1
|
Kołodziej-Sobczak D, Sobczak Ł, Łączkowski KZ. Protein Tyrosine Phosphatase 1B (PTP1B): A Comprehensive Review of Its Role in Pathogenesis of Human Diseases. Int J Mol Sci 2024; 25:7033. [PMID: 39000142 PMCID: PMC11241624 DOI: 10.3390/ijms25137033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Overexpression of protein tyrosine phosphatase 1B (PTP1B) disrupts signaling pathways and results in numerous human diseases. In particular, its involvement has been well documented in the pathogenesis of metabolic disorders (diabetes mellitus type I and type II, fatty liver disease, and obesity); neurodegenerative diseases (Alzheimer's disease, Parkinson's disease); major depressive disorder; calcific aortic valve disease; as well as several cancer types. Given this multitude of therapeutic applications, shortly after identification of PTP1B and its role, the pursuit to introduce safe and selective enzyme inhibitors began. Regrettably, efforts undertaken so far have proved unsuccessful, since all proposed PTP1B inhibitors failed, or are yet to complete, clinical trials. Intending to aid introduction of the new generation of PTP1B inhibitors, this work collects and organizes the current state of the art. In particular, this review intends to elucidate intricate relations between numerous diseases associated with the overexpression of PTP1B, as we believe that it is of the utmost significance to establish and follow a brand-new holistic approach in the treatment of interconnected conditions. With this in mind, this comprehensive review aims to validate the PTP1B enzyme as a promising molecular target, and to reinforce future research in this direction.
Collapse
Affiliation(s)
- Dominika Kołodziej-Sobczak
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089 Bydgoszcz, Poland;
| | - Łukasz Sobczak
- Hospital Pharmacy, Multidisciplinary Municipal Hospital in Bydgoszcz, Szpitalna 19, 85-826 Bydgoszcz, Poland
| | - Krzysztof Z. Łączkowski
- Department of Chemical Technology and Pharmaceuticals, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Jurasza 2, 85-089 Bydgoszcz, Poland;
| |
Collapse
|
2
|
Liu J, Zuo X, Huang M, Fang J, Li W, Shi Q, Wang Q, Liang Y. Multifunctional Gomisin B enhances cognitive function in APP/PS1 transgenic mice by regulating Aβ clearance and neuronal apoptosis. Biomed Pharmacother 2023; 166:115423. [PMID: 37673021 DOI: 10.1016/j.biopha.2023.115423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023] Open
Abstract
This study aimed to investigate the potential effects of Gomisin B, a natural compound known for its inhibition of CYP3A4, on cognitive dysfunction in APP/PS1 transgenic mice with Alzheimer's disease (AD). Additionally, the study explored the combined effects of Gomisin B and Osthole (OST). The research involved male wild-type (WT) mice and 7-month-old APP/PS1 transgenic AD mice. The assessment of behavioral changes included the use of the open field test (OFT) and the Morris water maze (MWM). OST levels in brain tissue were quantified using LC-MS/MS, while levels of oxidative stress were measured through an assay kit. Neuronal apoptosis was studied using Nissl staining, RT-qPCR, and immunofluorescence. Amyloid plaque clearance was assessed using thioflavine-S (Th-S) staining, RT-qPCR, and ELISA. The results of the study revealed that Gomisin B led to a significant improvement in cognitive dysfunction in APP/PS1 mice. Moreover, the simultaneous administration of OST and Gomisin B demonstrated enhanced therapeutic effects. These effects were attributed to the inhibition of β-site APP-Cleaving Enzyme 1 (BACE1) and oxidative stress by Gomisin B, along with its anti-apoptotic properties. The combined use of OST and Gomisin B exhibited a synergistic impact, resulting in more pronounced anti-oxidant and anti-apoptotic effects. In summary, this study pioneers the exploration of Gomisin B's multifunctional anti-AD properties in APP/PS1 mice. The findings provide a solid groundwork for the development of anti-Alzheimer's drugs based on natural active ingredients.
Collapse
Affiliation(s)
- Jinman Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Xue Zuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Mingjun Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiansong Fang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Weirong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qing Shi
- Affiliated Jiangmen TCM Hospital of Ji'nan University, Jiangmen 529099, China
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| | - Yong Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China; Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou 510405, China.
| |
Collapse
|
3
|
Makarov M, Kushnireva L, Papa M, Korkotian E. Presenilins and mitochondria-an intriguing link: mini-review. Front Neurosci 2023; 17:1249815. [PMID: 37575294 PMCID: PMC10416233 DOI: 10.3389/fnins.2023.1249815] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/15/2023] Open
Abstract
This review uncovers the intricate relationship between presenilins, calcium, and mitochondria in the context of Alzheimer's disease (AD), with a particular focus on the involvement of presenilin mutations in mitochondrial dysfunction. So far, it is unclear whether the impairment of mitochondrial function arises primarily from damage inflicted by β-amyloid upon mitochondria or from the disruption of calcium homeostasis due to presenilins dysfunctions. The roles of presenilins in mitophagy, autophagy, mitochondrial dynamics, and many other functions, non-γ-secretase related, also require close attention in future research. Resolution of contradictions in understanding of presenilins cellular functions are needed for new effective therapeutic strategies for AD.
Collapse
Affiliation(s)
- Mark Makarov
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Liliia Kushnireva
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Michele Papa
- Department of Mental and Physical Health and Preventive Medicine, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Eduard Korkotian
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
4
|
Yang Y, Bagyinszky E, An SSA. Presenilin-1 (PSEN1) Mutations: Clinical Phenotypes beyond Alzheimer's Disease. Int J Mol Sci 2023; 24:8417. [PMID: 37176125 PMCID: PMC10179041 DOI: 10.3390/ijms24098417] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/01/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Presenilin 1 (PSEN1) is a part of the gamma secretase complex with several interacting substrates, including amyloid precursor protein (APP), Notch, adhesion proteins and beta catenin. PSEN1 has been extensively studied in neurodegeneration, and more than 300 PSEN1 mutations have been discovered to date. In addition to the classical early onset Alzheimer's disease (EOAD) phenotypes, PSEN1 mutations were discovered in several atypical AD or non-AD phenotypes, such as frontotemporal dementia (FTD), Parkinson's disease (PD), dementia with Lewy bodies (DLB) or spastic paraparesis (SP). For example, Leu113Pro, Leu226Phe, Met233Leu and an Arg352 duplication were discovered in patients with FTD, while Pro436Gln, Arg278Gln and Pro284Leu mutations were also reported in patients with motor dysfunctions. Interestingly, PSEN1 mutations may also impact non-neurodegenerative phenotypes, including PSEN1 Pro242fs, which could cause acne inversa, while Asp333Gly was reported in a family with dilated cardiomyopathy. The phenotypic diversity suggests that PSEN1 may be responsible for atypical disease phenotypes or types of disease other than AD. Taken together, neurodegenerative diseases such as AD, PD, DLB and FTD may share several common hallmarks (cognitive and motor impairment, associated with abnormal protein aggregates). These findings suggested that PSEN1 may interact with risk modifiers, which may result in alternative disease phenotypes such as DLB or FTD phenotypes, or through less-dominant amyloid pathways. Next-generation sequencing and/or biomarker analysis may be essential in clearly differentiating the possible disease phenotypes and pathways associated with non-AD phenotypes.
