201
|
Lazard D, Vardi P, Bloch K. Anti-diabetic and neuroprotective effects of pancreatic islet transplantation into the central nervous system. Diabetes Metab Res Rev 2016; 32:11-20. [PMID: 25708430 DOI: 10.1002/dmrr.2644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/19/2015] [Indexed: 12/19/2022]
Abstract
During the last decades, the central nervous system (CNS) was intensively tested as a site for islet transplantation in different animal models of diabetes. Immunoprivilege properties of intracranial and intrathecal sites were found to delay and reduce rejection of transplanted allo-islets and xeno-islets, especially in the form of dispersed single cells. Insulin released from islets grafted in CNS was shown to cross the blood-brain barrier and to act as a regulator of peripheral glucose metabolism. In diabetic animals, sufficient nutrition and oxygen supply to islets grafted in the CNS provide adequate insulin response to increase glucose level resulting in rapid normoglycemia. In addition to insulin, pancreatic islets produce and secrete several other hormones, as well as neurotrophic and angiogenic factors with potential neuroprotective properties. Recent experimental studies and clinical trials provide a strong support for delivery of islet-derived macromolecules to CNS as a promising strategy to treat various brain disorders. This review article focuses mainly on analysis of current status of intracranial and intrathecal islet transplantations for treatment of experimental diabetes and discusses the possible neuroprotective properties of grafted islets into CNS as a novel therapeutic approach to brain disorders with cognitive dysfunctions characterized by impaired brain insulin signalling. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier
- Brain
- Central Nervous System
- Diabetes Mellitus, Type 1/blood
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/surgery
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Diabetic Neuropathies/prevention & control
- Disease Models, Animal
- Humans
- Hyperglycemia/prevention & control
- Hypoglycemia/prevention & control
- Insulin/metabolism
- Insulin Resistance
- Insulin Secretion
- Islets of Langerhans Transplantation/adverse effects
- Spinal Cord
- Subarachnoid Space
- Transplantation, Heterologous/adverse effects
- Transplantation, Heterotopic/adverse effects
Collapse
Affiliation(s)
- Daniel Lazard
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Pnina Vardi
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| | - Konstantin Bloch
- Laboratory of Diabetes and Obesity Research, Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv University, Petah Tikva, Israel
| |
Collapse
|
202
|
Abdul-Hay SO, Bannister TD, Wang H, Cameron MD, Caulfield TR, Masson A, Bertrand J, Howard EA, McGuire MP, Crisafulli U, Rosenberry TR, Topper CL, Thompson CR, Schürer SC, Madoux F, Hodder P, Leissring MA. Selective Targeting of Extracellular Insulin-Degrading Enzyme by Quasi-Irreversible Thiol-Modifying Inhibitors. ACS Chem Biol 2015; 10:2716-24. [PMID: 26398879 PMCID: PMC10127574 DOI: 10.1021/acschembio.5b00334] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Many therapeutically important enzymes are present in multiple cellular compartments, where they can carry out markedly different functions; thus, there is a need for pharmacological strategies to selectively manipulate distinct pools of target enzymes. Insulin-degrading enzyme (IDE) is a thiol-sensitive zinc-metallopeptidase that hydrolyzes diverse peptide substrates in both the cytosol and the extracellular space, but current genetic and pharmacological approaches are incapable of selectively inhibiting the protease in specific subcellular compartments. Here, we describe the discovery, characterization, and kinetics-based optimization of potent benzoisothiazolone-based inhibitors that, by virtue of a unique quasi-irreversible mode of inhibition, exclusively inhibit extracellular IDE. The mechanism of inhibition involves nucleophilic attack by a specific active-site thiol of the enzyme on the inhibitors, which bear an isothiazolone ring that undergoes irreversible ring opening with the formation of a disulfide bond. Notably, binding of the inhibitors is reversible under reducing conditions, thus restricting inhibition to IDE present in the extracellular space. The identified inhibitors are highly potent (IC50(app) = 63 nM), nontoxic at concentrations up to 100 μM, and appear to preferentially target a specific cysteine residue within IDE. These novel inhibitors represent powerful new tools for clarifying the physiological and pathophysiological roles of this poorly understood protease, and their unusual mechanism of action should be applicable to other therapeutic targets.
Collapse
Affiliation(s)
- Samer O. Abdul-Hay
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | | | | | | | - Thomas R. Caulfield
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Amandine Masson
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Juliette Bertrand
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Erin A. Howard
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Michael P. McGuire
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Umberto Crisafulli
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Terrone R. Rosenberry
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
| | - Caitlyn L. Topper
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| | - Caroline R. Thompson
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| | - Stephan C. Schürer
- Department
of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, Florida 33136, United States
| | | | | | - Malcolm A. Leissring
- Department
of Neuroscience, Mayo Clinic Florida, Jacksonville, Florida 32224, United States
- Institute
for Memory Impairments and Neurological Disorders (UCI MIND), University of California, Irvine, California 92697, United States
| |
Collapse
|
203
|
Murakami K, Watanabe T, Koike T, Kamata M, Igari T, Kondo S. Pharmacological properties of a novel and potent γ-secretase modulator as a therapeutic option for the treatment of Alzheimer's disease. Brain Res 2015; 1633:73-86. [PMID: 26707977 DOI: 10.1016/j.brainres.2015.12.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 12/03/2015] [Accepted: 12/11/2015] [Indexed: 11/17/2022]
Abstract
Previous studies of γ-secretase inhibitors (GSIs) and Notch-sparing GSIs have shown reduced amyloid-β (Aβ) peptide levels but increased Notch-related and -unrelated adverse effects. In this study, we examined the effects of compound-1 on Aβ processing and cognitive function and assessed Notch-related and -unrelated adverse effects. Compound-1 reduced Aβ40 and Aβ42 levels but inversely increased Aβ37 in Neuro2a cells, leading to no net changes in total Aβ levels, indicating that compound-1 is a γ-secretase modulator (GSM). In time-course experiments, compound-1 reduced Aβ40 and Aβ42 levels in tris-soluble fractions, with peak reduction at approximately 3h after oral administration in C57BL mice. Moreover, at >1mg/kg, compound-1 dose dependently reduced Aβ40 and Aβ42 levels in Tg2576 mice. Chronic treatment with compound-1 in Tg2576 mice for 4 months significantly reduced both soluble and insoluble Aβ42 levels and ameliorated cognitive impairments, even after drug withdrawal for 10 days following oral administration for 2 months. In contrast with compound-1, at 100-fold higher doses (100mg/kg), the GSI LY450139 decreased HES1 mRNA expression in thymus tissues and increased the intensity of periodic acid-Schiff (PAS)-positive areas in the intestine. Moreover, the Notch-sparing GSI BMS708163 led to amyloid precursor protein (APP)-β-C-terminal fragment accumulation in mouse primary neurons. BMS708163 significantly hampered cognitive function in normal mice 1 month after administration, whereas compound-1 significantly improved cognitive function. Taken together, the present novel and orally active GSM is a promising molecule for the treatment of pathologies associated with Aβ42 and Aβ40.
Collapse
Affiliation(s)
- Koji Murakami
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomomichi Watanabe
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tatsuki Koike
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Makoto Kamata
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Tomoko Igari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan
| | - Shinichi Kondo
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Kanagawa 251-8555, Japan.
| |
Collapse
|
204
|
Hughes TM, Craft S. The role of insulin in the vascular contributions to age-related dementia. Biochim Biophys Acta Mol Basis Dis 2015; 1862:983-91. [PMID: 26657615 DOI: 10.1016/j.bbadis.2015.11.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/17/2015] [Accepted: 11/29/2015] [Indexed: 01/12/2023]
Abstract
In addition to its well-known role in energy metabolism in the body, insulin is a vasoactive hormone that regulates peripheral and cerebral blood flow and neuronal function. Vascular and metabolic dysfunctions are emerging risk factors for Alzheimer's disease (AD) and age-related dementias, and recent evidence suggests that the two pathways are constitutive and interrelated. As a result, an emphasis on correcting metabolic disorders is emerging as an important strategy in the treatment and prevention of age-related cognitive impairment and AD. We review the evidence regarding the unique and interactive effects of vascular and metabolic disorders in pathological brain aging, with special consideration of the role of insulin dysregulation in promoting AD pathologic processes and vascular brain injury. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Timothy M Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Wake Forest University, USA.
| |
Collapse
|
205
|
Inflammation and Oxidative Stress: The Molecular Connectivity between Insulin Resistance, Obesity, and Alzheimer's Disease. Mediators Inflamm 2015; 2015:105828. [PMID: 26693205 PMCID: PMC4674598 DOI: 10.1155/2015/105828] [Citation(s) in RCA: 335] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 09/29/2015] [Indexed: 12/13/2022] Open
Abstract
Type 2 diabetes (T2DM), Alzheimer's disease (AD), and insulin resistance are age-related conditions and increased prevalence is of public concern. Recent research has provided evidence that insulin resistance and impaired insulin signalling may be a contributory factor to the progression of diabetes, dementia, and other neurological disorders. Alzheimer's disease (AD) is the most common subtype of dementia. Reduced release (for T2DM) and decreased action of insulin are central to the development and progression of both T2DM and AD. A literature search was conducted to identify molecular commonalities between obesity, diabetes, and AD. Insulin resistance affects many tissues and organs, either through impaired insulin signalling or through aberrant changes in both glucose and lipid (cholesterol and triacylglycerol) metabolism and concentrations in the blood. Although epidemiological and biological evidence has highlighted an increased incidence of cognitive decline and AD in patients with T2DM, the common molecular basis of cell and tissue dysfunction is rapidly gaining recognition. As a cause or consequence, the chronic inflammatory response and oxidative stress associated with T2DM, amyloid-β (Aβ) protein accumulation, and mitochondrial dysfunction link T2DM and AD.
Collapse
|
206
|
Amemori T, Jendelova P, Ruzicka J, Urdzikova LM, Sykova E. Alzheimer's Disease: Mechanism and Approach to Cell Therapy. Int J Mol Sci 2015; 16:26417-51. [PMID: 26556341 PMCID: PMC4661820 DOI: 10.3390/ijms161125961] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/26/2015] [Accepted: 10/26/2015] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. The risk of AD increases with age. Although two of the main pathological features of AD, amyloid plaques and neurofibrillary tangles, were already recognized by Alois Alzheimer at the beginning of the 20th century, the pathogenesis of the disease remains unsettled. Therapeutic approaches targeting plaques or tangles have not yet resulted in satisfactory improvements in AD treatment. This may, in part, be due to early-onset and late-onset AD pathogenesis being underpinned by different mechanisms. Most animal models of AD are generated from gene mutations involved in early onset familial AD, accounting for only 1% of all cases, which may consequently complicate our understanding of AD mechanisms. In this article, the authors discuss the pathogenesis of AD according to the two main neuropathologies, including senescence-related mechanisms and possible treatments using stem cells, namely mesenchymal and neural stem cells.
