1
|
Ding Y, Palecek SP, Shusta EV. iPSC-derived blood-brain barrier modeling reveals APOE isoform-dependent interactions with amyloid beta. Fluids Barriers CNS 2024; 21:79. [PMID: 39394110 PMCID: PMC11468049 DOI: 10.1186/s12987-024-00580-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Three common isoforms of the apolipoprotein E (APOE) gene - APOE2, APOE3, and APOE4 - hold varying significance in Alzheimer's Disease (AD) risk. The APOE4 allele is the strongest known genetic risk factor for late-onset Alzheimer's Disease (AD), and its expression has been shown to correlate with increased central nervous system (CNS) amyloid deposition and accelerated neurodegeneration. Conversely, APOE2 is associated with reduced AD risk and lower CNS amyloid burden. Recent clinical data have suggested that increased blood-brain barrier (BBB) leakage is commonly observed among AD patients and APOE4 carriers. However, it remains unclear how different APOE isoforms may impact AD-related pathologies at the BBB. METHODS To explore potential impacts of APOE genotypes on BBB properties and BBB interactions with amyloid beta, we differentiated isogenic human induced pluripotent stem cell (iPSC) lines with different APOE genotypes into both brain microvascular endothelial cell-like cells (BMEC-like cells) and brain pericyte-like cells. We then compared the effect of different APOE isoforms on BBB-related and AD-related phenotypes. Statistical significance was determined via ANOVA with Tukey's post hoc testing as appropriate. RESULTS Isogenic BMEC-like cells with different APOE genotypes had similar trans-endothelial electrical resistance, tight junction integrity and efflux transporter gene expression. However, recombinant APOE4 protein significantly impeded the "brain-to-blood" amyloid beta 1-40 (Aβ40) transport capabilities of BMEC-like cells, suggesting a role in diminished amyloid clearance. Conversely, APOE2 increased amyloid beta 1-42 (Aβ42) transport in the model. Furthermore, we demonstrated that APOE-mediated amyloid transport by BMEC-like cells is dependent on LRP1 and p-glycoprotein pathways, mirroring in vivo findings. Pericyte-like cells exhibited similar APOE secretion levels across genotypes, yet APOE4 pericyte-like cells showed heightened extracellular amyloid deposition, while APOE2 pericyte-like cells displayed the least amyloid deposition, an observation in line with vascular pathologies in AD patients. CONCLUSIONS While APOE genotype did not directly impact general BMEC or pericyte properties, APOE4 exacerbated amyloid clearance and deposition at the model BBB. Conversely, APOE2 demonstrated a potentially protective role by increasing amyloid transport and decreasing deposition. Our findings highlight that iPSC-derived BBB models can potentially capture amyloid pathologies at the BBB, motivating further development of such in vitro models in AD modeling and drug development.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, 1415 Engineering Drive, Madison, WI, 53706, USA.
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Boese AC, Hamblin MH, Lee JP. Neural stem cell therapy for neurovascular injury in Alzheimer's disease. Exp Neurol 2019; 324:113112. [PMID: 31730762 DOI: 10.1016/j.expneurol.2019.113112] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/02/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by progressive neurodegeneration leading to severe cognitive decline and eventual death. AD pathophysiology is complex, but neurotoxic accumulation of amyloid-β (Aβ) and hyperphosphorylation of Tau are believed to be main drivers of neurodegeneration in AD. The formation and deposition of Aβ plaques occurs in the brain parenchyma as well as in the cerebral vasculature. Thus, proper blood-brain barrier (BBB) and cerebrovascular functioning are crucial for clearance of Aβ from the brain, and neurovascular dysfunction may be a critical component of AD development. Further, neuroinflammation and dysfunction of angiogenesis, neurogenesis, and neurorestorative capabilities play a role in AD pathophysiology. Currently, there is no effective treatment to prevent or restore loss of brain tissue and cognitive decline in patients with AD. Based on multifactorial and complex pathophysiological cascades in multiple Alzheimer's disease stages, effective AD therapies need to focus on targeting early AD pathology and preserving cerebrovascular function. Neural stem cells (NSCs) participate extensively in mammalian brain homeostasis and repair and exhibit pleiotropic intrinsic properties that likely make them attractive candidates for the treatment of AD. In the review, we summarize the current advances in knowledge regarding neurovascular aspects of AD-related neurodegeneration and discuss multiple actions of NSCs from preclinical studies of AD to evaluate their potential for future clinical treatment of AD.
Collapse
Affiliation(s)
- Austin C Boese
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Milton H Hamblin
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Jean-Pyo Lee
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, USA; Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA.
| |
Collapse
|
3
|
Morris AWJ, Carare RO, Schreiber S, Hawkes CA. The Cerebrovascular Basement Membrane: Role in the Clearance of β-amyloid and Cerebral Amyloid Angiopathy. Front Aging Neurosci 2014; 6:251. [PMID: 25285078 PMCID: PMC4168721 DOI: 10.3389/fnagi.2014.00251] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 09/05/2014] [Indexed: 11/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), the accumulation of β-amyloid (Aβ) peptides in the walls of cerebral blood vessels, is observed in the majority of Alzheimer’s disease (AD) brains and is thought to be due to a failure of the aging brain to clear Aβ. Perivascular drainage of Aβ along cerebrovascular basement membranes (CVBMs) is one of the mechanisms by which Aβ is removed from the brain. CVBMs are specialized sheets of extracellular matrix that provide structural and functional support for cerebral blood vessels. Changes in CVBM composition and structure are observed in the aged and AD brain and may contribute to the development and progression of CAA. This review summarizes the properties of the CVBM, its role in mediating clearance of interstitial fluids and solutes from the brain, and evidence supporting a role for CVBM in the etiology of CAA.