Collapse
Affiliation(s)
- Youngsoon Yang
- Department of Neurology, Soonchunhyang University College of Medicine, Cheonan Hospital, Cheonan 31151, Republic of Korea;
| | - Eva Bagyinszky
- Graduate School of Environment Department of Industrial and Environmental Engineering, Gachon University, Seongnam 13120, Republic of Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
5
|
Raval M, Mishra S, Tiwari AK. Epigenetic regulons in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 198:185-247. [DOI: 10.1016/bs.pmbts.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
6
|
Liu M, Zhang X, Wang Y. Curcumin Alleviates Aβ 42-Induced Neuronal Metabolic Dysfunction via the Thrb/SIRT3 Axis and Improves Cognition in APP TG Mice. Neurochem Res 2021; 46:3166-3178. [PMID: 34401962 DOI: 10.1007/s11064-021-03414-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 12/06/2022]
Abstract
Curcumin has been reported to have a therapeutic effect on Alzheimer's disease (AD), but the specific mechanism remains to be elucidated. In the present research, we aimed to investigate the effect and molecular mechanism of curcumin on AD. Mouse primary hippocampal neuron cells were treated with various concentrations of beta-amyloid 42 (Aβ42) and the results found that Aβ42 inhibited cell viability in a dose-dependent manner. Compared with 50 ng/mL Aβ42, 500 ng/mL Aβ42 could further promote cell apoptosis, reduce the ratio of Nicotinamide adenine dinucleotide (NAD(+))/Nicotinamide adenine diphosphate hydride (NADH) and Adenosine 5'-triphosphate (ATP) level, and inhibit Sirtuins 3 (SIRT3) deacetylation activity and protein expression of Thyroid hormone receptor beta (Thrb) and SIRT3. Hence, 500 ng/mL Aβ42 was used to establish a cell model of AD. Curcumin significantly reversed the inhibitory effects of Aβ42 on cell viability, SIRT3 deacetylation activity, the ratio of NAD+/NADH, ATP level and the protein expression of Thrb and SIRT3, and the promotive effect on apoptosis. ChIPBase was used to predict the binding region of Thrb and SIRT3. Dual luciferase reporter gene and Chromatin immune precipitation (ChIP) assays were employed to verify the relationship between Thrb and promoter of SIRT3 mRNA. Overexpression of Thrb recovered Aβ42 induced metabolic dysfunction, while Thrb silence aggravated Aβ42 induced metabolic dysfunction. Moreover, Thrb silence or 3-TYP (a selective inhibitor of SIRT3) treatment abolished the amelioration of curcumin on Aβ42 induced metabolic dysfunction. Additionally, curcumin attenuated memory deficits in Amyloid precursor protein transgenic (APPTG) mice. Collectively, curcumin alleviated Aβ42-induced neuronal metabolic dysfunction through increasing Thrb expression and SIRT3 activity and improved cognition in APPTG mice.
Collapse
Affiliation(s)
- Min Liu
- Department of Basic Disciplines, Jiangxi Health Vocational College, Nanchang, 330052, China
| | - Xiaodan Zhang
- Department of Basic Disciplines, Jiangxi Health Vocational College, Nanchang, 330052, China
| | - Ying Wang
- Department of Recuperation No.1, Dalian Rehabilitation and Recuperation Center, Dalian, 116016, China.
| |
Collapse
|
7
|
Electroacupuncture Ameliorates Neuroinflammation-Mediated Cognitive Deficits through Inhibition of NLRP3 in Presenilin1/2 Conditional Double Knockout Mice. Neural Plast 2021; 2021:8814616. [PMID: 33505459 PMCID: PMC7806385 DOI: 10.1155/2021/8814616] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 12/16/2020] [Accepted: 12/27/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroinflammation is considered as one of the crucial pathogenesis in promoting neurodegenerative progress of Alzheimer's disease (AD). As complementary and alternative therapy, electroacupuncture (EA) stimulation has been widely used in clinical practice for anti-inflammation. However, whether EA promotes the cognitive deficits resulting from neuroinflammation in AD remains unclear. In this study, the presenilin 1 and 2 conditional double knockout (PS cDKO) mice, exhibited a series of AD-like pathology, robust neuroinflammatory responses, and memory deficits, were used to evaluate the potential neuroprotective effect of EA at Baihui (GV 20) and Shenting (GV 24) by behavioral testing, electrophysiology recording, and molecular biology analyzing. First, we observed that EA improved memory deficits and impaired synaptic plasticity. Moreover, EA possesses an ability to suppress the hyperphosphorylated tau and robust elevated NLRP3, ASC, Caspase-1, IL-1β, and IL-18 in PS cDKO mice. Importantly, MCC950, a potent and selective inhibitor of NLPR3 inflammasome, has similar effects on inhibiting the hyperphosphorylated tau and the robust elevated NLRP3 components and neuroinflammatory responses of PS cDKO mice as well as EA treatment. Furthermore, EA treatment is not able to further improve the AD-like phenotypes of PS cDKO mice in combination with the MCC950 administration. Therefore, EA stimulation at GV 20 and GV 24 acupoints may be a potential alternative therapy for deterring cognitive deficits in AD through suppression of NLRP3 inflammasome activation.
Collapse
|
8
|
Martínez-González K, Islas-Hernández A, Martínez-Ezquerro JD, Bermúdez-Rattoni F, Garcia-delaTorre P. Telomere length and oxidative stress variations in a murine model of Alzheimer's disease progression. Eur J Neurosci 2020; 52:4863-4874. [PMID: 32594585 DOI: 10.1111/ejn.14877] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 11/30/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, and ageing is its major risk factor. Changes in telomere length have been associated with ageing and some degenerative diseases. Our aim was to explore some of the molecular changes caused by the progression of AD in a transgenic murine model (3xTg-AD; B6; 129-Psen1 <tm1Mpm> Tg (APPSwe, tauP301L) 1Lfa). Telomere length was assessed by qPCR in both brain tissue and peripheral blood cells and compared between three age groups: 5, 9 and 13 months. In addition, a possible effect of oxidative stress on telomere length and AD progression was explored. Shorter telomeres were found in blood cells of older transgenic mice compared to younger and wild-type mice but no changes in telomere length in the hippocampus. An increase in oxidative stress with age was found for all strains, but no correlation was found between oxidative stress and shorter telomere length for transgenic mice. Telomere length and oxidative stress are affected by AD progression in the 3xTg-AD murine model. Changes in blood cells are more noticeable than changes in brain tissue, suggesting that systemic changes can be detected early in the disease in this murine model.