Collapse
Affiliation(s)
- Takashi Amemori
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Pavla Jendelova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| | - Jiri Ruzicka
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Lucia Machova Urdzikova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
| | - Eva Sykova
- Department of Neuroscience, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic.
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, V Uvalu 84, 150 06 Prague 5, Czech Republic.
| |
Collapse
|
207
|
Son SM, Kang S, Choi H, Mook-Jung I. Statins induce insulin-degrading enzyme secretion from astrocytes via an autophagy-based unconventional secretory pathway. Mol Neurodegener 2015; 10:56. [PMID: 26520569 PMCID: PMC4628355 DOI: 10.1186/s13024-015-0054-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 10/23/2015] [Indexed: 11/21/2022] Open
Abstract
Background Insulin degrading enzyme (IDE) is a major protease of amyloid beta peptide (Aβ), a prominent toxic protein in Alzheimer’s disease (AD) pathogenesis. Previous studies suggested that statins promote IDE secretion; however, the underlying mechanism is unknown, as IDE has no signal sequence. Results In this study, we found that simvastatin (0.2 μM for 12 h) induced the degradation of extracellular Aβ40, which depended on IDE secretion from primary astrocytes. In addition, simvastatin increased IDE secretion from astrocytes in a time- and dose-dependent manner. Moreover, simvastatin-mediated IDE secretion was mediated by an autophagy-based unconventional secretory pathway, and autophagic flux regulated simvastatin-mediated IDE secretion. Finally, simvastatin activated autophagy via the LKB1-AMPK-mTOR signaling pathway in astrocytes. Conclusions These results demonstrate a novel pathway for statin-mediated IDE secretion from astrocytes. Modulation of this pathway could provide a potential therapeutic target for treatment of Aβ pathology by enhancing extracellular clearance of Aβ. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0054-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sung Min Son
- Department of Biochemistry & Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| | - Seokjo Kang
- Department of Biochemistry & Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Heesun Choi
- Department of Biochemistry & Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea.
| | - Inhee Mook-Jung
- Department of Biochemistry & Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongro-gu, Seoul, 110-799, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
208
|
Catalytic site inhibition of insulin-degrading enzyme by a small molecule induces glucose intolerance in mice. Nat Commun 2015; 6:8250. [PMID: 26394692 PMCID: PMC4580987 DOI: 10.1038/ncomms9250] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 07/31/2015] [Indexed: 01/22/2023] Open
Abstract
Insulin-degrading enzyme (IDE) is a protease that cleaves insulin and other bioactive peptides such as amyloid-β. Knockout and genetic studies have linked IDE to Alzheimer's disease and type-2 diabetes. As the major insulin-degrading protease, IDE is a candidate drug target in diabetes. Here we have used kinetic target-guided synthesis to design the first catalytic site inhibitor of IDE suitable for in vivo studies (BDM44768). Crystallographic and small angle X-ray scattering analyses show that it locks IDE in a closed conformation. Among a panel of metalloproteases, BDM44768 selectively inhibits IDE. Acute treatment of mice with BDM44768 increases insulin signalling and surprisingly impairs glucose tolerance in an IDE-dependent manner. These results confirm that IDE is involved in pathways that modulate short-term glucose homeostasis, but casts doubt on the general usefulness of the inhibition of IDE catalytic activity to treat diabetes. Inhibiting insulin-degrading enzyme (IDE) has been proposed as a potential therapeutic strategy for the treatment of patients with diabetes. Here, the authors develop a novel IDE inhibitor but find that, surprisingly, IDE inhibition has negative effects on glucose tolerance in mice.
Collapse
|
209
|
Grasso G, Lanza V, Malgieri G, Fattorusso R, Pietropaolo A, Rizzarelli E, Milardi D. The insulin degrading enzyme activates ubiquitin and promotes the formation of K48 and K63 diubiquitin. Chem Commun (Camb) 2015; 51:15724-7. [PMID: 26364617 DOI: 10.1039/c5cc06786c] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
We report an ATP-dependent ubiquitin conjugation with IDE which, in turn, promotes Ub-Ub linkages in tube tests. We propose a novel function for IDE as a non-canonical ubiquitin activating enzyme.
Collapse
Affiliation(s)
- G Grasso
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, V.le A. Doria 6, 95125 Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
210
|
Neprilysin Confers Genetic Susceptibility to Alzheimer's Disease in Han Chinese. Mol Neurobiol 2015; 53:4883-92. [PMID: 26362309 DOI: 10.1007/s12035-015-9411-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 08/26/2015] [Indexed: 02/05/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, with increasing incidence all over the world. Amyloid-β (Aβ) was considered to be the original cause to AD, and many reported pathogenic or risk genes for AD were located in the Aβ generation and degradation pathways. Neprilysin (NEP), insulin-degrading enzyme (IDE), and matrix metalloprotease-9 (MMP-9) are the most important Aβ-degrading proteases. Accumulating genetic evidence suggested that single nucleotide polymorphisms (SNPs) of these genes confer susceptibility to AD in Caucasian populations. In this study, we screened eight SNPs within these three Aβ-degrading protease genes in 1475 individuals of two independent Han Chinese case-control cohorts. SNP rs1816558 of NEP was found to be significantly associated with AD after adjustment for ε4 allele of the apolipoprotein E gene (APOEε4) and the Bonferroni correction. The remaining variants were not associated with risk of AD in Han Chinese sample set. Further data mining revealed that messenger RNA (mRNA) level of NEP substantially increased during the development of AD and was positively correlated with APP expression. The combined results indicated that NEP confers genetic susceptibility to AD in Han Chinese populations.
Collapse
|
211
|
Tarasoff-Conway JM, Carare RO, Osorio RS, Glodzik L, Butler T, Fieremans E, Axel L, Rusinek H, Nicholson C, Zlokovic BV, Frangione B, Blennow K, Ménard J, Zetterberg H, Wisniewski T, de Leon MJ. Clearance systems in the brain-implications for Alzheimer disease. Nat Rev Neurol 2015; 11:457-70. [PMID: 26195256 PMCID: PMC4694579 DOI: 10.1038/nrneurol.2015.119] [Citation(s) in RCA: 1150] [Impact Index Per Article: 115.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Accumulation of toxic protein aggregates-amyloid-β (Aβ) plaques and hyperphosphorylated tau tangles-is the pathological hallmark of Alzheimer disease (AD). Aβ accumulation has been hypothesized to result from an imbalance between Aβ production and clearance; indeed, Aβ clearance seems to be impaired in both early and late forms of AD. To develop efficient strategies to slow down or halt AD, it is critical to understand how Aβ is cleared from the brain. Extracellular Aβ deposits can be removed from the brain by various clearance systems, most importantly, transport across the blood-brain barrier. Findings from the past few years suggest that astroglial-mediated interstitial fluid (ISF) bulk flow, known as the glymphatic system, might contribute to a larger portion of extracellular Aβ (eAβ) clearance than previously thought. The meningeal lymphatic vessels, discovered in 2015, might provide another clearance route. Because these clearance systems act together to drive eAβ from the brain, any alteration to their function could contribute to AD. An understanding of Aβ clearance might provide strategies to reduce excess Aβ deposits and delay, or even prevent, disease onset. In this Review, we describe the clearance systems of the brain as they relate to proteins implicated in AD pathology, with the main focus on Aβ.
Collapse
Affiliation(s)
| | - Roxana O Carare
- University of Southampton, Faculty of Medicine, Institute for Life Sciences, Southampton General Hospital, Southampton Hampshire, SO16 6YD, UK
| | - Ricardo S Osorio
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Lidia Glodzik
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Tracy Butler
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Els Fieremans
- New York University School of Medicine, 660 First Avenue, New York, NY 10016, USA
| | - Leon Axel
- New York University School of Medicine, 660 First Avenue, New York, NY 10016, USA
| | - Henry Rusinek
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Charles Nicholson
- New York University School of Medicine, 660 First Avenue, New York, NY 10016, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute at Keck School of Medicine of University of Southern California, 1501 San Pablo Street Los Angeles, CA 90089, USA
| | - Blas Frangione
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Kaj Blennow
- The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Joël Ménard
- Université Paris-Descartes, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Henrik Zetterberg
- The Sahlgrenska Academy at the University of Gothenburg, S-431 80 Mölndal, Sweden
| | - Thomas Wisniewski
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| | - Mony J de Leon
- New York University School of Medicine, 145 East 32nd Street, New York, NY 10016, USA
| |
Collapse
|
212
|
Proteasome Activity Is Affected by Fluctuations in Insulin-Degrading Enzyme Distribution. PLoS One 2015; 10:e0132455. [PMID: 26186340 PMCID: PMC4506093 DOI: 10.1371/journal.pone.0132455] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 06/11/2015] [Indexed: 01/13/2023] Open
Abstract
Insulin-Degrading-Enzyme (IDE) is a Zn2+-dependent peptidase highly conserved throughout evolution and ubiquitously distributed in mammalian tissues wherein it displays a prevalent cytosolic localization. We have recently demonstrated a novel Heat Shock Protein-like behaviour of IDE and its association with the 26S proteasome. In the present study, we examine the mechanistic and molecular features of IDE-26S proteasome interaction in a cell experimental model, extending the investigation also to the effect of IDE on the enzymatic activities of the 26S proteasome. Further, kinetic investigations indicate that the 26S proteasome activity undergoes a functional modulation by IDE through an extra-catalytic mechanism. The IDE-26S proteasome interaction was analyzed during the Heat Shock Response and we report novel findings on IDE intracellular distribution that might be of critical relevance for cell metabolism.
Collapse
|
213
|
An Extended Polyanion Activation Surface in Insulin Degrading Enzyme. PLoS One 2015; 10:e0133114. [PMID: 26186535 PMCID: PMC4506039 DOI: 10.1371/journal.pone.0133114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022] Open
Abstract
Insulin degrading enzyme (IDE) is believed to be the major enzyme that metabolizes insulin and has been implicated in the degradation of a number of other bioactive peptides, including amyloid beta peptide (Aβ), glucagon, amylin, and atrial natriuretic peptide. IDE is activated toward some substrates by both peptides and polyanions/anions, possibly representing an important control mechanism and a potential therapeutic target. A binding site for the polyanion ATP has previously been defined crystallographically, but mutagenesis studies suggest that other polyanion binding modes likely exist on the same extended surface that forms one wall of the substrate-binding chamber. Here we use a computational approach to define three potential ATP binding sites and mutagenesis and kinetic studies to confirm the relevance of these sites. Mutations were made at four positively charged residues (Arg 429, Arg 431, Arg 847, Lys 898) within the polyanion-binding region, converting them to polar or hydrophobic residues. We find that mutations in all three ATP binding sites strongly decrease the degree of activation by ATP and can lower basal activity and cooperativity. Computational analysis suggests conformational changes that result from polyanion binding as well as from mutating residues involved in polyanion binding. These findings indicate the presence of multiple polyanion binding modes and suggest the anion-binding surface plays an important conformational role in controlling IDE activity.
