning, attention, judgment, comprehension, memory, and language. OBJECTIVE In this article, we have focused on the recent progress that has been achieved in the development of an effective AD vaccine. SUMMARY Currently, available treatment options of AD are limited to deliver short-term symptomatic relief only. A number of strategies targeting amyloid-beta (Aβ) have been developed in order to treat or prevent AD. In order to exert an effective immune response, an AD vaccine should contain adjuvants that can induce an effective anti-inflammatory T helper 2 (Th2) immune response. AD vaccines should also possess the immunogens which have the capacity to stimulate a protective immune response against various cytotoxic Aβ conformers. The induction of an effective vaccine's immune response would necessitate the parallel delivery of immunogen to dendritic cells (DCs) and their priming to stimulate a Th2-polarized response. The aforesaid immune response is likely to mediate the generation of neutralizing antibodies against the neurotoxic Aβ oligomers (AβOs) and also anti-inflammatory cytokines, thus preventing the AD-related inflammation. CONCLUSION Since there is an age-related decline in the immune functions, therefore vaccines are more likely to prevent AD instead of providing treatment. AD vaccines might be an effective and convenient approach to avoid the treatment-related huge expense.
Collapse
Affiliation(s)
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh.,Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, India
| | | | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Saharanpur, India
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
7
|
Hinteregger B, Loeffler T, Flunkert S, Neddens J, Bayer TA, Madl T, Hutter-Paier B. Metabolic, Phenotypic, and Neuropathological Characterization of the Tg4-42 Mouse Model for Alzheimer's Disease. J Alzheimers Dis 2021; 80:1151-1168. [PMID: 33646155 PMCID: PMC8150512 DOI: 10.3233/jad-201204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Preclinical Alzheimer's disease (AD) research strongly depends on transgenic mouse models that display major symptoms of the disease. Although several AD mouse models have been developed representing relevant pathologies, only a fraction of available mouse models, like the Tg4-42 mouse model, display hippocampal atrophy caused by the death of neurons as the key feature of AD. The Tg4-42 mouse model is therefore very valuable for use in preclinical research. Furthermore, metabolic biomarkers which have the potential to detect biochemical changes, are crucial to gain deeper insights into the pathways, the underlying pathological mechanisms and disease progression. OBJECTIVE We thus performed an in-depth characterization of Tg4-42 mice by using an integrated approach to analyze alterations of complex biological networks in this AD in vivo model. METHODS Therefore, untargeted NMR-based metabolomic phenotyping was combined with behavioral tests and immunohistological and biochemical analyses. RESULTS Our in vivo experiments demonstrate a loss of body weight increase in homozygous Tg4-42 mice over time as well as severe impaired learning behavior and memory deficits in the Morris water maze behavioral test. Furthermore, we found significantly altered metabolites in two different brain regions and metabolic changes of the glutamate/4-aminobutyrate-glutamine axis. Based on these results, downstream effects were analyzed showing increased Aβ42 levels, increased neuroinflammation as indicated by increased astro- and microgliosis as well as neuronal degeneration and neuronal loss in homozygous Tg4-42 mice. CONCLUSION Our study provides a comprehensive characterization of the Tg4-42 mouse model which could lead to a deeper understanding of pathological features of AD. Additionally this study reveals changes in metabolic biomarker which set the base for future preclinical studies or drug development.
Collapse
Affiliation(s)
- Barbara Hinteregger
- QPS Austria GmbH, Grambach, Austria
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
| | | | | | | | - Thomas A. Bayer
- Department of Psychiatry and Psychotherapy, Division of Molecular Psychiatry, University Medical Center, Göttingen (UMG), Göttingen, Germany
| | - Tobias Madl
- Gottfried Schatz Research Center (for Cell Signaling, Metabolism and Aging), Division of Molecular Biology and Biochemistry, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | | |
Collapse
|
8
|
Qß Virus-like particle-based vaccine induces robust immunity and protects against tauopathy. NPJ Vaccines 2019; 4:26. [PMID: 31231552 PMCID: PMC6547647 DOI: 10.1038/s41541-019-0118-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/10/2019] [Indexed: 12/14/2022] Open
Abstract
Tauopathies, including frontotemporal dementia (FTD) and Alzheimer’s disease (AD) are progressive neurodegenerative diseases clinically characterized by cognitive decline and could be caused by the aggregation of hyperphosphorylated pathological tau (pTau) as neurofibrillary tangles (NFTs) inside neurons. There is currently no FDA-approved treatment that cures, slows or prevents tauopathies. Current immunotherapy strategies targeting pTau have generated encouraging data but may pose concerns about scalability, affordability, and efficacy. Here, we engineered a virus-like particle (VLP)-based vaccine in which tau peptide, phosphorylated at threonine 181, was linked at high valency to Qß bacteriophage VLPs (pT181-Qß). We demonstrate that vaccination with pT181-Qß is sufficient to induce a robust and long-lived anti-pT181 antibody response in the sera and the brains of both Non-Tg and rTg4510 mice. Only sera from pT181-Qß vaccinated mice are reactive to classical somatodendritic pTau in human FTD and AD post-mortem brain sections. Finally, we demonstrate that pT181-Qß vaccination reduces both soluble and insoluble species of hyperphosphorylated pTau in the hippocampus and cortex, avoids a Th1-mediated pro-inflammatory cell response, prevents hippocampal and corpus callosum atrophy and rescues cognitive dysfunction in a 4-month-old rTg4510 mouse model of FTD. These studies provide a valid scientific premise for the development of VLP-based immunotherapy to target pTau and potentially prevent Alzheimer’s diseases and related tauopathies. Tauopathies such as fronto-temporal dementia or Alzheimer’s disease are characterized by the accumulation of phosphorylated Tau (pTau) protein into pathogenic neurofibrillary tangles (NFT). Kiran Bhaskar and colleagues at the University of New Mexico investigate the efficacy of an active vaccine approach in the treatment of rTg4510 mice—an aggressive model of tauopathy. Mice receive 3 intramuscular doses of a disease-relevant pTau peptide (pT181) multivalently conjugated to an immunostimulatory bacteriophage virus-like particle (pT181-Qß). Vaccination induces high titers of anti-pTau—stable to at least 20 weeks—that is also able to bind human disease samples, but importantly does not react to unphosphorylated physiological Tau protein. Antibody can enter the brain and bind both soluble and intraneuronal pTau. Vaccination of mice reduces brain NFT, pathology, indicators of neuroinflammation and improves cognitive function in two different models of memory.
Collapse
|
9
|
Marciani DJ. Promising Results from Alzheimer's Disease Passive Immunotherapy Support the Development of a Preventive Vaccine. RESEARCH 2019; 2019:5341375. [PMID: 31549066 PMCID: PMC6750119 DOI: 10.34133/2019/5341375] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/18/2019] [Indexed: 12/23/2022]
Abstract
The apparently near-term effects of the monoclonal antibody BAN2401 in slowing the progression of prodromal Alzheimer's disease (AD) has created cautious optimism about the therapeutic use of antibodies that neutralize cytotoxic soluble amyloid-β aggregates, rather than removing plaque. Plaque being protective, as it immobilizes cytotoxic amyloid-β, rather than AD's causative agent. The presence of natural antibodies against cytotoxic amyloid-β implies the existence of a protective anti-AD immunity. Hence, for vaccines to induce a similar immunoresponse that prevents and/or delays the onset of AD, they must have adjuvants that stimulate a sole anti-inflammatory Th2 immunity, plus immunogens that induce a protective immunoresponse against diverse cytotoxic amyloid-β conformers. Indeed, amyloid-β pleomorphism may explain the lack of long-term protection by monoclonal antibodies that neutralize single conformers, like aducanumab. A situation that would allow new cytotoxic conformers to escape neutralization by previously effective monoclonal antibodies. Stimulation of a vaccine's effective immunoresponse would require the concurrent delivery of immunogen to dendritic cells and their priming, to induce a polarized Th2 immunity. An immunoresponse that would produce besides neutralizing antibodies against neurotoxic amyloid-β oligomers, anti-inflammatory cytokines; preventing inflammation that aggravates AD. Because of age-linked immune decline, vaccines would be significantly more effective in preventing, rather than treating AD. Considering the amyloid-β's role in tau's pathological hyperphosphorylation and their synergism in AD, the development of preventive vaccines against both amyloid-β and tau should be considered. Due to convenience and cost, vaccines may be the only option available to many countries to forestall the impending AD epidemic.
