1
|
Lv W, Jiang X, Zhang Y. The role of platelets in the blood-brain barrier during brain pathology. Front Cell Neurosci 2024; 17:1298314. [PMID: 38259501 PMCID: PMC10800710 DOI: 10.3389/fncel.2023.1298314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Platelets play critical roles in maintaining hemostasis. The blood brain barrier (BBB), a significant physical and metabolic barrier, helps maintain physiological stability by limiting transportations between the blood and neural tissues. When the brain undergoes inflammation, tumor, trauma, or bleeding, the platelet responses to help with maintaining BBB homeostasis. In the traditional point of view, activated platelets aggregate to form thrombi which cover the gaps of the blood vessels to protect BBB. However, increasing evidences indicate that platelets may harm BBB by enhancing vascular permeability. Hereby, we reviewed recently published articles with a special focus on the platelet-mediated damage of BBB. Factors released by platelets can induce BBB permeability, which involve platelet-activating factors (PAF), P-selectin, ADP, platelet-derived growth factors (PDGF) superfamily proteins, especially PDGF-AA and PDGF-CC, etc. Platelets can also secrete Amyloid-β (Aβ), which triggers neuroinflammation and downregulates the expression of tight junction molecules such as claudin-5 to damage BBB. Additionally, platelets can form aggregates with neutrophils to release reactive oxygen species (ROS), which can destroy the DNA, proteins, and lipids of endothelial cells (ECs). Moreover, platelets participate in neuroinflammation to affect BBB. Conversely, some of the platelet released factors such as PDGF-BB, protects BBB. In summary, platelets play dual roles in BBB integrity and the related mechanisms are reviewed.
Collapse
Affiliation(s)
| | - Xiaofan Jiang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yanyu Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
2
|
Rudge JD. A New Hypothesis for Alzheimer's Disease: The Lipid Invasion Model. J Alzheimers Dis Rep 2022; 6:129-161. [PMID: 35530118 PMCID: PMC9028744 DOI: 10.3233/adr-210299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023] Open
Abstract
This paper proposes a new hypothesis for Alzheimer's disease (AD)-the lipid invasion model. It argues that AD results from external influx of free fatty acids (FFAs) and lipid-rich lipoproteins into the brain, following disruption of the blood-brain barrier (BBB). The lipid invasion model explains how the influx of albumin-bound FFAs via a disrupted BBB induces bioenergetic changes and oxidative stress, stimulates microglia-driven neuroinflammation, and causes anterograde amnesia. It also explains how the influx of external lipoproteins, which are much larger and more lipid-rich, especially more cholesterol-rich, than those normally present in the brain, causes endosomal-lysosomal abnormalities and overproduction of the peptide amyloid-β (Aβ). This leads to the formation of amyloid plaques and neurofibrillary tangles, the most well-known hallmarks of AD. The lipid invasion model argues that a key role of the BBB is protecting the brain from external lipid access. It shows how the BBB can be damaged by excess Aβ, as well as by most other known risk factors for AD, including aging, apolipoprotein E4 (APOE4), and lifestyle factors such as hypertension, smoking, obesity, diabetes, chronic sleep deprivation, stress, and head injury. The lipid invasion model gives a new rationale for what we already know about AD, explaining its many associated risk factors and neuropathologies, including some that are less well-accounted for in other explanations of AD. It offers new insights and suggests new ways to prevent, detect, and treat this destructive disease and potentially other neurodegenerative diseases.
Collapse
Affiliation(s)
- Jonathan D’Arcy Rudge
- School of Biological Sciences, University of Reading, Reading, Berkshire, United Kingdom
| |
Collapse
|
3
|
Pan Y, Nicolazzo JA. Impact of aging, Alzheimer's disease and Parkinson's disease on the blood-brain barrier transport of therapeutics. Adv Drug Deliv Rev 2018; 135:62-74. [PMID: 29665383 DOI: 10.1016/j.addr.2018.04.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/17/2018] [Accepted: 04/07/2018] [Indexed: 01/01/2023]
Abstract
Older people are at a greater risk of medicine-induced toxicity resulting from either increased drug sensitivity or age-related pharmacokinetic changes. The scenario is further complicated with the two most prevalent age-related neurodegenerative diseases, Alzheimer's disease (AD) and Parkinson's disease (PD). With aging, AD and PD, there is growing evidence of altered structure and function of the blood-brain barrier (BBB), including modifications to tight junctions and efflux transporters, such as P-glycoprotein. The subsequent impact on CNS drug exposure and risk of neurotoxicity from systemically-acting medicines is less well characterized. The purpose of this review, therefore, is to provide an overview of the multiple changes that occur to the BBB as a result of aging, AD and PD, and the impact that such changes have on CNS exposure of drugs, based on studies conducted in aged rodents or rodent models of disease, and in elderly people with and without AD or PD.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
4
|
Lee AY, Hwang BR, Lee MH, Lee S, Cho EJ. Perilla frutescens var. japonica and rosmarinic acid improve amyloid-β25-35 induced impairment of cognition and memory function. Nutr Res Pract 2016; 10:274-81. [PMID: 27247723 PMCID: PMC4880726 DOI: 10.4162/nrp.2016.10.3.274] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/28/2015] [Accepted: 12/04/2015] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND/OBJECTIVES The accumulation of amyloid-β (Aβ) in the brain is a hallmark of Alzheimer's disease (AD) and plays a key role in cognitive dysfunction. Perilla frutescens var. japonica extract (PFE) and its major compound, rosmarinic acid (RA), have shown antioxidant and anti-inflammatory activities. We investigated whether administration of PFE and RA contributes to cognitive improvement in an Aβ25-35-injected mouse model. MATERIALS/METHODS Male ICR mice were intracerebroventricularly injected with aggregated Aβ25-35 to induce AD. Aβ25-35-injected mice were fed PFE (50 mg/kg/day) or RA (0.25 mg/kg/day) for 14 days and examined for learning and memory ability through the T-maze, object recognition, and Morris water maze test. RESULTS Our present study demonstrated that PFE and RA administration significantly enhanced cognition function and object discrimination, which were impaired by Aβ25-35, in the T-maze and object recognition tests, respectively. In addition, oral administration of PFE and RA decreased the time to reach the platform and increased the number of crossings over the removed platform when compared with the Aβ25-35-induced control group in the Morris water maze test. Furthermore, PFE and RA significantly decreased the levels of nitric oxide (NO) and malondialdehyde (MDA) in the brain, kidney, and liver. In particular, PFE markedly attenuated oxidative stress by inhibiting production of NO and MDA in the Aβ25-35-injected mouse brain. CONCLUSIONS These results suggest that PFE and its active compound RA have beneficial effects on cognitive improvement and may help prevent AD induced by Aβ.
Collapse
Affiliation(s)
- Ah Young Lee
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Bo Ra Hwang
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| | - Myoung Hee Lee
- Department of Southern Area Crop Science, National Institute of Crop Science, Rural Development Administration, Gyeongnam 50424, Korea
| | - Sanghyun Lee
- Department of Integrative Plant Science, Chung-Ang University, Anseong, Gyeonggi 17546, Korea
| | - Eun Ju Cho
- Department of Food Science and Nutrition & Kimchi Research Institute, Pusan National University, 2 Busandaehak-ro 63 beon-gil, Geumjeong-gu, Busan 46241, Korea
| |
Collapse
|
5
|
Cho SM, Lee S, Yang SH, Kim HY, Lee MJ, Kim HV, Kim J, Baek S, Yun J, Kim D, Kim YK, Cho Y, Woo J, Kim TS, Kim Y. Age-dependent inverse correlations in CSF and plasma amyloid-β(1-42) concentrations prior to amyloid plaque deposition in the brain of 3xTg-AD mice. Sci Rep 2016; 6:20185. [PMID: 26830653 PMCID: PMC4735736 DOI: 10.1038/srep20185] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/23/2015] [Indexed: 12/12/2022] Open
Abstract
Amyloid-β (Aβ) plays a critical role as a biomarker in Alzheimer's disease (AD) diagnosis. In addition to its diagnostic potential in the brain, recent studies have suggested that changes of Aβ level in the plasma can possibly indicate AD onset. In this study, we found that plasma Aβ(1-42) concentration increases with age, while the concentration of Aβ(1-42) in the cerebrospinal fluid (CSF) decreases in APPswe, PS1M146V and TauP301L transgenic (3xTg-AD) mice, if measurements were made before formation of ThS-positive plaques in the brain. Our data suggests that there is an inverse correlations between the plasma and CSF Aβ(1-42) levels until plaques form in transgenic mice's brains and that the plasma Aβ concentration possesses the diagnostic potential as a biomarker for diagnosis of early AD stages.
