201
|
Abstract
Cognitive decline is an increasingly important public health problem, with more than 100 million adults worldwide projected to develop dementia by 2050. Accordingly, there has been an increased interest in preventive strategies that diminish this risk. It has been recognized that lifestyle factors including dietary patterns, may be important in the prevention of cognitive decline and dementia in later life. Several dietary components have been examined, including antioxidants, fatty acids, and B vitamins. In addition, whole dietary eating plans, including the Mediterranean diet (MeDi), and the Dietary Approaches to Stop Hypertension (DASH) diet, with and without weight loss, have become areas of increasing interest. Although prospective epidemiological studies have observed that antioxidants, fatty acids, and B vitamins are associated with better cognitive functioning, randomized clinical trials have generally failed to confirm the value of any specific dietary component in improving neurocognition. Several randomized trials have examined the impact of changing 'whole' diets on cognitive outcomes. The MeDi and DASH diets offer promising preliminary results, but data are limited and more research in this area is needed.
Collapse
Affiliation(s)
- P J Smith
- Department of Psychiatry and Behavioral Sciences at Duke University Medical Center, Durham, USA
| | - J A Blumenthal
- Department of Psychiatry and Behavioral Sciences at Duke University Medical Center, Durham, USA
| |
Collapse
|
202
|
Currò M, Risitano R, Ferlazzo N, Cirmi S, Gangemi C, Caccamo D, Ientile R, Navarra M. Citrus bergamia Juice Extract Attenuates β-Amyloid-Induced Pro-Inflammatory Activation of THP-1 Cells Through MAPK and AP-1 Pathways. Sci Rep 2016; 6:20809. [PMID: 26853104 PMCID: PMC4745106 DOI: 10.1038/srep20809] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/08/2016] [Indexed: 12/19/2022] Open
Abstract
Flavonoids have been shown to be effective in protecting against age-related cognitive and motor decline in both in vitro and in vivo models. Recently, a flavonoid-rich extract of Citrus bergamia juice (BJe) has been shown to display anti-oxidant and anti-inflammatory properties against LPS-induced activation of human THP-1 monocytes. In the light of these observations, we wondered whether BJe may be beneficial against neuroinflammatory processes, such as those observed in Alzheimer’s disease. To this aim we used THP-1 monocytes to investigate the mechanisms underlying the beneficial potential of BJe against amyloid-beta1–42 (Aβ1−42) -mediated inflammation. Exposure of THP-1 cells to Aβ1−42 significantly induced the expression and secretion of IL-6 and IL-1β in THP-1 cells and increased the phosphorylation of ERK 1/2 as well as p46 and p54 members of JNK family. Moreover, Aβ1−42 raises AP-1 DNA binding activity in THP-1-treated cells. Interestingly, all these effects were reduced in the presence of BJe. Our data indicate that BJe may effectively counteract the pro-inflammatory activation of monocytes/microglial cells exposed to amyloid fibrils, suggesting a promising role as a natural drug against neuroinflammatory processes.
Collapse
Affiliation(s)
- Monica Currò
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, I-98100, Italy
| | - Roberto Risitano
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, I-98100, Italy
| | - Nadia Ferlazzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, I-98168, Italy
| | - Santa Cirmi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, I-98168, Italy
| | - Chiara Gangemi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, I-98100, Italy
| | - Daniela Caccamo
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, I-98100, Italy
| | - Riccardo Ientile
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Messina, I-98100, Italy
| | - Michele Navarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, I-98168, Italy
| |
Collapse
|
203
|
Skene NG, Grant SGN. Identification of Vulnerable Cell Types in Major Brain Disorders Using Single Cell Transcriptomes and Expression Weighted Cell Type Enrichment. Front Neurosci 2016; 10:16. [PMID: 26858593 PMCID: PMC4730103 DOI: 10.3389/fnins.2016.00016] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/12/2016] [Indexed: 11/13/2022] Open
Abstract
The cell types that trigger the primary pathology in many brain diseases remain largely unknown. One route to understanding the primary pathological cell type for a particular disease is to identify the cells expressing susceptibility genes. Although this is straightforward for monogenic conditions where the causative mutation may alter expression of a cell type specific marker, methods are required for the common polygenic disorders. We developed the Expression Weighted Cell Type Enrichment (EWCE) method that uses single cell transcriptomes to generate the probability distribution associated with a gene list having an average level of expression within a cell type. Following validation, we applied EWCE to human genetic data from cases of epilepsy, Schizophrenia, Autism, Intellectual Disability, Alzheimer's disease, Multiple Sclerosis and anxiety disorders. Genetic susceptibility primarily affected microglia in Alzheimer's and Multiple Sclerosis; was shared between interneurons and pyramidal neurons in Autism and Schizophrenia; while intellectual disabilities and epilepsy were attributable to a range of cell-types, with the strongest enrichment in interneurons. We hypothesized that the primary cell type pathology could trigger secondary changes in other cell types and these could be detected by applying EWCE to transcriptome data from diseased tissue. In Autism, Schizophrenia and Alzheimer's disease we find evidence of pathological changes in all of the major brain cell types. These findings give novel insight into the cellular origins and progression in common brain disorders. The methods can be applied to any tissue and disorder and have applications in validating mouse models.
Collapse
Affiliation(s)
- Nathan G Skene
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| |
Collapse
|
204
|
Skene NG, Grant SGN. Identification of Vulnerable Cell Types in Major Brain Disorders Using Single Cell Transcriptomes and Expression Weighted Cell Type Enrichment. Front Neurosci 2016; 10:16. [PMID: 26858593 DOI: 10.3389/fnins.2016.00016/bibtex] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 01/12/2016] [Indexed: 05/23/2023] Open
Abstract
The cell types that trigger the primary pathology in many brain diseases remain largely unknown. One route to understanding the primary pathological cell type for a particular disease is to identify the cells expressing susceptibility genes. Although this is straightforward for monogenic conditions where the causative mutation may alter expression of a cell type specific marker, methods are required for the common polygenic disorders. We developed the Expression Weighted Cell Type Enrichment (EWCE) method that uses single cell transcriptomes to generate the probability distribution associated with a gene list having an average level of expression within a cell type. Following validation, we applied EWCE to human genetic data from cases of epilepsy, Schizophrenia, Autism, Intellectual Disability, Alzheimer's disease, Multiple Sclerosis and anxiety disorders. Genetic susceptibility primarily affected microglia in Alzheimer's and Multiple Sclerosis; was shared between interneurons and pyramidal neurons in Autism and Schizophrenia; while intellectual disabilities and epilepsy were attributable to a range of cell-types, with the strongest enrichment in interneurons. We hypothesized that the primary cell type pathology could trigger secondary changes in other cell types and these could be detected by applying EWCE to transcriptome data from diseased tissue. In Autism, Schizophrenia and Alzheimer's disease we find evidence of pathological changes in all of the major brain cell types. These findings give novel insight into the cellular origins and progression in common brain disorders. The methods can be applied to any tissue and disorder and have applications in validating mouse models.
Collapse
Affiliation(s)
- Nathan G Skene
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| | - Seth G N Grant
- Centre for Clinical Brain Sciences, Edinburgh University Edinburgh, UK
| |
Collapse
|
205
|
Banks WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat Rev Drug Discov 2016; 15:275-92. [PMID: 26794270 DOI: 10.1038/nrd.2015.21] [Citation(s) in RCA: 753] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the biggest challenges in the development of therapeutics for central nervous system (CNS) disorders is achieving sufficient blood-brain barrier (BBB) penetration. Research in the past few decades has revealed that the BBB is not only a substantial barrier for drug delivery to the CNS but also a complex, dynamic interface that adapts to the needs of the CNS, responds to physiological changes, and is affected by and can even promote disease. This complexity confounds simple strategies for drug delivery to the CNS, but provides a wealth of opportunities and approaches for drug development. Here, I review some of the most important areas that have recently redefined the BBB and discuss how they can be applied to the development of CNS therapeutics.
Collapse
Affiliation(s)
- William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center and Department of Medicine, University of Washington School of Medicine, Division of Gerontology and Geriatric Medicine, 1660 South Columbian Way, Seattle, Washington 98108, USA
| |
Collapse
|
206
|
Ielacqua GD, Schlegel F, Füchtemeier M, Xandry J, Rudin M, Klohs J. Magnetic Resonance Q Mapping Reveals a Decrease in Microvessel Density in the arcAβ Mouse Model of Cerebral Amyloidosis. Front Aging Neurosci 2016; 7:241. [PMID: 26834622 PMCID: PMC4717293 DOI: 10.3389/fnagi.2015.00241] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/07/2015] [Indexed: 12/04/2022] Open
Abstract
Alterations in density and morphology of the cerebral microvasculature have been reported to occur in Alzheimer's disease patients and animal models of the disease. In this study we compared magnetic resonance imaging (MRI) techniques for their utility to detect age-dependent changes of the cerebral vasculature in the arcAβ mouse model of cerebral amyloidosis. Dynamic susceptibility contrast (DSC)-MRI was performed by tracking the passage of a superparamagnetic iron oxide nanoparticle in the brain with dynamic gradient echo planar imaging (EPI). From this measurements relative cerebral blood volume [rCBV(DSC)] and relative cerebral blood flow (rCBF) were estimated. For the same animal maps of the relaxation shift index Q were computed from high resolution gradient echo and spin echo data that were acquired before and after superparamagnetic iron oxide (SPIO) nanoparticle injection. Q-values were used to derive estimates of microvessel density. The change in the relaxation rates ΔR2* obtained from pre- and post-contrast gradient echo data was used for the alternative determination of rCBV [rCBV(ΔR2*)]. Linear mixed effects modeling found no significant association between rCBV(DSC), rCBV(ΔR2*), rCBF, and Q with genotype in 13-month old mice [compared to age-matched non-transgenic littermates (NTLs)] for any of the evaluated brain regions. In 24-month old mice there was a significant association for rCBV(DSC) with genotype in the cerebral cortex, and for rCBV(ΔR2*) in the cerebral cortex and cerebellum. For rCBF there was a significant association in the cerebellum but not in other brain regions. Q-values in the olfactory bulb, cerebral cortex, striatum, hippocampus, and cerebellum in 24-month old mice were significantly associated with genotype. In those regions Q-values were reduced between 11 and 26% in arcAβ mice compared to age-matched NTLs. Vessel staining with CD31 immunohistochemistry confirmed a reduction of microvessel density in the old arcAβ mice. We further demonstrated a region-specific association between parenchymal and vascular deposition of β-amyloid and decreased vascular density, without a correlation with the amount of Aβ deposition. We found that Q mapping was more suitable than the hemodynamic read-outs to detect amyloid-related degeneration of the cerebral microvasculature.