Collapse
Affiliation(s)
- Alan W J Morris
- Faculty of Medicine, Clinical and Experimental Sciences, University of Southampton , Southampton , UK
| | - Roxana O Carare
- Faculty of Medicine, Clinical and Experimental Sciences, University of Southampton , Southampton , UK
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University , Magdeburg , Germany ; German Center for Neurodegenerative Diseases (DZNE), Helmholtz Association , Magdeburg , Germany
| | - Cheryl A Hawkes
- Faculty of Medicine, Clinical and Experimental Sciences, University of Southampton , Southampton , UK
| |
Collapse
|
4
|
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: 3.9] [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
|
5
|
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: 5.9] [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
|
6
|
|
7
|
Kolialexi A, Tsangaris GT, Papantoniou N, Anagnostopoulos AK, Vougas K, Bagiokos V, Antsaklis A, Mavrou A. Application of proteomics for the identification of differentially expressed protein markers for Down syndrome in maternal plasma. Prenat Diagn 2008; 28:691-8. [PMID: 18551720 DOI: 10.1002/pd.2040] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Despite the large impact of ultrasonographic and biochemical markers on prenatal screening, the ability to accurately diagnose Down syndrome (DS) is still limited and better diagnostic testing is needed. METHODS Plasma from 8 women carrying a DS foetus and 12 with non-DS foetuses matched for gestational age, maternal age and ethnicity, in the second trimester of pregnancy, was analysed by two-dimensional gel electrophoresis (2-DE) and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS) in order to identify biomarkers for DS. RESULTS Gel comparison revealed nine proteins differentially expressed in maternal plasma in women with DS foetuses. Eight proteins, transthyretin (TTHY), ceruloplasmin (CERU), afamin (AFAM), alpha-1-microglobulin (AMBP), apolipoprotein E (APOE), serum amyloid P-component (SAMP), histidine-rich glycoprotein (HRG) and alpha-1-antitrypsin (A1AT) were up-regulated and one, clusterin (CLUS), down-regulated. All nine proteins are known to be involved in foetal growth and development. APOE, SAMP, AFAM and CLUS are associated with the DS phenotype. Western blot and densitometric analysis of APOE and SAMP confirmed the increase of both proteins by 19 and 48% respectively. CONCLUSIONS All differentially expressed proteins are candidate biomarkers for DS, providing opportunities for the development of non-invasive prenatal diagnosis. As these are preliminary findings, follow-up experiments are needed for their evaluation.
Collapse
|
8
|
Lee JA, Cole GJ. Generation of Transgenic Zebrafish Expressing Green Fluorescent Protein Under Control of Zebrafish Amyloid Precursor Protein Gene Regulatory Elements. Zebrafish 2007; 4:277-86. [DOI: 10.1089/zeb.2007.0516] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ju-Ahng Lee
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
| | - Gregory J. Cole
- Julius L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, North Carolina
- Department of Biology, North Carolina Central University, Durham, North Carolina
| |
Collapse
|
9
|
Rostagno A, Lashley T, Ng D, Meyerson J, Braendgaard H, Plant G, Bojsen-Møller M, Holton J, Frangione B, Revesz T, Ghiso J. Preferential association of serum amyloid P component with fibrillar deposits in familial British and Danish dementias: Similarities with Alzheimer's disease. J Neurol Sci 2007; 257:88-96. [PMID: 17374542 DOI: 10.1016/j.jns.2007.01.050] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Two hereditary forms of cerebrovascular amyloidosis, familial British and Danish dementias (FBD and FDD), share striking similarities with Alzheimer's disease (AD) despite structural differences among their amyloid subunits (ABri in FBD, ADan in FDD, and Abeta in AD). Neuropathological lesions in these disorders include neurofibrillary tangles, parenchymal amyloid and pre-amyloid deposits and overwhelming cerebral amyloid angiopathy co-localizing with reactive microglia and multiple amyloid associated proteins including activation products of the complement cascade. Immunohistochemical analysis of FBD and FDD brain lesions unveiled the presence of serum amyloid P-component (SAP) primarily associated with thioflavin positive amyloid deposits in spite of the significant pre-amyloid burden existing in both disorders. Using affinity chromatography and ELISA binding assays we demonstrated specific, calcium-dependent, saturable, high affinity binding interactions between SAP and ABri/ADan peptides, with dissociation constant values in the sub-nanomolar range and within the same order of magnitude as those resulting from the interaction of SAP with Alzheimer's Abeta1-40 and Abeta1-42. The preferential association of SAP with fibrillar amyloid lesions and not with non-fibrillar pre-amyloid deposits is puzzling, suggesting that SAP modulates the assembly and stability of the final fibril rather than participating in the early steps of protein misfolding and oligomerization.
Collapse
Affiliation(s)
- Agueda Rostagno
- Department of Pathology, New York University School of Medicine, New York 10016, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kisilevsky R, Ancsin JB, Szarek WA, Petanceska S. Heparan sulfate as a therapeutic target in amyloidogenesis: prospects and possible complications. Amyloid 2007; 14:21-32. [PMID: 17453622 DOI: 10.1080/13506120601116419] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Amyloid formation in vivo is a much more complicated process than studies of in vitro protein/peptide fibrillogenesis would lead one to believe. Amyloidogenesis in vivo involves multiple components, some no less important than the amyloidogenic protein/peptides themselves, and each of these components, and its role in the pathogenetic steps toward amyloid deposition could, theoretically, be a therapeutic target. Herein we use the definition of amyloid as it was originally described, discuss the similarities and differences between amyloid in vivo and in vitro, address the potential role of the extracellular matrix in in vivo amyloidogenesis by focusing on a specific component, namely heparan sulfate proteoglycan, and describe studies illustrating that heparan sulfate is a valid target for anti-amyloid therapy. In light of experimental and recent clinical results obtained from studies addressing heparan sulfate's role in amyloid deposition additional novel anti-amyloid therapeutic targets will be proposed. Lastly, given the multiple roles that heparan sulfate plays in organ development, and organ and cell function, potential side effects of targeting heparan sulfate biosynthesis for therapeutic purposes are considered.
Collapse
Affiliation(s)
- Robert Kisilevsky
- Department of Pathology and Molecular Medicine,Queen's University, Kingston General Hospital, Ontario, Canada.