Collapse
Affiliation(s)
- Soo Min Cho
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Sejin Lee
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Seung-Hoon Yang
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea
| | - Hye Yun Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea.,GoshenBiotech, 83-2 Wolmun-ri, Wabu-eup, Namyangju-si, Gyeonggi-do, Republic of Korea
| | - Michael Jisoo Lee
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Department of Medical Education, California Northstate University College of Medicine, 9700 W Taron Drive, Elk Grove, CA, USA
| | - Hyunjin Vincent Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jiyoon Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Seungyeop Baek
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Jin Yun
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Dohee Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Department of Biotechnology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea
| | - Yun Kyung Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| | - Yakdol Cho
- Center for Neuroscience and Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea
| | - Jiwan Woo
- Center for Neuroscience and Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea
| | - Tae Song Kim
- Center for BioMicroSystems, Brain Science Institute, Korea Institute of Science and Technology, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea
| | - YoungSoo Kim
- Center for Neuro-Medicine, Hwarangno 14-gil 5, Seongbuk-gu, Seoul, Republic of Korea.,Biological Chemistry Program, Korea University of Science and Technology, 217 Gajungro, Yuseong-gu, Daejeon, Republic of Korea
| |
Collapse
|
6
|
|
7
|
Abstract
Alzheimer's disease (AD), the most common form of dementia, is now representing one of the largest unmet medical needs. However, no effective treatment is now available to impede the progression of AD or delay its onset. There are two major challenges for the development of effective therapy for AD. First, the exact cause for AD onset is still unknown. Second, brain drug delivery is significantly hindered by the blood-brain barrier (BBB). In this review, we will summarize the pathological understanding about AD and the related treatments, compare BBB and its effect on brain drug delivery under normal and AD conditions and review the nanotherapeutic strategies that have been developed for AD therapy in recent years.
Collapse
|
8
|
Drug Access to the Central Nervous System in Alzheimer’s Disease: Preclinical and Clinical Insights. Pharm Res 2014; 32:819-39. [DOI: 10.1007/s11095-014-1522-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/12/2014] [Indexed: 12/12/2022]
|
9
|
Pardridge WM. Blood-brain barrier drug delivery of IgG fusion proteins with a transferrin receptor monoclonal antibody. Expert Opin Drug Deliv 2014; 12:207-22. [PMID: 25138991 DOI: 10.1517/17425247.2014.952627] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Biologic drugs are large molecules that do not cross the blood- brain barrier (BBB). Brain penetration is possible following the re-engineering of the biologic drug as an IgG fusion protein. The IgG domain is a MAb against an endogenous BBB receptor such as the transferrin receptor (TfR). The TfRMAb acts as a molecular Trojan horse to ferry the fused biologic drug into the brain via receptor-mediated transport on the endogenous BBB TfR. AREAS COVERED This review discusses TfR isoforms, models of BBB transport of transferrin and TfRMAbs, and the genetic engineering of TfRMAb fusion proteins, including BBB penetrating IgG-neurotrophins, IgG-decoy receptors, IgG-lysosomal enzyme therapeutics and IgG-avidin fusion proteins, as well as BBB transport of bispecific antibodies formed by fusion of a therapeutic antibody to a TfRMAb targeting antibody. Also discussed are quantitative aspects of the plasma pharmacokinetics and brain uptake of TfRMAb fusion proteins, as compared to the brain uptake of small molecules, and therapeutic applications of TfRMAb fusion proteins in mouse models of neural disease, including Parkinson's disease, stroke, Alzheimer's disease and lysosomal storage disorders. The review covers the engineering of TfRMAb-avidin fusion proteins for BBB targeted delivery of biotinylated peptide radiopharmaceuticals, low-affinity TfRMAb Trojan horses and the safety pharmacology of chronic administration of TfRMAb fusion proteins. EXPERT OPINION The BBB delivery of biologic drugs is possible following re-engineering as a fusion protein with a molecular Trojan horse such as a TfRMAb. The efficacy of this technology will be determined by the outcome of future clinical trials.
Collapse
Affiliation(s)
- William M Pardridge
- ArmaGen Technologies, Inc. , 26679 Agoura Road, Calabasas, CA 91302 , USA +1 818 252 8202 ; +1 818 252 8214 ;
| |
Collapse
|
10
|
Abstract
Current advances in nanotechnology have paved the way for the early detection, prevention and treatment of various diseases such as vascular disorders and cancer. These advances have provided novel approaches or modalities of incorporating or adsorbing therapeutic, biosensor and targeting agents into/on nanoparticles. With significant progress, nanomedicine for vascular therapy has shown significant advantages over traditional medicine because of its ability to selectively target the disease site and reduce adverse side effects. Targeted delivery of nanoparticles to vascular endothelial cells or the vascular wall provides an effective and more efficient way for early detection and/or treatment of vascular diseases such as atherosclerosis, thrombosis and Cerebrovascular Amyloid Angiopathy (CAA). Clinical applications of biocompatible and biodegradable polymers in areas such as vascular graft, implantable drug delivery, stent devices and tissue engineering scaffolds have advanced the candidature of polymers as potential nano-carriers for vascular-targeted delivery of diagnostic agents and drugs. This review focuses on the basic aspects of the vasculature and its associated diseases and relates them to polymeric nanoparticle-based strategies for targeting therapeutic agents to diseased vascular site.
Collapse
Affiliation(s)
- Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL ; Division of Radiation Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - Karunyna Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
11
|
Sumbria RK, Hui EKW, Lu JZ, Boado RJ, Pardridge WM. Disaggregation of amyloid plaque in brain of Alzheimer's disease transgenic mice with daily subcutaneous administration of a tetravalent bispecific antibody that targets the transferrin receptor and the Abeta amyloid peptide. Mol Pharm 2013; 10:3507-13. [PMID: 23924247 DOI: 10.1021/mp400348n] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Anti-amyloid antibodies (AAA) are under development as new therapeutics that disaggregate the amyloid plaque in brain in Alzheimer's disease (AD). However, the AAAs are large molecule drugs that do not cross the blood-brain barrier (BBB), in the absence of BBB disruption. In the present study, an AAA was re-engineered for receptor-mediated transport across the BBB via the endogenous BBB transferrin receptor (TfR). A single chain Fv (ScFv) antibody form of an AAA was fused to the carboxyl terminus of each heavy chain of a chimeric monoclonal antibody (mAb) against the mouse TfR, and this produced a tetravalent bispecific antibody designated the cTfRMAb-ScFv fusion protein. Unlike a conventional AAA, which has a plasma half-time of weeks, the cTfRMAb-ScFv fusion protein is cleared from plasma in mice with a mean residence time of about 3 h. Therefore, a novel protocol was developed for the treatment of one year old presenilin (PS)-1/amyloid precursor protein (APP) AD double transgenic PSAPP mice, which were administered daily subcutaneous (sc) injections of 5 mg/kg of the cTfRMAb-ScFv fusion protein for 12 consecutive weeks. At the end of the treatment, brain amyloid plaques were quantified with confocal microscopy using both Thioflavin-S staining and immunostaining with the 6E10 antibody against Abeta amyloid fibrils. Fusion protein treatment caused a 57% and 61% reduction in amyloid plaque in the cortex and hippocampus, respectively. No increase in plasma immunoreactive Abeta amyloid peptide, and no cerebral microhemorrhage, was observed. Chronic daily sc treatment of the mice with the fusion protein caused no immune reactions and only a low titer antidrug antibody response. In conclusion, re-engineering AAAs for receptor-mediated BBB transport allows for reduction in brain amyloid plaque without cerebral microhemorrhage following daily sc treatment for 12 weeks.
Collapse
Affiliation(s)
- Rachita K Sumbria
- Department of Medicine, UCLA, Los Angeles, California 90024, United States
| | | | | | | | | |
Collapse
|
12
|
Drummond ES, Muhling J, Martins RN, Wijaya LK, Ehlert EM, Harvey AR. Pathology associated with AAV mediated expression of beta amyloid or C100 in adult mouse hippocampus and cerebellum. PLoS One 2013; 8:e59166. [PMID: 23516609 PMCID: PMC3596293 DOI: 10.1371/journal.pone.0059166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 02/13/2013] [Indexed: 11/18/2022] Open
Abstract
Accumulation of beta amyloid (Aβ) in the brain is a primary feature of Alzheimer’s disease (AD) but the exact molecular mechanisms by which Aβ exerts its toxic actions are not yet entirely clear. We documented pathological changes 3 and 6 months after localised injection of recombinant, bi-cistronic adeno-associated viral vectors (rAAV2) expressing human Aβ40-GFP, Aβ42-GFP, C100-GFP or C100V717F-GFP into the hippocampus and cerebellum of 8 week old male mice. Injection of all rAAV2 vectors resulted in wide-spread transduction within the hippocampus and cerebellum, as shown by expression of transgene mRNA and GFP protein. Despite the lack of accumulation of Aβ protein after injection with AAV vectors, injection of rAAV2-Aβ42-GFP and rAAV2- C100V717F-GFP into the hippocampus resulted in significantly increased microgliosis and altered permeability of the blood brain barrier, the latter revealed by high levels of immunoglobulin G (IgG) around the injection site and the presence of IgG positive cells. In comparison, injection of rAAV2-Aβ40-GFP and rAAV2-C100-GFP into the hippocampus resulted in substantially less neuropathology. Injection of rAAV2 vectors into the cerebellum resulted in similar types of pathological changes, but to a lesser degree. The use of viral vectors to express different types of Aβ and C100 is a powerful technique with which to examine the direct in vivo consequences of Aβ expression in different regions of the mature nervous system and will allow experimentation and analysis of pathological AD-like changes in a broader range of species other than mouse.