Collapse
Affiliation(s)
- Giovanna D Ielacqua
- Institute for Biomedical Engineering, ETH and University of Zurich Zurich, Switzerland
| | - Felix Schlegel
- Institute for Biomedical Engineering, ETH and University of ZurichZurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH ZurichZurich, Switzerland
| | - Martina Füchtemeier
- German Center for Neurodegenerative DiseasesBerlin, Germany; Department of Experimental Neurology, Charité - University Medicine BerlinBerlin, Germany
| | - Jael Xandry
- Institute for Biomedical Engineering, ETH and University of Zurich Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, ETH and University of ZurichZurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH ZurichZurich, Switzerland; Institute of Pharmacology and Toxicology, University of ZurichZurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, ETH and University of ZurichZurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH ZurichZurich, Switzerland
| |
Collapse
|
207
|
Wolters FJ, de Bruijn RF, Hofman A, Koudstaal PJ, Ikram MA. Cerebral Vasoreactivity, Apolipoprotein E, and the Risk of Dementia. Arterioscler Thromb Vasc Biol 2016; 36:204-10. [DOI: 10.1161/atvbaha.115.306768] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/06/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Frank J. Wolters
- From the Departments of Neurology (F.J.W., R.F.A.G.d.B., P.J.K., M.A.I.), Epidemiology (F.J.W., R.F.A.G.d.B., A.H., M.A.I.), and Radiology (M.A.I.), Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Renée F.A.G. de Bruijn
- From the Departments of Neurology (F.J.W., R.F.A.G.d.B., P.J.K., M.A.I.), Epidemiology (F.J.W., R.F.A.G.d.B., A.H., M.A.I.), and Radiology (M.A.I.), Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Albert Hofman
- From the Departments of Neurology (F.J.W., R.F.A.G.d.B., P.J.K., M.A.I.), Epidemiology (F.J.W., R.F.A.G.d.B., A.H., M.A.I.), and Radiology (M.A.I.), Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Peter J. Koudstaal
- From the Departments of Neurology (F.J.W., R.F.A.G.d.B., P.J.K., M.A.I.), Epidemiology (F.J.W., R.F.A.G.d.B., A.H., M.A.I.), and Radiology (M.A.I.), Erasmus Medical Centre, Rotterdam, The Netherlands
| | - M. Arfan Ikram
- From the Departments of Neurology (F.J.W., R.F.A.G.d.B., P.J.K., M.A.I.), Epidemiology (F.J.W., R.F.A.G.d.B., A.H., M.A.I.), and Radiology (M.A.I.), Erasmus Medical Centre, Rotterdam, The Netherlands
| |
Collapse
|
208
|
Kumar A, Ekavali, Mishra J, Chopra K, Dhull DK. Possible role of P-glycoprotein in the neuroprotective mechanism of berberine in intracerebroventricular streptozotocin-induced cognitive dysfunction. Psychopharmacology (Berl) 2016; 233:137-52. [PMID: 26446867 DOI: 10.1007/s00213-015-4095-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/21/2015] [Indexed: 01/07/2023]
Abstract
RATIONALE The therapeutic potential of berberine has been well documented in various neurological problems. However, the neurological mechanism of berberine remains untapped in the light of its P-glycoprotein (P-gp)-mediated gut efflux properties responsible for reduced bioavailability. Verapamil, a well known L-type calcium channel blocker, has additional inhibitory activity against P-gp efflux pump. Thus, there is a strong scientific rationale to explore the interaction of berberine with verapamil as a possible neuroprotective strategy. OBJECTIVE The present study was designed to evaluate the effect of berberine, verapamil, and their combination on behavioral alterations, oxidative stress, mitochondrial dysfunction, neuroinflammation, and histopathological modifications in intracerebroventricular streptozocin (ICV-STZ)-induced sporadic dementia of Alzheimer's type in rats. METHODS Single bilateral ICV-STZ (3 mg/kg) administration was used as an experimental model of sporadic dementia of Alzheimer's type. RESULTS Berberine (25, 50, and 100 mg/kg, oral gavage) or verapamil (2.5 and 5 mg/kg, intraperitoneally) were used as treatment drugs, and memantine (5 mg/kg, intraperitoneally) was used as a standard. Berberine and verapamil significantly attenuated behavioral, biochemical, cellular, and histological alterations, suggesting their neuroprotective potential. Further, treatment of berberine (25 and 50 mg/kg) with verapamil (2.5 and 5.0 mg/kg) combinations respectively significantly potentiated their neuroprotective effect which was significant as compared to their effect per se in ICV-STZ-treated animals. CONCLUSION The augmentative outcome of verapamil on the neuroprotective effect of berberine can be speculated due to the inhibition of P-gp efflux mechanism and the prevention of calcium homeostasis alteration. Additionally, anti-inflammatory and antioxidant effects of both berberine and verapamil could also contribute in their protective effect.
Collapse
Affiliation(s)
- Anil Kumar
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| | - Ekavali
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Jitendriya Mishra
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Kanwaljit Chopra
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Dinesh K Dhull
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| |
Collapse
|
209
|
Sato F, Omura S, Jaffe S, Tsunoda I. Role of CD4+ T Cells in the Pathophysiology of Multiple Sclerosis. MULTIPLE SCLEROSIS 2016. [PMCID: PMC7150304 DOI: 10.1016/b978-0-12-800763-1.00004-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Although the precise etiology of MS remains unclear, CD4+ T cells have been proposed to play not only effector but also regulatory roles in MS. CD4+ T cells can be divided into four subsets: pro-inflammatory helper T (Th) 1 and Th17 cells, anti-inflammatory Th2 cells and regulatory T cells (Tregs). The roles of CD4+ T cells in MS have been clarified by either “loss-of-function” or “gain-of-function” methods, which have been carried out mainly in autoimmune and viral models of MS: experimental autoimmune encephalomyelitis and Theiler's murine encephalomyelitis virus infection, respectively. Observations in MS patients were consistent with the mechanisms found in the MS models, that is, increased pro-inflammatory Th1 and Th17 activity is associated with disease exacerbation, while anti-inflammatory Th2 cells and Tregs appear to play a protective role.
Collapse
|
210
|
Blood-Brain Barrier Disruption Is an Early Event That May Persist for Many Years After Traumatic Brain Injury in Humans. J Neuropathol Exp Neurol 2015; 74:1147-57. [DOI: 10.1097/nen.0000000000000261] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
211
|
Hay JR, Johnson VE, Young AM, Smith DH, Stewart W. Blood-Brain Barrier Disruption Is an Early Event That May Persist for Many Years After Traumatic Brain Injury in Humans. J Neuropathol Exp Neurol 2015. [DOI: 10.1093/jnen/74.12.1147] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
212
|
Sedeyn JC, Wu H, Hobbs RD, Levin EC, Nagele RG, Venkataraman V. Histamine Induces Alzheimer's Disease-Like Blood Brain Barrier Breach and Local Cellular Responses in Mouse Brain Organotypic Cultures. BIOMED RESEARCH INTERNATIONAL 2015; 2015:937148. [PMID: 26697497 PMCID: PMC4677161 DOI: 10.1155/2015/937148] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/30/2015] [Accepted: 11/08/2015] [Indexed: 11/18/2022]
Abstract
Among the top ten causes of death in the United States, Alzheimer's disease (AD) is the only one that cannot be cured, prevented, or even slowed down at present. Significant efforts have been exerted in generating model systems to delineate the mechanism as well as establishing platforms for drug screening. In this study, a promising candidate model utilizing primary mouse brain organotypic (MBO) cultures is reported. For the first time, we have demonstrated that the MBO cultures exhibit increased blood brain barrier (BBB) permeability as shown by IgG leakage into the brain parenchyma, astrocyte activation as evidenced by increased expression of glial fibrillary acidic protein (GFAP), and neuronal damage-response as suggested by increased vimentin-positive neurons occur upon histamine treatment. Identical responses-a breakdown of the BBB, astrocyte activation, and neuronal expression of vimentin-were then demonstrated in brains from AD patients compared to age-matched controls, consistent with other reports. Thus, the histamine-treated MBO culture system may provide a valuable tool in combating AD.