Collapse
Affiliation(s)
- D J Marciani
- Qantu Therapeutics, Inc., 612 E. Main Street, Lewisville, TX 75057, USA
| |
Collapse
|
10
|
Cao J, Hou J, Ping J, Cai D. Advances in developing novel therapeutic strategies for Alzheimer's disease. Mol Neurodegener 2018; 13:64. [PMID: 30541602 PMCID: PMC6291983 DOI: 10.1186/s13024-018-0299-8] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's Disease (AD), the most prevalent neurodegenerative disease of aging, affects one in eight older Americans. Nearly all drug treatments tested for AD today have failed to show any efficacy. There is a great need for therapies to prevent and/or slow the progression of AD. The major challenge in AD drug development is lack of clarity about the mechanisms underlying AD pathogenesis and pathophysiology. Several studies support the notion that AD is a multifactorial disease. While there is abundant evidence that amyloid plays a role in AD pathogenesis, other mechanisms have been implicated in AD such as tangle formation and spread, dysregulated protein degradation pathways, neuroinflammation, and loss of support by neurotrophic factors. Therefore, current paradigms of AD drug design have been shifted from single target approach (primarily amyloid-centric) to developing drugs targeted at multiple disease aspects, and from treating AD at later stages of disease progression to focusing on preventive strategies at early stages of disease development. Here, we summarize current strategies and new trends of AD drug development, including pre-clinical and clinical trials that target different aspects of disease (mechanism-based versus non-mechanism based, e.g. symptomatic treatments, lifestyle modifications and risk factor management).
Collapse
Affiliation(s)
- Jiqing Cao
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Jianwei Hou
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Jing Ping
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| | - Dongming Cai
- James J Peters VA Medical Center, Research & Development, Bronx, NY 10468 USA
- Department of Neurology, Alzheimer Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- The Central Hospital of The Hua Zhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Jena BP, Gatti DL, Arslanturk S, Pernal S, Taatjes DJ. Human skeletal muscle cell atlas: Unraveling cellular secrets utilizing 'muscle-on-a-chip', differential expansion microscopy, mass spectrometry, nanothermometry and machine learning. Micron 2018; 117:55-59. [PMID: 30472499 DOI: 10.1016/j.micron.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/14/2018] [Accepted: 11/14/2018] [Indexed: 01/28/2023]
Abstract
The 'Human Cell Atlas' project has been launched to obtain a comprehensive understanding of all cell types, the fundamental living units that constitute the human body. This is a global partnership and effort involving experts from many disciplines, from computer science, engineering to medicine, and is supported by several private and public organizations, among them, the Chan Zuckerberg Foundation, the National Institutes of Health, and Google, that will greatly benefit humanity. Nearly 37 trillion cells of various shapes, sizes, and composition, are precisely organized to constitute the human body. Humans, like all other living organisms, are dynamic, and therefore a comprehensive understanding of different cells in their various dynamic states is required to provide a reference map for the early diagnosis and various preventive approach to disease, and in the development of precision therapeutics. Skeletal muscles being the most abundant tissue and the largest locomotor and metabolic organ in the human body, requires a global understanding of its structure, composition, and function. The objective of creating a 'Human Skeletal Muscle Cell Atlas', necessitates therefore a comprehensive understanding of the emergent properties of skeletal muscle cell growth, development, structure, function and chemistry, under conditions of activity and inactivity. To achieve this objective would require a very precise yet rapid and cost-effective approach of combined multimodal imaging, including our new and novel 'Differential Expansion Microscopy', our 'Nanoscale Thermometry', combined with 'Mass Spectrometry', 'Motor Protein Motility Assay' and 'Machine Learning' tools.
Collapse
Affiliation(s)
- Bhanu P Jena
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| | - Domenico L Gatti
- Department of Biochemistry, Microbiology and Immunology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Suzan Arslanturk
- Department of Computer Science, College of Engineering, Wayne State University, Detroit, MI 48201, USA
| | - Sebastian Pernal
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Douglas J Taatjes
- Department of Pathology and Laboratory Medicine, Microscopy Imaging Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| |
Collapse
|
12
|
Coder B, Wang W, Wang L, Wu Z, Zhuge Q, Su DM. Friend or foe: the dichotomous impact of T cells on neuro-de/re-generation during aging. Oncotarget 2018; 8:7116-7137. [PMID: 27738345 PMCID: PMC5351694 DOI: 10.18632/oncotarget.12572] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 10/05/2016] [Indexed: 12/15/2022] Open
Abstract
The interaction between T cells and the central nervous system (CNS) in homeostasis and injury has been recognized being both pathogenic (CD4+ T-helper 1 - Th1, Th17 and γδT) and ameliorative (Th2 and regulatory T cells - Tregs). However, in-depth studies aimed to elucidate the precise in the aged microenvironment and the dichotomous role of Tregs have just begun and many aspects remain unclear. This is due, not only to a mutual dependency and reciprocal causation of alterations and diseases between the nervous and T cell immune systems, but also to an inconsistent aging of the two systems, which dynamically changes with CNS injury/recovery and/or aging process. Cellular immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution - sources of chronic inflammation in the elderly (termed inflammaging), potentially induces an acceleration of brain aging and memory loss. In turn, aging of the brain via neuro-endocrine-immune network drives total body systemic aging, including that of the immune system. Therefore, immunotherapeutics including vaccination and “protective autoimmunity” provide promising means to rejuvenate neuro-inflammatory disorders and repair CNS acute injury and chronic neuro-degeneration. We review the current understanding and recent discoveries linking the aging immune system with CNS injury and neuro-degeneration. Additionally, we discuss potential recovery and rejuvenation strategies, focusing on targeting the aging T cell immune system in an effort to alleviate acute brain injury and chronic neuro-degeneration during aging, via the “thymus-inflammaging-neurodegeneration axis”.
Collapse
Affiliation(s)
- Brandon Coder
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Weikan Wang
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang, P. R. China
| | - Liefeng Wang
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA.,Department of Biotechnology, Gannan Medical University, Ganzhou, P. R. China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P. R. China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disease Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang, P. R. China
| | - Dong-Ming Su
- Institute of Molecular Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
13
|
Kuang X, Zhou HJ, Thorne AH, Chen XN, Li LJ, Du JR. Neuroprotective Effect of Ligustilide through Induction of α-Secretase Processing of Both APP and Klotho in a Mouse Model of Alzheimer's Disease. Front Aging Neurosci 2017; 9:353. [PMID: 29163135 PMCID: PMC5673635 DOI: 10.3389/fnagi.2017.00353] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that alpha-processing single transmembrane proteins, amyloid precursor protein (APP) and anti-aging protein Klotho, are likely to be involved in the progression of Alzheimer's disease (AD). The natural phthalide Ligustilide (LIG) has been demonstrated to protect against aging- and amyloid-β (Aβ)-induced brain dysfunction in animal models. The present study is to investigate the effects of LIG on cognitive deficits and metabolism of both APP and Klotho and its underlying mechanism in AD double-transgenic (APP/PS1) mice and cultured human cells. Our results show that treatment with LIG significantly ameliorated memory impairment and Aβ levels and plaques burden. Specifically, LIG might act as a potent enhancer of α-secretase, disintegrin, and metalloprotease 10 (ADAM10), leading to upregulation of alpha-processing of both APP and Klotho and subsequent increases in the levels of both soluble APP fragment (sAPPα) and soluble Klotho (sKL) with inhibition of IGF-1/Akt/mTOR signaling in AD mice and cultured cells. Moreover, the specific ADAM10 inhibitor (G1254023X) effectively reversed LIG-induced alpha-processing of both APP and Klotho in vitro, while Klotho gene knockdown by small interfering RNA significantly blunted LIG-mediated inhibition of IGF-1/Akt/mTOR signaling in vitro. Taken together with the reported neuroprotective effects of both sAPPα and sKL as well as autophagy induction by Akt/mTOR pathway inhibition, our findings suggest that neuroprotection of LIG against AD is associated with induction alpha-processing of APP and Klotho and potential Aβ clearance. Whether LIG might induce Aβ autophagic clearance and the underlying mechanisms need to be further studied.
Collapse
Affiliation(s)
- Xi Kuang
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hong-Jing Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Amy H. Thorne
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, United States
| | - Xi-Nan Chen
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Lin-Jiao Li
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun-Rong Du
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Jun-Rong Du,
| |
Collapse
|
14
|
de la Monte SM. Insulin Resistance and Neurodegeneration: Progress Towards the Development of New Therapeutics for Alzheimer's Disease. Drugs 2017; 77:47-65. [PMID: 27988872 PMCID: PMC5575843 DOI: 10.1007/s40265-016-0674-0] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) should be regarded as a degenerative metabolic disease caused by brain insulin resistance and deficiency, and overlapping with the molecular, biochemical, pathophysiological, and metabolic dysfunctions in diabetes mellitus, non-alcoholic fatty liver disease, and metabolic syndrome. Although most of the diagnostic and therapeutic approaches over the past several decades have focused on amyloid-beta (Aβ42) and aberrantly phosphorylated tau, which could be caused by consequences of brain insulin resistance, the broader array of pathologies including white matter atrophy with loss of myelinated fibrils and leukoaraiosis, non-Aβ42 microvascular disease, dysregulated lipid metabolism, mitochondrial dysfunction, astrocytic gliosis, neuro-inflammation, and loss of synapses vis-à-vis growth of dystrophic neurites, is not readily accounted for by Aβ42 accumulations, but could be explained by dysregulated insulin/IGF-1 signaling with attendant impairments in signal transduction and gene expression. This review covers the diverse range of brain abnormalities in AD and discusses how insulins, incretins, and insulin sensitizers could be utilized to treat at different stages of neurodegeneration.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Pierre Galletti Research Building, 55 Claverick Street, Room 419, Providence, RI, 02903, USA.