| | | | | | | |
Collapse
|
11
|
Yin KJ, Hsu CY, Hu XY, Chen H, Chen SW, Xu J, Lee JM. Protein phosphatase 2A regulates bim expression via the Akt/FKHRL1 signaling pathway in amyloid-beta peptide-induced cerebrovascular endothelial cell death. J Neurosci 2006; 26:2290-9. [PMID: 16495456 PMCID: PMC6674809 DOI: 10.1523/jneurosci.5103-05.2006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Amyloid-beta peptide (Abeta)-induced death in cerebral endothelial cells (CECs) is preceded by mitochondrial dysfunction and signaling events characteristic of apoptosis. Mitochondria-dependent apoptosis engages Bcl-2 family proteins, especially the BH3-only homologues, which play a key role in initiating the apoptotic cascade. Here, we report that the expression of bim, but not other BH3-only members, was selectively increased in cerebral microvessels isolated from 18-month-old APPsw (Tg2576) mice, a model of cerebral amyloid angiopathy (CAA), suggesting a pivotal role for Bim in Abeta-induced cerebrovascular degeneration in vivo. A similar expression profile was observed in Abeta-treated CECs. Furthermore, Abeta induction of bim expression involved a pro-apoptotic transcription factor, FKHRL1. FKHRL1 bound to a consensus sequence in the bim promoter region and was activated by Abeta before bim expression. FKHRL1 activity was negatively regulated by phosphorylation catalyzed by Akt, an anti-apoptotic kinase. Akt upregulation by adenoviral gene transfer inhibited Abeta-induced FKHRL1 activation and bim induction. In addition, Abeta increased the activity of protein phosphatase 2A (PP2A), a ceramide-activated protein phosphatase. Suppression of PP2A activity by RNA interference or a specific inhibitor, okadaic acid, effectively suppressed Abeta-induced Akt inactivation and FKHRL1 activation, leading to an attenuation of bim expression and cell death in CECs. Coimmunoprecipitation experiments revealed that Abeta enhanced the binding of the PP2A regulatory subunit PP2ACalphabeta to Akt. These results implicate PP2A as an early regulator of Abeta-induced bim expression and CEC apoptosis via the Akt/FKHRL1 signaling pathway. We raise the possibility that this pathway may play a role in cerebrovascular degeneration in CAA.
Collapse
|
12
|
Yin KJ, Lee JM, Chen H, Xu J, Hsu CY. Abeta25-35 alters Akt activity, resulting in Bad translocation and mitochondrial dysfunction in cerebrovascular endothelial cells. J Cereb Blood Flow Metab 2005; 25:1445-55. [PMID: 15973355 DOI: 10.1038/sj.jcbfm.9600139] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The amyloid-beta peptide (Abeta) induces apoptosis in cerebrovascular endothelial cells (CECs), contributing to the pathogenesis of cerebral amyloid angiopathy. We have previously shown that Abeta induces apoptosis in CECs. In the present study, we report that Abeta25-35-induced CEC apoptosis involves the inactivation of Akt, a signaling kinase important in maintaining cell viability. Akt prevents the activation of death-signaling events by facilitating the inactivation of proapoptotic proteins such as Bad. We applied three strategies to show that Abeta25-35 inactivation of Akt is causally related to Abeta25-35-induced CEC death by preventing Bad activation and subsequent mitochondrial dysfunction (reflected by the release of endonuclease G and Smac, two proapoptotic intermembranous proteins of the mitochondria). Wortmannin, a PI3-kinase inhibitor, enhanced Abeta25-35-induced Bad activation, mitochondrial dysfunction and CEC death. Enhancement of Akt activity by a Tat-Akt fusion protein, or by viral gene transfer of a constitutively active mutant of akt, reduced Bad activation, mitochondrial dysfunction, and CEC death. Using a siRNA strategy to knock down the bad gene, we showed that Bad activation is causally related to Abeta25-35-induced mitochondrial dysfunction and CEC death. Together, these results establish that the Akt-Bad cascade is altered by Abeta25-35, resulting in CEC apoptosis.
Collapse
Affiliation(s)
- Ke-Jie Yin
- Department of Neurology, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
13
|
Rodrigo J, Fernández-Vizarra P, Castro-Blanco S, Bentura ML, Nieto M, Gómez-Isla T, Martínez-Murillo R, MartInez A, Serrano J, Fernández AP. Nitric oxide in the cerebral cortex of amyloid-precursor protein (SW) Tg2576 transgenic mice. Neuroscience 2004; 128:73-89. [PMID: 15450355 DOI: 10.1016/j.neuroscience.2004.06.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2004] [Indexed: 01/12/2023]
Abstract
Changes in the amyloid-peptide (Abeta), neuronal and inducible nitric oxide (NO)synthase (nNOS, iNOS), nitrotyrosine, glial fibrillary acidic protein, and lectin from Lycopersicon esculentum (tomato) were investigated in the cerebral cortex of transgenic mice (Tg2576) to amyloid precursor protein (APP), by immunohistochemistry (bright light, confocal, and electron microscopy). The expression of nitrergic proteins and synthesis of nitric oxide were analyzed by immunoblotting and NOS activity assays, respectively. The cerebral cortex of these transgenic mice showed an age-dependent progressive increase in intraneuronal aggregates of Abeta-peptide and extracellular formation of senile plaques surrounded by numerous microglial and reactive astrocytes. Basically, no changes to nNOS reactivity or expression were found in the cortical mantle of either wild or transgenic mice. This reactivity in wild mice corresponded to numerous large type I and small type II neurons. The transgenic mice showed swollen, twisted, and hypertrophic preterminal and terminal processes of type I neurons, and an increase of the type II neurons. The calcium-dependent NOS enzymatic activity was higher in wild than in the transgenic mice. The iNOS reactivity, expression and calcium-independent enzymatic activity increased in transgenic mice with respect to wild mice, and were related to cortical neurons and microglial cells. The progressive elevation of NO production resulted in a specific pattern of protein nitration in reactive astrocytes. The ultrastructural study carried out in the cortical mantle showed that the neurons contained intracellular aggregates of Abeta-peptide associated with the endoplasmic reticulum, mitochondria, and Golgi apparatus. The endothelial vascular cells also contained Abeta-peptide deposits. This transgenic model might contribute to understand the role of the nitrergic system in the biological changes related to neuropathological progression of Alzheimer's disease.