Collapse
|
214
|
Luchsinger JA, Cabral R, Eimicke JP, Manly JJ, Teresi J. Glycemia, Diabetes Status, and Cognition in Hispanic Adults Aged 55-64 Years. Psychosom Med 2015; 77:653-63. [PMID: 26163818 PMCID: PMC4503370 DOI: 10.1097/psy.0000000000000208] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To examine the association of glycemia and diabetes status with cognition among 600 Hispanics aged 55 to 64 years from Northern Manhattan. METHODS Diabetes was ascertained by history or hemoglobin A1c. Normal glucose tolerance and prediabetes were ascertained with hemoglobin A1c. Memory was assessed with the Selective Reminding Test. Executive abilities were assessed using the Color Trails 1 and 2 and verbal fluency test. The cross-sectional association of glycemia and diabetes status with cognitive performance was examined using linear regression. RESULTS Participants had a mean age of 59.2 (2.9) years, 76.7% were women, and more than 65% had prediabetes or diabetes. HbA1C (β = -0.97, p < .001) and diabetes (β = -2.06, p = .001) were related with lower Selective Reminding Test total recall after adjustment for demographics, education, and vascular risk factors. Prediabetes was associated with worse performance in Color Trail 2 (β = -6.45 p = .022) after full adjustment. CONCLUSIONS Higher glycemia and diabetes are related to worse memory and executive abilities in late middle age, whereas prediabetes is related only to worse executive abilities. Longitudinal follow-up is needed to understand the order and progression of these deficits.
Collapse
Affiliation(s)
- José A. Luchsinger
- Department of Medicine, Columbia University Medical Center, New York, NY
- Department of Epidemiology, Columbia University Medical Center, New York, NY
| | - Rafi Cabral
- Department of Medicine, Columbia University Medical Center, New York, NY
| | | | - Jennifer J. Manly
- Gertrude H. Sergievsky Center, Columbia University Medical Center, New York, NY
| | - Jeanne Teresi
- Department of Epidemiology, Columbia University Medical Center, New York, NY
| |
Collapse
|
215
|
Bedse G, Di Domenico F, Serviddio G, Cassano T. Aberrant insulin signaling in Alzheimer's disease: current knowledge. Front Neurosci 2015; 9:204. [PMID: 26136647 PMCID: PMC4468388 DOI: 10.3389/fnins.2015.00204] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/22/2015] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting elderly people. AD is a multifaceted pathology characterized by accumulation of extracellular neuritic plaques, intracellular neurofibrillary tangles (NFTs) and neuronal loss mainly in the cortex and hippocampus. AD etiology appears to be linked to a multitude of mechanisms that have not been yet completely elucidated. For long time, it was considered that insulin signaling has only peripheral actions but now it is widely accepted that insulin has neuromodulatory actions in the brain. Insulin signaling is involved in numerous brain functions including cognition and memory that are impaired in AD. Recent studies suggest that AD may be linked to brain insulin resistance and patients with diabetes have an increased risk of developing AD compared to healthy individuals. Indeed insulin resistance, increased inflammation and impaired metabolism are key pathological features of both AD and diabetes. However, the precise mechanisms involved in the development of AD in patients with diabetes are not yet fully understood. In this review we will discuss the role played by aberrant brain insulin signaling in AD. In detail, we will focus on the role of insulin signaling in the deposition of neuritic plaques and intracellular NFTs. Considering that insulin mitigates beta-amyloid deposition and phosphorylation of tau, pharmacological strategies restoring brain insulin signaling, such as intranasal delivery of insulin, could have significant therapeutic potential in AD treatment.
Collapse
Affiliation(s)
- Gaurav Bedse
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome Rome, Italy ; Department of Biochemical Sciences, Sapienza University of Rome Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences, Sapienza University of Rome Rome, Italy
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia Foggia, Italy
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia Foggia, Italy
| |
Collapse
|
216
|
Kim IH, Kim IJ, Wen Y, Park NY, Park J, Lee KW, Koh A, Lee JH, Koo SH, Kim KS. Vibrio vulnificus Secretes an Insulin-degrading Enzyme That Promotes Bacterial Proliferation in Vivo. J Biol Chem 2015; 290:18708-20. [PMID: 26041774 DOI: 10.1074/jbc.m115.656306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Indexed: 12/23/2022] Open
Abstract
We describe a novel insulin-degrading enzyme, SidC, that contributes to the proliferation of the human bacterial pathogen Vibrio vulnificus in a mouse model. SidC is phylogenetically distinct from other known insulin-degrading enzymes and is expressed and secreted specifically during host infection. Purified SidC causes a significant decrease in serum insulin levels and an increase in blood glucose levels in mice. A comparison of mice infected with wild type V. vulnificus or an isogenic sidC-deletion strain showed that wild type bacteria proliferated to higher levels. Additionally, hyperglycemia leads to increased proliferation of V. vulnificus in diabetic mice. Consistent with these observations, the sid operon was up-regulated in response to low glucose levels through binding of the cAMP-receptor protein (CRP) complex to a region upstream of the operon. We conclude that glucose levels are important for the survival of V. vulnificus in the host, and that this pathogen uses SidC to actively manipulate host endocrine signals, making the host environment more favorable for bacterial survival and growth.
Collapse
Affiliation(s)
- In Hwang Kim
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Ik-Jung Kim
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Yancheng Wen
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Na-Young Park
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Jinyoung Park
- the Division of Life Science, Korea University, Seoul 136-701, Korea
| | - Keun-Woo Lee
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Ara Koh
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea
| | - Ji-Hyun Lee
- the Division of Life Science, Korea University, Seoul 136-701, Korea
| | - Seung-Hoi Koo
- the Division of Life Science, Korea University, Seoul 136-701, Korea
| | - Kun-Soo Kim
- From the Department of Life Science, Sogang University, Seoul 121-742, Korea,
| |
Collapse
|
217
|
Abstract
Several studies have indicated that Diabetes Mellitus (DM) can increase the risk
of developing Alzheimer's disease (AD). This review briefly describes current
concepts in mechanisms linking DM and insulin resistance/deficiency to AD.
Insulin/insulin-like growth factor (IGF) resistance can contribute to
neurodegeneration by several mechanisms which involve: energy and metabolism
deficits, impairment of Glucose transporter-4 function, oxidative and
endoplasmic reticulum stress, mitochondrial dysfunction, accumulation of AGEs,
ROS and RNS with increased production of neuro-inflammation and activation of
pro-apoptosis cascade. Impairment in insulin receptor function and increased
expression and activation of insulin-degrading enzyme (IDE) have also been
described. These processes compromise neuronal and glial function, with a
reduction in neurotransmitter homeostasis. Insulin/IGF resistance causes the
accumulation of AβPP-Aβ oligomeric fibrils or insoluble larger
aggregated fibrils in the form of plaques that are neurotoxic. Additionally,
there is production and accumulation of hyper-phosphorylated insoluble fibrillar
tau which can exacerbate cytoskeletal collapse and synaptic disconnection.
Collapse
Affiliation(s)
- Maria Niures P S Matioli
- Pós-graduanda, nível de Doutorado, Departamento de Neurologia da Faculdade de Medicina da Universidade de São Paulo
| | - Ricardo Nitrini
- Professor Titular da Disciplina de Neurologia da Faculdade de Medicina da Universidade de São Paulo. Orientador e Professor Responsável pela Pós-graduação do Departamento de Neurologia da Faculdade de Medicina da Universidade de São Paulo
| |
Collapse
|
218
|
Grimm MOW, Stahlmann CP, Mett J, Haupenthal VJ, Zimmer VC, Lehmann J, Hundsdörfer B, Endres K, Grimm HS, Hartmann T. Vitamin E: Curse or Benefit in Alzheimer's Disease? A Systematic Investigation of the Impact of α-, γ- and δ-Tocopherol on Aß Generation and Degradation in Neuroblastoma Cells. J Nutr Health Aging 2015; 19:646-56. [PMID: 26054501 DOI: 10.1007/s12603-015-0506-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES The E vitamins are a class of lipophilic compounds including tocopherols, which have high antioxidative properties. Because of the elevated lipid peroxidation and increased reactive oxidative species in Alzheimer's disease (AD) many attempts have been made to slow down the progression of AD by utilizing the antioxidative action of vitamin E. Beside the mixed results of these studies nothing is known about the impact of vitamin E on the mechanisms leading to amyloid-β production and degradation being responsible for the plaque formation, one of the characteristic pathological hallmarks in AD. Here we systematically investigate the influence of different tocopherols on Aβ production and degradation in neuronal cell lines. MEASUREMENTS Beside amyloid-β level the mechanisms leading to Aβ production and degradation are examined. RESULTS Surprisingly, all tocopherols have shown to increase Aβ level by enhancing the Aβ production and decreasing the Aβ degradation. Aβ production is enhanced by an elevated activity of the involved enzymes, the β- and γ-secretase. These secretases are not directly affected, but tocopherols increase their protein level and expression. We could identify significant differences between the single tocopherols; whereas α-tocopherol had only minor effects on Aβ production, δ-tocopherol showed the highest potency to increase Aβ generation. Beside Aβ production, Aβ clearance was decreased by affecting IDE, one of the major Aβ degrading enzymes. CONCLUSIONS Our results suggest that beside the beneficial antioxidative effects of vitamin E, tocopherol has in respect to AD also a potency to increase the amyloid-β level, which differ for the analysed tocopherols. We therefore recommend that further studies are needed to clarify the potential role of these various vitamin E species in respect to AD and to identify the form which comprises an antioxidative property without having an amyloidogenic potential.
Collapse
Affiliation(s)
- M O W Grimm
- Marcus Grimm, Kirrberger Str.1, Building 90.1, 66421 Homburg/Saar, Germany; Tel: +49-6841-1647919; Fax: +49-6841-1624137; E-mail:
| | | | | | | | | | | | | | | | | | | |
Collapse
|
219
|
The role of type 2 diabetes in neurodegeneration. Neurobiol Dis 2015; 84:22-38. [PMID: 25926349 DOI: 10.1016/j.nbd.2015.04.008] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/18/2015] [Accepted: 04/21/2015] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence links type-2 diabetes (T2D) with dementia and neurodegenerative diseases such as Alzheimer's disease (AD). AD is the most common form of dementia and is characterised neuropathologically by the accumulation of extracellular beta amyloid (Aβ) peptide aggregates and intracellular hyper-phosphorylated tau protein, which are thought to drive and/or accelerate inflammatory and oxidative stress processes leading to neurodegeneration. Although the precise mechanism remains unclear, T2D can exacerbate these neurodegenerative processes. Brain atrophy, reduced cerebral glucose metabolism and CNS insulin resistance are features of both AD and T2D. Cell culture and animal studies have indicated that the early accumulation of Aβ may play a role in CNS insulin resistance and impaired insulin signalling. From the viewpoint of insulin resistance and impaired insulin signalling in the brain, these are also believed to initiate other aspects of brain injury, including inflammatory and oxidative stress processes. Here we review the clinical and experimental pieces of evidence that link these two chronic diseases of ageing, and discuss underlying mechanisms. The evaluation of treatments for the management of diabetes in preclinical, and clinical studies and trials for AD will also be discussed.