Collapse
Affiliation(s)
- Eleanor S Drummond
- School of Anatomy, Physiology and Human Biology, The University of Western Australia, Western Australia, Australia.
| | | | | | | | | | | |
Collapse
|
13
|
Banks WA. Drug delivery to the brain in Alzheimer's disease: consideration of the blood-brain barrier. Adv Drug Deliv Rev 2012; 64:629-39. [PMID: 22202501 DOI: 10.1016/j.addr.2011.12.005] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 12/09/2011] [Accepted: 12/09/2011] [Indexed: 12/21/2022]
Abstract
The successful treatment of Alzheimer's disease (AD) will require drugs that can negotiate the blood-brain barrier (BBB). However, the BBB is not simply a physical barrier, but a complex interface that is in intimate communication with the rest of the central nervous system (CNS) and influenced by peripheral tissues. This review examines three aspects of the BBB in AD. First, it considers how the BBB may be contributing to the onset and progression of AD. In this regard, the BBB itself is a therapeutic target in the treatment of AD. Second, it examines how the BBB restricts drugs that might otherwise be useful in the treatment of AD and examines strategies being developed to deliver drugs to the CNS for the treatment of AD. Third, it considers how drug penetration across the AD BBB may differ from the BBB of normal aging. In this case, those differences can complicate the treatment of CNS diseases such as depression, delirium, psychoses, and pain control in the AD population.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research, Education, and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA , USA.
| |
Collapse
|
14
|
A novel method for the rapid determination of beta-amyloid toxicity on acute hippocampal slices using MTT and LDH assays. Brain Res Bull 2012; 87:521-5. [DOI: 10.1016/j.brainresbull.2012.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 01/31/2023]
|
15
|
Bulbarelli A, Lonati E, Brambilla A, Orlando A, Cazzaniga E, Piazza F, Ferrarese C, Masserini M, Sancini G. Aβ42 production in brain capillary endothelial cells after oxygen and glucose deprivation. Mol Cell Neurosci 2012; 49:415-22. [DOI: 10.1016/j.mcn.2012.01.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 12/06/2011] [Accepted: 01/25/2012] [Indexed: 12/30/2022] Open
|
16
|
|
17
|
Sharma HS, Castellani RJ, Smith MA, Sharma A. The blood-brain barrier in Alzheimer's disease: novel therapeutic targets and nanodrug delivery. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2012; 102:47-90. [PMID: 22748826 DOI: 10.1016/b978-0-12-386986-9.00003-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Treatment strategies for Alzheimer's disease (AD) are still elusive. Thus, new strategies are needed to understand the pathogenesis of AD in order to provide suitable therapeutic measures. Available evidences suggest that in AD, passage across the blood-brain barrier (BBB) and transport exchanges for amyloid-β-peptide (ABP) between blood and the central nervous system (CNS) compartments play an important regulatory role for the deposition of brain ABP. New evidences suggest that BBB is altered in AD. Studies favoring transport theory clearly show that ABP putative receptors at the BBB control the level of soluble isoform of ABP in brain. This is achieved by regulating influx of circulating ABP into brain via specific receptor for advanced glycation end products (RAGE) and gp330/megalin-mediated transcytosis. On the other hand, the efflux of brain-derived ABP into the circulation across the vascular system via BBB is accomplished by low-density receptor-related protein-1 (LRP1). Furthermore, an increased BBB permeability in AD is also likely since structural damage of endothelial cells is quite frequent in AD brain. Thus, enhanced drug delivery in AD is needed to induce neuroprotection and therapeutic success. For this purpose, nanodrug delivery could be one of the available options that require active consideration for novel therapeutic strategies to treat AD cases. This review is focused on these aspects and provides new data showing that BBB plays an important role in AD-induced neurodegeneration and neurorepair.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- Cerebrovascular Research Laboratory, Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
18
|
Zhou QH, Fu A, Boado RJ, Hui EKW, Lu JZ, Pardridge WM. Receptor-mediated abeta amyloid antibody targeting to Alzheimer's disease mouse brain. Mol Pharm 2010; 8:280-5. [PMID: 21141969 DOI: 10.1021/mp1003515] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The goal of this work is the reduction in the Abeta amyloid peptide burden in brain of Alzheimer's disease (AD) transgenic mice without the concomitant elevation in plasma Abeta amyloid peptide. An anti-Abeta amyloid antibody (AAA) was re-engineered as a fusion protein with a blood-brain barrier (BBB) molecular Trojan horse. The AAA was engineered as a single chain Fv (ScFv) antibody, and the ScFv was fused to the heavy chain of a chimeric monoclonal antibody (mAb) against the mouse transferrin receptor (TfR), and this fusion protein was designated cTfRMAb-ScFv. The cTfRMAb-ScFv protein penetrates mouse brain from blood via transport on the BBB TfR, and the brain uptake is 3.5% of injected dose/gram brain following an intravenous administration. Double transgenic APPswe,PSEN1dE9 mice were studied at 12 months of age. The mice were shown to have extensive Abeta amyloid plaques in cerebral cortex based on immunocytochemistry. The mice were treated every 3-4 days by intravenous injections of either saline or the cTfRMAb-ScFv fusion protein at an injection dose of 1 mg/kg for 12 consecutive weeks. The brain Aβ¹⁻⁴² concentration was reduced 40% in the fusion protein treated mice, without any elevation in plasma Aβ¹⁻⁴² concentration. No cerebral microhemorrhage was observed in the treated mice. These results show that brain-penetrating antibody pharmaceutics can be developed for brain disorders such as AD following the re-engineering of the antibody as a fusion protein that is transported across the BBB via receptor-mediated transport.
Collapse
Affiliation(s)
- Qing-Hui Zhou
- Department of Medicine, UCLA, Los Angeles, California 90024, USA
| | | | | | | | | | | |
Collapse
|
19
|
Bachmeier C, Mullan M, Paris D. Characterization and use of human brain microvascular endothelial cells to examine β-amyloid exchange in the blood-brain barrier. Cytotechnology 2010; 62:519-29. [PMID: 20954006 DOI: 10.1007/s10616-010-9313-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Accepted: 10/03/2010] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by excessive cerebrovascular deposition of the β-amyloid peptide (Aβ). The investigation of Aβ transport across the blood-brain barrier (BBB) has been hindered by inherent limitations in the cellular systems currently used to model the BBB, such as insufficient barrier properties and poor reproducibility. In addition, many of the existing models are not of human or brain origin and are often arduous to establish and maintain. Thus, we characterized an in vitro model of the BBB employing human brain microvascular endothelial cells (HBMEC) and evaluated its utility to investigate Aβ exchange at the blood-brain interface. Our HBMEC model offers an ease of culture compared with primary isolated or coculture BBB models and is more representative of the human brain endothelium than many of the cell lines currently used to study the BBB. In our studies, the HBMEC model exhibited barrier properties comparable to existing BBB models as evidenced by the restricted permeability of a known paracellular marker. In addition, using a simple and rapid fluormetric assay, we showed that antagonism of key Aβ transport proteins significantly altered the bi-directional transcytosis of fluorescein-Aβ (1-42) across the HBMEC model. Moreover, the magnitude of these effects was consistent with reports in the literature using the same ligands in existing in vitro models of the BBB. These studies establish the HBMEC as a representative in vitro model of the BBB and offer a rapid fluorometric method of assessing Aβ exchange between the periphery and the brain.
Collapse
Affiliation(s)
- Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL, 34243, USA,
| | | | | |
Collapse
|
20
|
Abstract
Alzheimer’s disease, a neurodegenerative disorder, is associated with various pathological alterations to the blood–brain barrier, including disruption to the inter-endothelial tight junction proteins, altered expression of transport proteins involved in drug efflux, a reduction in cerebral blood flow and a thickening of the brain capillary basement membrane. There are many conflicting reports on whether such changes alter the ability of endogenous proteins to extravasate into the brain parenchyma, and there are even fewer reports focusing on the potential impact of these changes on drug transport into the CNS. The purpose of this review is to critically evaluate how the reported changes to the blood–brain barrier in Alzheimer’s disease have (or have not) resulted in altered CNS drug delivery, and to highlight the requirement for more rigorous and systematic studies in this field for the benefit of drug discovery and delivery scientists.