Collapse
Affiliation(s)
- J Rodrigo
- Department of Neuroanatomy and Cell Biology, Instituto Cajal, CSIC, Doctor Arce Avenue 37, 28002 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
BACKGROUND The main stumbling block in the clinical management and in the search for a cure of Alzheimer disease (AD) is that the cause of this disorder has remained uncertain until now. SUMMARY OF REVIEW Evidence that sporadic (nongenetic) AD is primarily a vascular rather than a neurodegenerative disorder is reviewed. This conclusion is based on the following evidence: (1) epidemiological studies showing that practically all risk factors for AD reported thus far have a vascular component that reduces cerebral perfusion; (2) risk factor association between AD and vascular dementia (VaD); (3) improvement of cerebral perfusion obtained from most pharmacotherapy used to reduce the symptoms or progression of AD; (4) detection of regional cerebral hypoperfusion with the use of neuroimaging techniques to preclinically identify AD candidates; (5) presence of regional brain microvascular abnormalities before cognitive and neurodegenerative changes; (6) common overlap of clinical AD and VaD cognitive symptoms; (7) similarity of cerebrovascular lesions present in most AD and VaD patients; (8) presence of cerebral hypoperfusion preceding hypometabolism, cognitive decline, and neurodegeneration in AD; and (9) confirmation of the heterogeneous and multifactorial nature of AD, likely resulting from the diverse presence of vascular risk factors or indicators of vascular disease. CONCLUSIONS Since the value of scientific evidence generally revolves around probability and chance, it is concluded that the data presented here pose a powerful argument in support of the proposal that AD should be classified as a vascular disorder. According to elementary statistics, the probability or chance that all these findings are due to an indirect pathological effect or to coincidental circumstances related to the disease process of AD seems highly unlikely. The collective data presented in this review strongly support the concept that sporadic AD is a vascular disorder. It is recommended that current clinical management of patients, treatment targets, research designs, and disease prevention efforts need to be critically reassessed and placed in perspective in light of these important findings.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neuropathology, University of California at San Diego, CA 92026, USA.
| |
Collapse
|
15
|
Yu PH. Involvement of cerebrovascular semicarbazide-sensitive amine oxidase in the pathogenesis of Alzheimer's disease and vascular dementia. Med Hypotheses 2001; 57:175-9. [PMID: 11461168 DOI: 10.1054/mehy.2001.1329] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Fibrillary tangles and senile plaques resulting from advanced aggregation of beta-amyloid and other proteins are pathological characteristics of Alzheimer's disease (AD). Cerebral amyloid angiopathy is quite common in AD. In fact, amyloid fibrils fuse to and emanate from the vascular basement membrane. Semicarbazide-sensitive amine oxidase (SSAO), located in outer membranes of vascular smooth muscles and endothelia, catalyzes deamination of methylamine-producing formaldehyde and hydrogen peroxide. SSAO is also involved in lymphocyte adhesion and is up-regulated in response to inflammation. SSAO-mediated generation of formaldehyde can induce protein (i.e. beta-amyloid) cross-linkage, deposition and subsequently plaque formation in the compartment adjacent to the cerebrovessels. Formaldehyde may cause cytotoxicity, which induces inflammation and release of more SSAO, producing a cascade of toxic cycle. Increased SSAO-mediated reaction may be chronically involved in the pathogenesis of vascular dementia and AD.
Collapse
Affiliation(s)
- P H Yu
- Neuropsychiatry Research Unit, Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
16
|
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: 3.6] [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
|
17
|
de la Torre JC. Impaired cerebromicrovascular perfusion. Summary of evidence in support of its causality in Alzheimer's disease. Ann N Y Acad Sci 2001; 924:136-52. [PMID: 11193790 DOI: 10.1111/j.1749-6632.2000.tb05572.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
After nearly a century of inquiry, the cause of Alzheimer's disease (AD) remains to be found. In this review, basic and clinical evidence is presented that assembles and hypothetically explains most of the key pathologic events associated with the development of AD. These pathologic events are triggered in AD by impaired cerebral perfusion originating in the microvasculature that affects the optimal delivery of glucose and oxygen and results in an energy metabolic breakdown of brain cell biosynthetic and synaptic pathways. We propose that two factors must be present before cognitive dysfunction and neurodegeneration is expressed in the AD brain: (1) advanced aging, (2) presence of a condition that lowers cerebral perfusion, such as a vascular risk factor. The first factor introduces a normal but potentially menacing process that lowers cerebral blood flow in proportion to increased aging, while the second factor adds a crucial burden that further lowers brain perfusion and places vulnerable neurons in a state of metabolic compromise leading to a death pathway. These two factors will lead to a critically attained threshold of cerebral hypoperfusion (CATCH). CATCH is a self-sustaining and progressive circulatory insufficiency that will destabilize neurons, synapses, neurotransmission, and cognitive function, creating in its wake a neurodegenerative process characterized by the formation of senile plaques, neurofibrillary tangles, amyloid angiopathy, and, in some cases, Lewy bodies. Since any of a considerable number of vessel-related conditions must be present in the aging individual for cognition to be affected, CATCH supports the heterogeneic disease profile assumed to be characteristic of the AD syndrome. A brief discussion of target therapy based on the proposed pathogenesis of AD is also reviewed.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neuroscience, University of California, San Diego, La Jolla, California 92093, USA.
| |
Collapse
|
18
|
de la Torre JC, Stefano GB. Evidence that Alzheimer's disease is a microvascular disorder: the role of constitutive nitric oxide. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2000; 34:119-36. [PMID: 11113503 DOI: 10.1016/s0165-0173(00)00043-6] [Citation(s) in RCA: 209] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Evidence is fast accumulating which indicates that Alzheimer's disease is a vascular disorder with neurodegenerative consequences rather than a neurodegenerative disorder with vascular consequences. It is proposed that two factors need to be present for AD to develop: (1) advanced ageing, (2) presence of a condition that lowers cerebral perfusion, such as a vascular-risk factor. The first factor introduces a normal but potentially insidious process that lowers cerebral blood flow in inverse relation to increased ageing; the second factor adds a crucial burden which further lowers brain perfusion and places vulnerable neurons in a state of high energy compromise leading to a cascade of neuronal metabolic turmoil. Convergence of the two factors above will culminate in a critically attained threshold of cerebral hypoperfusion (CATCH). CATCH is a hemodynamic microcirculatory insufficiency that will destabilize neurons, synapses, neurotransmission and cognitive function, creating in its wake a neurodegenerative state characterized by the formation of senile plaques, neurofibrillary tangles, amyloid angiopathy and in some cases, Lewy bodies. Since any of a considerable number of vascular-related conditions must be present in the ageing individual for cognition to be disturbed, CATCH identifies an important aspect of the heterogeneic disease profile assumed to be present in the AD syndrome. It is proposed that CATCH initiates AD by distorting regional brain capillary structure involving endothelial cell shape changes and impairment of nitric oxide (NO) release which affect signaling between the immune, cardiovascular and nervous systems. Evidence is presented that in many tissues there is a basal level of NO being produced and that the actions of several signaling molecules may initiate increases in basal NO levels. Moreover, these temporary increases in basal NO levels exert inhibitory cellular actions, via cellular conformational changes. Findings indicate that (a) constitutive NO is responsible for a basal or 'tonal' level of NO; (b) this NO keeps particular types of cells in a state of inhibition and (c) activation of these cells occurs through disinhibition. Consequently, tissues not maintaining a basal NO level are more prone to excitatory, immune, vascular and neural influences. Under such circumstances, these tissues cannot be down-regulated to normal basal levels, thus prolonging their excitatory state. Thus, the clinical convergence of advanced ageing in the presence of a chronic, pre-morbid vascular risk factor, can, in time, contribute to an endotheliopathy involving basal NO deficit, to the degree where regional metabolic dysfunction leads to cognitive meltdown and to progressive neurodegeneration characteristic of Alzheimer's disease.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Pathology, University of California, San Diego, 1363 Shinly, Suite 100, Escondido, CA 92026, USA.