Collapse
Affiliation(s)
- Katia Martínez-González
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, CDMX, Coyoacán, México
| | - Azul Islas-Hernández
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México.,Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, CDMX, Coyoacán, México
| | - José Darío Martínez-Ezquerro
- Unidad de Investigación Epidemiológica y en Servicios de Salud, Área de Envejecimiento, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, México City, México
| | - Federico Bermúdez-Rattoni
- Instituto de Fisiología Celular, División de Neurociencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Paola Garcia-delaTorre
- Unidad de Investigación Médica en Enfermedades Neurológicas, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| |
Collapse
|
9
|
Stenzel J, Rühlmann C, Lindner T, Polei S, Teipel S, Kurth J, Rominger A, Krause BJ, Vollmar B, Kuhla A. [ 18F]-florbetaben PET/CT Imaging in the Alzheimer's Disease Mouse Model APPswe/PS1dE9. Curr Alzheimer Res 2020; 16:49-55. [PMID: 30345916 DOI: 10.2174/1567205015666181022095904] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/07/2018] [Accepted: 10/15/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Positron-emission-tomography (PET) using 18F labeled florbetaben allows noninvasive in vivo-assessment of amyloid-beta (Aβ), a pathological hallmark of Alzheimer's disease (AD). In preclinical research, [18F]-florbetaben-PET has already been used to test the amyloid-lowering potential of new drugs, both in humans and in transgenic models of cerebral amyloidosis. The aim of this study was to characterize the spatial pattern of cerebral uptake of [18F]-florbetaben in the APPswe/ PS1dE9 mouse model of AD in comparison to histologically determined number and size of cerebral Aβ plaques. METHODS Both, APPswe/PS1dE9 and wild type mice at an age of 12 months were investigated by smallanimal PET/CT after intravenous injection of [18F]-florbetaben. High-resolution magnetic resonance imaging data were used for quantification of the PET data by volume of interest analysis. The standardized uptake values (SUVs) of [18F]-florbetaben in vivo as well as post mortem cerebral Aβ plaque load in cortex, hippocampus and cerebellum were analyzed. RESULTS Visual inspection and SUVs revealed an increased cerebral uptake of [18F]-florbetaben in APPswe/ PS1dE9 mice compared with wild type mice especially in the cortex, the hippocampus and the cerebellum. However, SUV ratios (SUVRs) relative to cerebellum revealed only significant differences in the hippocampus between the APPswe/PS1dE9 and wild type mice but not in cortex; this differential effect may reflect the lower plaque area in the cortex than in the hippocampus as found in the histological analysis. CONCLUSION The findings suggest that histopathological characteristics of Aβ plaque size and spatial distribution can be depicted in vivo using [18F]-florbetaben in the APPswe/PS1dE9 mouse model.
Collapse
Affiliation(s)
- J Stenzel
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - C Rühlmann
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - T Lindner
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - S Polei
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany
| | - S Teipel
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany, Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| | - J Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - A Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland
| | - B J Krause
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany.,Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany
| | - B Vollmar
- Core Facility Multimodal Small Animal Imaging, Rostock University Medical Center, Rostock, Germany.,Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| | - A Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
10
|
Sanna GD, Nusdeo G, Piras MR, Forteleoni A, Murru MR, Saba PS, Dore S, Sotgiu G, Parodi G, Ganau A. Cardiac Abnormalities in Alzheimer Disease: Clinical Relevance Beyond Pathophysiological Rationale and Instrumental Findings? JACC-HEART FAILURE 2020; 7:121-128. [PMID: 30704603 DOI: 10.1016/j.jchf.2018.10.022] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 10/25/2018] [Accepted: 10/25/2018] [Indexed: 12/22/2022]
Abstract
OBJECTIVES This case control study sought to assess the presence and characteristics of cardiac abnormalities in patients with Alzheimer disease (AD). BACKGROUND Protein misfolding is involved in the pathophysiology of neurodegenerative disorders such as AD. Recently, amyloid-beta (Aβ) aggregates were identified within the cardiomyocytes and interstitium of patients with AD, suggesting that Aβ oligomers may reach and damage the heart. METHODS The authors studied 32 patients with AD and 34 controls matched by age and sex, all of whom were free from cardiac or systemic diseases. A clinical evaluation, an electrocardiogram, and an echocardiogram were performed in all subjects. Furthermore, patients with AD underwent genetic analyses (of the PSEN1, PSEN2, APP, and APOE genes). RESULTS Compared to the control group, patients with AD had a higher prevalence of low-voltage electrocardiographic QRS complexes (28% vs. 3%, respectively; p = 0.004), a lower voltage/mass ratio (p = 0.05), a greater echocardiographic interventricular septum (10.1 ± 1.3 mm vs. 9.3 ± 1.1 mm, respectively; p = 0.01), a greater maximum wall thickness (10.8 ± 1.7 mm vs. 9.3 ± 1.1 mm, respectively; p = 0.0001), and a 2-fold higher prevalence of diastolic dysfunction (70% vs. 35%, respectively; p = 0.007). Symptoms and signs of heart failure were absent in all patients with AD. CONCLUSIONS This study shows that electrocardiographic and echocardiographic abnormalities, including diastolic dysfunction, are present in patients with AD and that these studies reproduce the pattern of cardiac amyloidosis. These findings suggest that, in AD, there may be subclinical cardiac involvement likely associated with Aβ amyloid deposition. The clinical relevance of these cardiac abnormalities should be evaluated in larger prospective studies.
Collapse
Affiliation(s)
- Giuseppe D Sanna
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy.
| | - Giuseppe Nusdeo
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy
| | - Maria Rita Piras
- Unità di valutazione Alzheimer, Azienda Ospedaliero Universitaria Sassari, Ospedale San Camillo, Sassari, Italy
| | | | - Maria Rita Murru
- Laboratorio Centro Sclerosi Multipla, Università di Cagliari, Cagliari, Italy
| | - Pier Sergio Saba
- Clinical and Interventional Cardiology, Sassari University Hospital, Sassari, Italy
| | - Simone Dore
- Clinical Epidemiology and Medical Statistics Unit, Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Sotgiu
- Clinical Epidemiology and Medical Statistics Unit, Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | | | | |
Collapse
|
11
|
Nudelman KNH, McDonald BC, Lahiri DK, Saykin AJ. Biological Hallmarks of Cancer in Alzheimer's Disease. Mol Neurobiol 2019; 56:7173-7187. [PMID: 30993533 PMCID: PMC6728183 DOI: 10.1007/s12035-019-1591-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/01/2019] [Indexed: 11/26/2022]
Abstract
Although Alzheimer's disease (AD) is an international health research priority for our aging population, little therapeutic progress has been made. This lack of progress may be partially attributable to disease heterogeneity. Previous studies have identified an inverse association of cancer and AD, suggesting that cancer history may be one source of AD heterogeneity. These findings are particularly interesting in light of the number of common risk factors and two-hit models hypothesized to commonly drive both diseases. We reviewed the ten hallmark biological alterations of cancer cells to investigate overlap with the AD literature and identified overlap of all ten hallmarks in AD, including (1) potentially common underlying risk factors, such as increased inflammation, deregulated cellular energetics, and genome instability; (2) inversely regulated mechanisms, including cell death and evading growth suppressors; and (3) functions with more complex, pleiotropic mechanisms, some of which may be stage-dependent in AD, such as cell adhesion/contact inhibition and angiogenesis. Additionally, we discuss the recent observation of a biological link between cancer and AD neuropathology. Finally, we address the therapeutic implications of this topic. The significant overlap of functional pathways and molecules between these diseases, some similarly and some oppositely regulated or functioning in each disease, supports the need for more research to elucidate cancer-related AD genetic and functional heterogeneity, with the aims of better understanding AD risk mediators, as well as further exploring the potential for some types of drug repurposing towards AD therapeutic development.