Collapse
|
21
|
Chen X, Ghribi O, Geiger JD. Caffeine protects against disruptions of the blood-brain barrier in animal models of Alzheimer's and Parkinson's diseases. J Alzheimers Dis 2010; 20 Suppl 1:S127-41. [PMID: 20164568 DOI: 10.3233/jad-2010-1376] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sporadic Alzheimer's disease (AD) and Parkinson's disease (PD) are two of the most common neurodegenerative diseases and as such they represent major public health problems. Finding effective treatments for AD and PD represents an unmet and elusive goal largely because these diseases are chronic and progressive, and have a complicated and ill-understood pathogenesis. Although the underlying mechanisms are not fully understood, caffeine, the most commonly ingested psychoactive drug in the world, has been shown in human and animal studies to be protective against AD and PD. One mechanism implicated in the pathogenesis of AD and PD is blood-brain barrier (BBB) dysfunction and we reported recently that caffeine exerts protective effects against AD and PD at least in part by keeping the BBB intact. The present review focuses on the role of BBB dysfunction in the pathogenesis of AD and PD, caffeine's protective effects against AD and PD, and potential mechanisms whereby caffeine protects against BBB leakage.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, USA
| | | | | |
Collapse
|
22
|
Boado RJ, Lu JZ, Hui EKW, Pardridge WM. IgG-single chain Fv fusion protein therapeutic for Alzheimer's disease: Expression in CHO cells and pharmacokinetics and brain delivery in the rhesus monkey. Biotechnol Bioeng 2010; 105:627-35. [PMID: 19816967 DOI: 10.1002/bit.22576] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Monoclonal antibodies (MAb) directed against the Abeta amyloid peptide of Alzheimer's disease (AD) are potential new therapies for AD, since these antibodies disaggregate brain amyloid plaque. However, the MAb is not transported across the blood-brain barrier (BBB). To enable BBB transport, a single chain Fv (ScFv) antibody against the Abeta peptide of AD was re-engineered as a fusion protein with the MAb against the human insulin receptor (HIR). The HIRMAb acts as a molecular Trojan horse to ferry the ScFv therapeutic antibody across the BBB. Chinese hamster ovary (CHO) cells were stably transfected with a tandem vector encoding the heavy and light chains of the HIRMAb-ScFv fusion protein. A high secreting line was isolated following methotrexate amplification and dilutional cloning. The HIRMAb-ScFv fusion protein in conditioned serum-free medium was purified by protein A affinity chromatography. The fusion protein was stable as a liquid formulation, and retained high-affinity binding of both the HIR and the Abeta amyloid peptide. The HIRMAb-ScFv fusion protein was radiolabeled with the (125)I-Bolton-Hunter reagent, followed by measurement of the pharmacokinetics of plasma clearance and brain uptake in the adult Rhesus monkey. The HIRMAb-ScFv fusion protein was rapidly cleared from plasma and was transported across the primate BBB in vivo. In conclusion, the HIRMAb-ScFv fusion protein is a new class of antibody-based therapeutic for AD that has been specifically engineered to cross the human BBB.
Collapse
Affiliation(s)
- Ruben J Boado
- ArmaGen Technologies, Inc., Santa Monica, California, USA
| | | | | | | |
Collapse
|
23
|
Pardridge WM. Alzheimer's disease drug development and the problem of the blood-brain barrier. Alzheimers Dement 2009; 5:427-32. [PMID: 19751922 DOI: 10.1016/j.jalz.2009.06.003] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 06/26/2009] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) drug development is limited by the presence of the blood-brain barrier (BBB). More than 98% of all small-molecule drugs, and approximately 100% of all large-molecule drugs, do not cross the BBB. Although the vast majority of AD drug candidates do not cross the BBB, the present-day AD drug-development effort is characterized by an imbalance wherein >99% of the drug-development effort is devoted to central nervous system (CNS) drug discovery, and <1% of drug development is devoted to CNS drug delivery. Future AD drug development needs a concerted effort to incorporate BBB sciences early in the CNS drug discovery process. This goal can be achieved by a reallocation of resources, and an expansion of research efforts in the pure science of BBB biology and the applied science of brain drug-targeting technology.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Nagababu E, Usatyuk PV, Enika D, Natarajan V, Rifkind JM. Vascular endothelial barrier dysfunction mediated by amyloid-beta proteins. J Alzheimers Dis 2009; 17:845-54. [PMID: 19542618 PMCID: PMC2852470 DOI: 10.3233/jad-2009-1104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neuronal inflammation is very common in Alzheimer's disease (AD). This inflammation can be caused by infiltration of neutrophils across the blood brain barrier. Endothelial permeability changes are required for the infiltration of high molecular weight components to the brain. Deposition of toxic amyloid-beta (A beta) fibrils in the cerebral vasculature, as well as in brain neurons, has been implicated in the development of AD. This study investigates the effect of A beta fibrils on the permeability of the endothelium and the mechanism for the observed permeability changes. A beta(1-40) and A beta(1-42) fibrils, but not monomers, were found to increase permeability of bovine pulmonary arterial endothelial cells in a dose- and time dependent manner as detected by transendothelial electrical resistance. This increase in permeability is only partially (25%) inhibited by catalase and is not associated with an increase in cytosolic Ca+2 or tyrosine phosphorylation. These results indicate that hydrogen peroxide is not the primary mediator for the permeability changes. Treatment of cells with both amyloid fibrils resulted in stress fiber formation, disruption and aggregation of actin filaments, and cellular gap formation. The results of this study reveal that A beta increases the permeability of endothelium by inducing change in the cytoskeleton network.
Collapse
Affiliation(s)
- Enika Nagababu
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| | - Peter V. Usatyuk
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Divya Enika
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Viswanathan Natarajan
- Pulmonary and Critical Care Section, Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Joseph M. Rifkind
- Molecular Dynamics Section, National Institute on Aging, National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21224
| |
Collapse
|
25
|
Gonzalez-Velasquez FJ, Kotarek JA, Moss MA. Soluble aggregates of the amyloid-beta protein selectively stimulate permeability in human brain microvascular endothelial monolayers. J Neurochem 2008; 107:466-77. [PMID: 18702666 DOI: 10.1111/j.1471-4159.2008.05618.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Cerebral amyloid angiopathy associated with Alzheimer's disease is characterized by cerebrovascular deposition of the amyloid-beta protein (Abeta). Abeta elicits a number of morphological and biochemical alterations in the cerebral microvasculature, which culminate in hemorrhagic stroke. Among these changes, compromise of the blood-brain barrier has been described in Alzheimer's disease brain, transgenic animal models of Alzheimer's disease, and cell culture experiments. In the current study, presented data illustrates that isolated soluble Abeta(1-40) aggregates, but not unaggregated monomer or mature fibril, enhance permeability in human brain microvascular endothelial monolayers. Abeta(1-40)-induced changes in permeability are paralleled by both a decrease in transendothelial electrical resistance and a re-localization of the tight junction-associated protein zonula occludin-1 away from cell borders and into the cytoplasm. Small soluble Abeta(1-40) aggregates are confirmed to be the most potent stimulators of endothelial monolayer permeability by establishing an inverse relationship between average aggregate size and stimulated changes in diffusional permeability coefficients. These results support previous findings demonstrating that small soluble Abeta(1-40) aggregates are also primarily responsible for endothelial activation, suggesting that these same species may elicit other changes in the cerebrovasculature associated with cerebral amyloid angiopathy and Alzheimer's disease.
Collapse
Affiliation(s)
- Francisco J Gonzalez-Velasquez
- Department of Chemical Engineering, University of South Carolina, Swearingen Engineering Center, Columbia, South Carolina, USA
| | | | | |
Collapse
|
26
|
Abstract
The blood-brain barrier (BBB), together with the blood-cerebrospinal-fluid barrier, protects and regulates the homeostasis of the brain. However, these barriers also limit the transport of small-molecule and, particularly, biopharmaceutical drugs such as proteins, genes and interference RNA to the brain, thereby limiting the treatment of many brain diseases. As a result, various drug delivery and targeting strategies are currently being developed to enhance the transport and distribution of drugs into the brain. In this review, we discuss briefly the biology and physiology of the BBB as the most important barrier for drug transport to the brain and, in more detail, the possibilities for delivering large-molecule drugs, particularly genes, by receptor-mediated nonviral drug delivery to the (human) brain. In addition, the systemic and intracellular pharmacokinetics of nonviral gene delivery, together with targeted brain imaging, are reviewed briefly.