Collapse
Affiliation(s)
- Jonathan C. Sedeyn
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Hao Wu
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
| | - Reilly D. Hobbs
- Department of Cell Biology, Rowan School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Eli C. Levin
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Robert G. Nagele
- Biomarker Discovery Center, New Jersey Institute for Successful Aging, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
- Department of Geriatrics and Gerontology, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Venkat Venkataraman
- Graduate School of Biomedical Sciences, Rowan University, Stratford, NJ 08084, USA
- Department of Cell Biology, Rowan School of Osteopathic Medicine, Stratford, NJ 08084, USA
| |
Collapse
|
213
|
Currais A, Goldberg J, Farrokhi C, Chang M, Prior M, Dargusch R, Daugherty D, Armando A, Quehenberger O, Maher P, Schubert D. A comprehensive multiomics approach toward understanding the relationship between aging and dementia. Aging (Albany NY) 2015; 7:937-55. [PMID: 26564964 PMCID: PMC4694064 DOI: 10.18632/aging.100838] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/30/2015] [Indexed: 04/14/2023]
Abstract
Because age is the greatest risk factor for sporadic Alzheimer's disease (AD), phenotypic screens based upon old age-associated brain toxicities were used to develop the potent neurotrophic drug J147. Since certain aspects of aging may be primary cause of AD, we hypothesized that J147 would be effective against AD-associated pathology in rapidly aging SAMP8 mice and could be used to identify some of the molecular contributions of aging to AD. An inclusive and integrative multiomics approach was used to investigate protein and gene expression, metabolite levels, and cognition in old and young SAMP8 mice. J147 reduced cognitive deficits in old SAMP8 mice, while restoring multiple molecular markers associated with human AD, vascular pathology, impaired synaptic function, and inflammation to those approaching the young phenotype. The extensive assays used in this study identified a subset of molecular changes associated with aging that may be necessary for the development of AD.
Collapse
Affiliation(s)
- Antonio Currais
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Joshua Goldberg
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | | | - Max Chang
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Marguerite Prior
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Richard Dargusch
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Daniel Daugherty
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Aaron Armando
- Department of Medicine, University of California San Diego, CA 92093-0601, USA
| | - Oswald Quehenberger
- Department of Medicine, University of California San Diego, CA 92093-0601, USA
| | - Pamela Maher
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - David Schubert
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
214
|
Sun P, Esteban G, Inokuchi T, Marco-Contelles J, Weksler BB, Romero IA, Couraud PO, Unzeta M, Solé M. Protective effect of the multitarget compound DPH-4 on human SSAO/VAP-1-expressing hCMEC/D3 cells under oxygen-glucose deprivation conditions: an in vitro experimental model of cerebral ischaemia. Br J Pharmacol 2015; 172:5390-402. [PMID: 26362823 DOI: 10.1111/bph.13328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 08/31/2015] [Accepted: 09/03/2015] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Stroke and Alzheimer's disease (AD) are related pathologies in which the cerebrovascular system is involved. Plasma levels of semicarbazide-sensitive amine oxidase/vascular adhesion protein 1 (SSAO/VAP-1, also known as Primary Amine Oxidase -PrAO) are increased in both stroke and AD patients and contribute to the vascular damage. During inflammation, its enzymatic activity mediates leukocyte recruitment to the injured tissue, inducing damage in the blood-brain barrier (BBB) and neuronal tissue. We hypothesized that by altering cerebrovascular function, SSAO/VAP-1 might play a role in the stroke-AD transition. Therefore, we evaluated the protective effect of the novel multitarget-directed ligand DPH-4, initially designed for AD therapy, on the BBB. EXPERIMENTAL APPROACH A human microvascular brain endothelial cell line expressing human SSAO/VAP-1 was generated, as the expression of SSAO/VAP-1 is lost in cultured cells. To simulate ischaemic damage, these cells were subjected to oxygen and glucose deprivation (OGD) and re-oxygenation conditions. The protective role of DPH-4 was then evaluated in the presence of methylamine, an SSAO substrate, and/or β-amyloid (Aβ). KEY RESULTS Under our conditions, DPH-4 protected brain endothelial cells from OGD and re-oxygenation-induced damage, and also decreased SSAO-dependent leukocyte adhesion. DPH-4 was also effective at preventing the damage induced by OGD and re-oxygenation in the presence of Aβ as a model of AD pathology. CONCLUSIONS AND IMPLICATIONS From these results, we concluded that the multitarget compound DPH-4 might be of therapeutic benefit to delay the onset and/or progression of the neurological pathologies associated with stroke and AD, which appear to be linked.
Collapse
Affiliation(s)
- P Sun
- Institut de Neurociències i Departament de Bioquímica i Biologia Molecular. Edifici M, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - G Esteban
- Institut de Neurociències i Departament de Bioquímica i Biologia Molecular. Edifici M, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - T Inokuchi
- Division of Chemistry and Biotechnology, Graduate School of Natural Science and Technology, Okayama University, 3.1.1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan
| | - J Marco-Contelles
- Instituto de Química Orgánica General (CSIC), 3 Juan de la Cierva, Madrid, 28006, Spain
| | - B B Weksler
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - I A Romero
- Department of Life, Health and Chemical Sciences, Open University, Milton Keynes, UK
| | - P O Couraud
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - M Unzeta
- Institut de Neurociències i Departament de Bioquímica i Biologia Molecular. Edifici M, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - M Solé
- Institut de Neurociències i Departament de Bioquímica i Biologia Molecular. Edifici M, Facultat de Medicina, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
215
|
Zhang H, Sachdev PS, Wen W, Crawford JD, Brodaty H, Baune BT, Kochan NA, Slavin MJ, Reppermund S, Kang K, Trollor JN. The relationship between inflammatory markers and voxel-based gray matter volumes in nondemented older adults. Neurobiol Aging 2015; 37:138-146. [PMID: 26559883 DOI: 10.1016/j.neurobiolaging.2015.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/24/2015] [Accepted: 10/08/2015] [Indexed: 11/26/2022]
Abstract
Ageing is characterized by chronically elevated inflammatory markers (IMs). Peripheral IM levels have been found in negative correlations with brain structural measures including global and lobar volumes and the hippocampus. This study investigated the relationship between 10 peripheral IMs and voxel-based gray matter (GM) volumes in nondemented older adults (n = 463). Two proinflammatory cytokines (tumor necrosis factor-α [TNF-α] and interleukin-1β) and 2 vascular IMs (vascular cellular adhesion molecule-1 and plasminogen activator inhibitor-1) were negatively correlated with regional GM volumes. TNF-α and interleukin-1β were both significantly correlated with GM volumes in the left occipitotemporal area, left superior occipital gyrus, and left inferior parietal lobule; TNF-α was also significantly correlated with the bilateral medial prefrontal cortices and approached significance for the correlations with the bilateral hippocampi. Significant GM correlations with vascular cellular adhesion molecule-1 were located in the bilateral anterior cingulate cortices, and with plasminogen activator inhibitor-1 in the cerebellum and right hippocampus. The neuroanatomical correlation patterns of 2 proinflammatory cytokines and 2 vascular IMs might be reflective of the effects of neurodegenerative and vascular pathological processes in the ageing brain.
Collapse
Affiliation(s)
- Haobo Zhang
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - John D Crawford
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Henry Brodaty
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Academic Department for Old Age Psychiatry, Prince of Wales Hospital, Randwick, New South Wales, Australia; Dementia Collaborative Research Centre, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Bernard T Baune
- Department of Psychiatry, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole A Kochan
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Melissa J Slavin
- Dementia Collaborative Research Centre, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Simone Reppermund
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Kristan Kang
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Julian N Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia; Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, Australia.
| |
Collapse
|
216
|
Different Brain Regions are Infected with Fungi in Alzheimer's Disease. Sci Rep 2015; 5:15015. [PMID: 26468932 PMCID: PMC4606562 DOI: 10.1038/srep15015] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022] Open
Abstract
The possibility that Alzheimer's disease (AD) has a microbial aetiology has been proposed by several researchers. Here, we provide evidence that tissue from the central nervous system (CNS) of AD patients contain fungal cells and hyphae. Fungal material can be detected both intra- and extracellularly using specific antibodies against several fungi. Different brain regions including external frontal cortex, cerebellar hemisphere, entorhinal cortex/hippocampus and choroid plexus contain fungal material, which is absent in brain tissue from control individuals. Analysis of brain sections from ten additional AD patients reveals that all are infected with fungi. Fungal infection is also observed in blood vessels, which may explain the vascular pathology frequently detected in AD patients. Sequencing of fungal DNA extracted from frozen CNS samples identifies several fungal species. Collectively, our findings provide compelling evidence for the existence of fungal infection in the CNS from AD patients, but not in control individuals.
Collapse
|
217
|
Poggesi A, Inzitari D, Pantoni L. Atrial Fibrillation and Cognition: Epidemiological Data and Possible Mechanisms. Stroke 2015; 46:3316-21. [PMID: 26396028 DOI: 10.1161/strokeaha.115.008225] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/31/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Anna Poggesi
- From the Neuroscience Section, NEUROFARBA Department, University of Florence, Florence, Italy (A.P., D.I., L.P.); and Institute of Neuroscience, Italian National Research Council, Florence, Italy (D.I.).
| | - Domenico Inzitari
- From the Neuroscience Section, NEUROFARBA Department, University of Florence, Florence, Italy (A.P., D.I., L.P.); and Institute of Neuroscience, Italian National Research Council, Florence, Italy (D.I.)
| | - Leonardo Pantoni
- From the Neuroscience Section, NEUROFARBA Department, University of Florence, Florence, Italy (A.P., D.I., L.P.); and Institute of Neuroscience, Italian National Research Council, Florence, Italy (D.I.)
| |
Collapse
|
218
|
|
219
|
Di Marco LY, Venneri A, Farkas E, Evans PC, Marzo A, Frangi AF. Vascular dysfunction in the pathogenesis of Alzheimer's disease--A review of endothelium-mediated mechanisms and ensuing vicious circles. Neurobiol Dis 2015; 82:593-606. [PMID: 26311408 DOI: 10.1016/j.nbd.2015.08.014] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/23/2015] [Accepted: 08/17/2015] [Indexed: 12/17/2022] Open
Abstract
Late-onset dementia is a major health concern in the ageing population. Alzheimer's disease (AD) accounts for the largest proportion (65-70%) of dementia cases in the older population. Despite considerable research effort, the pathogenesis of late-onset AD remains unclear. Substantial evidence suggests that the neurodegenerative process is initiated by chronic cerebral hypoperfusion (CCH) caused by ageing and cardiovascular conditions. CCH causes reduced oxygen, glucose and other nutrient supply to the brain, with direct damage not only to the parenchymal cells, but also to the blood-brain barrier (BBB), a key mediator of cerebral homeostasis. BBB dysfunction mediates the indirect neurotoxic effects of CCH by promoting oxidative stress, inflammation, paracellular permeability, and dysregulation of nitric oxide, a key regulator of regional blood flow. As such, BBB dysfunction mediates a vicious circle in which cerebral perfusion is reduced further and the neurodegenerative process is accelerated. Endothelial interaction with pericytes and astrocytes could also play a role in the process. Reciprocal interactions between vascular dysfunction and neurodegeneration could further contribute to the development of the disease. A comprehensive overview of the complex scenario of interacting endothelium-mediated processes is currently lacking, and could prospectively contribute to the identification of adequate therapeutic interventions. This study reviews the current literature of in vitro and ex vivo studies on endothelium-mediated mechanisms underlying vascular dysfunction in AD pathogenesis, with the aim of presenting a comprehensive overview of the complex network of causative relationships. Particular emphasis is given to vicious circles which can accelerate the process of neurovascular degeneration.