| | | |
Collapse
|
19
|
Castillo GM, Lukito W, Peskind E, Raskind M, Kirschner DA, Yee AG, Snow AD. Laminin inhibition of beta-amyloid protein (Abeta) fibrillogenesis and identification of an Abeta binding site localized to the globular domain repeats on the laminin a chain. J Neurosci Res 2000; 62:451-62. [PMID: 11054814 DOI: 10.1002/1097-4547(20001101)62:3<451::aid-jnr15>3.0.co;2-f] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
beta-Amyloid protein (Abeta) is a major component of neuritic plaques and cerebrovascular amyloid deposits in the brains of patients with Alzheimer's disease (AD). Inhibitors of Abeta fibrillogenesis are currently sought as potential future therapeutics for AD and related disorders. In the present study, the basement membrane protein laminin was found to bind Abeta 1-40 with a single dissociation constant, K(d) = 2.7 x 10(-9) M, and serve as a potent inhibitor of Abeta fibril formation. 25 microM of Abeta 1-40 was incubated at 37 degrees C for 1 week in the presence of 100 nM of laminin or other basement membrane components, including perlecan, type IV collagen, and fibronectin to determine their effects on Abeta fibril formation as evaluated by thioflavin T fluorometry. Of all the basement membrane components tested, laminin demonstrated the greatest inhibitory effect on Abeta-amyloid fibril formation, causing a ninefold inhibition at 1 and 3 days and a 21-fold inhibition at 1 week. The inhibitory effects of laminin on Abeta fibrillogenesis occurred in a dose-dependent manner and were still effective at lower concentrations. The inhibitory effects of laminin on Abeta 1-40 fibril formation was confirmed by negative stain electron microscopy, whereby laminin caused an almost complete inhibition of Abeta fibril formation and assembly by 3 days, resulting in the appearance of primarily amorphous nonfibrillar material. Laminin also caused partial disassembly of preformed Abeta-amyloid fibrils following 4 days of coincubation. Laminin was not effective as an inhibitor of islet amyloid polypeptide fibril formation, suggesting that laminin's amyloid inhibitory effects were Abeta-specific. To identify a potential Abeta-binding site(s) on laminin, laminin was first digested with V8, trypsin, or elastase. An Abeta-binding elastase digestion product of approximately 120-130 kDa was found. In addition, a approximately 55 kDa fragment derived from V8 and elastase-digested laminin interacted with biotinylated Abeta 1-40. Amino acid sequencing of the approximately 55 kDa fragment identified a conformationally dependent Abeta-binding site within laminin localized to the globular repeats on the laminin A chain. These studies demonstrate that laminin not only binds Abeta with relatively high affinity but is a potent inhibitor of Abeta-amyloid fibril formation. In addition, further identification of an Abeta-binding domain within the globular repeats on the laminin A chain may lead to the design of new therapeutics for the inhibition of Abeta fibrillogenesis.
Collapse
Affiliation(s)
- G M Castillo
- ProteoTech Inc., Kirkland, Washington 98034, USA.
| | | | | | | | | | | | | |
Collapse
|
20
|
de la Torre JC. Cerebral hypoperfusion, capillary degeneration, and development of Alzheimer disease. Alzheimer Dis Assoc Disord 2000; 14 Suppl 1:S72-81. [PMID: 10850734 DOI: 10.1097/00002093-200000001-00012] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Considerable clinical and experimental data have shown that cerebral perfusion is progressively decreased during increased aging and that this decrease in brain blood flow is significantly greater in Alzheimer disease (AD). The authors propose that advanced aging with a comorbid condition, such as a vascular risk factor, which further decreases cerebral perfusion, promotes a critically attained threshold of cerebral hypoperfusion (CATCH). With time, CATCH induces brain capillary degeneration and suboptimal delivery of energy substrates to neuronal tissue. Because glucose is the main fuel of brain cells, its impaired delivery, with the deficient delivery of oxygen, compromises neuronal stability because the supply for aerobic glycolysis fails to meet brain tissue demand. The outcome of CATCH is a metabolic cascade that involves, among other things, mitochondrial dysfunction, oxidative stress, decreased adenosine triphosphate production, abnormal protein synthesis, cell ionic pump deficiency, signal transduction defects, and neurotransmission failure. These events contribute to the progressive cognitive decline characteristic of patients with AD, as well as regional anatomic pathology, consisting of synaptic loss, senile plaques, neurofibrillary tangles, tissue atrophy, and neurodegeneration. CATCH identifies the clinical heterogeneic pattern that characterizes AD because it provides compelling evidence that any of a multitude of different etiopathophysiologic vascular risk factors, in the presence of advanced aging, can lead to AD. The evidence in support of CATCH as the pathogenic trigger of AD is crystallized in this review.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neuroscience, University of California, San Diego, La Jolla 92093, USA
| |
Collapse
|
21
|
Abstract
Current assumptions and conclusions in several active areas of amyloid research are examined to see how consistent the data from chosen in vitro and in vivo model systems are with clinical and anatomic observations. These areas include the assembly of amyloid-like fibrils in vitro, the nucleation phenomenon, amyloid fibril structure in vivo and in vitro, common structural components of the amyloids, and the regression of tissue amyloid and proteolysis of amyloid proteins. Divergences and congruencies are highlighted, which in turn suggests caution in the interpretation of present data, greater collaboration and communication among investigators, and, additional areas and techniques for investigation.