Collapse
Affiliation(s)
- Kelly N. H. Nudelman
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, IN, USA
| | - Brenna C. McDonald
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, IN, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, IN, USA
| | - Debomoy K. Lahiri
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, IN, USA
- Department of Psychiatry, Indiana University School of Medicine, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| | - Andrew J. Saykin
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, IN, USA
- Indiana Alzheimer Disease Center, Indiana University School of Medicine, IN, USA
- Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| |
Collapse
|
12
|
Yan T, Wang L, Gao J, Siedlak SL, Huntley ML, Termsarasab P, Perry G, Chen SG, Wang X. Rab10 Phosphorylation is a Prominent Pathological Feature in Alzheimer's Disease. J Alzheimers Dis 2019; 63:157-165. [PMID: 29562525 DOI: 10.3233/jad-180023] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly, characterized by neurofibrillary tangles (NFTs), senile plaques (SPs), and a progressive loss of neuronal cells in selective brain regions. Rab10, a small Rab GTPase involved in vesicular trafficking, has recently been identified as a novel protein associated with AD. Interestingly, Rab10 is a key substrate of leucine-rich repeat kinase 2 (LRRK2), a serine/threonine protein kinase genetically associated with the second most common neurodegenerative disease Parkinson's disease. However, the phosphorylation state of Rab10 has not yet been investigated in AD. Here, using a specific antibody recognizing LRRK2-mediated Rab10 phosphorylation at the amino acid residue threonine 73 (pRab10-T73), we performed immunocytochemical analysis of pRab10-T73 in hippocampal tissues of patients with AD. pRab10-T73 was prominent in NFTs in neurons within the hippocampus in all cases of AD examined, whereas immunoreactivity was very faint in control cases. Other characteristic AD pathological structures including granulovacuolar degeneration, dystrophic neurites and neuropil threads also contained pRab10-T73. The pRab10-T73 immunoreactivity was diminished greatly following dephosphorylation with alkaline phosphatase. pRab10-T73 was further found to be highly co-localized with hyperphosphorylated tau (pTau) in AD, and demonstrated similar pathological patterns as pTau in Down syndrome and progressive supranuclear palsy. Although pRab10-T73 immunoreactivity could be noted in dystrophic neurites surrounding SPs, SPs were largely negative for pRab10-T73. These findings indicate that Rab10 phosphorylation could be responsible for aberrations in the vesicle trafficking observed in AD leading to neurodegeneration.
Collapse
Affiliation(s)
- Tingxiang Yan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Luwen Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Ju Gao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sandra L Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Mikayla L Huntley
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Pichet Termsarasab
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA
| | - Shu G Chen
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xinglong Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
13
|
DNA methylation analysis on purified neurons and glia dissects age and Alzheimer's disease-specific changes in the human cortex. Epigenetics Chromatin 2018; 11:41. [PMID: 30045751 PMCID: PMC6058387 DOI: 10.1186/s13072-018-0211-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/17/2018] [Indexed: 12/30/2022] Open
Abstract
Background Epigenome-wide association studies (EWAS) based on human brain samples allow a deep and direct understanding of epigenetic dysregulation in Alzheimer’s disease (AD). However, strong variation of cell-type proportions across brain tissue samples represents a significant source of data noise. Here, we report the first EWAS based on sorted neuronal and non-neuronal (mostly glia) nuclei from postmortem human brain tissues. Results We show that cell sorting strongly enhances the robust detection of disease-related DNA methylation changes even in a relatively small cohort. We identify numerous genes with cell-type-specific methylation signatures and document differential methylation dynamics associated with aging specifically in neurons such as CLU, SYNJ2 and NCOR2 or in glia RAI1,CXXC5 and INPP5A. Further, we found neuron or glia-specific associations with AD Braak stage progression at genes such as MCF2L, ANK1, MAP2, LRRC8B, STK32C and S100B. A comparison of our study with previous tissue-based EWAS validates multiple AD-associated DNA methylation signals and additionally specifies their origin to neuron, e.g., HOXA3 or glia (ANK1). In a meta-analysis, we reveal two novel previously unrecognized methylation changes at the key AD risk genes APP and ADAM17. Conclusions Our data highlight the complex interplay between disease, age and cell-type-specific methylation changes in AD risk genes thus offering new perspectives for the validation and interpretation of large EWAS results. Electronic supplementary material The online version of this article (10.1186/s13072-018-0211-3) contains supplementary material, which is available to authorized users.
Collapse
|
14
|
Novel interaction between Alzheimer's disease-related protein presenilin 1 and glutamate transporter 1. Sci Rep 2018; 8:8718. [PMID: 29880815 PMCID: PMC5992168 DOI: 10.1038/s41598-018-26888-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 05/18/2018] [Indexed: 12/28/2022] Open
Abstract
Neuronal hyperactivity is one of the earliest events observed in Alzheimer’s disease (AD). Moreover, alterations in the expression of glutamate transporters have been reported to exacerbate amyloid pathology and cognitive deficits in transgenic AD mouse models. However, the molecular links between these pathophysiological changes remain largely unknown. Here, we report novel interaction between presenilin 1 (PS1), the catalytic component of the amyloid precursor protein-processing enzyme, γ-secretase, and a major glutamate transporter-1 (GLT-1). Our data demonstrate that the interaction occurs between PS1 and GLT-1 expressed at their endogenous levels in vivo and in vitro, takes place in both neurons and astrocytes, and is independent of the PS1 autoproteolysis and γ-secretase activity. This intriguing discovery may shed light on the molecular crosstalk between the proteins linked to the maintenance of glutamate homeostasis and Aβ pathology.