Collapse
Affiliation(s)
- Luigi Yuri Di Marco
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK.
| | - Annalena Venneri
- Department of Neuroscience, Medical School, University of Sheffield, Sheffield, UK; IRCCS San Camillo Foundation Hospital, Venice, Italy
| | - Eszter Farkas
- Department of Medical Physics and Informatics, Faculty of Medicine and Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Paul C Evans
- Department of Cardiovascular Science, Medical School, University of Sheffield, Sheffield, UK
| | - Alberto Marzo
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Alejandro F Frangi
- Centre for Computational Imaging and Simulation Technologies in Biomedicine (CISTIB), Department of Electronic and Electrical Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
220
|
Ryu JK, Cho T, Choi HB, Jantaratnotai N, McLarnon JG. Pharmacological antagonism of interleukin-8 receptor CXCR2 inhibits inflammatory reactivity and is neuroprotective in an animal model of Alzheimer's disease. J Neuroinflammation 2015; 12:144. [PMID: 26255110 PMCID: PMC4529987 DOI: 10.1186/s12974-015-0339-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/04/2015] [Indexed: 12/17/2022] Open
Abstract
Background The chemokine interleukin-8 (IL-8) and its receptor CXCR2 contribute to chemotactic responses in Alzheimer’s disease (AD); however, properties of the ligand and receptor have not been characterized in animal models of disease. The primary aim of our study was to examine effects of pharmacological antagonism of CXCR2 as a strategy to inhibit receptor-mediated inflammatory reactivity and enhance neuronal viability in animals receiving intrahippocampal injection of amyloid-beta (Aβ1–42). Methods In vivo studies used an animal model of Alzheimer’s disease incorporating injection of full-length Aβ1–42 into rat hippocampus. Immunohistochemical staining of rat brain was used to measure microgliosis, astrogliosis, neuronal viability, and oxidative stress. Western blot and Reverse Transcription PCR (RT-PCR) were used to determine levels of CXCR2 in animal tissue with the latter also used to determine expression of pro-inflammatory mediators. Immunostaining of human AD and non-demented (ND) tissue was also undertaken. Results We initially determined that in the human brain, AD relative to ND tissue exhibited marked increases in expression of CXCR2 with cell-specific receptor expression prominent in microglia. In Aβ1–42-injected rat brain, CXCR2 and IL-8 showed time-dependent increases in expression, concomitant with enhanced gliosis, relative to controls phosphate-buffered saline (PBS) or reverse peptide Aβ42–1 injection. Administration of the competitive CXCR2 antagonist SB332235 to peptide-injected rats significantly reduced expression of CXCR2 and microgliosis, with astrogliosis unchanged. Double staining studies demonstrated localization of CXCR2 and microglial immunoreactivity nearby deposits of Aβ1–42 with SB332235 effective in inhibiting receptor expression and microgliosis. The numbers of neurons in granule cell layer (GCL) were reduced in rats receiving Aβ1–42, compared with PBS, with administration of SB332235 to peptide-injected animals conferring neuroprotection. Oxidative stress was indicated in the animal model since both 4-hydroxynonenal (4-HNE) and hydroethidine (HEt) were markedly elevated in Aβ1–42 vs PBS-injected rat brain and diminished with SB332235 treatment. Conclusion Overall, the findings suggest critical roles for CXCR2-dependent inflammatory responses in an AD animal model with pharmacological modulation of the receptor effective in inhibiting inflammatory reactivity and conferring neuroprotection against oxidative damage.
Collapse
Affiliation(s)
- Jae K Ryu
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Science Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| | - T Cho
- Brain Research Centre, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada.
| | - Hyun B Choi
- Brain Research Centre, University of British Columbia, 2211 Wesbrook Mall, Vancouver, British Columbia, Canada.
| | - N Jantaratnotai
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| | - James G McLarnon
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2176 Health Science Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
221
|
Appraising the Role of Iron in Brain Aging and Cognition: Promises and Limitations of MRI Methods. Neuropsychol Rev 2015; 25:272-87. [PMID: 26248580 DOI: 10.1007/s11065-015-9292-y] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/24/2015] [Indexed: 12/11/2022]
Abstract
Age-related increase in frailty is accompanied by a fundamental shift in cellular iron homeostasis. By promoting oxidative stress, the intracellular accumulation of non-heme iron outside of binding complexes contributes to chronic inflammation and interferes with normal brain metabolism. In the absence of direct non-invasive biomarkers of brain oxidative stress, iron accumulation estimated in vivo may serve as its proxy indicator. Hence, developing reliable in vivo measurements of brain iron content via magnetic resonance imaging (MRI) is of significant interest in human neuroscience. To date, by estimating brain iron content through various MRI methods, significant age differences and age-related increases in iron content of the basal ganglia have been revealed across multiple samples. Less consistent are the findings that pertain to the relationship between elevated brain iron content and systemic indices of vascular and metabolic dysfunction. Only a handful of cross-sectional investigations have linked high iron content in various brain regions and poor performance on assorted cognitive tests. The even fewer longitudinal studies indicate that iron accumulation may precede shrinkage of the basal ganglia and thus predict poor maintenance of cognitive functions. This rapidly developing field will benefit from introduction of higher-field MRI scanners, improvement in iron-sensitive and -specific acquisition sequences and post-processing analytic and computational methods, as well as accumulation of data from long-term longitudinal investigations. This review describes the potential advantages and promises of MRI-based assessment of brain iron, summarizes recent findings and highlights the limitations of the current methodology.
Collapse
|
222
|
Abstract
The past two decades of research into the pathogenesis of Alzheimer disease (AD) have been driven largely by the amyloid hypothesis; the neuroinflammation that is associated with AD has been assumed to be merely a response to pathophysiological events. However, new data from preclinical and clinical studies have established that immune system-mediated actions in fact contribute to and drive AD pathogenesis. These insights have suggested both novel and well-defined potential therapeutic targets for AD, including microglia and several cytokines. In addition, as inflammation in AD primarily concerns the innate immune system - unlike in 'typical' neuroinflammatory diseases such as multiple sclerosis and encephalitides - the concept of neuroinflammation in AD may need refinement.
Collapse
|
223
|
Winkler EA, Sagare AP, Zlokovic BV. The pericyte: a forgotten cell type with important implications for Alzheimer's disease? Brain Pathol 2015; 24:371-86. [PMID: 24946075 DOI: 10.1111/bpa.12152] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/13/2014] [Indexed: 12/13/2022] Open
Abstract
Pericytes are cells in the blood-brain barrier (BBB) that degenerate in Alzheimer's disease (AD), a neurodegenerative disorder characterized by early neurovascular dysfunction, elevation of amyloid β-peptide (Aβ), tau pathology and neuronal loss, leading to progressive cognitive decline and dementia. Pericytes are uniquely positioned within the neurovascular unit between endothelial cells of brain capillaries, astrocytes and neurons. Recent studies have shown that pericytes regulate key neurovascular functions including BBB formation and maintenance, vascular stability and angioarchitecture, regulation of capillary blood flow, and clearance of toxic cellular by-products necessary for normal functioning of the central nervous system (CNS). Here, we review the concept of the neurovascular unit and neurovascular functions of CNS pericytes. Next, we discuss vascular contributions to AD and review new roles of pericytes in the pathogenesis of AD such as vascular-mediated Aβ-independent neurodegeneration, regulation of Aβ clearance and contributions to tau pathology, neuronal loss and cognitive decline. We conclude that future studies should focus on molecular mechanisms and pathways underlying aberrant signal transduction between pericytes and its neighboring cells within the neurovascular unit, that is, endothelial cells, astrocytes and neurons, which could represent potential therapeutic targets to control pericyte degeneration in AD and the resulting secondary vascular and neuronal degeneration.