- Department of Neurosurgery, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Neuropathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
- Department of Pathology, Rhode Island Hospital, and the Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
15
|
Zhang ZX, Zhao RP, Wang DS, Wang AN. Fuzhisan ameliorates Aβ production and tau phosphorylation in hippocampal of 11 month old APP/PS1 transgenic mice: A Western blot study. Exp Gerontol 2016; 84:88-95. [DOI: 10.1016/j.exger.2016.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 08/24/2016] [Accepted: 09/05/2016] [Indexed: 12/30/2022]
|
16
|
Abstract
Nonhuman animal (“animal”) experimentation is typically defended by arguments that it is reliable, that animals provide sufficiently good models of human biology and diseases to yield relevant information, and that, consequently, its use provides major human health benefits. I demonstrate that a growing body of scientific literature critically assessing the validity of animal experimentation generally (and animal modeling specifically) raises important concerns about its reliability and predictive value for human outcomes and for understanding human physiology. The unreliability of animal experimentation across a wide range of areas undermines scientific arguments in favor of the practice. Additionally, I show how animal experimentation often significantly harms humans through misleading safety studies, potential abandonment of effective therapeutics, and direction of resources away from more effective testing methods. The resulting evidence suggests that the collective harms and costs to humans from animal experimentation outweigh potential benefits and that resources would be better invested in developing human-based testing methods.
Collapse
|
17
|
Banik A, Brown RE, Bamburg J, Lahiri DK, Khurana D, Friedland RP, Chen W, Ding Y, Mudher A, Padjen AL, Mukaetova-Ladinska E, Ihara M, Srivastava S, Padma Srivastava MV, Masters CL, Kalaria RN, Anand A. Translation of Pre-Clinical Studies into Successful Clinical Trials for Alzheimer's Disease: What are the Roadblocks and How Can They Be Overcome? J Alzheimers Dis 2016; 47:815-43. [PMID: 26401762 DOI: 10.3233/jad-150136] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Preclinical studies are essential for translation to disease treatments and effective use in clinical practice. An undue emphasis on single approaches to Alzheimer's disease (AD) appears to have retarded the pace of translation in the field, and there is much frustration in the public about the lack of an effective treatment. We critically reviewed past literature (1990-2014), analyzed numerous data, and discussed key issues at a consensus conference on Brain Ageing and Dementia to identify and overcome roadblocks in studies intended for translation. We highlight various factors that influence the translation of preclinical research and highlight specific preclinical strategies that have failed to demonstrate efficacy in clinical trials. The field has been hindered by the domination of the amyloid hypothesis in AD pathogenesis while the causative pathways in disease pathology are widely considered to be multifactorial. Understanding the causative events and mechanisms in the pathogenesis are equally important for translation. Greater efforts are necessary to fill in the gaps and overcome a variety of confounds in the generation, study design, testing, and evaluation of animal models and the application to future novel anti-dementia drug trials. A greater variety of potential disease mechanisms must be entertained to enhance progress.
Collapse
Affiliation(s)
- Avijit Banik
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - James Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Debomoy K Lahiri
- Departments of Psychiatry and of Medical & Molecular Genetics, Indiana University School of Medicine, Neuroscience Research Center, Indianapolis, IN, USA
| | - Dheeraj Khurana
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Robert P Friedland
- Department of Neurology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Wei Chen
- Division of Pulmonary Medicine, Allergy and Immunology, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh, 318C Parran Hall, Pittsburgh, PA, USA
| | - Amritpal Mudher
- Southampton Neurosciences Group, University of Southampton, Southampton, UK
| | - Ante L Padjen
- Department of Pharmacology & Therapeutics, McGill University, Montreal, QC, Canada
| | - Elizabeta Mukaetova-Ladinska
- Institute of Neuroscience, Newcastle University, NIHR Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Sudhir Srivastava
- Division of Toxicology, Central Drug Research Institute, Lucknow, India
| | - M V Padma Srivastava
- Department of Neurology, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Colin L Masters
- Mental Health Research Institute, University of Melbourne, Royal Parade, The VIC, Australia
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, NIHR Biomedical Research Building, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
18
|
Li Y, Yuan X, Shen Y, Zhao J, Yue R, Liu F, He W, Wang R, Shan L, Zhang W. Bacopaside I ameliorates cognitive impairment in APP/PS1 mice via immune-mediated clearance of β-amyloid. Aging (Albany NY) 2016; 8:521-33. [PMID: 26946062 PMCID: PMC4833143 DOI: 10.18632/aging.100913] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/20/2016] [Indexed: 06/05/2023]
Abstract
Standardized extracts of Bacopa monniera (BME) have been shown to exert a neuroprotective effect against mental diseases, such as depression, anxiety and Alzheimer's disease (AD), in chronic administration studies. However, its mechanism of action has remained unclear. In this study, we evaluated the therapeutic effect of Bacopaside I (BS-I), a major triterpenoid saponin of BME, on the cognitive impairment and neuropathology in APP/PS1 transgenic mice and explored the possible mechanism from a biological systems perspective. We found that BS-I treatment significantly ameliorated learning deficits, improved long-term spatial memory, and reduced plaque load in APP/PS1 mice. We constructed BS-I's therapeutic effect network by mapping the nodes onto the protein-protein interaction (PPI) network constructed according to their functional categories based on genomic and proteomic data. Because many of the top enrichment categories related to the processes of the immune system and phagocytosis were detected, we proposed that BS-I promotes amyloid clearance via the induction of a suitable degree of innate immune stimulation and phagocytosis. Our research may help to clarify the neuroprotective effect of BME and indicated that natural saponins target the immune system, which may offer new research avenues to discover novel treatments for AD.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Xing Yuan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Yunheng Shen
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Jing Zhao
- Department of Mathematics, Logistical Engineering University, Chongqing 401311, China
| | - Rongcai Yue
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Fang Liu
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Weiwei He
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Rui Wang
- School of Pharmacy, East China University of Science and Technology, Shanghai 200237, P.R. China
| | - Lei Shan
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
- Shanghai Institute of Pharmaceutical Industry, Shanghai 200040, P.R. China
| |
Collapse
|
19
|
Ritter A, Cummings J. Fluid Biomarkers in Clinical Trials of Alzheimer's Disease Therapeutics. Front Neurol 2015; 6:186. [PMID: 26379620 PMCID: PMC4553391 DOI: 10.3389/fneur.2015.00186] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/10/2015] [Indexed: 02/02/2023] Open
Abstract
With the demographic shift of the global population toward longer life expectancy, the number of people living with Alzheimer’s disease (AD) has rapidly expanded and is projected to triple by the year 2050. Current treatments provide symptomatic relief but do not affect the underlying pathology of the disease. Therapies that prevent or slow the progression of the disease are urgently needed to avoid this growing public health emergency. Insights gained from decades of research have begun to unlock the pathophysiology of this complex disease and have provided targets for disease-modifying therapies. In the last decade, few therapeutic agents designed to modify the underlying disease process have progressed to clinical trials and none have been brought to market. With the focus on disease modification, biomarkers promise to play an increasingly important role in clinical trials. Six biomarkers have now been included in diagnostic criteria for AD and are regularly incorporated into clinical trials. Three biomarkers are neuroimaging measures – hippocampal atrophy measured by magnetic resonance imaging (MRI), amyloid uptake as measured by Pittsburg compound B positron emission tomography (PiB-PET), and decreased fluorodeoxyglucose (18F) uptake as measured by PET (FDG-PET) – and three are sampled from fluid sources – cerebrospinal fluid levels of amyloid β42 (Aβ42), total tau, and phosphorylated tau. Fluid biomarkers are important because they can provide information regarding the underlying biochemical processes that are occurring in the brain. The purpose of this paper is to review the literature regarding the existing and emerging fluid biomarkers and to examine how fluid biomarkers have been incorporated into clinical trials.
Collapse
Affiliation(s)
- Aaron Ritter
- Cleveland Clinic Lou Ruvo Center for Brain Health , Las Vegas, NV , USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health , Las Vegas, NV , USA
| |
Collapse
|
20
|
Marciani DJ. Alzheimer's disease vaccine development: A new strategy focusing on immune modulation. J Neuroimmunol 2015; 287:54-63. [PMID: 26439962 DOI: 10.1016/j.jneuroim.2015.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/31/2022]
Abstract
Despite significant advances in the development of Alzheimer's disease (AD) vaccines effective in animal models, these prototypes have been clinically unsuccessful; apparently the result of using immunogens modified to prevent inflammation. Hence, a new paradigm is needed that uses entire AD-associated immunogens, a notion supported by recent successful passive immunotherapy results, with adjuvants that induce Th2-only while inhibiting without abrogating Th1 immunity. Here, we discuss the obstacles to AD vaccine development and Th2-adjuvants that by acting on dendritic and T cells, would elicit regardless of the antigen a safe and effective antibody response, while preventing damaging neuroinflammation and ameliorating immunosenescence.