Collapse
|
220
|
Murakami K. Conformation-specific antibodies to target amyloid β oligomers and their application to immunotherapy for Alzheimer's disease. Biosci Biotechnol Biochem 2015; 78:1293-305. [PMID: 25130729 DOI: 10.1080/09168451.2014.940275] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Amyloid β-protein (Aβ) oligomers, intermediates of Aβ aggregation, cause cognitive impairment and synaptotoxicity in the pathogenesis of Alzheimer's disease (AD). Immunotherapy using anti-Aβ antibody is one of the most promising approaches for AD treatment. However, most clinical trials using conventional sequence-specific antibodies have proceeded with difficulty. This is probably due to the unintended removal of the non-pathological monomer and fibrils of Aβ as well as the pathological oligomers by these antibodies that recognize Aβ sequence, which is not involved in synaptotoxicity. Several efforts have been made recently to develop conformation-specific antibodies that target the tertiary structure of Aβ oligomers. Here, we review the recent findings of Aβ oligomers and anti-Aβ antibodies including our own, and discuss their potential as therapeutic and diagnostic tools.
Collapse
Affiliation(s)
- Kazuma Murakami
- a Division of Food Science and Biotechnology , Graduate School of Agriculture, Kyoto University , Kyoto , Japan
| |
Collapse
|
221
|
Humpel C. Organotypic vibrosections from whole brain adult Alzheimer mice (overexpressing amyloid-precursor-protein with the Swedish-Dutch-Iowa mutations) as a model to study clearance of beta-amyloid plaques. Front Aging Neurosci 2015; 7:47. [PMID: 25914642 PMCID: PMC4391240 DOI: 10.3389/fnagi.2015.00047] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/24/2015] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a severe neurodegenerative disorder of the brain, pathologically characterized by extracellular beta-amyloid plaques, intraneuronal Tau inclusions, inflammation, reactive glial cells, vascular pathology and neuronal cell death. The degradation and clearance of beta-amyloid plaques is an interesting therapeutic approach, and the proteases neprilysin (NEP), insulysin and matrix metalloproteinases (MMP) are of particular interest. The aim of this project was to establish and characterize a simple in vitro model to study the degrading effects of these proteases. Organoytpic brain vibrosections (120 μm thick) were sectioned from adult (9 month old) wildtype and transgenic mice (expressing amyloid precursor protein (APP) harboring the Swedish K670N/M671L, Dutch E693Q, and Iowa D694N mutations; APP_SDI) and cultured for 2 weeks. Plaques were stained by immunohistochemistry for beta-amyloid and Thioflavin S. Our data show that plaques were evident in 2 week old cultures from 9 month old transgenic mice. These plaques were surrounded by reactive GFAP+ astroglia and Iba1+ microglia. Incubation of fresh slices for 2 weeks with 1-0.1-0.01 μg/ml of NEP, insulysin, MMP-2, or MMP-9 showed that NEP, insulysin, and MMP-9 markedly degraded beta-amyloid plaques but only at the highest concentration. Our data provide for the first time a potent and powerful living brain vibrosection model containing a high number of plaques, which allows to rapidly and simply study the degradation and clearance of beta-amyloid plaques in vitro.
Collapse
Affiliation(s)
- Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Department of Psychiatry and Psychotherapy, Medical University of Innsbruck Innsbruck, Austria
| |
Collapse
|
222
|
Donepezil improves learning and memory deficits in APP/PS1 mice by inhibition of microglial activation. Neuroscience 2015; 290:530-42. [DOI: 10.1016/j.neuroscience.2015.01.058] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 12/26/2014] [Accepted: 01/25/2015] [Indexed: 12/28/2022]
|
223
|
Thal DR. Clearance of amyloid β-protein and its role in the spreading of Alzheimer's disease pathology. Front Aging Neurosci 2015; 7:25. [PMID: 25805990 PMCID: PMC4353250 DOI: 10.3389/fnagi.2015.00025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 02/22/2015] [Indexed: 11/13/2022] Open
Affiliation(s)
- Dietmar R Thal
- Laboratory of Neuropathology - Institute of Pathology, Center for Biomedical Research, University of Ulm Ulm, Germany
| |
Collapse
|
224
|
Bohm C, Chen F, Sevalle J, Qamar S, Dodd R, Li Y, Schmitt-Ulms G, Fraser PE, St George-Hyslop PH. Current and future implications of basic and translational research on amyloid-β peptide production and removal pathways. Mol Cell Neurosci 2015; 66:3-11. [PMID: 25748120 PMCID: PMC4503820 DOI: 10.1016/j.mcn.2015.02.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/12/2023] Open
Abstract
Inherited variants in multiple different genes are associated with increased risk for Alzheimer's disease (AD). In many of these genes, the inherited variants alter some aspect of the production or clearance of the neurotoxic amyloid β-peptide (Aβ). Thus missense, splice site or duplication mutants in the presenilin 1 (PS1), presenilin 2 (PS2) or the amyloid precursor protein (APP) genes, which alter the levels or shift the balance of Aβ produced, are associated with rare, highly penetrant autosomal dominant forms of Familial Alzheimer's Disease (FAD). Similarly, the more prevalent late-onset forms of AD are associated with both coding and non-coding variants in genes such as SORL1, PICALM and ABCA7 that affect the production and clearance of Aβ. This review summarises some of the recent molecular and structural work on the role of these genes and the proteins coded by them in the biology of Aβ. We also briefly outline how the emerging knowledge about the pathways involved in Aβ generation and clearance can be potentially targeted therapeutically. This article is part of Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- C Bohm
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - F Chen
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - J Sevalle
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - S Qamar
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - R Dodd
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Y Li
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - G Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P H St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada; Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
225
|
Zhu H, Wang X, Wallack M, Li H, Carreras I, Dedeoglu A, Hur JY, Zheng H, Li H, Fine R, Mwamburi M, Sun X, Kowall N, Stern RA, Qiu WQ. Intraperitoneal injection of the pancreatic peptide amylin potently reduces behavioral impairment and brain amyloid pathology in murine models of Alzheimer's disease. Mol Psychiatry 2015; 20:252-62. [PMID: 24614496 PMCID: PMC4161670 DOI: 10.1038/mp.2014.17] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/06/2014] [Accepted: 01/31/2014] [Indexed: 12/13/2022]
Abstract
Amylin, a pancreatic peptide, and amyloid-beta peptides (Aβ), a major component of Alzheimer's disease (AD) brain, share similar β-sheet secondary structures, but it is not known whether pancreatic amylin affects amyloid pathogenesis in the AD brain. Using AD mouse models, we investigated the effects of amylin and its clinical analog, pramlintide, on AD pathogenesis. Surprisingly, chronic intraperitoneal (i.p.) injection of AD animals with either amylin or pramlintide reduces the amyloid burden as well as lowers the concentrations of Aβ in the brain. These treatments significantly improve their learning and memory assessed by two behavioral tests, Y maze and Morris water maze. Both amylin and pramlintide treatments increase the concentrations of Aβ1-42 in cerebral spinal fluid (CSF). A single i.p. injection of either peptide also induces a surge of Aβ in the serum, the magnitude of which is proportionate to the amount of Aβ in brain tissue. One intracerebroventricular injection of amylin induces a more significant surge in serum Aβ than one i.p. injection of the peptide. In 330 human plasma samples, a positive association between amylin and Aβ1-42 as well as Aβ1-40 is found only in patients with AD or amnestic mild cognitive impairment. As amylin readily crosses the blood-brain barrier, our study demonstrates that peripheral amylin's action on the central nervous system results in translocation of Aβ from the brain into the CSF and blood that could be an explanation for a positive relationship between amylin and Aβ in blood. As naturally occurring amylin may play a role in regulating Aβ in brain, amylin class peptides may provide a new avenue for both treatment and diagnosis of AD.
Collapse
Affiliation(s)
- H Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University Medical Campus, Boston, MA, USA
| | - X Wang
- Department of Pharmacology and Experimental Therapeutics, Boston University Medical Campus, Boston, MA, USA
| | - M Wallack
- Department of Pharmacology and Experimental Therapeutics, Boston University Medical Campus, Boston, MA, USA
| | - H Li
- Department of Pharmacology and Experimental Therapeutics, Boston University Medical Campus, Boston, MA, USA
| | - I Carreras
- Department of Neurology, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
| | - A Dedeoglu
- Department of Neurology, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
| | - J-Y Hur
- Memorial Sloan-Kettering Institute, New York, NY, USA
| | - H Zheng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - H Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - R Fine
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
- ENRM VA Medical Center, Bedford, MA, USA
| | - M Mwamburi
- Department of Public Health and Family Medicine, Tufts University, Boston, MA, USA
| | - X Sun
- Department of Neurology, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
| | - N Kowall
- Department of Neurology, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
| | - R A Stern
- Department of Neurology, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
- Department of Neural Surgery, Boston University Medical Campus, Boston, MA, USA
| | - W Q Qiu
- Department of Pharmacology and Experimental Therapeutics, Boston University Medical Campus, Boston, MA, USA
- Alzheimer's Disease Center, Boston University Medical Campus, Boston, MA, USA
- Department of Psychiatry, Boston University Medical Campus, Boston, MA, USA
| |
Collapse
|
226
|
Silverberg GD, Miller MC, Pascale CL, Caralopoulos IN, Agca Y, Agca C, Stopa EG. Kaolin-induced chronic hydrocephalus accelerates amyloid deposition and vascular disease in transgenic rats expressing high levels of human APP. Fluids Barriers CNS 2015; 12:2. [PMID: 25685319 PMCID: PMC4328504 DOI: 10.1186/2045-8118-12-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/14/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Normal pressure hydrocephalus (NPH) is most common in the elderly and has a high co-morbidity with Alzheimer's disease (AD) and cerebrovascular disease (CVD). To understand the relationship between NPH, AD and CVD, we investigated how chronic hydrocephalus impacts brain amyloid-beta peptide (Aβ) accumulation and vascular pathology in an AD transgenic rodent model. Previously we showed that the altered CSF physiology produced by kaolin-hydrocephalus in older wild-type Sprague-Dawley rats increased Aβ and hyperphosphorylated Tau (Silverberg et. al. Brain Res. 2010, 1317:286-296). We postulated that hydrocephalus would similarly affect an AD rat model. METHODS Thirty-five transgenic rats (tgAPP21) that express high levels of human APP and naturally overproduce Aβ40 were used. Six- (n = 7) and twelve-month-old (n = 9) rats had hydrocephalus induced by cisternal kaolin injection. We analyzed Aβ burden (Aβ40, Aβ42 and oligomeric Aβ) and vascular integrity (Masson trichrome and Verhoeff-Van Gieson) by immunohistochemistry and chemical staining at 10 weeks (n = 8) and 6 months (n = 5) post hydrocephalus induction. We also analyzed whether the vascular pathology seen in tgAPP21 rats, which develop amyloid angiopathy, was accelerated by hydrocephalus. Age-matched naïve and sham-operated tgAPP21 rats served as controls (n = 19). RESULTS In hydrocephalic tgAPP21 rats, compared to naïve and sham-operated controls, there was increased Aβ 40 and oligomeric Aβ in hippocampal and cortical neurons at 10 weeks and 6 months post-hydrocephalus induction. No dense-core amyloid plaques were seen, but diffuse Aβ immunoreactivity was evident in neurons. Vascular pathology was accelerated by the induction of hydrocephalus compared to controls. In the six-month-old rats, subtle degenerative changes were noted in vessel walls at 10 weeks post-kaolin, whereas at six months post-kaolin and in the 12-month-old hydrocephalic rats more pronounced amyloid angiopathic changes were seen, with frequent large areas of infarction noted. CONCLUSIONS Kaolin-hydrocephalus can accelerate intraneuronal Aβ40 accumulation and vascular pathology in tgAPP21 rats. In addition, disrupted CSF production and reduced CSF turnover results in impaired Aβ clearance and accelerated vascular pathology in chronic hydrocephalus. The high co-morbidity seen in NPH, AD and CVD is likely not to be an age-related coincidence, but rather a convergence of pathologies related to diminished CSF clearance.