Collapse
Affiliation(s)
- Ethan A Winkler
- Zilkha Neurogenetic Institute, University of Southern California Keck School of Medicine, Los Angeles, CA; Department of Neurosurgery, University of California San Francisco, San Francisco, CA
| | | | | |
Collapse
|
224
|
Brock AJ, Kasus-Jacobi A, Lerner M, Logan S, Adesina AM, Anne Pereira H. The antimicrobial protein, CAP37, is upregulated in pyramidal neurons during Alzheimer's disease. Histochem Cell Biol 2015; 144:293-308. [PMID: 26170148 PMCID: PMC4575391 DOI: 10.1007/s00418-015-1347-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2015] [Indexed: 01/02/2023]
Abstract
Inflammation is a well-defined factor in Alzheimer's disease (AD). There is a strong need to identify the molecules contributing to neuroinflammation so that therapies can be designed to prevent immune-mediated neurotoxicity. The cationic antimicrobial protein of 37 kDa (CAP37) is an inflammatory mediator constitutively expressed in neutrophils (PMNs). In addition to antibiotic activity, CAP37 exerts immunomodulatory effects on microglia. We hypothesize that CAP37 mediates the neuroinflammation associated with AD. However, PMNs are not customarily associated with the pathology of AD. This study was therefore designed to identify non-neutrophilic source(s) of CAP37 in brains of AD patients. Brain tissues from patients and age-matched controls were analyzed for CAP37 expression using immunohistochemistry (IHC). To determine factors that induce CAP37 in AD, HCN-1A primary human neurons were treated with tumor necrosis factor-alpha (TNF-α) or amyloid β1-40 (Aβ) and analyzed by IHC. Western blotting and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to confirm CAP37 expression in neurons and brain tissues. IHC revealed CAP37 in cortical neurons in temporal and parietal lobes as well as CA3 and CA4 hippocampal neurons in patients with AD. CAP37 was found in more neurons in AD patients compared with age-matched controls. qRT-PCR and Western blotting showed an increase in CAP37 transcript and protein in the AD temporal lobe, a brain region that is highly impacted in AD. qRT-PCR observations confirmed CAP37 expression in neurons. TNF-α and Aβ increased neuronal expression of CAP37. These findings support our hypothesis that neuronal CAP37 may modulate the neuroinflammatory response in AD.
Collapse
Affiliation(s)
- Amanda J Brock
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Anne Kasus-Jacobi
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA.,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA
| | - Megan Lerner
- Department of Surgery, University of Oklahoma Health Sciences Center, 1122 NE 13th St., ORB 350, Oklahoma City, OK, 73117, USA
| | - Sreemathi Logan
- Department of Geriatrics, University of Oklahoma Health Sciences Center, 975 NE 10th St., BRC 1303, Oklahoma City, OK, 73104, USA
| | - Adekunle M Adesina
- Department of Pathology, Baylor College of Medicine, One Baylor Plaza, Rm 286A, Houston, TX, 77030, USA
| | - H Anne Pereira
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 255, Oklahoma City, OK, 73117, USA. .,Department of Cell Biology, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 329, Oklahoma City, OK, USA. .,Department of Pathology, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Ave., CPB 329, Oklahoma City, OK, 73117, USA.
| |
Collapse
|
225
|
De Marco M, Venneri A. ‘O’ blood type is associated with larger grey-matter volumes in the cerebellum. Brain Res Bull 2015; 116:1-6. [DOI: 10.1016/j.brainresbull.2015.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 01/22/2023]
|
226
|
Jiang L, Zhong J, Dou X, Cheng C, Huang Z, Sun X. Effects of ApoE on intracellular calcium levels and apoptosis of neurons after mechanical injury. Neuroscience 2015; 301:375-83. [PMID: 26073697 DOI: 10.1016/j.neuroscience.2015.06.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 05/31/2015] [Accepted: 06/03/2015] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The current study aimed to explore the effects of apolipoprotein e (ApoE) on intracellular calcium ([Ca(2+)]i) and apoptosis of neurons after mechanical injury in vitro. METHODS A neuron mechanical injury model was established after primary neurons obtained from APOE knockout and wild-type (WT) mice, and four experimental groups were generated: Group-ApoE4, Group-ApoE3, Group-ApoE(-) and Group-WT. Recombinant ApoE4 and ApoE3 were added to Group-ApoE4 and Group-ApoE3 respectively, and Group-ApoE(-) and Group-WT were control groups. Intracellular calcium was labeled by fluo-3/AM and examined using laser scanning confocal microscope and flow cytometry, and the apoptosis of neurons was also evaluated. RESULTS The intracellular calcium levels and apoptosis rates of mice neurons were significantly higher in Group-ApoE4 than in Group-ApoE3 and Group-WT after mechanical injury. However, without mechanical injury on neurons, no significant differences in intracellular calcium levels and apoptosis rates were found among all four experimental groups. The effects of ApoE4 on intracellular calcium levels and apoptosis rates of injured neurons were partly decreased by EGTA treatment. CONCLUSION Compared with ApoE3-treatment and WT neurons, ApoE4 caused higher intracellular calcium levels and apoptosis rates of neurons after mechanical injury. This suggested APOE polymorphisms may affect neuron apoptosis after mechanical injury through different influences on intracellular calcium levels.
Collapse
Affiliation(s)
- L Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, PR China
| | - J Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, PR China
| | - X Dou
- Chongqing Medical University, PR China
| | - C Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, PR China
| | - Z Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, PR China
| | - X Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, PR China.
| |
Collapse
|
227
|
Currais A. Ageing and inflammation - A central role for mitochondria in brain health and disease. Ageing Res Rev 2015; 21:30-42. [PMID: 25684584 DOI: 10.1016/j.arr.2015.02.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 01/29/2015] [Accepted: 02/02/2015] [Indexed: 02/08/2023]
Abstract
To develop successful therapies that prevent or treat neurodegenerative diseases requires an understanding of the upstream events. Ageing is by far the greatest risk factor for most of these diseases, and to clarify their causes will require an understanding of the process of ageing itself. Starting with the question Why do we age as individual organisms, but the line of pluripotent embryonic stem cells and germ cells carried by individuals and transmitted to descendants is immortal? this review discusses how the process of cellular differentiation leads to the accumulation of biological imperfections with ageing, and how these imperfections may be the cause of chronic inflammatory responses to stress that undermine cellular function. Both differentiation and inflammation involve drastic metabolic changes associated with alterations in mitochondrial dynamics that shift the balance between aerobic glycolysis and oxidative phosphorylation. With ageing, mitochondrial dysfunction can be both the cause and consequence of inflammatory processes and elicit metabolic adaptations that might be either protective or become progressively detrimental. It is argued here that an understanding of the relationship between metabolism, differentiation and inflammation is essential to understand the pathological mechanisms governing brain health and disease during ageing.
Collapse
|
228
|
Alonso R, Pisa D, Marina AI, Morato E, Rábano A, Rodal I, Carrasco L. Evidence for fungal infection in cerebrospinal fluid and brain tissue from patients with amyotrophic lateral sclerosis. Int J Biol Sci 2015; 11:546-58. [PMID: 25892962 PMCID: PMC4400386 DOI: 10.7150/ijbs.11084] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/10/2015] [Indexed: 12/14/2022] Open
Abstract
Among neurogenerative diseases, amyotrophic lateral sclerosis (ALS) is a fatal illness characterized by a progressive motor neuron dysfunction in the motor cortex, brainstem and spinal cord. ALS is the most common form of motor neuron disease; yet, to date, the exact etiology of ALS remains unknown. In the present work, we have explored the possibility of fungal infection in cerebrospinal fluid (CSF) and in brain tissue from ALS patients. Fungal antigens, as well as DNA from several fungi, were detected in CSF from ALS patients. Additionally, examination of brain sections from the frontal cortex of ALS patients revealed the existence of immunopositive fungal antigens comprising punctate bodies in the cytoplasm of some neurons. Fungal DNA was also detected in brain tissue using PCR analysis, uncovering the presence of several fungal species. Finally, proteomic analyses of brain tissue demonstrated the occurrence of several fungal peptides. Collectively, our observations provide compelling evidence of fungal infection in the ALS patients analyzed, suggesting that this infection may play a part in the etiology of the disease or may constitute a risk factor for these patients.
Collapse
Affiliation(s)
- Ruth Alonso
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Diana Pisa
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Ana Isabel Marina
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Esperanza Morato
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| | - Alberto Rábano
- 2. Department of Neuropathology and Tissue Bank, Unidad de Investigación Proyecto Alzheimer, Fundación CIEN, Instituto de Salud Carlos III, Madrid. Spain
| | - Izaskun Rodal
- 2. Department of Neuropathology and Tissue Bank, Unidad de Investigación Proyecto Alzheimer, Fundación CIEN, Instituto de Salud Carlos III, Madrid. Spain
| | - Luis Carrasco
- 1. Centro de Biología Molecular "Severo Ochoa". c/Nicolás Cabrera, 1. Universidad Autónoma de Madrid. Cantoblanco. 28049 Madrid. Spain
| |
Collapse
|
229
|
|
230
|
The effect of depression on serum VEGF level in Alzheimer's disease. DISEASE MARKERS 2015; 2015:742612. [PMID: 25838619 PMCID: PMC4369941 DOI: 10.1155/2015/742612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 12/22/2022]
Abstract
Objective. Growing evidence suggests that angiogenesis might represent a new pathogenic mechanism involved in the progression of Alzheimer's disease (AD). Among angiogenic cytokines, vascular endothelial growth factor (VEGF) levels in AD patients have been evaluated, but the results are controversial among studies. We investigated serum levels of VEGF in AD patients with depression, AD patients without depression, and the controls, respectively. The aim of this study is to elucidate the relationship between VEGF, depression, and cognitive impairment in AD. Methods. The CDR (Clinical Dementia Rating), MMSE-KC (the Mini-Mental Status Examination-Korean version), and SGDS-K (the Korean version of the Geriatric Depression Scale-Short Form) were measured in the subjects. Serum VEGF levels were measured in 24 AD patients with depression, 25 AD patients without depression, and 26 controls, using an enzyme-linked immunosorbent assay kit. Results. Serum VEGF levels in AD patients with depression were significantly higher than AD patients without depression or the control. A correlation was observed between VEGF and scores on SGDS-K, but no correlation was detected between VEGF and MMSE-KC scores. Conclusion. Serum VEGF levels in AD patients with depression were higher than those without depression. Depression might be associated with changes in serum levels of VEGF in AD patients.