Collapse
Affiliation(s)
- Albertus G de Boer
- Blood-Brain-Barrier Research Group, Division of Pharmacology, Leiden-Amsterdam Center for Drug Research, University of Leiden, Leiden, The Netherlands.
| | | |
Collapse
|
27
|
Verma S, Nakaoke R, Dohgu S, Banks WA. Release of cytokines by brain endothelial cells: A polarized response to lipopolysaccharide. Brain Behav Immun 2006; 20:449-55. [PMID: 16309883 DOI: 10.1016/j.bbi.2005.10.005] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 09/30/2005] [Accepted: 10/13/2005] [Indexed: 12/31/2022] Open
Abstract
Brain endothelial cells (BECs) comprise the blood-brain barrier (BBB) and are an active part of the neuroimmune system, responding to and transporting cytokines. BECs also have the ability to secrete neuroimmune substances, including cytokines. A unique feature of the BEC is its polarization, with its luminal (blood-facing) and abluminal (brain-facing) cell membranes differing in their lipid, receptor, and transporter compositions. This polarization could have functional consequences for neuroimmune communication. We postulated (i) that cytokine secretion from the luminal or abluminal membranes could differ under baseline or stimulated conditions and (ii) that an immune challenge from one side of the BBB could result in cytokine release from the other. We used an in vitro BBB model of mouse BECs cultured as monolayers to investigate cytokine secretion into luminal and abluminal chambers. Our major findings in these studies were: (i) the first demonstration that interleukin (IL)-1alpha, IL-10, and granulocyte-macrophage colony-stimulating factor are secreted from BECs and confirmation of the secretions of IL-6 and tumor necrosis factor-alpha, (ii) that constitutive and lipopolysaccharide (LPS)-stimulated secretion of cytokines is polarized in favor of luminal secretion, and (iii) that response to neuroimmune stimulation is also polarized as exemplified by the finding that abluminal LPS more robustly induced secretion of IL-6 than did luminal LPS. Overall, these findings support the BBB as an important source of cytokines. Furthermore, the BBB can respond to immune challenges received from one side of the neuroimmune axis by releasing cytokines into the other.
Collapse
Affiliation(s)
- Sulekha Verma
- Geriatrics Research Educational and Clinical Center, Veterans Affairs Medical Center, St. Louis, MO, USA
| | | | | | | |
Collapse
|
28
|
Arshavsky YI. Alzheimer's disease, brain immune privilege and memory: a hypothesis. J Neural Transm (Vienna) 2006; 113:1697-707. [PMID: 16932992 DOI: 10.1007/s00702-006-0524-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2006] [Accepted: 04/28/2006] [Indexed: 12/13/2022]
Abstract
The most distinctive feature of Alzheimer's disease (AD) is the specific degeneration of the neurons involved in memory consolidation, storage, and retrieval. Patients suffering from AD forget basic information about their past, loose linguistic and calculative abilities and communication skills. Thus, understanding the etiology of AD may provide insights into the mechanisms of memory and vice versa. The brain is an immunologically privileged site protected from the organism's immune reactions by the blood-brain barrier (BBB). All risk factors for AD (both cardiovascular and genetic) lead to destruction of the BBB. Evidence emerging from recent literature indicates that AD may have an autoimmune nature associated with BBB impairments. This hypothesis suggests that the process of memory consolidation involves the synthesis of novel macromolecules recognized by the immune system as "non-self" antigens. The objective of this paper is to stimulate new approaches to studies of neural mechanisms underpinning memory consolidation and its breakdown during AD. If the hypothesis on the autoimmune nature of AD is correct, the identification of the putative antigenic macromolecules might be critical to understanding the etiology and prevention of AD, as well as for elucidating cellular mechanisms of learning and memory.
Collapse
Affiliation(s)
- Y I Arshavsky
- Institute for Nonlinear Science, University of California San Diego, La Jolla, CA 92093-0402, USA.
| |
Collapse
|
29
|
Roy S, Rauk A. Alzheimer's disease and the 'ABSENT' hypothesis: mechanism for amyloid beta endothelial and neuronal toxicity. Med Hypotheses 2005; 65:123-37. [PMID: 15893129 DOI: 10.1016/j.mehy.2004.08.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2004] [Accepted: 08/13/2004] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease [AD] is the most common cause of dementia among people age 65 and older. One of the biggest stumbling blocks in developing effective drug therapy for Alzheimer's disease has been the lack of a comprehensive hypothesis that explains the mechanism behind all of the histopathological changes seen in patients suffering from Alzheimer's disease. An overview of the currently popular 'amyloid' and 'vascular' hypotheses for AD demonstrates that neither hypothesis by itself can explain all the known histopathological and biochemical lesions seen in Alzheimer's disease. The paper presents a hypothesis that tries to explain the mechanism behind almost all the histopathological changes, and varying clinical manifestations seen in both diagnosed AD and Vascular Dementia [VaD]. The new hypothesis is based on the known dual toxicity of beta amyloid to both vascular and neuronal tissues, their synergy and the resultant net effect on the onset and progression of AD. The new hypothesis therefore will be known as the Amyloid Beta Synergistic Endothelial and Neuronal Toxicity [ABSENT] hypothesis. The ABSENT hypothesis will try to show the common chemical mechanism behind almost all of the pathological changes seen in AD. According to the ABSENT hypothesis, beta amyloid itself generates all the free radicals that cause both vascular dysfunction and the neuronal damage seen in AD. The chemical mechanism proposed is based on evidence from physical chemistry experiments, calculations as well as in vitro/in vivo experiments. The ABSENT hypothesis does not favor one mode of beta amyloid-induced brain damage over the other, rather it considers the net effects of the neuronal stress/damage caused by both the cerebrovascular dysfunction and direct neurotoxicity caused by beta amyloid. The hypothesis states that each patient has a different balance of predisposing factors that modulate the extent of neurotoxicity and cerebrovascular dysfunction caused by beta amyloid and thereby explains the wide range and mixed nature of damage and dysfunction seen in the studies done on patients diagnosed with AD, VaD or 'mixed dementias'. According to the hypothesis, beta amyloid peptides are necessary if not sufficient to cause AD, VaD and mixed senile dementias. The hypothesis, therefore, proposes the term Beta Amyloid Dementias [BAD] to describe the conditions currently covered by the diagnoses of 'AD', 'VaD' and 'Mixed [senile] Dementias'. Finally, the ABSENT hypothesis tries to put forth a direct chemical mechanism behind the apparent synergy and increased association between old age, pre- and coexisting vascular disease, diabetes and AD.
Collapse
Affiliation(s)
- Samir Roy
- Department of Chemistry, University of Calgary, 2500 University Drive, NW Alberta, Canada T2N 1N4.
| | | |
Collapse
|
30
|
Kálmán J, Palotás A, Kis G, Boda K, Túri P, Bari F, Domoki F, Dóda I, Argyelán M, Vincze G, Séra T, Csernay L, Janka Z, Pávics L. Regional cortical blood flow changes following sodium lactate infusion in Alzheimer's disease. Eur J Neurosci 2005; 21:1671-8. [PMID: 15845094 DOI: 10.1111/j.1460-9568.2005.03924.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bilateral temporoparietal hypoperfusion is a characteristic single photon emission computed tomography (SPECT) finding in Alzheimer's disease (AD). Lactate is a metabolic vasodilator and is known to provoke increased cerebral blood flow (CBF) in healthy adults. This work investigated whether lactate, which is present in high concentrations in AD cerebrospinal fluid, affects AD-specific perfusion abnormalities. Twenty mild-to-moderately demented AD probands participated in the self-controlled study. The regional CBF was examined utilizing (99m)Tc-HMPAO SPECT after sodium lactate infusion (0.5 M, 5 mL/kg body weight) and 0.9% NaCl infusion, one on each of two separate days. Despite the vasodilatator effects of sodium lactate, AD rCBF patterns did not show increase in temporo-parietal regions after its infusion. AD-specific bi-temporo-parietal reduction in CBF was accompanied by further hypoperfusion in the parieto-occipital areas after the sodium lactate infusion in seven patients, while no CBF changes were observed in the case of the remaining 13 probands. The pattern of the CBF abnormalities was not correlated with the apolipoprotein E genotype. The decreased vascular responsiveness to sodium lactate reflects disturbed vasoregulatory processes in AD and it is unlikely that lactate would have any relevance in the treatment of AD-related cerebral hypoperfusion, but could be used to improve the value of perfusion SPECT in the diagnosis of AD.
Collapse
Affiliation(s)
- János Kálmán
- Albert Szent-Györgyi Medical and Pharmaceutical Center, Faculty of Medicine, University of Szeged, H-6721 Szeged, Semmelweis u. 6, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
D'Andrea MR. Add Alzheimer’s disease to the list of autoimmune diseases. Med Hypotheses 2005; 64:458-63. [PMID: 15617848 DOI: 10.1016/j.mehy.2004.08.024] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Accepted: 08/18/2004] [Indexed: 11/19/2022]
Abstract
A sole pathological event leading to Alzheimer's disease (AD) remains undiscovered in spite of decades of costly research. In fact, it is more probable that the causes of AD are the result of a myriad of intertwining pathologies. However, hope remains that a single awry event could lead to the many pathological events observed in AD brain tissues thereby creating the presentation of simultaneous pathologies. Age-related vascular diseases, which include an impaired blood-brain barrier (BBB), are a common denominator associated with various degrees of dementia, including AD. Recently, a key finding not only demonstrated the anomalous presence of immunoglobulin (Ig) detection in the brain parenchyma of AD tissues but, most importantly, specific neurons that showed degenerative, apoptotic features contained these vascular-derived antibodies. In addition, subsequent studies detected classical complement components, C1q and C5b-9, in these Ig-positive neurons, which also were spatially more associated with reactive microglia over the Ig-negative neurons. Thus, it is possible that the mere presence of anti-neuronal autoantibodies in the serum, whose importance had been previously dismissed, may be without pathological consequence until there is a BBB dysfunction to allow the deleterious effects of these autoantibodies access on their targets. Hence, these observations suggest autoimmunity-induced cell death in AD.