Collapse
Affiliation(s)
- R Kisilevsky
- Department of Pathology, Queen's University, The Syl and Molly Apps Research Center, Kingston General Hospital, Ontario, Kingston, K7L 3N6, Canada
| |
Collapse
|
22
|
de la Torre JC. Critically attained threshold of cerebral hypoperfusion: can it cause Alzheimer's disease? Ann N Y Acad Sci 2000; 903:424-36. [PMID: 10818533 DOI: 10.1111/j.1749-6632.2000.tb06394.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
After nearly a century of inquiry, the cause of sporadic Alzheimer's disease (AD) remains to be found. On the subject of AD pathogenesis, recent basic and clinical evidence strongly argues in favor of the concept that AD is linked to brain circulatory pathology. This concept, when viewed from many different medical disciplines and from close pre-morbid similarities to vascular dementia, assembles and hypothetically explains most of the key pathologic events associated with the development of AD. These pathologic events are triggered in AD by impaired cerebral perfusion originating in the microvasculature which affects the optimal delivery of glucose and oxygen and results in an energy metabolic breakdown of brain cell biosynthetic and synaptic pathways. We propose that two factors converge to initiate cognitive dysfunction and neurodegeneration as expressed in AD brain: (1) advanced aging, and (2) the presence of a condition that lowers cerebral perfusion. The first factor introduces a normal but potentially deconstructing process that lowers cerebral blood flow in proportion to increased aging, whereas the second factor adds a crucial burden that further lowers brain perfusion to a critical threshold that triggers neuronal metabolic compromise. When age and a condition that lowers cerebral perfusion converge, critically attained threshold of cerebral hypoperfusion (CATCH) results. CATCH is a cyclical and progressive cerebrovascular insufficiency that will destabilize neurons, synapses, neurotransmission, and cognitive ability, eventually evolving into a neurodegenerative process characterized by the formation of senile plaques, neurofibrillary tangles, and amyloid angiopathy. The concept of impaired cerebral perfusion as the cause of this dementia also explains the heterogeneic profile observed in AD patients, because an extensive list of risk factors for AD are also reported to significantly diminish blood flow to the aging brain.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neurosciences, University of California, San Diego, La Jolla 92093, USA.
| |
Collapse
|
23
|
Abstract
The pathology of Alzheimer's disease (AD) is not limited to amyloid plaques and neurofibrillary tangles. Recent evidence suggests that more than 30% of AD cases exhibit cerebrovascular pathology, which involves the cellular elements that represent the blood-brain barrier. Certain vascular lesions such as microvascular degeneration affecting the cerebral endothelium, cerebral amyloid angiopathy and periventricular white matter lesions are evident in virtually all cases of AD. Furthermore, clinical studies have demonstrated blood-brain barrier dysfunction in AD patients who exhibit peripheral vascular abnormalities such as hypertension, cardiovascular disease and diabetes. Whether these vascular lesions along with perivascular denervation are coincidental or causal in the pathogenetic processes of AD remains to be defined. In this chapter, I review biochemical and morphological evidence in context with the variable but distinct cerebrovascular pathology described in AD. I also consider genetic influences such as apolipoprotein E in relation to cerebrovascular lesions that may shed light on the pathophysiology of the cerebral vasculature. The compelling vascular pathology associated with AD suggests that transient and focal breach of the blood-brain barrier occurs in late onset AD and may involve an interaction of several factors, which include perivascular mediators as well as peripheral circulation derived factors that perturb the endothelium. These vascular abnormalities are likely to worsen cognitive disability in AD.
Collapse
Affiliation(s)
- R N Kalaria
- Institute for Health of the Elderly, Newcastle General Hospital, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
24
|
de la Torre JC. Critically attained threshold of cerebral hypoperfusion: the CATCH hypothesis of Alzheimer's pathogenesis. Neurobiol Aging 2000; 21:331-42. [PMID: 10867218 DOI: 10.1016/s0197-4580(00)00111-1] [Citation(s) in RCA: 209] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This review discusses the experimental and clinical data which indicate that chronic cerebral hypoperfusion can affect metabolic, anatomic, and cognitive function adversely. In aged but not young animals, chronic brain hypoperfusion results in regional pre- and post-synaptic changes, protein synthesis abnormalities, energy metabolic dysregulation, reduced glucose utilization, cholinergic receptor loss, and visuo-spatial memory deficits. Additionally, aging animals that are kept for prolonged periods of time after chronic brain hypoperfusion, also develop brain capillary degeneration in CA1 hippocampus and neuronal damage extending from the hippocampal region to the temporo-parietal cortex where neurodegenerative tissue atrophy eventually forms. All these pathologic events occur in rodents in the absence of senile plaques and neurofibrillary tangles. Alzheimer brains reveal similar biochemical and structural changes as those experimentally induced in aging animals. Moreover, regional cerebral hypoperfusion is one of the earlier (if not the earliest) clinical manifestations in both the sporadic and familial forms of Alzheimer's disease. In addition, therapy that improves or increases cerebral perfusion is generally of some benefit to Alzheimer patients. Conversely, a variety of disorders with different etiologies that impair or diminish cerebral perfusion are reported to be risk factors for this dementia. These findings have prompted us to propose the concept that advanced aging in the presence of a vascular risk factor can converge to create a critically attained threshold of cerebral hypoperfusion (CATCH) that triggers regional brain microcirculatory disturbances and impairs optimal delivery of energy substrates needed for normal brain cell function. The outcome of this defect generates a chain of events leading to the progressive evolution of brain metabolic, cognitive and tissue pathology that characterize Alzheimer's disease. The possible role of CATCH in familial and early onset Alzheimer's disease is briefly discussed from a theoretical vantagepoint. The growing and most recent evidence in support of the CATCH concept is the focus of this review.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neurosciences (MTF-0624), University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0624, USA.
| |
Collapse
|
25
|
Steusloff K, Röcken C, Saeger W. Basement membrane proteins and apolipoprotein E in growth hormone secreting adenomas and their correlation to amyloid: an immunoelectron microscopic study. Endocr Pathol 2000; 11:49-56. [PMID: 15304839 DOI: 10.1385/ep:11:1:49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Having previously demonstrated the association of basement membrane components (BMC), as well as apolipoprotein E and amyloid in growth hormone (GH)-producing pituitary adenomas, the aim of this study was to further analyze this relationship at the ultrastructural level. Ultrathin sections from four amyloidotic sparsely granulated monohormonal GH-producing adenomas previously investigated light microscopically were selected. Immunoelectron microscopy was performed, using a single labeling postembedding on-grid Protein-A gold method with antisera directed against laminin, fibronectin and apolipoprotein E. In all four adenomas, anti-laminin, anti-fibronectin, and anti-apolipoprotein E reacted with amyloid fibrils. No BMC were demonstrated between amyloid deposits, making it likely that synthesis and deposition of BMC may be secondary to the deposition of amyloid. The intimate spatial relationship between BMC as well as apolipoprotein E and amyloid fibrils may indicate morphological evidence of a particular arrangement of amyloid components in GH-secreting adenoma amyloid and a pathophysiological link.