Collapse
Affiliation(s)
- Gerald D Silverberg
- />Department of Neurosurgery, The Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
- />Stanford University, 710 Frenchmans Rd, Stanford, CA 94305 USA
| | - Miles C Miller
- />Department of Neurosurgery, The Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
| | - Crissey L Pascale
- />Department of Neurosurgery, The Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
| | - Ilias N Caralopoulos
- />Department of Neurosurgery, The Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
| | - Yuksel Agca
- />Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO 65211 USA
| | - Cansu Agca
- />Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO 65211 USA
| | - Edward G Stopa
- />Department of Neurosurgery, The Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
- />Department of Pathology (Neuropathology), Warren Alpert Medical School of Brown University and the Aldrich Laboratories at Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903 USA
| |
Collapse
|
227
|
Calvo-Ochoa E, Arias C. Cellular and metabolic alterations in the hippocampus caused by insulin signalling dysfunction and its association with cognitive impairment during aging and Alzheimer's disease: studies in animal models. Diabetes Metab Res Rev 2015; 31:1-13. [PMID: 24464982 DOI: 10.1002/dmrr.2531] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 12/03/2013] [Accepted: 01/15/2014] [Indexed: 01/09/2023]
Abstract
A growing body of animal and epidemiological studies suggest that metabolic diseases such as obesity, insulin resistance, metabolic syndrome and type 2 diabetes mellitus are associated with the development of cognitive impairment, dementia and Alzheimer's disease, particularly in aging. Several lines of evidence suggest that insulin signalling dysfunction produces these metabolic alterations and underlie the development of these neurodegenerative diseases. In this article, we address normal insulin function in the synapse; we review and discuss the physiopathological hallmarks of synaptic insulin signalling dysfunction associated with metabolic alterations. Additionally, we describe and review the major animal models of obesity, insulin resistance, metabolic syndrome and type 2 diabetes mellitus. The comprehensive knowledge of the molecular mechanisms behind the association of metabolic alterations and cognitive impairment could facilitate the early detection of neurodegenerative diseases in patients with metabolic alterations, with treatment that focus on neuroprotection. It could also help in the development of metabolic-based therapies and drugs for using in dementia and Alzheimer's disease patients to alleviate their symptoms in a more efficient and comprehensive way.
Collapse
Affiliation(s)
- Erika Calvo-Ochoa
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF, Mexico
| | | |
Collapse
|
228
|
Glycemia and cognitive function in metabolic syndrome and coronary heart disease. Am J Med 2015; 128:46-55. [PMID: 25220612 PMCID: PMC4306431 DOI: 10.1016/j.amjmed.2014.08.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/19/2014] [Accepted: 08/19/2014] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Higher hemoglobin A1c (HbA1c) is associated with lower cognitive function in type 2 diabetes. To determine whether associations persist at lower levels of dysglycemia in patients who have established cardiovascular disease, cognitive performance was assessed in the Targeting INflammation Using SALsalate in CardioVascular Disease (TINSAL-CVD) trial. METHODS The age-adjusted relationships between HbA1c and cognitive performance measured by the Mini-Mental State Examination, Digit Symbol Substitution Test, Rey Auditory Verbal Learning Test, Trail Making Test, and Categorical Verbal Fluency were assessed in 226 men with metabolic syndrome and established stable coronary artery disease. RESULTS Of the participants, 61.5% had normoglycemia, 20.8% had impaired fasting glucose, and 17.7% had type 2 diabetes. HbA1c was associated with cognitive function tests of Digit Symbol Substitution Test, Rey Auditory Verbal Learning Test, Trail Making Test, and Categorical Verbal Fluency (all P < .02), but not the Mini-Mental State Examination. In an age-adjusted model, a 1% (11 mmol/mol) higher HbA1c value was associated with a 5.9 lower Digit Symbol Substitution Test score (95% confidence interval [CI], -9.58 to -2.21; P < .0001); a 2.44 lower Rey Auditory Verbal Learning Test score (95% CI, -4.00 to -0.87; P < .0001); a 15.6 higher Trail Making Test score (95% CI, 5.73 to 25.6; P < .0001); and a 3.71 lower Categorical Verbal Fluency score (95% CI, -6.41 to -1.01; P < .02). In a multivariate model adjusting for age, education, and cardiovascular covariates, HbA1c remained associated with cognitive function tests of Rey Auditory Verbal Learning Test (R(2) = 0.27, P < .0001), Trail Making Test (R(2) = 0.18, P < .0001), and Categorical Verbal Fluency (R(2) = 0.20, P < .0001), although association with the Digit Symbol Substitution Test was reduced. CONCLUSIONS Higher HbA1c is associated with lower cognitive function performance scores across multiple domain tests in men with metabolic syndrome and coronary artery disease. Future studies may demonstrate whether glucose lowering within the normative range improves cognitive health.
Collapse
|
229
|
Oberstein TJ, Spitzer P, Klafki HW, Linning P, Neff F, Knölker HJ, Lewczuk P, Wiltfang J, Kornhuber J, Maler JM. Astrocytes and microglia but not neurons preferentially generate N-terminally truncated Aβ peptides. Neurobiol Dis 2015; 73:24-35. [DOI: 10.1016/j.nbd.2014.08.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 08/05/2014] [Accepted: 08/31/2014] [Indexed: 12/30/2022] Open
|
230
|
Baranello RJ, Bharani KL, Padmaraju V, Chopra N, Lahiri DK, Greig NH, Pappolla MA, Sambamurti K. Amyloid-beta protein clearance and degradation (ABCD) pathways and their role in Alzheimer's disease. Curr Alzheimer Res 2015; 12:32-46. [PMID: 25523424 PMCID: PMC4820400 DOI: 10.2174/1567205012666141218140953] [Citation(s) in RCA: 233] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 11/16/2014] [Accepted: 12/05/2014] [Indexed: 11/22/2022]
Abstract
Amyloid-β proteins (Aβ) of 42 (Aβ42) and 40 aa (Aβ40) accumulate as senile plaques (SP) and cerebrovascular amyloid protein deposits that are defining diagnostic features of Alzheimer's disease (AD). A number of rare mutations linked to familial AD (FAD) on the Aβ precursor protein (APP), Presenilin-1 (PS1), Presenilin- 2 (PS2), Adamalysin10, and other genetic risk factors for sporadic AD such as the ε4 allele of Apolipoprotein E (ApoE-ε4) foster the accumulation of Aβ and also induce the entire spectrum of pathology associated with the disease. Aβ accumulation is therefore a key pathological event and a prime target for the prevention and treatment of AD. APP is sequentially processed by β-site APP cleaving enzyme (BACE1) and γ-secretase, a multisubunit PS1/PS2-containing integral membrane protease, to generate Aβ. Although Aβ accumulates in all forms of AD, the only pathways known to be affected in FAD increase Aβ production by APP gene duplication or via base substitutions on APP and γ-secretase subunits PS1 and PS2 that either specifically increase the yield of the longer Aβ42 or both Aβ40 and Aβ42. However, the vast majority of AD patients accumulate Aβ without these known mutations. This led to proposals that impairment of Aβ degradation or clearance may play a key role in AD pathogenesis. Several candidate enzymes, including Insulin-degrading enzyme (IDE), Neprilysin (NEP), Endothelin-converting enzyme (ECE), Angiotensin converting enzyme (ACE), Plasmin, and Matrix metalloproteinases (MMPs) have been identified and some have even been successfully evaluated in animal models. Several studies also have demonstrated the capacity of γ-secretase inhibitors to paradoxically increase the yield of Aβ and we have recently established that the mechanism is by skirting Aβ degradation. This review outlines major cellular pathways of Aβ degradation to provide a basis for future efforts to fully characterize the panel of pathways responsible for Aβ turnover.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kumar Sambamurti
- Department of Neurosciences, Medical University of South Carolina, 173 Ashley Avenue, BSB 403, Charleston, SC 29425, USA.
| |
Collapse
|
231
|
Seto SW, Yang GY, Kiat H, Bensoussan A, Kwan YW, Chang D. Diabetes Mellitus, Cognitive Impairment, and Traditional Chinese Medicine. Int J Endocrinol 2015; 2015:810439. [PMID: 26060494 PMCID: PMC4427766 DOI: 10.1155/2015/810439] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/15/2015] [Indexed: 12/30/2022] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder affecting a large number of people worldwide. Numerous studies have demonstrated that DM can cause damage to multiple systems, leading to complications such as heart disease, cancer, and cerebrovascular disorders. Numerous epidemiological studies have shown that DM is closely associated with dementia and cognition dysfunction, with recent research focusing on the role of DM-mediated cerebrovascular damage in dementia. Despite the therapeutic benefits of antidiabetic agents for the treatment of DM-mediated cognitive dysfunction, most of these pharmaceutical agents are associated with various undesirable side-effects and their long-term benefits are therefore in doubt. Early evidence exists to support the use of traditional Chinese medicine (TCM) interventions, which tend to have minimal toxicity and side-effects. More importantly, these TCM interventions appear to offer significant effects in reducing DM-related complications beyond blood glucose control. However, more research is needed to further validate these claims and to explore their relevant mechanisms of action. The aims of this paper are (1) to provide an updated overview on the association between DM and cognitive dysfunction and (2) to review the scientific evidence underpinning the use of TCM interventions for the treatment and prevention of DM-induced cognitive dysfunction and dementia.