Collapse
|
231
|
Canobbio I, Abubaker AA, Visconte C, Torti M, Pula G. Role of amyloid peptides in vascular dysfunction and platelet dysregulation in Alzheimer's disease. Front Cell Neurosci 2015; 9:65. [PMID: 25784858 PMCID: PMC4347625 DOI: 10.3389/fncel.2015.00065] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 02/11/2015] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative cause of dementia in the elderly. AD is accompanied by the accumulation of amyloid peptides in the brain parenchyma and in the cerebral vessels. The sporadic form of AD accounts for about 95% of all cases. It is characterized by a late onset, typically after the age of 65, with a complex and still poorly understood aetiology. Several observations point towards a central role of cerebrovascular dysfunction in the onset of sporadic AD (SAD). According to the "vascular hypothesis", AD may be initiated by vascular dysfunctions that precede and promote the neurodegenerative process. In accordance to this, AD patients show increased hemorrhagic or ischemic stroke risks. It is now clear that multiple bidirectional connections exist between AD and cerebrovascular disease, and in this new scenario, the effect of amyloid peptides on vascular cells and blood platelets appear to be central to AD. In this review, we analyze the effect of amyloid peptides on vascular function and platelet activation and its contribution to the cerebrovascular pathology associated with AD and the progression of this disease.
Collapse
Affiliation(s)
- Ilaria Canobbio
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Aisha Alsheikh Abubaker
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Caterina Visconte
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Mauro Torti
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| | - Giordano Pula
- Department of Biology and Biotechnology, Unit of Biochemistry, University of Pavia Pavia, Italy
| |
Collapse
|
232
|
Solé M, Miñano-Molina AJ, Unzeta M. A cross-talk between Aβ and endothelial SSAO/VAP-1 accelerates vascular damage and Aβ aggregation related to CAA-AD. Neurobiol Aging 2015; 36:762-75. [DOI: 10.1016/j.neurobiolaging.2014.09.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 09/18/2014] [Accepted: 09/29/2014] [Indexed: 02/07/2023]
|
233
|
Fonseca ACR, Resende R, Cardoso SM, Pereira CF. The role of proteotoxic stress in vascular dysfunction in the pathogenesis of Alzheimer’s disease. ENDOPLASMIC RETICULUM STRESS IN DISEASES 2015. [DOI: 10.1515/ersc-2015-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractAlzheimer’s disease (AD) is the principal cause of dementia in the elderly; however, its prevalence is increasing due to the fact that current pharmaceuticals used to manage the symptoms are not capable of preventing, halting, or reversing disease progression. In the last decade, evidence has accumulated to support the hypothesis that a primary cerebral vascular dysfunction initiates the cascade of events that leads to neuronal injury and the subsequent cognitive decline observed in AD. The mechanisms underlying these vascular defects and their relationship with neurodegeneration are still poorly understood however. It is pathologically known that cerebrovascular dysfunctions can induce the deposition of amyloid-β (Aβ), an amyloidogenic and toxic peptide that in turn causes cerebrovascular degeneration. Mammalian cells regulate proteostasis and the functioning of intracellular organelles through diverse mechanisms such as the Unfolded Protein Response, the Ubiquitin-Proteasome System and autophagy; however, when these mechanisms cannot compensate for perturbations in homeostasis, the cell undergoes programmed death via apoptosis. This review summarizes recent studies that together correlate the deregulation of protein quality control pathways with dysfunction of vascular endothelial cells of the brain in AD, thus supporting the hypothesis that it is the vicious, progressive failure of the proteostatic network and endothelial activation that underlies the cerebrovascular changes that symptomize AD.
Collapse
|
234
|
Palavra F, Díaz EC, Sena A. Cardiometabolic Risk, Inflammation, and Neurodegenerative Disorders. BIOMARKERS OF CARDIOMETABOLIC RISK, INFLAMMATION AND DISEASE 2015:133-159. [DOI: 10.1007/978-3-319-16018-4_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
235
|
González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Metabolite profiling for the identification of altered metabolic pathways in Alzheimer's disease. J Pharm Biomed Anal 2014; 107:75-81. [PMID: 25575172 DOI: 10.1016/j.jpba.2014.10.010] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 10/08/2014] [Accepted: 10/10/2014] [Indexed: 01/05/2023]
Abstract
Gas chromatography coupled to mass spectrometry is the most frequent tool for metabolomic profiling of low molecular weight metabolites. Its suitability in health survey is beyond doubt, given that primary metabolites involved in central pathways of metabolism are usually altered in diseases. The objective of this work is to investigate metabolic differences in serum between Alzheimer's disease patients and healthy controls in order to elucidate pathological mechanisms underlying to disease. Alterations in levels of 23 metabolites were detected, including increased lactic acid, α-ketoglutarate, isocitric acid, glucose, oleic acid, adenosine and cholesterol, as well as decreased urea, valine, aspartic acid, pyroglutamate, glutamine, phenylalanine, asparagine, ornithine, pipecolic acid, histidine, tyrosine, palmitic and uric acid, tryptophan, stearic acid and cystine. Metabolic pathway analysis revealed the involvement of multiple affected pathways, such as energy deficiencies, oxidative stress, hyperammonemia, and others. Moreover, it is noteworthy that some of these compounds have not been previously described in AD research, such as α-ketoglutarate, isocitrate pipecolic acid, pyroglutamate and adenosine, confirming the potential of this metabolomic approach in the search of novel potential markers for early detection of Alzheimer's disease.
Collapse
Affiliation(s)
- Raúl González-Domínguez
- Department of Chemistry and CC.MM, Faculty of Experimental Science, University of Huelva, Campus de El Carmen, 21007 Huelva, Spain; Campus of Excellence International ceiA3, University of Huelva, Spain; Research Center of Health and Environment (CYSMA), University of Huelva, Campus de El Carmen, 21007 Huelva, Spain.
| | - Tamara García-Barrera
- Department of Chemistry and CC.MM, Faculty of Experimental Science, University of Huelva, Campus de El Carmen, 21007 Huelva, Spain; Campus of Excellence International ceiA3, University of Huelva, Spain; Research Center of Health and Environment (CYSMA), University of Huelva, Campus de El Carmen, 21007 Huelva, Spain.
| | - José Luis Gómez-Ariza
- Department of Chemistry and CC.MM, Faculty of Experimental Science, University of Huelva, Campus de El Carmen, 21007 Huelva, Spain; Campus of Excellence International ceiA3, University of Huelva, Spain; Research Center of Health and Environment (CYSMA), University of Huelva, Campus de El Carmen, 21007 Huelva, Spain.
| |
Collapse
|
236
|
da Fonseca ACC, Matias D, Garcia C, Amaral R, Geraldo LH, Freitas C, Lima FRS. The impact of microglial activation on blood-brain barrier in brain diseases. Front Cell Neurosci 2014; 8:362. [PMID: 25404894 PMCID: PMC4217497 DOI: 10.3389/fncel.2014.00362] [Citation(s) in RCA: 386] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 10/13/2014] [Indexed: 12/16/2022] Open
Abstract
The blood-brain barrier (BBB), constituted by an extensive network of endothelial cells (ECs) together with neurons and glial cells, including microglia, forms the neurovascular unit (NVU). The crosstalk between these cells guarantees a proper environment for brain function. In this context, changes in the endothelium-microglia interactions are associated with a variety of inflammation-related diseases in brain, where BBB permeability is compromised. Increasing evidences indicate that activated microglia modulate expression of tight junctions, which are essential for BBB integrity and function. On the other hand, the endothelium can regulate the state of microglial activation. Here, we review recent advances that provide insights into interactions between the microglia and the vascular system in brain diseases such as infectious/inflammatory diseases, epilepsy, ischemic stroke and neurodegenerative disorders.
Collapse
Affiliation(s)
- Anna Carolina Carvalho da Fonseca
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Diana Matias
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Celina Garcia
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Rackele Amaral
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Luiz Henrique Geraldo
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Catarina Freitas
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| | - Flavia Regina Souza Lima
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Centro de Ciências da Saúde, Bloco F, Universidade Federal do Rio de Janeiro Rio de Janeiro, RJ, Brazil
| |
Collapse
|
237
|
Gil-Montoya JA, Sanchez-Lara I, Carnero-Pardo C, Fornieles F, Montes J, Vilchez R, Burgos JS, Gonzalez-Moles MA, Barrios R, Bravo M. Is periodontitis a risk factor for cognitive impairment and dementia? A case-control study. J Periodontol 2014; 86:244-53. [PMID: 25345338 DOI: 10.1902/jop.2014.140340] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Dementia is a multi-etiologic syndrome characterized by multiple cognitive deficits but not always by the presence of cognitive impairment. Cognitive impairment is associated with multiple non-modifiable risk factors but few modifiable factors. Epidemiologic studies have shown an association between periodontitis, a potentially modifiable risk factor, and cognitive impairment. The objective of this study is to determine whether clinical periodontitis is associated with the diagnosis of cognitive impairment/dementia after controlling for known risk factors, including age, sex, and education level. METHODS A case-control study was conducted in Granada, Spain, in two groups of dentate individuals aged >50 years: 1) cases with a firm diagnosis of mild cognitive impairment or dementia of any type or severity and 2) controls with no subjective memory loss complaints and a score >30 in the Phototest cognitive test (screening test for cognitive impairment). Periodontitis was evaluated by measuring tooth loss, plaque and bleeding indexes, probing depths, and clinical attachment loss (AL). RESULTS The study included 409 dentate adults, 180 with cognitive impairment and 229 without. A moderate and statistically significant association was observed between AL and cognitive impairment after controlling for age, sex, education level, oral hygiene habits, and hyperlipidemia (P = 0.049). No significant association was found between tooth loss and cognitive impairment. CONCLUSION Periodontitis appears to be associated with cognitive impairment after controlling for confounders such as age, sex, and education level.