Collapse
Affiliation(s)
- K Steusloff
- Institute of Pathology, Marienkrankenhaus, Hamburg, Germany
| | | | | |
Collapse
|
26
|
Tong XK, Hamel E. Regional cholinergic denervation of cortical microvessels and nitric oxide synthase-containing neurons in Alzheimer's disease. Neuroscience 1999; 92:163-75. [PMID: 10392839 DOI: 10.1016/s0306-4522(98)00750-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
In the present study, we investigated in the human cerebral cortex whether, as in the rat, basal forebrain cholinergic neurons innervate cortical microvessels and nitric oxide synthase-containing neurons and, further, we compared the status of this innervation between aged controls and neuropathologically confirmed cases of Alzheimer's disease. Using immunocytochemistry of choline acetyltransferase coupled to reduced nicotinamide adenine dinucleotide phosphate-diaphorase histochemistry, we show in young human subjects the presence of a cholinergic input to the cortical microcirculation, and of numerous perisomatic and peridendritic contacts between cholinergic nerve terminals and reduced nicotinamide adenine dinucleotide phosphate-diaphorase neurons. A regional cholinergic denervation of both cortical microvessels and reduced nicotinamide adenine dinucleotide phosphate-diaphorase neurons was found in Alzheimer's disease patients as compared to aged controls, and it paralleled the loss of total cholinergic nerve terminals in the corresponding areas of the cerebral cortex. The vascular denervation was more severe in the temporal (77%, P < 0.05) than in the frontal (48%, not significant) cortex, and the reduced nicotinamide adenine dinucleotide phosphate-diaphorase intracortical neurons were similarly deprived of their cholinergic input (P < 0.01) in both regions. Interestingly, a significant increase in luminal diameter (48%, P < 0.01) and area (> 160%, P < 0.01) of perfused microvessels was found in Alzheimer's tissues, possibly a consequence of both loss of neurogenic input and structural changes in blood vessel walls. The data indicate that intracortical microvessels and nitric oxide neurons in Alzheimer's disease are deprived of a cholinergic neurogenic control, a situation which is likely to result in a compromised ability to adapt cortical perfusion to neuronal activation during functional tasks related to cognition, arousal and attention. We conclude that such deficits in neurovascular regulation are likely to be an important pathogenic factor underlying cerebral blood flow dysfunctions in Alzheimer's disease.
Collapse
Affiliation(s)
- X K Tong
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Québec, Canada
| | | |
Collapse
|
27
|
Woodrow SI, Stewart RJ, Kisilevsky R, Gore J, Young ID. Experimental AA amyloidogenesis is associated with differential expression of extracellular matrix genes. Amyloid 1999; 6:22-30. [PMID: 10211408 DOI: 10.3109/13506129908993284] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
An abnormality in basement membrane metabolism has been postulated to play an important role in the pathogenesis of experimental murine AA amyloidosis. The potential contribution of the structural basement membrane proteins laminin, type IV collagen and entactin to amyloidogenesis in this model was investigated with a kinetic analysis of the expression of the corresponding genes during amyloid formation. Splenic AA amyloid deposition was stimulated by the concomitant administration of subcutaneous silver nitrate, as an inflammatory stimulus, and intravenous amyloid enhancing factor. Using a reverse transcription-polymerase chain reaction assay, a differential pattern of expression of these genes was observed at the mRNA level. Whereas laminin B1 mRNA levels did not change at any time during amyloidogenesis, a 2.2 to 3 fold induction of laminin B2, entactin and alpha 1-type IV collagen mRNAs coincided with the initial detection of splenic amyloid deposits at 48 hours post-stimulation, as detected by immunohistochemistry. Temporal and spatial codeposition of laminin and type IV collagen with amyloid was demonstrated by immunohistochemistry. A 1.4, 2.3 and 2.2-fold increase in laminin B2, entactin and alpha 1-type IV collagen mRNA levels, respectively, was detected at 24 hours post-stimulation, a point at which amyloid deposits could not be detected. Neither inflammation nor amyloid enhancing factor alone influenced laminin, entactin or type IV collagen expression at the protein or mRNA level. These observations suggest that the laminin B2 chain and alpha 1-type IV collagen chain account, at least in part, for the observed laminin and collagen IV immunoreactivity in AA amyloid deposits and that entactin may also be a component of the amyloid deposit. The onset of the induction of laminin B2, entactin and alpha 1-type IV collagen gene expression prior to the appearance of amyloid deposits, and our previous data with the heparan sulfate proteoglycan, perlecan, suggests these basement membrane proteins may play a role in the initial stages of AA fibrillogenesis.
Collapse
Affiliation(s)
- S I Woodrow
- Department of Biochemistry, Queen's University Kingston, Ontario, Canada
| | | | | | | | | |
Collapse
|
28
|
Flynn BL. Pharmacologic management of Alzheimer disease, Part I: Hormonal and emerging investigational drug therapies. Ann Pharmacother 1999; 33:178-87. [PMID: 10084414 DOI: 10.1345/aph.16378] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To provide information about current genetic research in Alzheimer disease (AD) and potential pharmacologic interventions. Investigational pharmacologic management of AD, including serenics, hormonal therapy, neurotransmitter augmentation, neurobiologic agents, nootropics, and ampakines are also reviewed. DATA SOURCES Studies, review articles, and editorials identified from MEDLINE searches (from January 1993 to December 1996), and bibliographies of identified articles. STUDY SELECTION Studies, review articles, and editorials addressing current areas of AD pharmacotherapy research, including hormonal therapy and select investigational agents. DATA EXTRACTION Pertinent information was selected and the data were synthesized into a review format. DATA SYNTHESIS AD is a devastating disease characterized by progressive memory loss, cognitive decline, and behavioral disturbances. The behavioral problems associated with AD can present a difficult clinical challenge. Many patients with AD are intolerant of traditional pharmacologic management, including antipsychotics, antidepressants, and anxiolytics. Hormonal agents, including estrogen, medroxyprogesterone, and cyproterone acetate, may be efficacious therapeutic alternatives in the management of sexual behavioral disturbances in men. Research regarding estrogen's role in AD prevention and effect on cognitive function and behavioral symptoms in women with AD are evaluated. Studies evaluating neurotransmitter augmentation and neurobiologic agents in AD are reviewed. CONCLUSIONS Environmental, genetic, neurobiologic, hormonal, and neurotransmitter influences, and their respective roles in AD pathology, are being investigated. Researchers concur that it is imperative to recognize the correlation of these factors in the etiology of AD to design effective prevention and treatment strategies. Additional studies are essential to elucidate the most efficacious treatments for AD and the attendant behavioral disturbances.