Collapse
Affiliation(s)
- S. W. Seto
- National Institute of Complementary Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia
| | - G. Y. Yang
- National Institute of Complementary Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia
| | - H. Kiat
- Faculty of Medicine, University of New South Wales, Kensington, NSW 2052, Australia
- School of Medicine, University of Western Sydney, Locked Bag 1797, Penrith, NSW 2751, Australia
- Faculty of Medicine and Health Sciences, Macquarie University, NSW 2109, Australia
| | - A. Bensoussan
- National Institute of Complementary Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia
| | - Y. W. Kwan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - D. Chang
- National Institute of Complementary Medicine, University of Western Sydney, Campbelltown, NSW 2560, Australia
- *D. Chang:
| |
Collapse
|
232
|
Sharma S, Taliyan R. Synergistic effects of GSK-3β and HDAC inhibitors in intracerebroventricular streptozotocin-induced cognitive deficits in rats. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:337-49. [PMID: 25547373 DOI: 10.1007/s00210-014-1081-2] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/12/2014] [Indexed: 11/24/2022]
Abstract
Recent studies suggest the importance of combined treatment of glycogen synthase kinase-3β (GSK-3β) and histone deacetylase (HDAC) inhibition in various in vitro and in vivo models of neurological diseases. Lithium chloride (LiCl) and valproate (VPA), two well-known mood stabilizers, have been reported to act through GSK-3β and HDAC inhibition, respectively. The present study was designed to investigate the potential of low-dose combination of LiCl and VPA in intracerebroventricular streptozotocin (ICV-STZ)-induced cognitive deficits in rats. STZ was injected twice (3 mg/kg ICV) on alternate days (day 1 and day 3) in rats. The ICV-STZ-treated rats received LiCl (60 mg/kg, i.p.), VPA (200 mg/kg, i.p.), and combination of both LiCl (60 mg/kg, i.p.) and VPA (200 mg/kg, i.p.) drugs for a period of 3 weeks. The ICV-STZ administration results in significant memory impairment, elevated oxidative-nitrosative stress, and reduced brain-derived neurotrophic factor (BDNF) levels. Using a battery of behavioral and biochemical tests, we observed that co-treatment of both drugs showed synergistic effect in improving the spatial learning and memory impairment as well as significantly attenuated the oxidative stress markers in STZ-treated rats as compared to either drug alone. Moreover, the combination of both drugs reversed the hyperinsulinemic brain condition and improved the BDNF levels in STZ-treated rats. Based upon these results, it could be suggested that a low-dose combination of LiCl and VPA produces synergistic and more consistent neuroprotective effects in ICV-STZ-induced cognitive deficits in rats.
Collapse
Affiliation(s)
- Sorabh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, 333031, Rajasthan, India
| | | |
Collapse
|
233
|
Abstract
Members of the low-density lipoprotein (LDL) receptor gene family have a diverse set of biological functions that transcend lipid metabolism. Lipoprotein receptors have broad effects in both the developing and adult brain and participate in synapse development, cargo trafficking, and signal transduction. In addition, several family members play key roles in Alzheimer's disease (AD) pathogenesis and neurodegeneration. This Review summarizes our current understanding of the role lipoprotein receptors play in CNS function and AD pathology, with a special emphasis on amyloid-independent roles in endocytosis and synaptic dysfunction.
Collapse
|
234
|
Saitoh Y, Fujikake N, Okamoto Y, Popiel HA, Hatanaka Y, Ueyama M, Suzuki M, Gaumer S, Murata M, Wada K, Nagai Y. p62 plays a protective role in the autophagic degradation of polyglutamine protein oligomers in polyglutamine disease model flies. J Biol Chem 2014; 290:1442-53. [PMID: 25480790 DOI: 10.1074/jbc.m114.590281] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oligomer formation and accumulation of pathogenic proteins are key events in the pathomechanisms of many neurodegenerative diseases, such as Alzheimer disease, ALS, and the polyglutamine (polyQ) diseases. The autophagy-lysosome degradation system may have therapeutic potential against these diseases because it can degrade even large oligomers. Although p62/sequestosome 1 plays a physiological role in selective autophagy of ubiquitinated proteins, whether p62 recognizes and degrades pathogenic proteins in neurodegenerative diseases has remained unclear. In this study, to elucidate the role of p62 in such pathogenic conditions in vivo, we used Drosophila models of neurodegenerative diseases. We found that p62 predominantly co-localizes with cytoplasmic polyQ protein aggregates in the MJDtr-Q78 polyQ disease model flies. Loss of p62 function resulted in significant exacerbation of eye degeneration in these flies. Immunohistochemical analyses revealed enhanced accumulation of cytoplasmic aggregates by p62 knockdown in the MJDtr-Q78 flies, similarly to knockdown of autophagy-related genes (Atgs). Knockdown of both p62 and Atgs did not show any additive effects in the MJDtr-Q78 flies, implying that p62 function is mediated by autophagy. Biochemical analyses showed that loss of p62 function delays the degradation of the MJDtr-Q78 protein, especially its oligomeric species. We also found that loss of p62 function exacerbates eye degeneration in another polyQ disease fly model as well as in ALS model flies. We therefore conclude that p62 plays a protective role against polyQ-induced neurodegeneration, by the autophagic degradation of polyQ protein oligomers in vivo, indicating its therapeutic potential for the polyQ diseases and possibly for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuji Saitoh
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan, the Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan, the Graduate School of Medical and Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan
| | - Nobuhiro Fujikake
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yuma Okamoto
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - H Akiko Popiel
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yusuke Hatanaka
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Morio Ueyama
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Mari Suzuki
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Sébastien Gaumer
- the Laboratoire de Génétique et Biologie Cellulaire, EA4589, Université de Versailles-Saint-Quentin-en-Yvelines, École Pratique des Hautes Etudes, 45 Avenue des Etats-Unis, 78035 Versailles Cedex, France, and
| | - Miho Murata
- the Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo 187-8551, Japan, the Graduate School of Medical and Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, Chiba 260-8670, Japan
| | - Keiji Wada
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan
| | - Yoshitaka Nagai
- From the Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo 187-8502, Japan, Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
235
|
Proitsi P, Lupton MK, Velayudhan L, Hunter G, Newhouse S, Lin K, Fogh I, Tsolaki M, Daniilidou M, Pritchard M, Craig D, Todd S, Johnston JA, McGuinness B, Kloszewska I, Soininen H, Mecocci P, Vellas B, Passmore PA, Sims R, Williams J, Brayne C, Stewart R, Sham P, Lovestone S, Powell JF. Alleles that increase risk for type 2 diabetes mellitus are not associated with increased risk for Alzheimer's disease. Neurobiol Aging 2014; 35:2883.e3-2883.e10. [PMID: 25150574 DOI: 10.1016/j.neurobiolaging.2014.07.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 07/20/2014] [Indexed: 11/26/2022]
Abstract
Although epidemiological studies suggest that type 2 diabetes mellitus (T2DM) increases the risk of late-onset Alzheimer's disease (LOAD), the biological basis of this relationship is not well understood. The aim of this study was to examine the genetic comorbidity between the 2 disorders and to investigate whether genetic liability to T2DM, estimated by a genotype risk scores based on T2DM associated loci, is associated with increased risk of LOAD. This study was performed in 2 stages. In stage 1, we combined genotypes for the top 15 T2DM-associated polymorphisms drawn from approximately 3000 individuals (1349 cases and 1351 control subjects) with extracted and/or imputed data from 6 genome-wide studies (>10,000 individuals; 4507 cases, 2183 controls, 4989 population controls) to form a genotype risk score and examined if this was associated with increased LOAD risk in a combined meta-analysis. In stage 2, we investigated the association of LOAD with an expanded T2DM score made of 45 well-established variants drawn from the 6 genome-wide studies. Results were combined in a meta-analysis. Both stage 1 and stage 2 T2DM risk scores were not associated with LOAD risk (odds ratio = 0.988; 95% confidence interval, 0.972-1.004; p = 0.144 and odds ratio = 0.993; 95% confidence interval, 0.983-1.003; p = 0.149 per allele, respectively). Contrary to expectation, genotype risk scores based on established T2DM candidates were not associated with increased risk of LOAD. The observed epidemiological associations between T2DM and LOAD could therefore be a consequence of secondary disease processes, pleiotropic mechanisms, and/or common environmental risk factors. Future work should focus on well-characterized longitudinal cohorts with extensive phenotypic and genetic data relevant to both LOAD and T2DM.
Collapse
Affiliation(s)
- Petroula Proitsi
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK; Department of Psychiatry, State Key Laboratory of Brain and Cognitive Sciences, and Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong.
| | - Michelle K Lupton
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Latha Velayudhan
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Gillian Hunter
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Stephen Newhouse
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Kuang Lin
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Isabella Fogh
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Magda Tsolaki
- Memory and Dementia Centre, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Makrina Daniilidou
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Megan Pritchard
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - David Craig
- Ageing group, Centre for Public Health, School of Medicine and Dentistry, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Stephen Todd
- Ageing group, Centre for Public Health, School of Medicine and Dentistry, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Janet A Johnston
- Ageing group, Centre for Public Health, School of Medicine and Dentistry, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Bernadette McGuinness
- Ageing group, Centre for Public Health, School of Medicine and Dentistry, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Iwona Kloszewska
- Department of Old Age Psychiatry & Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Hilkka Soininen
- Department of Neurology, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| | - Patrizia Mecocci
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, Perugia, Italy
| | - Bruno Vellas
- Department of Internal and Geriatrics Medicine, INSERM U 1027, Gerontopole, Hôpitaux de Toulouse, Toulouse, France
| | - Peter A Passmore
- Ageing group, Centre for Public Health, School of Medicine and Dentistry, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Rebecca Sims
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Julie Williams
- MRC Centre for Neuropsychiatric Genetics and Genomics, Department of Psychological Medicine and Neurology, School of Medicine, Cardiff University, Cardiff, UK
| | - Carol Brayne
- Department of Public Health and Primary Care, Cambridge Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Robert Stewart
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Pak Sham
- Department of Psychiatry, State Key Laboratory of Brain and Cognitive Sciences, and Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Simon Lovestone
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK; Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford OX3 7JX, UK
| | - John F Powell
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| |
Collapse
|
236
|
Brundel M, Kappelle LJ, Biessels GJ. Brain imaging in type 2 diabetes. Eur Neuropsychopharmacol 2014; 24:1967-81. [PMID: 24726582 DOI: 10.1016/j.euroneuro.2014.01.023] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 01/23/2014] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with cognitive dysfunction and dementia. Brain imaging may provide important clues about underlying processes. This review focuses on the relationship between T2DM and brain abnormalities assessed with different imaging techniques: both structural and functional magnetic resonance imaging (MRI), including diffusion tensor imaging and magnetic resonance spectroscopy, as well as positron emission tomography and single-photon emission computed tomography. Compared to people without diabetes, people with T2DM show slightly more global brain atrophy, which increases gradually over time compared with normal aging. Moreover, vascular lesions are seen more often, particularly lacunar infarcts. The association between T2DM and white matter hyperintensities and microbleeds is less clear. T2DM has been related to diminished cerebral blood flow and cerebrovascular reactivity, particularly in more advanced disease. Diffusion tensor imaging is a promising technique with respect to subtle white matter involvement. Thus, brain imaging studies show that T2DM is associated with both degenerative and vascular brain damage, which develops slowly over the course of many years. The challenge for future studies will be to further unravel the etiology of brain damage in T2DM, and to identify subgroups of patients that will develop distinct progressive brain damage and cognitive decline.