Collapse
|
15
|
Gao J, Wang L, Huntley ML, Perry G, Wang X. Pathomechanisms of TDP-43 in neurodegeneration. J Neurochem 2018; 146:10.1111/jnc.14327. [PMID: 29486049 PMCID: PMC6110993 DOI: 10.1111/jnc.14327] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 02/08/2018] [Accepted: 02/20/2018] [Indexed: 12/11/2022]
Abstract
Neurodegeneration, a term that refers to the progressive loss of structure and function of neurons, is a feature of many neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). There is no cure or treatment available that can prevent or reverse neurodegenerative conditions. The causes of neurodegeneration in these diseases remain largely unknown; yet, an extremely small proportion of these devastating diseases are associated with genetic mutations in proteins involved in a wide range of cellular pathways and processes. Over the past decade, it has become increasingly clear that the most notable neurodegenerative diseases, such as ALS, FTLD, and AD, share a common prominent pathological feature known as TAR DNA-binding protein 43 (TDP-43) proteinopathy, which is usually characterized by the presence of aberrant phosphorylation, ubiquitination, cleavage and/or nuclear depletion of TDP-43 in neurons and glial cells. The role of TDP-43 as a neurotoxicity trigger has been well documented in different in vitro and in vivo experimental models. As such, the investigation of TDP-43 pathomechanisms in various major neurodegenerative diseases is on the rise. Here, after a discussion of stages of TDP-43 proteinopathy during disease progression in various major neurodegenerative diseases, we review previous and most recent studies about the potential pathomechanisms with a particular emphasis on ALS, FTLD, and AD, and discuss the possibility of targeting TDP-43 as a common therapeutic approach to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Ju Gao
- Departments of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Luwen Wang
- Departments of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mikayla L. Huntley
- Departments of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Xinglong Wang
- Departments of Pathology, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Šimić G, Babić Leko M, Wray S, Harrington CR, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik CM, Hof PR. Monoaminergic neuropathology in Alzheimer's disease. Prog Neurobiol 2017; 151:101-138. [PMID: 27084356 PMCID: PMC5061605 DOI: 10.1016/j.pneurobio.2016.04.001] [Citation(s) in RCA: 184] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 03/09/2016] [Accepted: 04/05/2016] [Indexed: 01/02/2023]
Abstract
None of the proposed mechanisms of Alzheimer's disease (AD) fully explains the distribution patterns of the neuropathological changes at the cellular and regional levels, and their clinical correlates. One aspect of this problem lies in the complex genetic, epigenetic, and environmental landscape of AD: early-onset AD is often familial with autosomal dominant inheritance, while the vast majority of AD cases are late-onset, with the ε4 variant of the gene encoding apolipoprotein E (APOE) known to confer a 5-20 fold increased risk with partial penetrance. Mechanisms by which genetic variants and environmental factors influence the development of AD pathological changes, especially neurofibrillary degeneration, are not yet known. Here we review current knowledge of the involvement of the monoaminergic systems in AD. The changes in the serotonergic, noradrenergic, dopaminergic, histaminergic, and melatonergic systems in AD are briefly described. We also summarize the possibilities for monoamine-based treatment in AD. Besides neuropathologic AD criteria that include the noradrenergic locus coeruleus (LC), special emphasis is given to the serotonergic dorsal raphe nucleus (DRN). Both of these brainstem nuclei are among the first to be affected by tau protein abnormalities in the course of sporadic AD, causing behavioral and cognitive symptoms of variable severity. The possibility that most of the tangle-bearing neurons of the LC and DRN may release amyloid β as well as soluble monomeric or oligomeric tau protein trans-synaptically by their diffuse projections to the cerebral cortex emphasizes their selective vulnerability and warrants further investigations of the monoaminergic systems in AD.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | | | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb, Croatia
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, Lille, France
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Claude M Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
17
|
Iatrou A, Kenis G, Rutten BPF, Lunnon K, van den Hove DLA. Epigenetic dysregulation of brainstem nuclei in the pathogenesis of Alzheimer's disease: looking in the correct place at the right time? Cell Mol Life Sci 2017; 74:509-523. [PMID: 27628303 PMCID: PMC5241349 DOI: 10.1007/s00018-016-2361-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 08/15/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
Even though the etiology of Alzheimer's disease (AD) remains unknown, it is suggested that an interplay among genetic, epigenetic and environmental factors is involved. An increasing body of evidence pinpoints that dysregulation in the epigenetic machinery plays a role in AD. Recent developments in genomic technologies have allowed for high throughput interrogation of the epigenome, and epigenome-wide association studies have already identified unique epigenetic signatures for AD in the cortex. Considerable evidence suggests that early dysregulation in the brainstem, more specifically in the raphe nuclei and the locus coeruleus, accounts for the most incipient, non-cognitive symptomatology, indicating a potential causal relationship with the pathogenesis of AD. Here we review the advancements in epigenomic technologies and their application to the AD research field, particularly with relevance to the brainstem. In this respect, we propose the assessment of epigenetic signatures in the brainstem as the cornerstone of interrogating causality in AD. Understanding how epigenetic dysregulation in the brainstem contributes to AD susceptibility could be of pivotal importance for understanding the etiology of the disease and for the development of novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- A Iatrou
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - G Kenis
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - B P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - K Lunnon
- University of Exeter Medical School, RILD, University of Exeter, Barrack Road, Devon, UK
| | - D L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands.
- Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, 97080, Würzburg, Germany.
| |
Collapse
|
18
|
Abstract
Alzheimer's disease (AD) is the primary cause of dementia in the elderly. It remains incurable and poses a huge socio-economic challenge for developed countries with an aging population. AD manifests by progressive decline in cognitive functions and alterations in behaviour, which are the result of the extensive degeneration of brain neurons. The AD pathogenic mechanism involves the accumulation of amyloid beta peptide (Aβ), an aggregating protein fragment that self-associates to form neurotoxic fibrils that trigger a cascade of cellular events leading to neuronal injury and death. Researchers from academia and the pharmaceutical industry have pursued a rational approach to AD drug discovery and targeted the amyloid cascade. Schemes have been devised to prevent the overproduction and accumulation of Aβ in the brain. The extensive efforts of the past 20 years have been translated into bringing new drugs to advanced clinical trials. The most progressed mechanism-based therapies to date consist of immunological interventions to clear Aβ oligomers, and pharmacological drugs to inhibit the secretase enzymes that produce Aβ, namely β-site amyloid precursor-cleaving enzyme (BACE) and γ-secretase. After giving an update on the development and current status of new AD therapeutics, this review will focus on BACE inhibitors and, in particular, will discuss the prospects of verubecestat (MK-8931), which has reached phase III clinical trials.