Collapse
Affiliation(s)
- Michael R D'Andrea
- Johnson and Johnson Pharmaceutical, Research and Development, Drug Discovery, Spring House, PA 19477-0776, USA.
| |
Collapse
|
32
|
Yang DI, Yeh CH, Chen S, Xu J, Hsu CY. Neutral sphingomyelinase activation in endothelial and glial cell death induced by amyloid beta-peptide. Neurobiol Dis 2004; 17:99-107. [PMID: 15350970 DOI: 10.1016/j.nbd.2004.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2004] [Revised: 05/19/2004] [Accepted: 06/02/2004] [Indexed: 12/16/2022] Open
Abstract
We have explored the molecular mechanism underlying amyloid beta-peptide (Abeta)-mediated cytotoxicity in vitro. Exposure of murine cerebral endothelial cells (CECs) or C6 glioma cells to Abeta25-35 resulted in dose-dependent cell death. Ceramide is a pro-apoptotic lipid mediator. Forced elevation of cellular ceramide levels, either by application of an exogenous C2 ceramide analogue or bacterial sphingomyelinase that induces endogenous ceramide release from sphingomyelin, mimicked Abeta25-35 cytotoxicity in both CECs and C6 glioma cells. Abeta25-35-induced synthesis of ceramide was selectively mediated by activation of neutral sphingomyelinase (nSMase), but not acidic sphingomyelinase (aSMase) or ceramide synthase. Both 3-O-Me-SM and N-acetyl-L-cysteine, the selective and nonselective pharmacological inhibitors of nSMase, respectively, suppressed nSMase activation, ceramide production, and cytotoxic action induced by Abeta25-35 in CECs. Furthermore, genetic knockdown of nSMase by an antisense strategy rendered C6 glioma cells specifically resistant to Abeta25-35 cytotoxicity without affecting their vulnerability to serum deprivation. Together, nSMase activation with subsequent ceramide production may contribute, at least partially, to Abeta25-35 cytotoxicity in cell types with cerebral endothelial and glial lineage.
Collapse
Affiliation(s)
- Ding-I Yang
- Institute of Neuroscience, Tzu Chi University, Hualien 970, Taiwan, ROC
| | | | | | | | | |
Collapse
|
33
|
Farkas IG, Czigner A, Farkas E, Dobó E, Soós K, Penke B, Endrész V, Mihály A. Beta-amyloid peptide-induced blood-brain barrier disruption facilitates T-cell entry into the rat brain. Acta Histochem 2004; 105:115-25. [PMID: 12831163 DOI: 10.1078/0065-1281-00696] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Activated T-lymphocytes can migrate through the blood-brain barrier (BBB) and are able to invade the central nervous system (CNS). In the present study, we investigated whether disruption of the BBB leads to enhanced T-cell migration into the CNS. Amyloid-beta peptide 25-35 (A beta) or tumor necrosis factor-alpha (TNFalpha) were administered into the right common carotid artery of adult male Wistar rats. The agents were administered either alone, or were followed by a cell suspension of exogenously activated T-cells. Rats of other groups received activated or non-stimulated T-lymphocytes only. Sagittal brain sections were analyzed with immunohistochemistry of CD3 to reveal the presence of T-lymphocytes within the CNS parenchyma. Administration of activated T-cells alone led to T-cell migration into the brain. Infusion of either substances (A beta or TNFalpha) resulted in T-cell invasion of the CNS even when no exogenous T-cells were added. Infusion of either of the agents together with T-lymphocytes generated a more intense T-lymphocyte migration than in the other groups. Electron microscopic analysis and Evans-blue extravasation studies confirmed parallel disruption of the BBB. Our study demonstrates that A beta and TNFalpha induce enhanced T-lymphocyte migration towards the brain. This effect may be attributed at least partly to dysfunctioning of the BBB, but other mechanisms are also possible.
Collapse
Affiliation(s)
- Ibolya G Farkas
- Department of Anatomy, Histology and Embryology, University of Szeged, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Cerebral amyloid angiopathy (CAA) is the result of the deposition of an amyloidogenic protein in cortical and leptomeningeal vessels. The most common type of CAA is caused by amyloid beta-protein (Abeta), which is particularly associated with Alzheimer's disease (AD). Excessive Abeta-CAA formation can be caused by several mutations in the Abeta precursor protein and presenilin genes. The origin of Abeta in CAA is likely to be neuronal, although cerebrovascular cells or the circulation cannot be excluded as a source. Despite the apparent similarity, the pathogenesis of CAA appears to differ from that of senile plaques in several aspects, including the mechanism of Abeta-induced cellular toxicity, the extent of inflammatory reaction and the role of oxidative stress. Therefore, therapeutic strategies for AD should, at least in part, also target CAA. Moreover, CAA and cerebrovascular disease (CVD) may set a lower threshold for AD-like changes to cause dementia and may even cause dementia on its own, since patients with AD and CAA and/or CVD appear to be more cognitively impaired than patients with only AD. In conclusion, the precise impact of CAA on AD or dementia remains unclear, however, its role may have been underestimated in the past, and more extensive studies of in vitro and in vivo models for CAA will be needed to elucidate the importance of CAA-specific approaches in designing intervention strategies for AD.
Collapse
Affiliation(s)
- Annemieke A M Rensink
- Department of Neurology, Laboratory of Pediatrics and Neurology, University Medical Center, 319, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
35
|
de Boer AG, van der Sandt ICJ, Gaillard PJ. The role of drug transporters at the blood-brain barrier. Annu Rev Pharmacol Toxicol 2003; 43:629-56. [PMID: 12415123 DOI: 10.1146/annurev.pharmtox.43.100901.140204] [Citation(s) in RCA: 140] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic interface between the blood and the brain. It eliminates (toxic) substances from the endothelial compartment and supplies the brain with nutrients and other (endogenous) compounds. It can be considered as an organ protecting the brain and regulating its homeostasis. Until now, many transport systems have been discovered that play an important role in maintaining BBB integrity and brain homeostasis. In this review, we focus on the role of carrier- and receptor-mediated transport systems (CMT, RMT) at the BBB. These include CMT systems, such as P-glycoprotein, multidrug-resistance proteins 1-7, nucleoside transporters, organic anion transporters, and large amino-acid transporters; RMT systems, such as the transferrin-1 and -2 receptors; and the scavenger receptors SB-AI and SB-BI.
Collapse
Affiliation(s)
- A G de Boer
- Blood-Brain Barrier Research Group, Division of Pharmacology, Leiden/Amsterdam Center for Drug Research, University of Leiden, The Netherlands.
| | | | | |
Collapse
|
36
|
Abstract
A new therapeutic approach is being developed for the treatment of Alzheimer's disease (AD). This approach involves the deliberate induction of an autoimmune response to amyloid-beta (Abeta) peptide, the constituent of neuritic plaques that is thought to cause the neurodegeneration and dementia in AD. If this approach is to be effective, antibodies must be produced that can selectively target the toxic forms of Abeta, while leaving the functionally-relevant forms of Abeta and its precursor protein untouched. Furthermore, an approach needs to be found that avoids provoking an acute neuroinflammatory response. The situation is made even more challenging by uncertainty regarding which isoforms of Abeta contribute to the pathogenesis of AD.
Collapse
Affiliation(s)
- Stephen R Robinson
- School of Psychology, Psychiatry and Psychological Medicine, Monash University, Australia.
| | | | | |
Collapse
|
37
|
Robinson SR, Bishop GM. Abeta as a bioflocculant: implications for the amyloid hypothesis of Alzheimer's disease. Neurobiol Aging 2002; 23:1051-72. [PMID: 12470802 DOI: 10.1016/s0197-4580(01)00342-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Research into Alzheimer's disease (AD) has been guided by the view that deposits of fibrillar amyloid-beta peptide (Abeta) are neurotoxic and are largely responsible for the neurodegeneration that accompanies the disease. This 'amyloid hypothesis' has claimed support from a wide range of molecular, genetic and animal studies. We critically review these observations and highlight inconsistencies between the predictions of the amyloid hypothesis and the published data. We show that the data provide equal support for a 'bioflocculant hypothesis', which posits that Abeta is normally produced to bind neurotoxic solutes (such as metal ions), while the precipitation of Abeta into plaques may be an efficient means of presenting these toxins to phagocytes. We conclude that if the deposition of Abeta represents a physiological response to injury then therapeutic treatments aimed at reducing the availability of Abeta may hasten the disease process and associated cognitive decline in AD.