Collapse
Affiliation(s)
- Manon Brundel
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands.
| | - L Jaap Kappelle
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
237
|
Katsumoto A, Lu H, Miranda AS, Ransohoff RM. Ontogeny and functions of central nervous system macrophages. THE JOURNAL OF IMMUNOLOGY 2014; 193:2615-21. [PMID: 25193935 DOI: 10.4049/jimmunol.1400716] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Microglia, the only nonneuroepithelial cells found in the parenchyma of the CNS, originate during embryogenesis from the yolk sac and enter the CNS quite early (embryonic day 9.5-10 in mice). Thereafter, microglia are maintained independently of any input from the blood and, in particular, do not require hematopoietic stem cells as a source of replacement for senescent cells. Monocytes are hematopoietic cells, derived from bone marrow. The ontogeny of microglia and monocytes is important for understanding CNS pathologies. Microglial functions are distinct from those of blood-derived monocytes, which invade the CNS only under pathological conditions. Recent data reveal that microglia play an important role in managing neuronal cell death, neurogenesis, and synaptic interactions. In this article, we discuss the physiology of microglia and the functions of monocytes in CNS pathology. We address the roles of microglia and monocytes in neurodegenerative diseases as an example of CNS pathology.
Collapse
Affiliation(s)
- Atsuko Katsumoto
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Haiyan Lu
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Aline S Miranda
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and
| | - Richard M Ransohoff
- Neuroinflammation Research Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195; and Mellen Center for Multiple Sclerosis Treatment and Research, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
238
|
Libertini G. The programmed aging paradigm: How we get old. BIOCHEMISTRY (MOSCOW) 2014; 79:1004-16. [DOI: 10.1134/s0006297914100034] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
239
|
Is Alzheimer's disease related to metabolic syndrome? A Wnt signaling conundrum. Prog Neurobiol 2014; 121:125-46. [PMID: 25084549 DOI: 10.1016/j.pneurobio.2014.07.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/17/2014] [Accepted: 07/23/2014] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, affecting more than 36 million people worldwide. AD is characterized by a progressive loss of cognitive functions. For years, it has been thought that age is the main risk factor for AD. Recent studies suggest that life style factors, including nutritional behaviors, play a critical role in the onset of dementia. Evidence about the relationship between nutritional behavior and AD includes the role of conditions such as obesity, hypertension, dyslipidemia and elevated glucose levels. The coexistence of some of these cardio-metabolic risk factors is generally known as metabolic syndrome (MS). Some clinical studies support the role of MS in the onset of AD. However, the cross-talk between the molecular signaling implicated in these disorders is unknown. In the present review, we focus on the molecular correlates that support the relationship between MS and the onset of AD. We also discuss relevant issues such as the role of leptin, insulin and renin-angiotensin signaling in the brain and the possible role of Wnt signaling in both MS and AD. We discuss the evidence supporting the use of ob/ob mice, high-fructose diets, aortic coarctation-induced hypertension and Octodon degus, which spontaneously develops β-amyloid deposits and metabolic derangements, as suitable animal models to address the relationships between MS and AD. Finally, we examine emergent data supporting the role of Wnt signaling in the modulation of AD and MS, implicating this pathway as a therapeutic target in both conditions.
Collapse
|
240
|
Qiu WQ, Zhu H. Amylin and its analogs: a friend or foe for the treatment of Alzheimer's disease? Front Aging Neurosci 2014; 6:186. [PMID: 25120481 PMCID: PMC4114192 DOI: 10.3389/fnagi.2014.00186] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 07/09/2014] [Indexed: 12/20/2022] Open
Abstract
Amylin, a gut-brain axis hormone, and amyloid-beta peptides (Aβ), a major component of the Alzheimer's disease (AD) brain, share several features, including similar β-sheet secondary structures, binding to the same receptor and being degraded by the same protease, insulin degrading enzyme (IDE). However, while amylin readily crosses the blood brain barrier (BBB) and mediates several activities including improving glucose metabolism, relaxing cerebrovascular structure, modulating inflammatory reaction and perhaps enhancing neural regeneration, Aβ has no known physiological functions. Thus, abundant Aβ in the AD brain could block or interfere with the binding of amylin to its receptor and hinder its functions. Recent studies using animal models for AD demonstrate that amylin and its analog reduce the AD pathology in the brain and improve cognitive impairment in AD. Given that, in addition to amyloid plaques and neurofibrillary tangles, perturbed cerebral glucose metabolism and cerebrovascular damage are the hallmarks of the AD brain, we propose that giving exogenous amylin type peptides have the potential to become a new avenue for the diagnosis and therapeutic of AD. Although amylin's property of self-aggregation may be a limitation to developing it as a therapeutic for AD, its clinical analog, pramlintide containing 3 amino acid differences from amylin, does not aggregate like human amylin, but more potently mediates amylin's activities in the brain. Pramlintide is an effective drug for diabetes with a favorable profile of safety. Thus a randomized, double-blind, placebo-controlled clinical trial should be conducted to examine the efficacy of pramlintide for AD. This review summarizes the knowledge and findings on amylin type peptides and discuss pros and cons for their potential for AD.
Collapse
Affiliation(s)
- Wei Qiao Qiu
- Department of Psychiatry, Boston University School of Medicine Boston, MA, USA ; Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine Boston, MA, USA ; Alzheimer's Disease Center, Boston University School of Medicine Boston, MA, USA
| | - Haihao Zhu
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine Boston, MA, USA
| |
Collapse
|
241
|
Kukreja L, Kujoth GC, Prolla TA, Van Leuven F, Vassar R. Increased mtDNA mutations with aging promotes amyloid accumulation and brain atrophy in the APP/Ld transgenic mouse model of Alzheimer's disease. Mol Neurodegener 2014; 9:16. [PMID: 24885175 PMCID: PMC4028006 DOI: 10.1186/1750-1326-9-16] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/03/2014] [Indexed: 11/14/2022] Open
Abstract
Background The role of mitochondrial dysfunction has long been implicated in age-related brain pathology, including Alzheimer’s disease (AD). However, the mechanism by which mitochondrial dysfunction may cause neurodegeneration in AD is unclear. To model mitochondrial dysfunction in vivo, we utilized mice that harbor a knockin mutation that inactivates the proofreading function of mitochondrial DNA polymerase γ (PolgA D257A), so that these mice accumulate mitochondrial DNA mutations with age. PolgA D257A mice develop a myriad of mitochondrial bioenergetic defects and physical phenotypes that mimic premature ageing, with subsequent death around one year of age. Results We crossed the D257A mice with a well-established transgenic AD mouse model (APP/Ld) that develops amyloid plaques. We hypothesized that mitochondrial dysfunction would affect Aβ synthesis and/or clearance, thus contributing to amyloidogenesis and triggering neurodegeneration. Initially, we discovered that Aβ42 levels along with Aβ42 plaque density were increased in D257A; APP/Ld bigenic mice compared to APP/Ld monogenic mice. Elevated Aβ production was not responsible for increased amyloid pathology, as levels of BACE1, PS1, C99, and C83 were unchanged in D257A; APP/Ld compared to APP/Ld mice. However, the levels of a major Aβ clearance enzyme, insulin degrading enzyme (IDE), were reduced in mice with the D257A mutation, suggesting this as mechanism for increased amyloid load. In the presence of the APP transgene, D257A mice also exhibited significant brain atrophy with apparent cortical thinning but no frank neuron loss. D257A; APP/Ld mice had increased levels of 17 kDa cleaved caspase-3 and p25, both indicative of neurodegeneration. Moreover, D257A; APP/Ld neurons appeared morphologically disrupted, with swollen and vacuolated nuclei. Conclusions Overall, our results implicate synergism between the effects of the PolgA D257A mutation and Aβ in causing neurodegeneration. These findings provide insight into mechanisms of mitochondrial dysfunction that may contribute to the pathogenesis of AD via decreased clearance of Aβ.
Collapse
Affiliation(s)
| | | | | | | | - Robert Vassar
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
242
|
Mosher KI, Wyss-Coray T. Microglial dysfunction in brain aging and Alzheimer's disease. Biochem Pharmacol 2014; 88:594-604. [PMID: 24445162 PMCID: PMC3972294 DOI: 10.1016/j.bcp.2014.01.008] [Citation(s) in RCA: 453] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 01/06/2014] [Accepted: 01/07/2014] [Indexed: 12/23/2022]
Abstract
Microglia, the immune cells of the central nervous system, have long been a subject of study in the Alzheimer's disease (AD) field due to their dramatic responses to the pathophysiology of the disease. With several large-scale genetic studies in the past year implicating microglial molecules in AD, the potential significance of these cells has become more prominent than ever before. As a disease that is tightly linked to aging, it is perhaps not entirely surprising that microglia of the AD brain share some phenotypes with aging microglia. Yet the relative impacts of both conditions on microglia are less frequently considered in concert. Furthermore, microglial "activation" and "neuroinflammation" are commonly analyzed in studies of neurodegeneration but are somewhat ill-defined concepts that in fact encompass multiple cellular processes. In this review, we have enumerated six distinct functions of microglia and discuss the specific effects of both aging and AD. By calling attention to the commonalities of these two states, we hope to inspire new approaches for dissecting microglial mechanisms.
Collapse
Affiliation(s)
- Kira Irving Mosher
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA; Neuroscience IDP Program, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California 94305, USA; Center for Tissue Regeneration, Repair and Restoration, Veterans Administration Palo Alto Health Care System, Palo Alto, California 94304, USA.
| |
Collapse
|
243
|
Abstract
Obesity, metabolic syndrome, and type 2 diabetes (T2D) are related disorders with widespread deleterious effects throughout the body. One important target of damage is the brain. Persons with metabolic disorders are at significantly increased risk for cognitive decline and the development of vascular dementia and Alzheimer's disease. Our review of available evidence from epidemiologic, clinical, and basic research suggests that neural dysfunction from T2D-related disease results from several underlying mechanisms, including metabolic, inflammatory, vascular, and oxidative changes. The relationships between T2D and neural dysfunction are regulated by several modifiers. We emphasize 2 such modifiers, the genetic risk factor apolipoprotein E and an age-related endocrine change, low testosterone. Both factors are independent risk factors for Alzheimer's disease that may also cooperatively regulate pathologic interactions between T2D and dementia. Continued elucidation of the links between metabolic disorders and neural dysfunction promises to foster the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Anusha Jayaraman
- 3715 McClintock Avenue, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191 USA, , (213) 740-8244
| | - Christian J. Pike
- 3715 McClintock Avenue, Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089-0191 USA, , (213) 740-4205
| |
Collapse
|
244
|
Chen Y, Deng Y, Zhang B, Gong CX. Deregulation of brain insulin signaling in Alzheimer's disease. Neurosci Bull 2014; 30:282-94. [PMID: 24652456 PMCID: PMC5562654 DOI: 10.1007/s12264-013-1408-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/03/2014] [Indexed: 01/09/2023] Open
Abstract
Contrary to the previous belief that insulin does not act in the brain, studies in the last three decades have demonstrated important roles of insulin and insulin signal transduction in various functions of the central nervous system. Deregulated brain insulin signaling and its role in molecular pathogenesis have recently been reported in Alzheimer's disease (AD). In this article, we review the roles of brain insulin signaling in memory and cognition, the metabolism of amyloid β precursor protein, and tau phosphorylation. We further discuss deficiencies of brain insulin signaling and glucose metabolism, their roles in the development of AD, and recent studies that target the brain insulin signaling pathway for the treatment of AD. It is clear now that deregulation of brain insulin signaling plays an important role in the development of sporadic AD. The brain insulin signaling pathway also offers a promising therapeutic target for treating AD and probably other neurodegenerative disorders.