Collapse
Affiliation(s)
- Dante J Marciani
- Qantu Therapeutics, Inc., 612 E. Main Street, Lewisville, TX 75057, USA.
| |
Collapse
|
21
|
Autophagy is involved in oral rAAV/Aβ vaccine-induced Aβ clearance in APP/PS1 transgenic mice. Neurosci Bull 2015; 31:491-504. [PMID: 26254061 DOI: 10.1007/s12264-015-1546-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/14/2015] [Indexed: 12/17/2022] Open
Abstract
The imbalance between ß-amyloid (Aß) generation and clearance plays a fundamental role in the pathogenesis of Alzheimer's disease (AD). The sporadic form of AD is characterized by an overall impairment in Aß clearance. Immunotherapy targeting Aß clearance is believed to be a promising approach and is under active clinical investigation. Autophagy is a conserved pathway for degrading abnormal protein aggregates and is crucial for Aß clearance. We previously reported that oral vaccination with a recombinant AAV/Aß vaccine increased the clearance of Aß from the brain and improved cognitive ability in AD animal models, while the underlying mechanisms were not well understood. In this study, we first demonstrated that oral vaccination with rAAV/Aß decreased the p62 level and up-regulated the LC3B-II/LC3B-I ratio in APP/PS1 mouse brain, suggesting enhanced autophagy. Further, inhibition of the Akt/mTOR pathway may account for autophagy enhancement. We also found increased anti-Aß antibodies in the sera of APP/PS1 mice with oral vaccination, accompanied by elevation of complement factors C1q and C3 levels in the brain. Our results indicate that autophagy is closely involved in oral vaccination-induced Aß clearance, and modulating the autophagy pathway may be an important strategy for AD prevention and intervention.
Collapse
|
22
|
|
23
|
Portacolone E, Berridge C, K Johnson J, Schicktanz S. Time to reinvent the science of dementia: the need for care and social integration. Aging Ment Health 2014; 18:269-75. [PMID: 24180580 DOI: 10.1080/13607863.2013.837149] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVES The increasing number of older adults with dementia is a large and growing public health problem. Alzheimer's disease, the prevailing form of dementia, is projected to quadruple worldwide. To date, the care and social integration of individuals with dementia is complicated by limited collaborations between biomedicine and other disciplines. The objective of this paper is therefore to reflect on the orientation of biomedicine with regard to the science of dementia, and to articulate a path for moving forward. METHODS The authors drew upon, and expanded, the insights of an interdisciplinary, international workshop entitled 'Bioethics and the Science of Aging: The Case of Dementia' held in October 2012 at the University of California in Berkeley. RESULTS The care of individuals with dementia compels solid interdisciplinary collaborations. There are several issues affecting the care of individuals with dementia: (1) an evolving definition of dementia; (2) the ambiguous benefits of the diagnosis of dementia; (3) ethical conflicts concerning consent processes and clinical trials; and (4) a limited understanding of the perspective of the person with dementia. CONCLUSION We argue that it is time for a renewed dialogue between biomedicine and other disciplines -- particularly public health, the social sciences, the medical humanities and bioethics. This interdisciplinary dialogue would facilitate a process of self-reflection within biomedicine. This dialogue will also provide the foundation for equitable public health interventions and will further prioritize the values and preferences of individuals with dementia, as well as their care and social integration.
Collapse
Affiliation(s)
- Elena Portacolone
- a Institute for Health and Aging , University of California in San Francisco , CA , USA
| | | | | | | |
Collapse
|
24
|
Jiang T, Guo W, Sha S, Xing X, Guo R, Cao Y. Nasal mucosal inhalation of amyloid-beta peptide 3-10 defective adenovirus attenuates cytotoxicity induced by beta-amyloid (1-42). Neural Regen Res 2014; 9:872-7. [PMID: 25206904 PMCID: PMC4146261 DOI: 10.4103/1673-5374.131605] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2014] [Indexed: 11/06/2022] Open
Abstract
Three-month-old Alzheimer's disease model transgenic mice were immunized with Aβ1–42 Plp-Adenovirus [Ad]-X-CMV-(Aβ3–10)10-CpG [AdCpG-(Aβ3–10)10] or AdCpG virus fluid via nasal mucosal inhalation, respectively. ELISA analysis of serum showed Aβ42 antibody titers were significantly increased in mice immunized with Aβ1–42 and AdCpG-(Aβ3–10)10. Concanavalin A and AdCpG-(Aβ3–10)10 stimulation significantly increased the number of proliferating spleen cells cultured from AdCpG(Aβ3–10)10 and Aβ42 groups compared with the control group. In the AdCpG(Aβ3–10)10 group, levels of interleukin (IL)-4 and IL-10 were increased, while those of IL-2 and interferon-γ were decreased. In the Aβ42 group, levels of IL-4, IL-10, IL-2 and interferon-γ were all increased. Experimental findings indicate that AdCpG-(Aβ3–10)10 vaccine can produce strong T helper 2 (Th2) humoral immune responses in addition to the production of Aβ42 antibody. The cellular immunologic response was weak and avoided Aβ1–42-mediated cytotoxicity.
Collapse
Affiliation(s)
- Tongzi Jiang
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wanshu Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Sha Sha
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xiaona Xing
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Rong Guo
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yunpeng Cao
- Department of Neurology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
25
|
Abstract
Most infectious diseases are caused by pathogenic infiltrations from the mucosal tract. Therefore, vaccines delivered to the mucosal tissues can mimic natural infections and provide protection at the first site of infection. Thus, mucosal, especially, oral delivery is becoming the most preferred mode of vaccination. However, oral vaccines have to overcome several barriers such as the extremely low pH of the stomach, the presence of proteolytic enzymes and bile salts as well as low permeability in the intestine. Several formulations based on nanoparticle strategies are currently being explored to prepare stable oral vaccine formulations. This review briefly discusses several molecular mechanisms involved in intestinal immune cell activation and various aspects of oral nanoparticle-based vaccine design that should be considered for improved mucosal and systemic immune responses.
Collapse
Affiliation(s)
- Nirmal Marasini
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | | | | |
Collapse
|
26
|
Roy M, Cardoso C, Dorieux O, Malgorn C, Epelbaum S, Petit F, Kraska A, Brouillet E, Delatour B, Perret M, Aujard F, Dhenain M. Age-associated evolution of plasmatic amyloid in mouse lemur primates: relationship with intracellular amyloid deposition. Neurobiol Aging 2014; 36:149-56. [PMID: 25131002 DOI: 10.1016/j.neurobiolaging.2014.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 07/02/2014] [Accepted: 07/12/2014] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder. Amyloid-β peptide (Aβ) deposition in the brain is one of its hallmarks, and the measure of plasma Aβ is considered to be a biomarker for anti-amyloid drug efficacy in animal models of AD. However, age-associated plasmatic Aβ modulation in animal models is practically never addressed in the literature. Mouse lemur primates are used as a model of normal and AD-like cerebral aging. Here, we studied the effect of age on plasmatic Aβ in 58 mouse lemurs aged from 1 to 10 years. A subset of animals presented high plasmatic Aβ, and the proportion of animals with high plasmatic Aβ was higher in aged animals as compared with young ones. Histologic evaluation of the brain of some of these animals was carried out to assess extracellular and intracellular amyloid load. In aged lemurs, plasmatic Aβ was negatively correlated with the density of neurons accumulating deposits of Aβ.