Collapse
Affiliation(s)
- Stephen R Robinson
- Department of Psychology, Monash University, Clayton, Vic. 3800, Australia.
| | | |
Collapse
|
38
|
Pan W, Solomon B, Maness LM, Kastin AJ. Antibodies to beta-amyloid decrease the blood-to-brain transfer of beta-amyloid peptide. Exp Biol Med (Maywood) 2002; 227:609-15. [PMID: 12192102 DOI: 10.1177/153537020222700808] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Amyloid-beta peptides (Abeta) play an important role in the pathophysiology of dementia of the Alzheimer's type and in amyloid angiopathy. Abeta outside the CNS could contribute to plaque formation in the brain where its entry would involve interactions with the blood-brain barrier (BBB). Effective antibodies to Abeta have been developed in an effort to vaccinate against Alzheimer's disease. These antibodies could interact with Abeta in the peripheral blood, block the passage of Abeta across the BBB, or prevent Abeta deposition within the CNS. To determine whether the blocking antibodies act at the BBB level, we examined the influx of radiolabeled Abeta (125I-Abeta(1-40)) into the brain after ex-vivo incubation with the antibodies. Antibody mAb3D6 (élan Company) reduced the blood-to-brain influx of Abeta after iv bolus injection. It also significantly decreased the accumulation of Abeta in brain parenchyma. To confirm the in-vivo study and examine the specificity of mAb3D6, in-situ brain perfusion in serum-free buffer was performed after incubation of 125I-Abeta(1-40) with another antibody mAbmc1 (DAKO Company). The presence of mAbmc1 also caused significant reduction of the influx of Abeta into the brain after perfusion. Therefore, effective antibodies to Abeta can reduce the influx of Abeta(1-40) into the brain.
Collapse
Affiliation(s)
- Weihong Pan
- Veterans Affairs Medical Center and Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA.
| | | | | | | |
Collapse
|
39
|
Abstract
Effective pharmacological treatment of cognitive disorders in dementia is lacking despite extensive efforts to produce active therapy aimed at neuronal and vascular targets. In this review, the evidence for the involvement of vascular mechanisms in the pathology and evolution of dementia will be examined and the potential importance of age-related changes in cerebrovascular structure and cerebral blood flow (CBF) autoregulation will be discussed. With a description of recent clinical results (on statins, angiotensin-converting enzyme inhibitors and Ca(2+) channel blockers) and experimental results (on beta-amyloid), the impact of drugs on cerebrovascular targets is examined. The working hypothesis that targeting vascular mechanisms in dementia is an option for future therapy is proposed.
Collapse
Affiliation(s)
- J Atkinson
- Cardiovascular Research Group Nancy (EA 3116), Pharmacy Faculty, Henri Poincaré University, 54000, Nancy, France.
| |
Collapse
|
40
|
Abstract
The aging of the central nervous system and the development of incapacitating neurological diseases like Alzheimer's disease (AD) are generally associated with a wide range of histological and pathophysiological changes eventually leading to a compromised cognitive status. Although the diverse triggers of the neurodegenerative processes and their interactions are still the topic of extensive debate, the possible contribution of cerebrovascular deficiencies has been vigorously promoted in recent years. Various forms of cerebrovascular insufficiency such as reduced blood supply to the brain or disrupted microvascular integrity in cortical regions may occupy an initiating or intermediate position in the chain of events ending with cognitive failure. When, for example, vasoconstriction takes over a dominating role in the cerebral vessels, the perfusion rate of the brain can considerably decrease causing directly or through structural vascular damage a drop in cerebral glucose utilization. Consequently, cerebral metabolism can suffer a setback leading to neuronal damage and a concomitant suboptimal cognitive capacity. The present review focuses on the microvascular aspects of neurodegenerative processes in aging and AD with special attention to cerebral blood flow, neural metabolic changes and the abnormalities in microvascular ultrastructure. In this context, a few of the specific triggers leading to the prominent cerebrovascular pathology, as well as the potential neurological outcome of the compromised cerebral microvascular system are also going to be touched upon to a certain extent, without aiming at total comprehensiveness. Finally, a set of animal models are going to be presented that are frequently used to uncover the functional relationship between cerebrovascular factors and the damage to neural networks.
Collapse
Affiliation(s)
- E Farkas
- Department of Animal Physiology, Graduate School of Behavioral and Cognitive Neurosciences, University of Groningen, P.O. Box 14, 9750 AA Haren, The Netherlands.
| | | |
Collapse
|
41
|
|
42
|
Poduslo JF, Curran GL, Wengenack TM, Malester B, Duff K. Permeability of proteins at the blood-brain barrier in the normal adult mouse and double transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2001; 8:555-67. [PMID: 11493021 DOI: 10.1006/nbdi.2001.0402] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The permeability of albumin, insulin, and human A beta 1--40 at the blood-brain barrier (BBB) was determined in the normal adult mouse (B6/SJL) and in the double transgenic Alzheimer mouse (APP, PS1) by using an I.V. bolus injection technique to quantify the permeability coefficient-surface area (PS) product for each protein after correction for the residual plasma volume (V(p)) occupied by the protein in the blood vessels of different brain regions using a second aliquot of the same protein radiolabeled with a different isotope of iodine ((125)I vs (131)I) as a vascular space marker. This technology for quantifying BBB permeability of proteins was adapted from the rat to the mouse and involved catheterizing the femoral artery and vein of the mouse instead of the brachial artery and vein as for the rat. Because of the smaller blood volume in the mouse, serial sampling (20 microl) of blood from the femoral artery of the mouse was performed and directly TCA precipitated to generate a whole blood washout curve for the intact protein. When similar blood sampling techniques were used in the rat, the PS values for albumin and insulin at the BBB were similar in these two species. In the double transgenic mouse, the V(p) values for albumin were significantly increased 1.4- to 1.6-fold in five of six brain regions compared to the normal adult mouse, which indicated increased adherence of albumin to vessel walls. As a result, the PS values were significantly decreased, from 1.4- to 3.2-fold, which likely reflected decreased transport of albumin by passive diffusion. In contrast, insulin, which is taken up into the brain by a receptor-mediated transport mechanism at the BBB, showed no significant difference in the V(p) values but a significant increase in the PS values in four of six brain regions. This suggests a compensatory mechanism in the Alzheimer's transgenic brain whereby there is an increased permeability to insulin at the BBB. Surprisingly, there was no significant difference in the V(p) or PS values for human A beta 1--40 at the BBB in the double transgenic Alzheimer mouse at 24, 32, or 52 weeks of age, when there is both significant A beta levels in the plasma and amyloid burden in the brains of these animals. These data suggest that there is not an alteration in permeability to human A beta 1--40 at the BBB with increasing amyloid burden in the double transgenic Alzheimer mouse. Although these observations suggest structural alterations at the BBB, they do not support the concept of extensive BBB damage with substantial increases in BBB permeability in Alzheimer's disease.
Collapse
Affiliation(s)
- J F Poduslo
- Molecular Neurobiology Laboratory, Department of Neurlogy, Mayo Clinic and Foundation, Rochester, Minnesota, 55905, USA.
| | | | | | | | | |
Collapse
|
43
|
Abstract
The amyloid precursor protein (APP) gene and its protein products have multiple functions in the central nervous system and fulfil criteria as neuractive peptides: presence, release and identity of action. There is increased understanding of the role of secretases (proteases) in the metabolism of APP and the production of its peptide fragments. The APP gene and its products have physiological roles in synaptic action, development of the brain, and in the response to stress and injury. These functions reveal the strategic importance of APP in the workings of the brain and point to its evolutionary significance.
Collapse
Affiliation(s)
- P K Panegyres
- Department of Neuropathology, Royal Perth Hospital, Western Australia.
| |
Collapse
|
44
|
Banks WA, Farr SA, Morley JE. Permeability of the blood-brain barrier to albumin and insulin in the young and aged SAMP8 mouse. J Gerontol A Biol Sci Med Sci 2000; 55:B601-6. [PMID: 11129390 DOI: 10.1093/gerona/55.12.b601] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The decrease in the insulin cerebrospinal fluid/serum ratio seen in Alzheimer's disease has been suggested as a mechanism by which brain glucose utilization could be perturbed. Insulin is transported across the blood-brain barrier (BBB) by a system that is altered by pathophysiological events. We used SAMP8 mice, a strain that by 8-12 months of age develops severe deficits in learning and memory, to determine whether the insulin transporter or BBB integrity was altered with aging. BBB integrity was measured by injecting radioactive albumin intravenously, washing out the vascular space up to 17 hours later, and measuring brain/serum ratios. This very sensitive method found no increase in the permeability of the BBB to albumin in young and aged SAMP8 mice. This compares with previous studies in humans with Alzheimer's disease and in other colonies of SAMP8 mice that have found evidence for BBB disruption. For radioactively labeled insulin, we used multiple-time regression analysis to measure both the unidirectional influx rate (Ki) and the reversible binding to brain endothelium (Vi). A non-significant decrease in the transport rate for whole brain occurred in aged SAMP8 mice. Ki and Vi values significantly varied among brain regions and the Ki for the thalamus and the Vi for the cerebellum and thalamus were higher in aged mice. We conclude that alterations in BBB integrity or the activity of the BBB insulin transporter do not underlie the deficits in learning and memory seen in the aged SAMP8 mouse.