Collapse
Affiliation(s)
- Genevieve Evin
- Florey Institute of Neuroscience and Mental Health, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
19
|
Kaufmann D, Kaur Dogra A, Tahrani A, Herrmann F, Wink M. Extracts from Traditional Chinese Medicinal Plants Inhibit Acetylcholinesterase, a Known Alzheimer's Disease Target. Molecules 2016; 21:molecules21091161. [PMID: 27589716 PMCID: PMC6273583 DOI: 10.3390/molecules21091161] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 08/24/2016] [Accepted: 08/27/2016] [Indexed: 12/20/2022] Open
Abstract
Inhibition of acetylcholinesterase (AChE) is a common treatment for early stages of the most general form of dementia, Alzheimer's Disease (AD). In this study, methanol, dichloromethane and aqueous crude extracts from 80 Traditional Chinese Medical (TCM) plants were tested for their in vitro anti-acetylcholinesterase activity based on Ellman's colorimetric assay. All three extracts of Berberis bealei (formerly Mahonia bealei), Coptis chinensis and Phellodendron chinense, which contain numerous isoquinoline alkaloids, substantially inhibited AChE. The methanol and aqueous extracts of Coptis chinensis showed IC50 values of 0.031 µg/mL and 2.5 µg/mL, therefore having an up to 100-fold stronger AChE inhibitory activity than the already known AChE inhibitor galantamine (IC50 = 4.33 µg/mL). Combinations of individual alkaloids berberine, coptisine and palmatine resulted in a synergistic enhancement of ACh inhibition. Therefore, the mode of AChE inhibition of crude extracts of Coptis chinensis, Berberis bealei and Phellodendron chinense is probably due to of this synergism of isoquinoline alkaloids. All extracts were also tested for their cytotoxicity in COS7 cells and none of the most active extracts was cytotoxic at the concentrations which inhibit AChE. Based on these results it can be stated that some TCM plants inhibit AChE via synergistic interaction of their secondary metabolites. The possibility to isolate pure lead compounds from the crude extracts or to administer these as nutraceuticals or as cheap alternative to drugs in third world countries make TCM plants a versatile source of natural inhibitors of AChE.
Collapse
Affiliation(s)
- Dorothea Kaufmann
- Institute of Pharmacy and Molecular Biotechnology, Department of Biology, Ruprecht Karls University Heidelberg, Heidelberg 69120, Germany.
| | - Anudeep Kaur Dogra
- Centre for Pharmacognosy and Phytotherapy, The School of Pharmacy, University of London, 29-39 Brunswick Square, London WC1N 1AX, UK.
| | - Ahmad Tahrani
- Institute of Pharmacy and Molecular Biotechnology, Department of Biology, Ruprecht Karls University Heidelberg, Heidelberg 69120, Germany.
| | - Florian Herrmann
- Institute of Pharmacy and Molecular Biotechnology, Department of Biology, Ruprecht Karls University Heidelberg, Heidelberg 69120, Germany.
| | - Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Department of Biology, Ruprecht Karls University Heidelberg, Heidelberg 69120, Germany.
| |
Collapse
|
20
|
Reinert J, Richard BC, Klafki HW, Friedrich B, Bayer TA, Wiltfang J, Kovacs GG, Ingelsson M, Lannfelt L, Paetau A, Bergquist J, Wirths O. Deposition of C-terminally truncated Aβ species Aβ37 and Aβ39 in Alzheimer's disease and transgenic mouse models. Acta Neuropathol Commun 2016; 4:24. [PMID: 26955942 PMCID: PMC4784385 DOI: 10.1186/s40478-016-0294-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/26/2016] [Indexed: 11/10/2022] Open
Abstract
In Alzheimer's disease (AD) a variety of amyloid β-peptides (Aβ) are deposited in the form of extracellular diffuse and neuritic plaques (NP), as well as within the vasculature. The generation of Aβ from its precursor, the amyloid precursor protein (APP), is a highly complex procedure that involves subsequent proteolysis of APP by β- and γ-secretases. Brain accumulation of Aβ due to impaired Aβ degradation and/or altered ratios between the different Aβ species produced is believed to play a pivotal role in AD pathogenesis. While the presence of Aβ40 and Aβ42 in vascular and parenchymal amyloid have been subject of extensive studies, the deposition of carboxyterminal truncated Aβ peptides in AD has not received comparable attention. In the current study, we for the first time demonstrate the immunohistochemical localization of Aβ37 and Aβ39 in human sporadic AD (SAD). Our study further included the analysis of familial AD (FAD) cases carrying the APP mutations KM670/671NL, E693G and I716F, as well as a case of the PSEN1 ΔExon9 mutation. Aβ37 and Aβ39 were found to be widely distributed within the vasculature in the brains of the majority of studied SAD and FAD cases, the latter also presenting considerable amounts of Aβ37 containing NPs. In addition, both peptides were found to be present in extracellular plaques but only scarce within the vasculature in brains of a variety of transgenic AD mouse models. Taken together, our study indicates the importance of C-terminally truncated Aβ in sporadic and familial AD and raises questions about how these species are generated and regulated.
Collapse
|
21
|
Somavarapu AK, Kepp KP. The dynamic mechanism of presenilin-1 function: Sensitive gate dynamics and loop unplugging control protein access. Neurobiol Dis 2016; 89:147-56. [PMID: 26852951 DOI: 10.1016/j.nbd.2016.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 12/20/2022] Open
Abstract
There is no molecular explanation for the many presenilin 1 (PSEN1) mutations causing Alzheimer's disease, but both gain of function relating to amyloid production and loss of isolated PSEN1 function have been implied. We report here the first detailed dynamic all-atom model of mature PSEN1 from molecular dynamics in an explicit membrane with particular account of the as yet unexplored loop dynamics. We find that mature PSEN1 contains multiple distinct conformational states whereas non-mature PSEN1 is a typical one-state protein. We confirm a previously suggested gating mechanism, and find that the 106-131 loop acts as a "hinge" for the TM2 and TM6 "doors". More importantly, we identify an unplugging mechanism of the Exon 9 loop associated only with mature PSEN1. Proper opening of both the "gate" and "plug" in the membrane produces channel-like morphologies and access to the catalytic aspartates. Dynamically, these features seem linked. The long-range sensitivity of this gate-plug system to subtle conformational changes can explain why so many PSEN1 mutants cause disease. Reduced access and imprecise substrate cleavage associated with impaired gate-plug dynamics is directly illustrated by the effect of maturation in our work and could explain the overall reduction in Aβ levels upon PSEN1 mutation and the increase in the Aβ 42/40 ratio. Yet, our PSEN1-only dynamics are particularly insightful in revealing PSEN1-only dynamics relating to e.g. its role as membrane channel. Thus, our identified gate-plug mechanism is relevant for designing PSEN1 modulating therapies for treatment of Alzheimer's disease within both the amyloid/γ-secretase hypothesis and within the PSEN1 loss of function paradigm.