Collapse
Affiliation(s)
- Yanxing Chen
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009 China
| | - Yanqiu Deng
- Department of Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070 China
| | - Baorong Zhang
- Department of Neurology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009 China
| | - Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York 10314 USA
| |
Collapse
|
245
|
Abstract
Plasma amyloid β-42 (Aβ42) and Aβ42/Aβ40 are increasingly recognized as biomarkers for dementia, with low levels indicating increased risk. Little is known about the demographic and medical correlates of plasma Aβ40 or Aβ42. In 997 community-dwelling, nondemented older adults from the Health, Aging, and Body Composition Study, we determined the cross-sectional association between a wide range of demographic and medical variables with Aβ40 and Aβ42. In multivariate stepwise linear regression models, Aβ40 was significantly associated with race (β=-14.70, F=22.01, P<0.0001), age (β=1.34, F=6.39, P=0.01), creatinine (β=52.91, F=151.77, P<0.0001), and the serum brain-derived neurotrophic factor (β=-0.0004, F=7.34, P=0.007); Aβ42 was significantly associated with race (β=-3.72, F=30.83, P<0.0001), sex (β=1.39, F=4.32, P=0.04), education (β=1.50, F=4.78, P=0.03), apolipoprotein E e4 genotype (β=-2.82, F=16.57, P<0.0001), and creatinine (β=9.32, F=120.09, P<0.0001). These correlates should be considered as potential confounders in future studies investigating plasma Aβ as a biomarker of dementia. Understanding fully how these correlates mediate or modify the association between plasma Aβ and dementia will be a fundamental step in determining the biological pathways through which plasma Aβ40 and Aβ42 are associated with dementia, and in determining their full potential as biomarkers.
Collapse
|
246
|
Dinkins MB, Dasgupta S, Wang G, Zhu G, Bieberich E. Exosome reduction in vivo is associated with lower amyloid plaque load in the 5XFAD mouse model of Alzheimer's disease. Neurobiol Aging 2014; 35:1792-800. [PMID: 24650793 DOI: 10.1016/j.neurobiolaging.2014.02.012] [Citation(s) in RCA: 370] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/06/2014] [Accepted: 02/10/2014] [Indexed: 11/19/2022]
Abstract
We present evidence here that exosomes stimulate aggregation of amyloid beta (Aβ)1-42 in vitro and in vivo and interfere with uptake of Aβ by primary cultured astrocytes and microglia in vitro. Exosome secretion is prevented by the inhibition of neutral sphingomyelinase 2 (nSMase2), a key regulatory enzyme generating ceramide from sphingomyelin, with GW4869. Using the 5XFAD mouse, we show that intraperitoneal injection of GW4869 reduces the levels of brain and serum exosomes, brain ceramide, and Aβ1-42 plaque load. Reduction of total Aβ1-42 as well as number of plaques in brain sections was significantly greater (40% reduction) in male than female mice. Our results suggest that GW4869 reduces amyloid plaque formation in vivo by preventing exosome secretion and identifies nSMase2 as a potential drug target in AD by interfering with exosome secretion.
Collapse
Affiliation(s)
- Michael B Dinkins
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, USA
| | - Somsankar Dasgupta
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, USA
| | - Guanghu Wang
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, USA
| | - Gu Zhu
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, USA
| | - Erhard Bieberich
- Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, GA, USA.
| |
Collapse
|
247
|
Qiu WQ, Wallack M, Dean M, Liebson E, Mwamburi M, Zhu H. Association between amylin and amyloid-β peptides in plasma in the context of apolipoprotein E4 allele. PLoS One 2014; 9:e88063. [PMID: 24520345 PMCID: PMC3919737 DOI: 10.1371/journal.pone.0088063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/06/2014] [Indexed: 01/10/2023] Open
Abstract
Amylin, a pancreatic peptide that readily crosses the blood brain barrier (BBB), and amyloid-beta peptide (Aβ), the main component of amyloid plaques and a major component of Alzheimer's disease (AD) pathology in the brain, share several features. These include having similar β-sheet secondary structures, binding to the same receptor, and being degraded by the same protease. Thus, amylin may be associated with Aβ, but the nature of their relationship remains unclear. In this study, we used human samples to study the relationship between plasma amylin and Aβ in the context of the apolipoprotein E alleles (ApoE). We found that concentrations of Aβ1-42 (P<0.0001) and Aβ1-40 (P<0.0001) increased with each quartile increase of amylin. Using multivariate regression analysis, the study sample showed that plasma amylin was associated with Aβ1-42 (β = +0.149, SE = 0.025, P<0.0001) and Aβ1-40 (β = +0.034, SE = 0.016, P = 0.04) as an outcome after adjusting for age, gender, ethnicity, ApoE4, BMI, diabetes, stroke, kidney function and lipid profile. This positive association between amylin and Aβ1-42 in plasma was found regardless of the ApoE genotype. In contrast, the relationship between amylin and Aβ1-40 in plasma seen in ApoE4 non-carriers disappeared in the presence of ApoE4. Using AD mouse models, our recent study demonstrates that intraperitoneal (i.p.) injection of synthetic amylin enhances the removal of Aβ from the brain into blood, thus resulting in increased blood levels of both amylin and Aβ. The positive association between amylin and Aβ, especially Aβ1-42, in human blood samples is probably relevant to the findings in the AD mouse models. The presence of ApoE4 may attenuate amylin's capacity to remove Aβ, especially Aβ1-40, from the AD brain.
Collapse
Affiliation(s)
- Wei Qiao Qiu
- Departments of Psychiatry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Max Wallack
- Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Alzheimer's Disease Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Michael Dean
- Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Elizabeth Liebson
- McLean Hospital, Harvard Medical School, Belmont, Massachusetts, United States of America
| | - Mkaya Mwamburi
- Department of Public Health and Family Medicine, Tufts University, Boston, Massachusetts, United States of America
| | - Haihao Zhu
- Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|
248
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
249
|
Qiu WQ, Li H, Zhu H, Scott T, Mwamburi M, Rosenberg I, Rosenzweig J. Plasma Amylin and Cognition in Diabetes in the Absence and the Presence of Insulin Treatment. ACTA ACUST UNITED AC 2014; 5. [PMID: 25750761 PMCID: PMC4350457 DOI: 10.4172/2155-6156.1000458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Background Plasma amylin is positively associated with cognitive function in humans. Amylin treatment improves memory in Alzheimer’s mouse models. However, the relationship between plasma amylin, diabetes and cognition is not clear. Objectives In this study we examined the concentration of plasma amylin, its relationship with diabetes and cognition. Material and Method A cross-sectional, homebound elderly population with data of plasma amylin under fasting condition and cognitive measurements was used. Results We found that subjects with a long and chronic duration of diabetes were more likely to take insulin treatment and have reduced secretion of amylin. Compared to non-diabetics, diabetic subjects without insulin treatment had a higher concentration, but those with insulin treatment had a lower concentration, of plasma amylin [median (Q1, Q3): 20 (11.0, 36.2) vs. 25.2 (13.2, 50.6) vs. 15.0 (4.9, 33.8), p<0.0001]. In the whole sample vs. in the absence of diabetes, plasma amylin was positively associated with logical memory delayed recall (β= +0.61, SE=0.25, p=0.02 vs. β=+0.80, SE=0.33, p=0.02) and block design (β=+0.62, SE=0.24, p=0.009 vs. β=+0.93, SE=0.31, p=0.003), and negatively associated with Trailmaking A scores (β= −6.21, SE=1.55, p<0.0001 vs. β=−7.51, SE=1.95, p=0.0001) and Trailmaking B (β= −4.32, SE=2.13, p=0.04 vs. β= −5.86, SE=2.73, p=0.04). All these relationships disappeared in the presence of diabetes regardless the treatment. Conclusion This study suggests that secretion of amylin by pancreas compensates and then deteriorates depending on the duration of diabetes. Amylin’s activities for cognition are impaired in the presence of diabetes.
Collapse
Affiliation(s)
- Wei Qiao Qiu
- Department of Psychiatry, Boston University School of Medicine, USA ; Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, USA ; Department of Medicine, Endocrinology, Boston University School of Medicine, USA
| | - Huajie Li
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, USA ; Department of Neurology, the First People's Hospital of Chang Zhou, China
| | - Haihao Zhu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, USA
| | | | | | | | - James Rosenzweig
- Department of Medicine, Endocrinology, Boston University School of Medicine, USA
| |
Collapse
|
250
|
Qiu WQ, Au R, Zhu H, Wallack M, Liebson E, Li H, Rosenzweig J, Mwamburi M, Stern RA. Positive association between plasma amylin and cognition in a homebound elderly population. J Alzheimers Dis 2014; 42:555-63. [PMID: 24898659 PMCID: PMC4834912 DOI: 10.3233/jad-140210] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Our recent study reported that amylin, a pancreatic peptide that readily crosses the blood-brain barrier, improves learning and memory in Alzheimer's disease mouse models. However, the relationship between peripheral amylin and cognition in humans is unknown. In this follow-up study, using a cross-sectional, homebound elderly population, improvement in cognitive function with increasing quartiles of plasma amylin was suggested by positive association with verbal memory (p = 0.0002) and visuoconstruction tasks (p = 0.004), and inverse association with timed measures of attention (p < 0.0001) and executive function (p = 0.04). After adjusting for demographic information, apolipoprotein E4 allele, diabetes, stroke, kidney function, and lipid profile, log10 of plasma amylin remained associated with these cognitive domains. In contrast, plasma amyloid-β peptide was not associated with these specific cognitive domains. Our study suggests that peripheral amylin may be protective for cognitive decline, especially in the domains affected by Alzheimer's disease.
Collapse
Affiliation(s)
- Wei Qiao Qiu
- Department of Psychiatry, Boston University School of Medicine, Boston, MA, USA
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Rhoda Au
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| | - Haihao Zhu
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - Max Wallack
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | | | - Huajie Li
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
- Department of Neurology, The First People’s Hospital of Chang Zhou, China
| | - James Rosenzweig
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Mkaya Mwamburi
- Department of Public Health and Family Medicine, Tufts University, Medford, MA, USA
| | - Robert A. Stern
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Department of Neurosurgery, Boston University School of Medicine, Boston, MA, USA
- Department of Alzheimer’s Disease Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|