Collapse
Affiliation(s)
- W A Banks
- GRECC, Veterans Affairs Medical Center-St. Louis and Saint Louis University School of Medicine, Department of Internal Medicine, Missouri, USA.
| | | | | |
Collapse
|
45
|
Harkany T, Penke B, Luiten PG. beta-Amyloid excitotoxicity in rat magnocellular nucleus basalis. Effect of cortical deafferentation on cerebral blood flow regulation and implications for Alzheimer's disease. Ann N Y Acad Sci 2000; 903:374-86. [PMID: 10818528 DOI: 10.1111/j.1749-6632.2000.tb06389.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease is the most common type of dementia with a still largely unclear etiopathology. One of the factors that may directly contribute to the development and progression of the disorder is the abundant accumulation of beta-amyloid peptides (A beta) in senile plaques. In the present account we review coherent in vivo experimental evidence that A beta infusion into the rat magnocellular nucleus basalis (MBN) induces abrupt and persistent behavioral dysfunctions, perturbations of sensory information processing, storage, and retrieval. These substantial behavioral changes are due to the loss of cholinergic neurons in the MBN and their ascending projections to the frontoparietal cortex. Both neuroanatomical and neurochemical observations pin-point that infusion of A beta into the rat basal forebrain significantly decreases choline-acetyltransferase and acetylcholinesterase activities and the population of--probably--M2 muscarinic acetylcholine receptors in the cerebral cortex. Neuropharmacological data indicate that A beta toxicity is mediated by an excitotoxic cascade involving blockade of astroglial glutamate uptake, sustained activation of N-methyl-D-aspartate receptors and an overt intracellular Ca2+ influx. These changes are associated with increased nitric oxide synthase activity in cortical target areas that may directly lead to the generation of free radicals. Besides, as microvessels of the neocortex receive direct input from the MBN we assume that the loss of cholinergic innervation and hence that of tonic cholinergic vasoregulation ultimately leads to disturbances of vascular (endothelial) function and nutrient supply that may directly enhance neuronal vulnerability during aging and in Alzheimer's disease.
Collapse
Affiliation(s)
- T Harkany
- Department of Animal Physiology, University of Groningen, Haren, The Netherlands.
| | | | | |
Collapse
|
46
|
Halliday G, Robinson SR, Shepherd C, Kril J. Alzheimer's disease and inflammation: a review of cellular and therapeutic mechanisms. Clin Exp Pharmacol Physiol 2000; 27:1-8. [PMID: 10696521 DOI: 10.1046/j.1440-1681.2000.03200.x] [Citation(s) in RCA: 141] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
1. Of the neurodegenerative diseases that cause dementia, Alzheimer's disease (AD) is the most common. Three major pathologies characterize the disease: senile plaques, neurofibrillary tangles and inflammation. We review the literature on events contributing to the inflammation and the treatments thought to target this pathology. 2. The senile plaques of AD consist primarily of complexes of the beta-amyloid protein. This protein is central to the pathogenesis of the disease. 3. Inflammatory microglia are consistently associated with senile plaques in AD, although the classic inflammatory response (immunoglobulin and leucocyte infiltration) is absent. beta-Amyloid fragments appear to mediate such inflammatory mechanisms by activating the complement pathway in a similar fashion to immunoglobulin. 4. Epidemiological studies have identified a reduced risk of AD in patients with arthritis and in leprosy patients treated with anti-inflammatory drugs. Longitudinal studies have shown that the consumption of anti-inflammatory medications reduces the risk of AD only in younger patients (< 75 years). 5. There is a considerable body of in vitro evidence indicating that the inflammatory response of microglial cells is reduced by non-steroidal anti-inflammatory drugs (NSAID). However, no published data are available concerning the effects of these medications on brain pathology in AD. 6. Cyclo-oxygenase 2 enzyme is constitutively expressed in neurons and is up-regulated in degenerative brain regions in AD. Non-steroidal anti-inflammatory drugs may reduce this expression. 7. Platelets are a source of beta-amyloid and increased platelet activation and increased circulating beta-amyloid have been identified in AD. Anti-platelet medication (including NSAID) would prevent such activation and its potentially harmful consequences. 8. Increased levels of luminal beta-amyloid permeabilizes the blood-brain barrier (BBB) and increases vasoconstriction of arterial vessels, paralleling the alterations observed with infection and inflammation. Cerebral amyloidosis is highly prevalent in AD, compromising the BBB and vasoactivity. Anti-inflammatory medications may alleviate these problems.
Collapse
Affiliation(s)
- G Halliday
- Prince of Wales Medical Research Institute, Randwick, Australia.
| | | | | | | |
Collapse
|
47
|
Harkany T, Hortobágyi T, Sasvári M, Kónya C, Penke B, Luiten PG, Nyakas C. Neuroprotective approaches in experimental models of beta-amyloid neurotoxicity: relevance to Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry 1999; 23:963-1008. [PMID: 10621945 DOI: 10.1016/s0278-5846(99)00058-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
1. beta-Amyloid peptides (A beta s) accumulate abundantly in the Alzheimer's disease (AD) brain in areas subserving information acquisition and processing, and memory formation. A beta fragments are produced in a process of abnormal proteolytic cleavage of their precursor, the amyloid precursor protein (APP). While conflicting data exist in the literature on the roles of A beta s in the brain, and particularly in AD, recent studies have provided firm experimental evidence for the direct neurotoxic properties of A beta. 2. Sequence analysis of A beta s revealed a high degree of evolutionary conservation and inter-species homology of the A beta amino acid sequence. In contrast, synthetic A beta fragments, even if modified fluorescent or isotope-labeled derivatives, are pharmacological candidates for in vitro and in vivo modeling of their cellular actions. During the past decade, acute injection, prolonged mini-osmotic brain perfusion approaches or A beta infusions into the blood circulation were developed in order to investigate the effects of synthetic A beta s, whereas transgenic models provided insight into the distinct molecular steps of pathological APP cleavage. 3. The hippocampus, caudate putamen, amygdala and neocortex all formed primary targets of acute neurotoxicity screening, but functional consequences of A beta infusions were primarily demonstrated following either intracerebroventricular or basal forebrain (medial septum or magnocellular basal nucleus (MBN)) infusions of A beta fragments. 4. In vivo investigations confirmed that, while the active core of A beta is located within the beta(25-35) sequence, the flanking peptide regions influence not only the folding properties of the A beta fragments, but also their in vivo neurotoxic potentials. 5. It has recently been established that A beta administration deranges neuron-glia signaling, affects the glial glutamate uptake and thereby induces noxious glutamatergic stimulation of nerve cells. In fact, a critical role for N-methyl-D-aspartate (NMDA) receptors was postulated in the neurotoxic processes. Additionally, A beta s might become internalized, either after their selective binding to cell-surface receptors or after membrane association in consequence of their highly lipophilic nature, and induce free radical generation and subsequent oxidative injury. Ca(2+)-mediated neurotoxic events and generation of oxygen free radicals may indeed potentiate each other, or even converge to the same neurotoxic events, leading to cell death. 6. Neuroprotection against A beta toxicity was achieved by both pre- and post-treatment with NMDA receptor channel antagonists. Moreover, direct radical-scavengers, such as vitamin E or vitamin C, attenuated A beta toxicity with high efficacy. Interestingly, combined drug treatments did not necessarily result in additive enhanced neuroprotection. 7. Similarly to the blockade of NMDA receptors, the neurotoxic action of A beta s could be markedly decreased by pharmacological manipulation of voltage-dependent Ca(2+)-channels, serotonergic IA or adenosine A1 receptors, and by drugs eliciting membrane hyperpolarization or indirect blockade of Ca(2+)-mediated intracellular consequences of intracerebral A beta infusions. 8. A beta neurotoxicity might be dose-dependently modulated by trace metals. In spite of the fact that zinc (Zn) may act as a potent inhibitor of the NMDA receptor channel, high Zn doses accelerate A beta fibril formation, stabilize the beta-sheet conformation and thereby potentiate A beta neurotoxicity. Combined trace element supplementation with Se, Mn, or Mg, which prevails over the expression of detoxifying enzymes or counteracts intracellular elevations of Ca2+, may reduce the neurotoxic impact of A beta s. 9. Alterations in the regulatory functions of the hypothalamo-pituitary-adrenal axis may contribute significantly to neurodegenerative changes in the brain. Furthermore, AD patients exhibit substantially increased circadia
Collapse
Affiliation(s)
- T Harkany
- Central Research Division of Clinical and Experimental Laboratory Medicine, Haynal Imre University of Health Sciences, Budapest, Hungary.
| | | | | | | | | | | | | |
Collapse
|