Collapse
Affiliation(s)
| | - Kasper P Kepp
- Technical University of Denmark, DTU Chemistry, DK-2800 Kongens Lyngby, Denmark.
| |
Collapse
|
22
|
Šimić G, Babić Leko M, Wray S, Harrington C, Delalle I, Jovanov-Milošević N, Bažadona D, Buée L, de Silva R, Di Giovanni G, Wischik C, Hof PR. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer's Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016; 6:6. [PMID: 26751493 PMCID: PMC4808800 DOI: 10.3390/biom6010006] [Citation(s) in RCA: 430] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 11/28/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022] Open
Abstract
Abnormal deposition of misprocessed and aggregated proteins is a common final pathway of most neurodegenerative diseases, including Alzheimer's disease (AD). AD is characterized by the extraneuronal deposition of the amyloid β (Aβ) protein in the form of plaques and the intraneuronal aggregation of the microtubule-associated protein tau in the form of filaments. Based on the biochemically diverse range of pathological tau proteins, a number of approaches have been proposed to develop new potential therapeutics. Here we discuss some of the most promising ones: inhibition of tau phosphorylation, proteolysis and aggregation, promotion of intra- and extracellular tau clearance, and stabilization of microtubules. We also emphasize the need to achieve a full understanding of the biological roles and post-translational modifications of normal tau, as well as the molecular events responsible for selective neuronal vulnerability to tau pathology and its propagation. It is concluded that answering key questions on the relationship between Aβ and tau pathology should lead to a better understanding of the nature of secondary tauopathies, especially AD, and open new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Goran Šimić
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Mirjana Babić Leko
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Selina Wray
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Charles Harrington
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Ivana Delalle
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston 02118, MA, USA.
| | - Nataša Jovanov-Milošević
- Department of Neuroscience, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia.
| | - Danira Bažadona
- Department of Neurology, University Hospital Center Zagreb, Zagreb 10000, Croatia.
| | - Luc Buée
- Laboratory Alzheimer & Tauopathies, Université Lille and INSERM U1172, Jean-Pierre Aubert Research Centre, Lille 59045, France.
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Molecular Neuroscience, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Giuseppe Di Giovanni
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta.
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK.
| | - Claude Wischik
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| | - Patrick R Hof
- Fishberg Department of Neuroscience, Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
23
|
Lardenoije R, Iatrou A, Kenis G, Kompotis K, Steinbusch HWM, Mastroeni D, Coleman P, Lemere CA, Hof PR, van den Hove DLA, Rutten BPF. The epigenetics of aging and neurodegeneration. Prog Neurobiol 2015; 131:21-64. [PMID: 26072273 PMCID: PMC6477921 DOI: 10.1016/j.pneurobio.2015.05.002] [Citation(s) in RCA: 246] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 05/13/2015] [Accepted: 05/13/2015] [Indexed: 12/14/2022]
Abstract
Epigenetics is a quickly growing field encompassing mechanisms regulating gene expression that do not involve changes in the genotype. Epigenetics is of increasing relevance to neuroscience, with epigenetic mechanisms being implicated in brain development and neuronal differentiation, as well as in more dynamic processes related to cognition. Epigenetic regulation covers multiple levels of gene expression; from direct modifications of the DNA and histone tails, regulating the level of transcription, to interactions with messenger RNAs, regulating the level of translation. Importantly, epigenetic dysregulation currently garners much attention as a pivotal player in aging and age-related neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease, where it may mediate interactions between genetic and environmental risk factors, or directly interact with disease-specific pathological factors. We review current knowledge about the major epigenetic mechanisms, including DNA methylation and DNA demethylation, chromatin remodeling and non-coding RNAs, as well as the involvement of these mechanisms in normal aging and in the pathophysiology of the most common neurodegenerative diseases. Additionally, we examine the current state of epigenetics-based therapeutic strategies for these diseases, which either aim to restore the epigenetic homeostasis or skew it to a favorable direction to counter disease pathology. Finally, methodological challenges of epigenetic investigations and future perspectives are discussed.
Collapse
Affiliation(s)
- Roy Lardenoije
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Artemis Iatrou
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Gunter Kenis
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Konstantinos Kompotis
- Center for Integrative Genomics, University of Lausanne, Genopode Building, 1015 Lausanne-Dorigny, Switzerland
| | - Harry W M Steinbusch
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands
| | - Diego Mastroeni
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands; L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, 10515 W. Santa Fe Drive, Sun City, AZ 85351, USA
| | - Paul Coleman
- L.J. Roberts Alzheimer's Disease Center, Banner Sun Health Research Institute, 10515 W. Santa Fe Drive, Sun City, AZ 85351, USA
| | - Cynthia A Lemere
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Daniel L A van den Hove
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands; Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Fuechsleinstrasse 15, 97080 Wuerzburg, Germany
| | - Bart P F Rutten
- School for Mental Health and Neuroscience (MHeNS), Department of Psychiatry and Neuropsychology, Maastricht University, Universiteitssingel 50, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
24
|
Freitas-Andrade M, Naus CC. Astrocytes in neuroprotection and neurodegeneration: The role of connexin43 and pannexin1. Neuroscience 2015; 323:207-21. [PMID: 25913636 DOI: 10.1016/j.neuroscience.2015.04.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 04/13/2015] [Accepted: 04/15/2015] [Indexed: 12/26/2022]
Abstract
The World Health Organization has predicted that by 2040 neurodegenerative diseases will overtake cancer to become the world's second leading cause of death after cardiovascular disease. This has sparked the development of several European and American brain research initiatives focusing on elucidating the underlying cellular and molecular mechanisms of neurodegenerative diseases. Connexin (Cx) and pannexin (Panx) membrane channel proteins are conduits through which neuronal, glial, and vascular tissues interact. In the brain, this interaction is highly critical for homeostasis and brain repair after injury. Understanding the molecular mechanisms by which these membrane channels function, in health and disease, might be particularly influential in establishing conceptual frameworks to develop new therapeutics against Cx and Panx channels. This review focuses on current insights and emerging concepts, particularly the impact of connexin43 and pannexin1, under neuroprotective and neurodegenerative conditions within the context of astrocytes.
Collapse
Affiliation(s)
- M Freitas-Andrade
- Department of Cellular and Physiological Sciences, The Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - C C Naus
- Department of Cellular and Physiological Sciences, The Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
25
|
Evin G, Barakat A. Critical analysis of the use of β-site amyloid precursor protein-cleaving enzyme 1 inhibitors in the treatment of Alzheimer's disease. Degener Neurol Neuromuscul Dis 2014; 4:1-19. [PMID: 32669897 PMCID: PMC7337240 DOI: 10.2147/dnnd.s41056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/06/2014] [Indexed: 01/18/2023] Open
Abstract
Alzheimer’s disease (AD) is the major cause of dementia in the elderly and an unmet clinical challenge. A variety of therapies that are currently under development are directed to the amyloid cascade. Indeed, the accumulation and toxicity of amyloid-β (Aβ) is believed to play a central role in the etiology of the disease, and thus rational interventions are aimed at reducing the levels of Aβ in the brain. Targeting β-site amyloid precursor protein-cleaving enzyme (BACE)-1 represents an attractive strategy, as this enzyme catalyzes the initial and rate-limiting step in Aβ production. Observation of increased levels of BACE1 and enzymatic activity in the brain, cerebrospinal fluid, and platelets of patients with AD and mild cognitive impairment supports the potential benefits of BACE1 inhibition. Numerous potent inhibitors have been generated, and many of these have been proved to lower Aβ levels in the brain of animal models. Over 10 years of intensive research on BACE1 inhibitors has now culminated in advancing half a dozen of these drugs into human trials, yet translating the in vitro and cellular efficacy of BACE1 inhibitors into preclinical and clinical trials represents a challenge. This review addresses the promises and also the potential problems associated with BACE1 inhibitors for AD therapy, as the complex biological function of BACE1 in the brain is becoming unraveled.