Collapse
Affiliation(s)
- Maggie Roy
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France; Research Center on Aging, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Cécile Cardoso
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France
| | - Olène Dorieux
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France; CNRS UMR 7179, MNHN, Brunoy, France
| | - Carole Malgorn
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France
| | - Stéphane Epelbaum
- Sorbonne Universités, Paris, France; UPMC Univ Paris 06 UMR S 1127, Paris, France; Inserm, U 1127, Paris, France; CNRS UMR 7225, Paris, France; ICM, Paris, France
| | - Fanny Petit
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France
| | - Audrey Kraska
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France
| | - Emmanuel Brouillet
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France
| | - Benoît Delatour
- Sorbonne Universités, Paris, France; UPMC Univ Paris 06 UMR S 1127, Paris, France; Inserm, U 1127, Paris, France; CNRS UMR 7225, Paris, France; ICM, Paris, France
| | | | | | - Marc Dhenain
- CEA, DSV, I2BM, MIRCen, URA CEA CNRS 2210, Fontenay aux Roses, France; CNRS, URA 2210, Fontenay aux Roses, France.
| |
Collapse
|
27
|
Focusing the amyloid cascade hypothesis on N-truncated Abeta peptides as drug targets against Alzheimer's disease. Acta Neuropathol 2014; 127:787-801. [PMID: 24803226 PMCID: PMC4024135 DOI: 10.1007/s00401-014-1287-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 02/07/2023]
Abstract
Although N-truncated Aβ variants are known to be the main constituent of amyloid plaques in the brains of patients with Alzheimer’s disease, their potential as targets for pharmacological intervention has only recently been investigated. In the last few years, the Alzheimer field has experienced a paradigm shift with the ever increasing understanding that targeting amyloid plaques has not led to a successful immunotherapy. On the other hand, there can be no doubt that the amyloid cascade hypothesis is central to the etiology of Alzheimer’s disease, raising the question as to why it is apparently failing to translate into the clinic. In this review, we aim to refocus the amyloid hypothesis integrating N-truncated Aβ peptides based on mounting evidence that they may represent better targets than full-length Aβ. In addition to Aβ peptides starting with an Asp at position 1, a variety of different N-truncated Aβ peptides have been identified starting with amino residue Ala-2, pyroglutamylated Glu-3, Phe-4, Arg-5, His-6, Asp-7, Ser-8, Gly-9, Tyr-10 and pyroglutamylated Glu-11. Certain forms of N-truncated species are better correlates for early pathological changes found pre-symptomatically more often than others. There is also evidence that, together with full-length Aβ, they might be physiologically detectable and are naturally secreted by neurons. Others are known to form soluble aggregates, which have neurotoxic properties in transgenic mouse models. It has been clearly demonstrated by several groups that some N-truncated Aβs dominate full-length Aβ in the brains of Alzheimer’s patients. We try to address which of the N-truncated variants may be promising therapeutic targets and which enzymes might be involved in the generation of these peptides
Collapse
|
28
|
Wyse RD, Dunbar GL, Rossignol J. Use of genetically modified mesenchymal stem cells to treat neurodegenerative diseases. Int J Mol Sci 2014; 15:1719-45. [PMID: 24463293 PMCID: PMC3958818 DOI: 10.3390/ijms15021719] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 12/18/2013] [Accepted: 01/14/2014] [Indexed: 01/01/2023] Open
Abstract
The transplantation of mesenchymal stem cells (MSCs) for treating neurodegenerative disorders has received growing attention recently because these cells are readily available, easily expanded in culture, and when transplanted, survive for relatively long periods of time. Given that such transplants have been shown to be safe in a variety of applications, in addition to recent findings that MSCs have useful immunomodulatory and chemotactic properties, the use of these cells as vehicles for delivering or producing beneficial proteins for therapeutic purposes has been the focus of several labs. In our lab, the use of genetic modified MSCs to release neurotrophic factors for the treatment of neurodegenerative diseases is of particular interest. Specifically, glial cell-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain derived neurotrophic factor (BDNF) have been recognized as therapeutic trophic factors for Parkinson's, Alzheimer's and Huntington's diseases, respectively. The aim of this literature review is to provide insights into: (1) the inherent properties of MSCs as a platform for neurotrophic factor delivery; (2) the molecular tools available for genetic manipulation of MSCs; (3) the rationale for utilizing various neurotrophic factors for particular neurodegenerative diseases; and (4) the clinical challenges of utilizing genetically modified MSCs.
Collapse
Affiliation(s)
- Robert D Wyse
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Gary L Dunbar
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| | - Julien Rossignol
- Field Neurosciences Institute Laboratory for Restorative Neurology, Brain Research and Integrative Neuroscience Center, Program in Neuroscience, Central Michigan University, Mount Pleasant, MI 48859, USA.
| |
Collapse
|
29
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
30
|
Bao XQ, Li N, Wang T, Kong XC, Tai WJ, Sun H, Zhang D. FLZ alleviates the memory deficits in transgenic mouse model of Alzheimer's disease via decreasing beta-amyloid production and tau hyperphosphorylation. PLoS One 2013; 8:e78033. [PMID: 24223757 PMCID: PMC3817172 DOI: 10.1371/journal.pone.0078033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/06/2013] [Indexed: 01/15/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia worldwide and mainly characterized by the aggregated β-amyloid (Aβ) and hyperphosphorylated tau. FLZ is a novel synthetic derivative of natural squamosamide and has been proved to improve memory deficits in dementia animal models. In this study, we aimed to investigate the mechanisms of FLZ’s neuroprotective effect in APP/PS1 double transgenic mice and SH-SY5Y (APPwt/swe) cells. The results showed that treatment with FLZ significantly improved the memory deficits of APP/PS1 transgenic mice and decreased apoptosis of SH-SY5Y (APPwt/swe) cells. FLZ markedly attenuated Aβ accumulation and tau phosphorylation both in vivo and in vitro. Mechanistic study showed that FLZ interfered APP processing, i.e., FLZ decreased β-amyloid precursor protein (APP) phosphorylation, APP-carboxy-terminal fragment (APP-CTF) production and β-amyloid precursor protein cleaving enzyme 1 (BACE1) expression. These results indicated that FLZ reduced Aβ production through inhibiting amyloidogenic pathway. The mechanistic study about FLZ’s inhibitory effect on tau phosphorylation revealed t the involvement of Akt/glycogen synthase kinase 3β (GSK3β) pathway. FLZ treatment increased Akt activity and inhibited GSK3β activity both in vivo and in vitro. The inhibitory effect of FLZ on GSK3β activity and tau phosphorylation was suppressed by inhibiting Akt activity, indicating that Akt/GSK3β pathway might be the possible mechanism involved in the inhibitory effect of FLZ on tau hyperphosphorylation. These results suggested FLZ might be a potential anti-AD drug as it not only reduced Aβ production via inhibition amyloidogenic APP processing pathway, but also attenuated tau hyperphosphoylation mediated by Akt/GSK3β.
Collapse
Affiliation(s)
- Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tao Wang
- Beijing Municipal Corps Hospital of Chinese People’s Armed Police Force, Beijing, China
| | | | - Wen-Jiao Tai
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hua Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail:
| |
Collapse
|
31
|
Klementieva O, Aso E, Filippini D, Benseny-Cases N, Carmona M, Juvés S, Appelhans D, Cladera J, Ferrer I. Effect of Poly(propylene imine) Glycodendrimers on β-Amyloid Aggregation in Vitro and in APP/PS1 Transgenic Mice, as a Model of Brain Amyloid Deposition and Alzheimer’s Disease. Biomacromolecules 2013; 14:3570-80. [DOI: 10.1021/bm400948z] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- O. Klementieva
- Institute of Neuropathology,
Pathologic Anatomy Service, IDIBELL-University Hospital Bellvitge, Feixa Llarga
sn, 08907 L’Hospitalet
de Llobregat, Spain
| | - E. Aso
- Institute of Neuropathology,
Pathologic Anatomy Service, IDIBELL-University Hospital Bellvitge, Feixa Llarga
sn, 08907 L’Hospitalet
de Llobregat, Spain
| | - D. Filippini
- Leibniz Institute of Polymer Research Dresden, Hohe Strasse 6, D-01069 Dresden, Germany
| | - N. Benseny-Cases
- Polygone
Scientifique Louis Néel, ESRF, 6 rue Jules Horowitz, 38000, Grenoble, France
| | - M. Carmona
- Institute of Neuropathology,
Pathologic Anatomy Service, IDIBELL-University Hospital Bellvitge, Feixa Llarga
sn, 08907 L’Hospitalet
de Llobregat, Spain
| | - S. Juvés
- Institute of Neuropathology,
Pathologic Anatomy Service, IDIBELL-University Hospital Bellvitge, Feixa Llarga
sn, 08907 L’Hospitalet
de Llobregat, Spain
| | - D. Appelhans
- Leibniz Institute of Polymer Research Dresden, Hohe Strasse 6, D-01069 Dresden, Germany
| | - J. Cladera
- Biophysics Unit
and Center of Studies in Biophysics, Department of Biochemistry
and Molecular Biology, Universitat Autònoma de Barcelona (UAB), 08193 Bellaterra, Spain
| | - I. Ferrer
- Institute of Neuropathology,
Pathologic Anatomy Service, IDIBELL-University Hospital Bellvitge, Feixa Llarga
sn, 08907 L’Hospitalet
de Llobregat, Spain
- Department
of Pathology and Experimental Therapy, School of Medicine, University of Barcelona, L’Hospitalet
de Llobregat, Spain
- CIBERNED, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Camboni M, Wang CM, Miranda C, Yoon JH, Xu R, Zygmunt D, Kaspar BK, Martin PT. Active and passive immunization strategies based on the SDPM1 peptide demonstrate pre-clinical efficacy in the APPswePSEN1dE9 mouse model for Alzheimer's disease. Neurobiol Dis 2013; 62:31-43. [PMID: 24021662 DOI: 10.1016/j.nbd.2013.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 07/18/2013] [Accepted: 09/01/2013] [Indexed: 12/31/2022] Open
Abstract
Recent clinical and pre-clinical studies suggest that both active and passive immunization strategies targeting Aβ amyloid may have clinical benefit in Alzheimer's disease. Here, we demonstrate that vaccination of APPswePSEN1dE9 mice with SDPM1, an engineered non-native Aβ amyloid-specific binding peptide, lowers brain Aβ amyloid plaque burden and brain Aβ1-40 and Aβ1-42 peptide levels, improves cognitive learning and memory in Morris water maze tests and increases the expression of synaptic brain proteins. This was the case in young mice immunized prior to development of significant brain amyloid burden, and in older mice, where brain amyloid was already present. Active immunization was optimized using ALUM as an adjuvant to stimulate production of anti-SDPM1 and anti-Aβ amyloid antibodies. Intracerebral injection of P4D6, an SDPM1 peptide-mimotope antibody, also lowered brain amyloid plaque burden in APPswePSEN1dE9 mice. Additionally, P4D6 inhibited Aβ amyloid-mediated toxicity in cultured neuronal cells. The protein sequence of the variable domain within the P4D6 heavy chain was found to mimic a multimer of the SDPM1 peptide motif. These data demonstrate the efficacy of active and passive vaccine strategies to target Aβ amyloid oligomers using an engineered peptide-mimotope strategy.