Collapse
Affiliation(s)
- B L Flynn
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy and Allied Health Professions, Creighton University, St. Joseph Villa Nursing Center, Omaha, NE 68178, USA
| |
Collapse
|
29
|
|
30
|
Abstract
Hypertension has been related to the development of dementia, including Alzheimer's disease, and cognitive dysfunction in middle-aged and elderly populations. One possible explanation is that hypertension is a risk factor for cerebral infarcts and ischaemic subcortical white-matter lesions. Hypertension may also give rise to a blood-brain barrier dysfunction, which has been suggested to be involved in the aetiology and pathogenesis of Alzheimer's disease. Other possible explanations for the association are shared risk factors, such as psychological stress, and the formation of free oxygen radicals. Low blood pressure has also been hypothesized to give rise to brain damage and dementia. However, the brain is involved in blood pressure regulation and dementia disorders may therefore influence the blood pressure. The findings of an association between hypertension and dementia may have implications for prevention and treatment.
Collapse
Affiliation(s)
- I Skoog
- Department of Psychiatry, Sahlgrenska Hospital, Göteborg University, Sweden
| |
Collapse
|
31
|
de la Torre JC. Hemodynamic consequences of deformed microvessels in the brain in Alzheimer's disease. Ann N Y Acad Sci 1997; 826:75-91. [PMID: 9329682 DOI: 10.1111/j.1749-6632.1997.tb48462.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The cause of sporadic Alzheimer's disease (AD) remains a mystery. Mounting clinical and experimental data, however, suggest that a cerebral hemodynamic role may affect neuronoglial metabolism. Light and electron microscopy have consistently revealed that the microvasculature in AD brains contains structurally deformed capillaries which create a distorted intraluminal conduit for blood flow. The cerebral capillary distortions can create "disturbed" rather than "laminar" blood flow. Chronically disturbed capillary blood flow will impair normal delivery of essential nutrients to brain neurons as well as impede catabolic outflow of CNS waste products. This condition will negatively affect cerebral metabolism, primarily because of impaired glucose delivery to neurons. Impaired glucose delivery to AD brain results in a patho-chemical cascade that will impair the Na+, K(+)-ATPase ion pump and affect the syntheses of ATP, acetylcholine, and other neurotransmitters. The outcome of this metabolic dysfunction can promote neurofibrillary tangle and senile plaque formation in AD brain.
Collapse
Affiliation(s)
- J C de la Torre
- University of New Mexico, Division of Neurosurgery, Albuquerque 87131, USA.
| |
Collapse
|
32
|
Age-dependent neuronal and synaptic degeneration in mice transgenic for the C terminus of the amyloid precursor protein. J Neurosci 1996. [PMID: 8824314 DOI: 10.1523/jneurosci.16-21-06732.1996] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The molecular basis for the degeneration of neurons and the deposition of amyloid in plaques and in the cerebrovasculature in Alzheimer's disease (AD) is incompletely understood. We have proposed that one molecule common to these abnormal processes is a fragment of the Alzheimer amyloid precursor protein (APP) comprising the C-terminal 100 amino acids of this molecule (APP-C100). We tested this hypothesis by creating transgenic mice expressing APP-C100 in the brain. We report here that aging (18-28 month) APP-C100 transgenic mice exhibit profound degeneration of neurons and synapses in Ammon's horn and the dentate gyrus of the hippocampal formation. Of the 106 transgenic mice between 8 and 28 months of age that were examined, all of those older than 18 months displayed severe hippocampal degeneration. The numerous degenerating axonal profiles contained increased numbers of neurofilaments, whorls of membrane, and accumulations of debris resembling secondary lysosomes near the cell body. The dendrites of degenerating granule and pyramidal cells contained disorganized, wavy microtubules. Cerebral blood vessels had thickened refractile basal laminae, and microglia laden with debris lay adjacent to larger venous vessels. Mice transgenic for Flag-APP-C100 (in which the hydrophilic Flag tag was fused to the N terminus of APP-C100) showed a similar degree of neurodegeneration in the hippocampal formation as early as 12 months of age. The 45 control mice displayed only occasional necrotic cells and no extensive cell degeneration in the same brain regions. These findings show that APP-C100 is capable of causing some of the neuropathological features of AD.
Collapse
|
33
|
Perlmutter LS. Microvascular pathology and vascular basement membrane components in Alzheimer's disease. Mol Neurobiol 1994; 9:33-40. [PMID: 7888105 DOI: 10.1007/bf02816103] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Several factors have highlighted the vasculature in Alzheimer's disease (AD): Cerebral amyloid angiopathy (CAA) is common, amyloid fibrils emanate from the vascular basement membrane (VBM), and similar forms of beta-amyloid are found in vascular and parenchymal amyloid accumulations. The present article discusses the presence of microvascular pathology in AD. Microangiopathy, in addition to neurofibrillary tangles, senile plaques, and CAA, is a common pathologic hallmark of AD. VBM components are associated with amyloid plaques, and nonamyloidotic alterations of the VBM occur in brain regions susceptible to AD lesions. Also, intra-VBM perivascular cells (traditionally called pericytes), a subset of which share the immunophenotype of microglia and other mononuclear phagocytic system (MPS) cells, have been implicated in vascular alterations and cerebrovascular amyloid deposition. Perivascular and parenchymal MPS cells have access to several sources of the beta-amyloid protein precursor, including platelets, circulating white cells, and neurons. MPS cells would thus be ideally situated to uptake and process the precursor, and deposit beta-amyloid in a fashion analogous to that seen in other forms of systemic and cerebral amyloidoses.
Collapse
Affiliation(s)
- L S Perlmutter
- Department of Neurology, University of Southern California School of Medicine, Los Angeles 90033
| |
Collapse
|