Collapse
|
238
|
Williams R, Münch G, Gyengesi E, Bennett L. Bacopa monnieri (L.) exerts anti-inflammatory effects on cells of the innate immune system in vitro. Food Funct 2014; 5:517-20. [PMID: 24452710 DOI: 10.1039/c3fo60467e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Bacopa monnieri (L., BM) is a traditional Ayurvedic medicinal herb recognised for its efficacy in relieving acute pain and inflammation, as related to selective inhibition of cyclo-oxygenase-2 (COX-2) enzyme and consequent reduction in COX-2-mediated prostanoid mediators. BM is also associated with cognitive enhancing (nootropic) activity including improving memory free recall, observed after prolonged intake (>3 months). It is likely that the time frame required to exert an effect in the brain reflects regulation by BM of chronic inflammation and oxidative stress associated with aging and chronic diseases, and other polypharmacological effects. We report down-regulation by BM of NO and TNF-α in stimulated RAW 246.7 macrophages and of IFN-γ in stimulated human blood cells. Furthermore, in human blood cells, IL-10 was slightly elevated indicating polarisation towards a regulatory T cell phenotype. These results provide further supportive evidence to justify the clinical evaluation of BM for managing diseases involving chronic systemic and brain inflammation driven by the innate immune system.
Collapse
Affiliation(s)
- Roderick Williams
- CSIRO Preventative Health Flagship, Animal, Food and Health Sciences, 671 Sneydes Road, Werribee, Victoria 3030, Australia
| | | | | | | |
Collapse
|
239
|
Coenzyme Q10 protects human endothelial cells from β-amyloid uptake and oxidative stress-induced injury. PLoS One 2014; 9:e109223. [PMID: 25272163 PMCID: PMC4182835 DOI: 10.1371/journal.pone.0109223] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 08/31/2014] [Indexed: 01/30/2023] Open
Abstract
Neuropathological symptoms of Alzheimer's disease appear in advances stages, once neuronal damage arises. Nevertheless, recent studies demonstrate that in early asymptomatic stages, ß-amyloid peptide damages the cerebral microvasculature through mechanisms that involve an increase in reactive oxygen species and calcium, which induces necrosis and apoptosis of endothelial cells, leading to cerebrovascular dysfunction. The goal of our work is to study the potential preventive effect of the lipophilic antioxidant coenzyme Q (CoQ) against ß-amyloid-induced damage on human endothelial cells. We analyzed the protective effect of CoQ against Aβ-induced injury in human umbilical vein endothelial cells (HUVECs) using fluorescence and confocal microscopy, biochemical techniques and RMN-based metabolomics. Our results show that CoQ pretreatment of HUVECs delayed Aβ incorporation into the plasma membrane and mitochondria. Moreover, CoQ reduced the influx of extracellular Ca2+, and Ca2+ release from mitochondria due to opening the mitochondrial transition pore after β-amyloid administration, in addition to decreasing O2.− and H2O2 levels. Pretreatment with CoQ also prevented ß-amyloid-induced HUVECs necrosis and apoptosis, restored their ability to proliferate, migrate and form tube-like structures in vitro, which is mirrored by a restoration of the cell metabolic profile to control levels. CoQ protected endothelial cells from Aβ-induced injury at physiological concentrations in human plasma after oral CoQ supplementation and thus could be a promising molecule to protect endothelial cells against amyloid angiopathy.
Collapse
|
240
|
González-Domínguez R, García-Barrera T, Gómez-Ariza JL. Using direct infusion mass spectrometry for serum metabolomics in Alzheimer’s disease. Anal Bioanal Chem 2014; 406:7137-48. [DOI: 10.1007/s00216-014-8102-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/04/2014] [Accepted: 08/11/2014] [Indexed: 12/15/2022]
|
241
|
The role of the blood-brain barrier in the pathogenesis of senile plaques in Alzheimer's disease. Int J Alzheimers Dis 2014; 2014:191863. [PMID: 25309772 PMCID: PMC4189524 DOI: 10.1155/2014/191863] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/03/2014] [Accepted: 09/08/2014] [Indexed: 11/30/2022] Open
Abstract
The accumulation of beta-amyloid [Aβ] within senile plaques [SP] is characteristic of these lesions in Alzheimer's disease. The accumulation of Aβ42, in particular, in the superior temporal [ST] cortex may result from an inability of the blood brain barrier (BBB) to regulate the trans-endothelial transport and clearance of the amyloid. Lipoprotein receptor-related protein [LRP] and P-glycoprotein [P-gp] facilitate the efflux of Aβ out of the brain, whereas receptor for advanced glycation end products [RAGE] facilitates Aβ influx. Additionally, vascular endothelial growth factor [VEGF] and endothelial nitric oxide synthase [eNOS] may influence the trans-BBB transport of Aβ. In this study we examined ST samples and compared SP burden of all types with the capillary expression of LRP, p-gp, RAGE, VEGF, and e-NOS in samples from 15 control and 15 Alzheimer brains. LRP, P-gp, RAGE, VEGF, and eNOS positive capillaries and Aβ42 plaques were quantified and statistical analysis of the nonparametric data was performed using the Mann-Whitney and Kruskal-Wallis tests. In the Alzheimer condition P-gp, VEGF, and eNOS positive capillaries were negatively correlated with SP burden, but LRP and RAGE were positively correlated with SP burden. These results indicate altered BBB function in the pathogenesis of SPs in Alzheimer brains.
Collapse
|
242
|
Metabolomic study of lipids in serum for biomarker discovery in Alzheimer's disease using direct infusion mass spectrometry. J Pharm Biomed Anal 2014; 98:321-6. [DOI: 10.1016/j.jpba.2014.05.023] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 04/12/2014] [Accepted: 05/08/2014] [Indexed: 12/22/2022]
|
243
|
Maarouf CL, Kokjohn TA, Walker DG, Whiteside CM, Kalback WM, Whetzel A, Sue LI, Serrano G, Jacobson SA, Sabbagh MN, Reiman EM, Beach TG, Roher AE. Biochemical assessment of precuneus and posterior cingulate gyrus in the context of brain aging and Alzheimer's disease. PLoS One 2014; 9:e105784. [PMID: 25166759 PMCID: PMC4148328 DOI: 10.1371/journal.pone.0105784] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 07/24/2014] [Indexed: 12/12/2022] Open
Abstract
Defining the biochemical alterations that occur in the brain during “normal” aging is an important part of understanding the pathophysiology of neurodegenerative diseases and of distinguishing pathological conditions from aging-associated changes. Three groups were selected based on age and on having no evidence of neurological or significant neurodegenerative disease: 1) young adult individuals, average age 26 years (n = 9); 2) middle-aged subjects, average age 59 years (n = 5); 3) oldest-old individuals, average age 93 years (n = 6). Using ELISA and Western blotting methods, we quantified and compared the levels of several key molecules associated with neurodegenerative disease in the precuneus and posterior cingulate gyrus, two brain regions known to exhibit early imaging alterations during the course of Alzheimer’s disease. Our experiments revealed that the bioindicators of emerging brain pathology remained steady or decreased with advancing age. One exception was S100B, which significantly increased with age. Along the process of aging, neurofibrillary tangle deposition increased, even in the absence of amyloid deposition, suggesting the presence of amyloid plaques is not obligatory for their development and that limited tangle density is a part of normal aging. Our study complements a previous assessment of neuropathology in oldest-old subjects, and within the limitations of the small number of individuals involved in the present investigation, it adds valuable information to the molecular and structural heterogeneity observed along the course of aging and dementia. This work underscores the need to examine through direct observation how the processes of amyloid deposition unfold or change prior to the earliest phases of dementia emergence.
Collapse
Affiliation(s)
- Chera L. Maarouf
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Tyler A. Kokjohn
- Department of Microbiology, Midwestern University, Glendale, Arizona, United States of America
| | - Douglas G. Walker
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Charisse M. Whiteside
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Walter M. Kalback
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alexis Whetzel
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Lucia I. Sue
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Geidy Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Sandra A. Jacobson
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Marwan N. Sabbagh
- Cleo Roberts Center for Clinical Research, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, Arizona, United States of America
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
| | - Alex E. Roher
- The Longtine Center for Neurodegenerative Biochemistry, Banner Sun Health Research Institute, Sun City, Arizona, United States of America
- * E-mail:
| |
Collapse
|
244
|
Nazeri A, Ganjgahi H, Roostaei T, Nichols T, Zarei M. Imaging proteomics for diagnosis, monitoring and prediction of Alzheimer's disease. Neuroimage 2014; 102 Pt 2:657-65. [PMID: 25173418 DOI: 10.1016/j.neuroimage.2014.08.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 08/18/2014] [Accepted: 08/22/2014] [Indexed: 01/18/2023] Open
Abstract
Proteomic and imaging markers have been widely studied as potential biomarkers for diagnosis, monitoring and prognosis of Alzheimer's disease. In this study, we used Alzheimer Disease Neuroimaging Initiative dataset and performed parallel independent component analysis on cross sectional and longitudinal proteomic and imaging data in order to identify the best proteomic model for diagnosis, monitoring and prediction of Alzheimer disease (AD). We used plasma proteins measurement and imaging data from AD and healthy controls (HC) at the baseline and 1 year follow-up. Group comparisons at baseline and changes over 1 year were calculated for proteomic and imaging data. The results were fed into parallel independent component analysis in order to identify proteins that were associated with structural brain changes cross sectionally and longitudinally. Regression model was used to find the best model that can discriminate AD from HC, monitor AD and to predict MCI converters from non-converters. We showed that five proteins are associated with structural brain changes in the brain. These proteins could discriminate AD from HC with 57% specificity and 89% sensitivity. Four proteins whose change over 1 year were associated with brain structural changes could discriminate AD from HC with sensitivity of 93%, and specificity of 92%. This model predicted MCI conversion to AD in 2 years with 94% accuracy. This model has the highest accuracy in prediction of MCI conversion to AD within the ADNI-1 dataset. This study shows that combination of selected plasma protein levels and MR imaging is a useful method in identifying potential biomarker.