Collapse
Affiliation(s)
- Marybeth Camboni
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Chiou-Miin Wang
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Carlos Miranda
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Jung Hae Yoon
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Rui Xu
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Deborah Zygmunt
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA
| | - Brian K Kaspar
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Neuroscience, The Ohio State University, USA
| | - Paul T Martin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, USA; Department of Pediatrics, The Ohio State University, USA; Department of Physiology and Cell Biology, The Ohio State University, USA.
| |
Collapse
|
33
|
Immunogenicity, efficacy, safety, and mechanism of action of epitope vaccine (Lu AF20513) for Alzheimer's disease: prelude to a clinical trial. J Neurosci 2013; 33:4923-34. [PMID: 23486963 DOI: 10.1523/jneurosci.4672-12.2013] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The Alzheimer's disease (AD) process is understood to involve the accumulation of amyloid plaques and tau tangles in the brain. However, attempts at targeting the main culprits, neurotoxic Aβ peptides, have thus far proven unsuccessful for improving cognitive function. Recent clinical trials with passively administrated anti-Aβ antibodies failed to slow cognitive decline in mild to moderate AD patients, but suggest that an immunotherapeutic approach could be effective in patients with mild AD. Using an AD mouse model (Tg2576), we tested the immunogenicity (cellular and humoral immune responses) and efficacy (AD-like pathology) of clinical grade Lu AF20513 vaccine. We found that Lu AF20513 induces robust "non-self" T-cell responses and the production of anti-Aβ antibodies that reduce AD-like pathology in the brains of Tg2576 mice without inducing microglial activation and enhancing astrocytosis or cerebral amyloid angiopathy. A single immunization with Lu AF20513 induced strong humoral immunity in mice with preexisting memory T-helper cells. In addition, Lu AF20513 induced strong humoral responses in guinea pigs and monkeys. These data support the translation of Lu AF20513 to the clinical setting with the aims of: (1) inducing therapeutically potent anti-Aβ antibody responses in patients with mild AD, particularly if they have memory T-helper cells generated after immunizations with conventional tetanus toxoid vaccine, and (2) preventing pathological autoreactive T-cell responses.
Collapse
|
34
|
Alving CR, Rao M, Steers NJ, Matyas GR, Mayorov AV. Liposomes containing lipid A: an effective, safe, generic adjuvant system for synthetic vaccines. Expert Rev Vaccines 2012; 11:733-44. [PMID: 22873129 DOI: 10.1586/erv.12.35] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liposomes containing monophosphoryl lipid A (MPLA) have previously exhibited considerable potency and safety in human trials with a variety of candidate vaccines, including vaccines to malaria, HIV-1 and several different types of cancer. The long history of research and development of MPLA and liposomal MPLA as vaccine adjuvants reveals that there are numerous opportunities for creation and development of generic (nonproprietary) adjuvant system formulations with these materials that are not only highly potent and safe, but also readily available as native materials or as synthetic compounds. They are easily manufactured as potentially inexpensive and easy to use adjuvant systems and might be effective even with synthetic peptides as antigens.
Collapse
Affiliation(s)
- Carl R Alving
- Laboratory of Adjuvant and Antigen Research, US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | | | | | | | | |
Collapse
|
35
|
Shuai H, Zhang J, Zhang J, Xie J, Zhang M, Ma J, Zhang L, Wang X. Role of stereotaxically injected IgG from db/db mice in the phosphorylation of the microtubule-associated protein tau in hippocampus. Brain Res 2012; 1486:14-26. [PMID: 23036273 DOI: 10.1016/j.brainres.2012.08.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/21/2012] [Accepted: 08/25/2012] [Indexed: 11/29/2022]
Abstract
People with type 2 diabetes (T2DM) mellitus are high risk for dementia and Alzheimer's disease (AD) via several plausible pathways. However, the underlying mechanisms have been still unclear, and the relation of immune injury to the pathogenesis of T2DM-related AD is not yet completely understood. Our present study aimed to elucidate the possible role of immunoglobulin IgG in the immune process of AD associated with T2DM in db/db mice. Hippocampi of 20 db/db mice and 20 C57BL/6 mice were subjected to immunohistochemistry and immunofluorescence assays. The phosphorylation of tau, glycogen synthase kinase (GSK)-3β and AKT activity was examined by Western blot analysis. IgG purified from the sera of IgG deposit-positive db/db mice was stereotaxically injected into the hippocampi of another 12 db/db mice and 12 C57BL/6 mice. The phosphorylation of tau, Abeta, GSK-3β and AKT activity was analyzed. Compared with the C57BL/6 control, 13 of the 20 db/db mice exhibited high levels of IgG deposits in the hippocampus. Treatment with IgG triggered tau hyperphosphorylations and Abeta deposition, which are likely major factors in AD. Meanwhile, IgG inhibited AKT phosphorylation and promoted GSK-3β activity. The IgG deposits observed in some db/db mice were possibly related to the impairment of T2DM-related AD development. Some autoimmune processes may be involved in AD in type 2 diabetes mellitus development at the level of the hippocampus.
Collapse
Affiliation(s)
- Hongxia Shuai
- Department of Endocrinology, XiangYang Central Hospital, XiangYang, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Proctor CJ, Pienaar IS, Elson JL, Kirkwood TBL. Aggregation, impaired degradation and immunization targeting of amyloid-beta dimers in Alzheimer's disease: a stochastic modelling approach. Mol Neurodegener 2012; 7:32. [PMID: 22748062 PMCID: PMC3583291 DOI: 10.1186/1750-1326-7-32] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 06/11/2012] [Indexed: 01/02/2023] Open
Abstract
Background Alzheimer’s disease (AD) is the most frequently diagnosed neurodegenerative disorder affecting humans, with advanced age being the most prominent risk factor for developing AD. Despite intense research efforts aimed at elucidating the precise molecular underpinnings of AD, a definitive answer is still lacking. In recent years, consensus has grown that dimerisation of the polypeptide amyloid-beta (Aß), particularly Aß42, plays a crucial role in the neuropathology that characterise AD-affected post-mortem brains, including the large-scale accumulation of fibrils, also referred to as senile plaques. This has led to the realistic hope that targeting Aß42 immunotherapeutically could drastically reduce plaque burden in the ageing brain, thus delaying AD onset or symptom progression. Stochastic modelling is a useful tool for increasing understanding of the processes underlying complex systems-affecting disorders such as AD, providing a rapid and inexpensive strategy for testing putative new therapies. In light of the tool’s utility, we developed computer simulation models to examine Aß42 turnover and its aggregation in detail and to test the effect of immunization against Aß dimers. Results Our model demonstrates for the first time that even a slight decrease in the clearance rate of Aß42 monomers is sufficient to increase the chance of dimers forming, which could act as instigators of protofibril and fibril formation, resulting in increased plaque levels. As the process is slow and levels of Aβ are normally low, stochastic effects are important. Our model predicts that reducing the rate of dimerisation leads to a significant reduction in plaque levels and delays onset of plaque formation. The model was used to test the effect of an antibody mediated immunological response. Our results showed that plaque levels were reduced compared to conditions where antibodies are not present. Conclusion Our model supports the current thinking that levels of dimers are important in initiating the aggregation process. Although substantial knowledge exists regarding the process, no therapeutic intervention is on offer that reliably decreases disease burden in AD patients. Computer modelling could serve as one of a number of tools to examine both the validity of reliable biomarkers and aid the discovery of successful intervention strategies.
Collapse
Affiliation(s)
- Carole J Proctor
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom.
| | | | | | | |
Collapse
|
37
|
Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F. New pharmacological strategies for treatment of Alzheimer's disease: focus on disease modifying drugs. Br J Clin Pharmacol 2012; 73:504-17. [PMID: 22035455 DOI: 10.1111/j.1365-2125.2011.04134.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Current approved drug treatments for Alzheimer disease (AD) include cholinesterase inhibitors (donepezil, rivastigmine, galantamine) and the NMDA receptor antagonist memantine. These drugs provide symptomatic relief but poorly affect the progression of the disease. Drug discovery has been directed, in the last 10 years, to develop 'disease modifying drugs' hopefully able to counteract the progression of AD. Because in a chronic, slow progressing pathological process, such as AD, an early start of treatment enhances the chance of success, it is crucial to have biomarkers for early detection of AD-related brain dysfunction, usable before clinical onset. Reliable early biomarkers need therefore to be prospectively tested for predictive accuracy, with specific cut off values validated in clinical practice. Disease modifying drugs developed so far include drugs to reduce β amyloid (Aβ) production, drugs to prevent Aβ aggregation, drugs to promote Aβ clearance, drugs targeting tau phosphorylation and assembly and other approaches. Unfortunately none of these drugs has demonstrated efficacy in phase 3 studies. The failure of clinical trials with disease modifying drugs raises a number of questions, spanning from methodological flaws to fundamental understanding of AD pathophysiology and biology. Recently, new diagnostic criteria applicable to presymptomatic stages of AD have been published. These new criteria may impact on drug development, such that future trials on disease modifying drugs will include populations susceptible to AD, before clinical onset. Specific problems with completed trials and hopes with ongoing trials are discussed in this review.
Collapse
Affiliation(s)
- Salvatore Salomone
- Department of Clinical and Molecular Biomedicine, Section of Pharmacology and Biochemistry Department of Formative Processes, University of Catania, Viale Andrea Doria 6, Catania, Italy
| | | | | | | | | |
Collapse
|
38
|
Consequences of inhibiting amyloid precursor protein processing enzymes on synaptic function and plasticity. Neural Plast 2012; 2012:272374. [PMID: 22792491 PMCID: PMC3390164 DOI: 10.1155/2012/272374] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 04/22/2012] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease, one of whose major pathological hallmarks is the accumulation of amyloid plaques comprised of aggregated β-amyloid (Aβ) peptides. It is now recognized that soluble Aβ oligomers may lead to synaptic dysfunctions early in AD pathology preceding plaque deposition. Aβ is produced by a sequential cleavage of amyloid precursor protein (APP) by the activity of β- and γ-secretases, which have been identified as major candidate therapeutic targets of AD. This paper focuses on how Aβ alters synaptic function and the functional consequences of inhibiting the activity of the two secretases responsible for Aβ generation. Abnormalities in synaptic function resulting from the absence or inhibition of the Aβ-producing enzymes suggest that Aβ itself may have normal physiological functions which are disrupted by abnormal accumulation of Aβ during AD pathology. This interpretation suggests that AD therapeutics targeting the β- and γ-secretases should be developed to restore normal levels of Aβ or combined with measures to circumvent the associated synaptic dysfunction(s) in order to have minimal impact on normal synaptic function.
Collapse
|
39
|
Small DH, Aguilar MI. Targeting pre-mRNA splicing: a BACE-ic strategy for AD drug development? J Neurochem 2012; 121:695-6. [DOI: 10.1111/j.1471-4159.2012.07679.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
de la Monte SM. Contributions of brain insulin resistance and deficiency in amyloid-related neurodegeneration in Alzheimer's disease. Drugs 2012; 72:49-66. [PMID: 22191795 PMCID: PMC4550303 DOI: 10.2165/11597760-000000000-00000] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in North America. Growing evidence supports the concept that AD is fundamentally a metabolic disease that results in progressive impairment in the brain's capacity to utilize glucose and respond to insulin and insulin-like growth factor (IGF) stimulation. Moreover, the heterogeneous nature of AD is only partly explained by the brain's propensity to accumulate aberrantly processed, misfolded and aggregated oligomeric structural proteins, including amyloid-β peptides and hyperphosphorylated tau. Evidence suggests that other factors, including impaired energy metabolism, oxidative stress, neuroinflammation, insulin and IGF resistance, and insulin/IGF deficiency in the brain should be incorporated into an overarching hypothesis to develop more realistic diagnostic and therapeutic approaches to AD. In this review, the interrelationship between impaired insulin and IGF signalling and amyloid-β pathology is discussed along with potential therapeutic approaches. Impairments in brain insulin/IGF signalling lead to increased expression of amyloid-β precursor protein (AβPP) and accumulation of AβPP-Aβ. In addition, they promote oxidative stress and deficits in energy metabolism, leading to the activation of pro-AβPP-Aβ-mediated neurodegeneration cascades. Although brain insulin/IGF resistance and deficiency can be induced by primary or secondary disease processes, the soaring rates of peripheral insulin resistance associated with obesity, diabetes mellitus and metabolic syndrome quite likely play major roles in the current AD epidemic. Both clinical and experimental data have linked chronic hyperinsulinaemia to cognitive impairment and neurodegeneration with increased AβPP-Aβ accumulation/reduced clearance in the CNS. Correspondingly, both the restoration of insulin responsiveness and the use of insulin therapy can lead to improved cognitive performance, although with variable effects on brain AβPP-Aβ load. On the other hand, experimental evidence supports the concept that the toxic effects of AβPP-Aβ can promote insulin resistance. Together, these findings suggest that a positive feedback loop of progressive neurodegeneration can develop whereby insulin resistance drives AβPP-Aβ accumulation, and AβPP-Aβ fibril toxicity drives brain insulin resistance. This phenomenon could explain why measuring AβPP-Aβ levels in cerebrospinal fluid or imaging of the brain has proven to be inadequate as a stand-alone biomarker for diagnosing AD, and why the clinical trial results of anti-AβPP-Aβ monotherapy have been disappointing. Instead, the aggregate data suggest that brain insulin resistance and deficiency must also be therapeutically targeted to halt AD progression or reverse its natural course. The positive therapeutic effects of different treatments that address the role of brain insulin/IGF resistance and deficiency, including the use of intranasal insulin delivery, incretins and insulin sensitizer agents are discussed along with potential benefits of lifestyle changes to modify risk for developing mild cognitive impairment or AD. Altogether, the data strongly support the notion that we must shift toward the implementation of multimodal rather than unimodal diagnostic and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| |
Collapse
|
41
|
Lane RF, Shineman DW, Steele JW, Lee LBH, Fillit HM. Beyond amyloid: the future of therapeutics for Alzheimer's disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 64:213-71. [PMID: 22840749 DOI: 10.1016/b978-0-12-394816-8.00007-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Currently, the field is awaiting the results of several pivotal Phase III clinical Alzheimer's disease (AD) trials that target amyloid-β (Aβ). In light of the recent biomarker studies that indicate Aβ levels are at their most dynamic 5-10 years before the onset of clinical symptoms, it is becoming uncertain whether direct approaches to target Aβ will achieve desired clinical efficacy. AD is a complex neurodegenerative disease caused by dysregulation of numerous neurobiological networks and cellular functions, resulting in synaptic loss, neuronal loss, and ultimately impaired memory. While it is clear that Aβ plays a key role in the pathogenesis of AD, it may be a challenging and inefficient target for mid-to-late stage AD intervention. Throughout the course of AD, multiple pathways become perturbed, presenting a multitude of possible therapeutic avenues for design of AD intervention and prophylactic therapies. In this chapter, we sought to first provide an overview of Aβ-directed strategies that are currently in development, and the pivotal Aβ-targeted trials that are currently underway. Next, we delve into the biology and therapeutic designs associated with other key areas of research in the field including tau, protein trafficking and degradation pathways, ApoE, synaptic function, neurotrophic/neuroprotective strategies, and inflammation and energy utilization. For each area we have provided a comprehensive and balanced overview of the therapeutic strategies currently in preclinical and clinical development, which will shape the future therapeutic landscape of AD.
Collapse
Affiliation(s)
- Rachel F Lane
- Alzheimer's Drug Discovery Foundation, New York, NY, USA
| | | | | | | | | |
Collapse
|
42
|
Lynch MA, Mills KHG. Immunology meets neuroscience--opportunities for immune intervention in neurodegenerative diseases. Brain Behav Immun 2012; 26:1-10. [PMID: 21664452 DOI: 10.1016/j.bbi.2011.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 05/05/2011] [Accepted: 05/24/2011] [Indexed: 12/18/2022] Open
Abstract
Neuroinflammatory changes are characteristic of many, if not all, neurodegenerative diseases but the extent to which the immune system is involved in the pathogenesis of these diseases is unclear. The findings of several studies during the past decade has established that there is a well-developed communication between the central nervous system (CNS) and the peripheral immune system, but also has revealed that the immune system in the CNS is much more sophisticated that previously acknowledged. In this mini-review, we discuss two major neurodegenerative disorders, Alzheimer's disease (AD) and multiple sclerosis (MS), and consider whether the therapies most likely to succeed are those that are identified by studying the marriage of neuroscience and immunology.