Collapse
Affiliation(s)
- Arash Nazeri
- Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Habib Ganjgahi
- National Brain Mapping Centre, and Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran 4739, Iran; Department of Statistics, University of Warwick, Coventry CV4 7AL, UK
| | - Tina Roostaei
- Interdisciplinary Neuroscience Research Program, Tehran University of Medical Sciences, Tehran, 1417614411, Iran
| | - Thomas Nichols
- Department of Statistics, University of Warwick, Coventry CV4 7AL, UK
| | - Mojtaba Zarei
- National Brain Mapping Centre, and Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran 4739, Iran.
| | | |
Collapse
|
245
|
Martin C. Contributions and complexities from the use of in vivo animal models to improve understanding of human neuroimaging signals. Front Neurosci 2014; 8:211. [PMID: 25191214 PMCID: PMC4137227 DOI: 10.3389/fnins.2014.00211] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 07/01/2014] [Indexed: 01/18/2023] Open
Abstract
Many of the major advances in our understanding of how functional brain imaging signals relate to neuronal activity over the previous two decades have arisen from physiological research studies involving experimental animal models. This approach has been successful partly because it provides opportunities to measure both the hemodynamic changes that underpin many human functional brain imaging techniques and the neuronal activity about which we wish to make inferences. Although research into the coupling of neuronal and hemodynamic responses using animal models has provided a general validation of the correspondence of neuroimaging signals to specific types of neuronal activity, it is also highlighting the key complexities and uncertainties in estimating neural signals from hemodynamic markers. This review will detail how research in animal models is contributing to our rapidly evolving understanding of what human neuroimaging techniques tell us about neuronal activity. It will highlight emerging issues in the interpretation of neuroimaging data that arise from in vivo research studies, for example spatial and temporal constraints to neuroimaging signal interpretation, or the effects of disease and modulatory neurotransmitters upon neurovascular coupling. We will also give critical consideration to the limitations and possible complexities of translating data acquired in the typical animals models used in this area to the arena of human fMRI. These include the commonplace use of anesthesia in animal research studies and the fact that many neuropsychological questions that are being actively explored in humans have limited homologs within current animal models for neuroimaging research. Finally we will highlighting approaches, both in experimental animals models (e.g. imaging in conscious, behaving animals) and human studies (e.g. combined fMRI-EEG), that mitigate against these challenges.
Collapse
Affiliation(s)
- Chris Martin
- Department of Psychology, The University of Sheffield Sheffield, UK
| |
Collapse
|
246
|
Banks WA, Abrass CK, Hansen KM. Differentiating the Influences of Aging and Adiposity on Brain Weights, Levels of Serum and Brain Cytokines, Gastrointestinal Hormones, and Amyloid Precursor Protein. J Gerontol A Biol Sci Med Sci 2014; 71:21-9. [PMID: 25128822 DOI: 10.1093/gerona/glu100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/02/2014] [Indexed: 12/23/2022] Open
Abstract
Aging and obesity exert important effects on disease. Differentiating these effects is difficult, however, because weight gain often accompanies aging. Here, we used a nested design of aged, calorically restricted, and refed rats to measure changes in brain and blood levels of cytokines and gastrointestinal hormones, brain amyloid precursor protein levels, and brain and body weights. By comparing groups and using path analysis, we found divergent influences of chronological aging versus body weight, our main findings being (i) changes in whole brain weight and serum macrophage colony-stimulating factor levels correlated better with body weight than with chronological aging, (ii) a decrease in brain cytokines and brain plasminogen activator inhibitor levels correlated better with chronological aging than with body weight, (iii) serum erythropoietin levels were influenced by both body weight and aging, (iv) serum plasminogen activator inhibitor, serum cytokines, and brain tumor necrosis factor were not influenced by aging or body weight, and (v) brain amyloid precursor protein more closely related to body weight and serum levels of gastrointestinal hormones than to brain weight, chronological aging, or cytokines. These findings show that although aging and body weight interact, their influences are distinct not only among various cytokines and hormones but also between the central nervous system and the peripheral tissue compartments.
Collapse
Affiliation(s)
- William A Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington. Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle.
| | - Christine K Abrass
- Primary and Specialty Care Medicine, Department of Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| | - Kim M Hansen
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington
| |
Collapse
|
247
|
Increased risk of dementia among chronic osteomyelitis patients. Eur J Clin Microbiol Infect Dis 2014; 34:153-159. [DOI: 10.1007/s10096-014-2200-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 06/27/2014] [Indexed: 10/24/2022]
|
248
|
Wang F, Guo X, Shen X, Kream RM, Mantione KJ, Stefano GB. Vascular dysfunction associated with type 2 diabetes and Alzheimer's disease: a potential etiological linkage. Med Sci Monit Basic Res 2014; 20:118-29. [PMID: 25082505 PMCID: PMC4138067 DOI: 10.12659/msmbr.891278] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The endothelium performs a crucial role in maintaining vascular integrity leading to whole organ metabolic homeostasis. Endothelial dysfunction represents a key etiological factor leading to moderate to severe vasculopathies observed in both Type 2 diabetic and Alzheimer’s Disease (AD) patients. Accordingly, evidence-based epidemiological factors support a compelling hypothesis stating that metabolic rundown encountered in Type 2 diabetes engenders severe cerebral vascular insufficiencies that are causally linked to long term neural degenerative processes in AD. Of mechanistic importance, Type 2 diabetes engenders an immunologically mediated chronic pro-inflammatory state involving interactive deleterious effects of leukocyte-derived cytokines and endothelial-derived chemotactic agents leading to vascular and whole organ dysfunction. The long term negative consequences of vascular pro-inflammatory processes on the integrity of CNS basal forebrain neuronal populations mediating complex cognitive functions establish a striking temporal comorbidity of AD with Type 2 diabetes. Extensive biomedical evidence supports the pivotal multi-functional role of constitutive nitric oxide (NO) production and release as a critical vasodilatory, anti-inflammatory, and anti-oxidant, mechanism within the vascular endothelium. Within this context, we currently review the functional contributions of dysregulated endothelial NO expression to the etiology and persistence of Type 2 diabetes-related and co morbid AD-related vasculopathies. Additionally, we provide up-to-date perspectives on critical areas of AD research with special reference to common NO-related etiological factors linking Type 2 diabetes to the pathogenesis of AD.
Collapse
Affiliation(s)
- Fuzhou Wang
- Department of Anesthesiology and Critical Care Medicine, Nanjing Maternit and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Xirong Guo
- Institutes of Pediatrics, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Xiaofeng Shen
- Department of Anesthesiology and Critical Care Medicine, Nanjing Maternity and Child Health Care Hospital, Nanjing Medical University, Nanjing, China (mainland)
| | - Richard M Kream
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| | - Kirk J Mantione
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| | - George B Stefano
- Neuroscience Research Institute, State University of New York - College at Old Westbury, Old Westbury, USA
| |
Collapse
|
249
|
Volume of the hippocampal subfields in healthy adults: differential associations with age and a pro-inflammatory genetic variant. Brain Struct Funct 2014; 220:2663-74. [PMID: 24947882 DOI: 10.1007/s00429-014-0817-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 06/05/2014] [Indexed: 12/14/2022]
Abstract
The hippocampus is one of the most age-sensitive brain regions, yet the mechanisms of hippocampal shrinkage remain unclear. Recent studies suggest that hippocampal subfields are differentially vulnerable to aging and differentially sensitive to vascular risk. Promoters of inflammation are frequently proposed as major contributors to brain aging and vascular disease but their effects on hippocampal subfields are unknown. We examined the associations of hippocampal subfield volumes with age, a vascular risk factor (hypertension), and genetic polymorphisms associated with variation in pro-inflammatory cytokines levels (IL-1β C-511T and IL-6 C-174G) and risk for Alzheimer's disease (APOEε4) in healthy adult volunteers (N = 80; age = 22-82 years). Volumes of three hippocampal subfields, cornu ammonis (CA) 1-2, CA3-dentate gyrus, and the subiculum were manually measured on high-resolution magnetic resonance images. Advanced age was differentially associated with smaller volume of CA1-2, whereas carriers of the T allele of IL-1β C-511T polymorphism had smaller volume of all hippocampal subfields than CC homozygotes did. Neither of the other genetic variants, nor diagnosis of hypertension, was associated with any of the measured volumes. The results support the notion that volumes of age-sensitive brain regions may be affected by pro-inflammatory factors that may be targeted by therapeutic interventions.
Collapse
|
250
|
Abstract
Vascular risk factors and cerebrovascular disease are common causes of dementia. Shared risk factors for vascular dementia and Alzheimer's disease, as well as frequent coexistence of these pathologies in cognitively impaired older people, suggests convergence of the aetiology, prevention and management of the commonest dementias affecting older people. In light of this understanding, the cognitive impairment associated with cerebrovascular disease is an increasingly important and recognised area of the medicine of older people. Although the incidence of cerebrovascular events is declining in many populations, the overall burden associated with brain vascular disease will continue to increase associated with population ageing. A spectrum of cognitive disorders related to cerebrovascular disease is now recognised. Cerebrovascular disease in older people is associated with specific clinical and imaging findings. Although prevention remains the cornerstone of management, the diagnosis of brain vascular disease is important because of the potential to improve clinical outcomes through clear diagnosis, enhanced control of risk factors, lifestyle interventions and secondary prevention. Specific pharmacological intervention may also be indicated for some patients with cognitive impairment and cerebrovascular disease. However the evidence base to guide intervention remains relatively sparse.
Collapse
|