Collapse
Affiliation(s)
- Marina A Lynch
- Trinity Institute for Neuroscience, Trinity College, Dublin, Ireland.
| | | |
Collapse
|
43
|
de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012. [PMID: 22201977 DOI: 10.2741/482] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growing evidence supports roles for brain insulin and insulin-like growth factor (IGF) resistance and metabolic dysfunction in the pathogenesis of Alzheimer's disease (AD). Whether the underlying problem stems from a primary disorder of central nervous system (CNS) neurons and glia, or secondary effects of systemic diseases such as obesity, Type 2 diabetes, or metabolic syndrome, the end-results include impaired glucose utilization, mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and the propagation of cascades that result in the accumulation of neurotoxic misfolded, aggregated, and ubiquitinated fibrillar proteins. This article reviews the roles of impaired insulin and IGF signaling to AD-associated neuronal loss, synaptic disconnection, tau hyperphosphorylation, amyloid-beta accumulation, and impaired energy metabolism, and discusses therapeutic strategies and lifestyle approaches that could be used to prevent, delay the onset, or reduce the severity of AD. Finally, it is critical to recognize that AD is heterogeneous and has a clinical course that fully develops over a period of several decades. Therefore, early and multi-modal preventive and treatment approaches should be regarded as essential.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
44
|
de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012; 4:1582-1605. [PMID: 22201977 PMCID: PMC4550311 DOI: 10.2741/e482] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Growing evidence supports roles for brain insulin and insulin-like growth factor (IGF) resistance and metabolic dysfunction in the pathogenesis of Alzheimer's disease (AD). Whether the underlying problem stems from a primary disorder of central nervous system (CNS) neurons and glia, or secondary effects of systemic diseases such as obesity, Type 2 diabetes, or metabolic syndrome, the end-results include impaired glucose utilization, mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and the propagation of cascades that result in the accumulation of neurotoxic misfolded, aggregated, and ubiquitinated fibrillar proteins. This article reviews the roles of impaired insulin and IGF signaling to AD-associated neuronal loss, synaptic disconnection, tau hyperphosphorylation, amyloid-beta accumulation, and impaired energy metabolism, and discusses therapeutic strategies and lifestyle approaches that could be used to prevent, delay the onset, or reduce the severity of AD. Finally, it is critical to recognize that AD is heterogeneous and has a clinical course that fully develops over a period of several decades. Therefore, early and multi-modal preventive and treatment approaches should be regarded as essential.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
45
|
Jeon S, Kang JH, Pak SC, Koo BS. Study on Protection Against β-Amyloid Peptide Toxicity With Oral Administration of Medicinal Herbs. J Evid Based Complementary Altern Med 2011. [DOI: 10.1177/2156587211429463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Seonghyangjeongkisan has been used as a therapeutic agent for cerebral disease in Korea, but its effectiveness in Alzheimer’s disease is not well known. In this study, we examined whether Seonghyangjeongkisan could protect against amyloid β–induced cytotoxicity in neuroblastoma cells and the brain. Seonghyangjeongkisan rescued amyloid β–induced cytotoxicity dose dependently and reduced amyloid β–induced apoptosis and reactive oxygen species. Injection of mice with amyloid β impaired performance on the passive avoidance task, but Seonghyangjeongkisan markedly improved memory impairment in mice, with it being more effective than tacrine treatment in mice. Moreover, the activation of stress-related kinases such as extracellular signal-regulated kinase, c-Jun NH2-terminal kinase, and p38 was suppressed, and the phosphorylation of τ protein, which is known as a marker of Alzheimer’s disease, was also suppressed by Seonghyangjeongkisan treatment in the hippocampus. These results demonstrate that Seonghyangjeongkisan reduces amyloid β-induced toxicity in the brain, suggesting that it may be a useful complementary therapy against Alzheimer’s disease.
Collapse
Affiliation(s)
- Songhee Jeon
- Dongguk University Research Institute of Biotechnology, Seoul, Republic of Korea
| | | | | | | |
Collapse
|
46
|
Jawhar S, Wirths O, Bayer TA. Pyroglutamate amyloid-β (Aβ): a hatchet man in Alzheimer disease. J Biol Chem 2011; 286:38825-32. [PMID: 21965666 DOI: 10.1074/jbc.r111.288308] [Citation(s) in RCA: 171] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pyroglutamate-modified amyloid-β (Aβ(pE3)) peptides are gaining considerable attention as potential key participants in the pathology of Alzheimer disease (AD) due to their abundance in AD brain, high aggregation propensity, stability, and cellular toxicity. Transgenic mice that produce high levels of Aβ(pE3-42) show severe neuron loss. Recent in vitro and in vivo experiments have proven that the enzyme glutaminyl cyclase catalyzes the formation of Aβ(pE3). In this minireview, we summarize the current knowledge on Aβ(pE3), discussing its discovery, biochemical properties, molecular events determining formation, prevalence in the brains of AD patients, Alzheimer mouse models, and potential as a target for therapy and as a diagnostic marker.
Collapse
Affiliation(s)
- Sadim Jawhar
- Department of Molecular Psychiatry, Georg-August-University Göttingen, University Medicine Göttingen, 37075 Göttingen, Germany
| | | | | |
Collapse
|
47
|
Ma Y, Li Y, Zong LX, Xing XN, Zhang WG, Cao YP. Improving memory and decreasing cognitive impairment in Tg-APPswe/PSEN1dE9 mice with Aβ3-10 repeat fragment plasmid by reducing Aβ deposition and inflammatory response. Brain Res 2011; 1400:112-24. [DOI: 10.1016/j.brainres.2011.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 05/04/2011] [Accepted: 05/15/2011] [Indexed: 02/06/2023]
|
48
|
Hoogerhout P, Kamphuis W, Brugghe HF, Sluijs JA, Timmermans HAM, Westdijk J, Zomer G, Boog CJP, Hol EM, van den Dobbelsteen GPJM. A cyclic undecamer peptide mimics a turn in folded Alzheimer amyloid β and elicits antibodies against oligomeric and fibrillar amyloid and plaques. PLoS One 2011; 6:e19110. [PMID: 21526148 PMCID: PMC3079747 DOI: 10.1371/journal.pone.0019110] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 03/17/2011] [Indexed: 01/17/2023] Open
Abstract
The 39- to 42-residue amyloid β (Aβ) peptide is deposited in extracellular fibrillar plaques in the brain of patients suffering from Alzheimer's Disease (AD). Vaccination with these peptides seems to be a promising approach to reduce the plaque load but results in a dominant antibody response directed against the N-terminus. Antibodies against the N-terminus will capture Aβ immediately after normal physiological processing of the amyloid precursor protein and therefore will also reduce the levels of non-misfolded Aβ, which might have a physiologically relevant function. Therefore, we have targeted an immune response on a conformational neo-epitope in misfolded amyloid that is formed in advance of Aβ-aggregation. A tetanus toxoid-conjugate of the 11-meric cyclic peptide Aβ(22-28)-YNGK' elicited specific antibodies in Balb/c mice. These antibodies bound strongly to the homologous cyclic peptide-bovine serum albumin conjugate, but not to the homologous linear peptide-conjugate, as detected in vitro by enzyme-linked immunosorbent assay. The antibodies also bound--although more weakly--to Aβ(1-42) oligomers as well as fibrils in this assay. Finally, the antibodies recognized Aβ deposits in AD mouse and human brain tissue as established by immunohistological staining. We propose that the cyclic peptide conjugate might provide a lead towards a vaccine that could be administered before the onset of AD symptoms. Further investigation of this hypothesis requires immunization of transgenic AD model mice.
Collapse
Affiliation(s)
- Peter Hoogerhout
- Department of Vaccinology, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Jacobson SA, Sabbagh MN. Investigational drugs for the treatment of AD: what can we learn from negative trials? ALZHEIMERS RESEARCH & THERAPY 2011; 3:14. [PMID: 21539725 PMCID: PMC3226276 DOI: 10.1186/alzrt73] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Given the level of interest and activity in the race to find a treatment for Alzheimer's disease, it is expected that a reasonably safe and effective drug will be identified within the next decade. It may be worthwhile to pause periodically during the course of this race to take stock of what we have learned. Over the past few years, a number of trials have been conducted with promising new compounds (including some with novel mechanisms of action) that failed to meet primary endpoints and so were discontinued from clinical development. This article reviews a set of molecules with a range of mechanisms that have been trialed but with negative results. This article also examines the reasons for the negative findings and summarizes some of what we have learned from these experiences.
Collapse
Affiliation(s)
- Sandra A Jacobson
- The Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, 10515 W, Santa Fe Drive, Sun City, AZ 85351, USA.
| | | |
Collapse
|
50
|
Axelsen TV, Holm A, Christiansen G, Birkelund S. Identification of the shortest Aβ-peptide generating highly specific antibodies against the C-terminal end of amyloid-β42. Vaccine 2011; 29:3260-9. [DOI: 10.1016/j.vaccine.2011.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 02/03/2011] [Accepted: 02/09/2011] [Indexed: 10/18/2022]
|