Collapse
Affiliation(s)
- Genevieve Evin
- Oxidation Biology Laboratory, Mental Health Research Institute, Florey Institute of Neuroscience and Mental Health, University of Melbourne.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Adel Barakat
- Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Dosunmu R, Wu J, Basha MR, Zawia NH. Environmental and dietary risk factors in Alzheimer’s disease. Expert Rev Neurother 2014; 7:887-900. [PMID: 17610395 DOI: 10.1586/14737175.7.7.887] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects millions in the aging population worldwide and will affect millions more in the next 20 years. Over 90% of all cases are sporadic, with genetics playing a minor role in the etiology of AD. Therefore, it is crucial to investigate the environment and diet as primary risk factors in AD pathology. This review considers epidemiologic case control studies, and in vitro and in vivo research to investigate the potential of environmental exposure to metals, air pollution and pesticides as well as diet as risk factors for AD. In some cases, the role of genetic mutations and environmental risk is discussed. The evidence examined in this review provides a brief overview of the current literature on selected, significant risk factors in promoting amyloid-beta accumulation and aggregation, thus contributing to neurodegeneration.
Collapse
Affiliation(s)
- Remi Dosunmu
- University of Rhode Island, Department of Biomedical & Pharmaceutical Sciences, Kingston, RI 02881, USA.
| | | | | | | |
Collapse
|
27
|
Gutiérrez M, Matus MF, Poblete T, Amigo J, Vallejos G, Astudillo L. Isoxazoles: synthesis, evaluation and bioinformatic design as acetylcholinesterase inhibitors. ACTA ACUST UNITED AC 2013; 65:1796-804. [PMID: 24180424 DOI: 10.1111/jphp.12180] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 10/10/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Inhibition of acetylcholinesterase (AChE) is a common treatment for early stages of Alzheimer's disease. In this study, nine isoxazoles derivatives were tested for their in-vitro AChE activity. The molecular docking showed the interaction of the compounds with the active site. METHODS The isoxazoles were synthesized using 1,3-dipolar cycloaddition in the presence of sodium hypochlorite. They were also isolated and characterized by spectroscopic methods. The in-vitro activity was measured by an adapted version of Ellman's assay. KEY FINDINGS The isoxazoles are described as inhibitors of AChE. The most potent compound in the series exhibited a moderate inhibitory activity (50% inhibitory concentration = 134.87 μm). The design of new compounds was created by using the RACHEL module of the SYBYL software. CONCLUSIONS Our research provided enough evidence of the efficacy of isoxazoles as AChE inhibitors. The isoxazoles were synthesized and evaluated as inhibitors of AChE. The docking study based on a novel series of complexes isoxazole with AChE from Electroporus electricus has demonstrated that the ligand bind is similar to the compounds used as reference. To find new candidates with the isoxazole core that act as inhibitors of AChE, part of the structure of the compound 9 was used for de-novo design. Molecular docking models of the ligand-AChE complexes suggest that the compound 10 is located on the periphery of the AChE active site.
Collapse
Affiliation(s)
- Margarita Gutiérrez
- Laboratorio Síntesis Orgánica, Instituto de Química de Recursos Naturales, Universidad de Talca, Talca
| | | | | | | | | | | |
Collapse
|
28
|
Karr JE, Alexander JE, Winningham RG. Omega-3 polyunsaturated fatty acids and cognition throughout the lifespan: A review. Nutr Neurosci 2013; 14:216-25. [DOI: 10.1179/1476830511y.0000000012] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
29
|
Adwan L, Zawia NH. Epigenetics: a novel therapeutic approach for the treatment of Alzheimer's disease. Pharmacol Ther 2013; 139:41-50. [PMID: 23562602 PMCID: PMC3693222 DOI: 10.1016/j.pharmthera.2013.03.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 03/19/2013] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia in the elderly. It is characterized by the deposition of two forms of aggregates within the brain, the amyloid β plaques and tau neurofibrillary tangles. Currently, no disease-modifying agent is approved for the treatment of AD. Approved pharmacotherapies target the peripheral symptoms but they do not prevent or slow down the progression of the disease. Although several disease-modifying immunotherapeutic agents are in clinical development, many have failed due to the lack of efficacy or serious adverse events. Epigenetic changes including DNA methylation and histone modifications are involved in learning and memory and have been recently highlighted for holding promise as potential targets for AD therapeutics. Dynamic and latent epigenetic alterations are incorporated in AD pathological pathways and present valuable reversible targets for AD and other neurological disorders. The approval of epigenetic drugs for cancer treatment has opened the door for the development of epigenetic drugs for other disorders including neurodegenerative diseases. In particular, methyl donors and histone deacetylase inhibitors are being investigated for possible therapeutic effects to rescue memory and cognitive decline found in such disorders. This review explores the area of epigenetics for potential AD interventions and presents the most recent findings in this field.
Collapse
Affiliation(s)
- Lina Adwan
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
30
|
Kolesnikova EÉ. Mitochondrial Dysfunction and Molecular Bases of Neurodegenerative Diseases. NEUROPHYSIOLOGY+ 2013. [DOI: 10.1007/s11062-013-9341-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
31
|
Ramamoorthy M, Sykora P, Scheibye-Knudsen M, Dunn C, Kasmer C, Zhang Y, Becker KG, Croteau DL, Bohr VA. Sporadic Alzheimer disease fibroblasts display an oxidative stress phenotype. Free Radic Biol Med 2012; 53:1371-80. [PMID: 22885031 PMCID: PMC4617209 DOI: 10.1016/j.freeradbiomed.2012.07.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2012] [Revised: 06/13/2012] [Accepted: 07/17/2012] [Indexed: 11/16/2022]
Abstract
Alzheimer disease (AD) is a major health problem in the United States, affecting one in eight Americans over the age of 65. The number of elderly suffering from AD is expected to continue to increase over the next decade, as the average age of the U.S. population increases. The risk factors for and etiology of AD are not well understood; however, recent studies suggest that exposure to oxidative stress may be a contributing factor. Here, microarray gene expression signatures were compared in AD-patient-derived fibroblasts and normal fibroblasts exposed to hydrogen peroxide or menadione (to simulate conditions of oxidative stress). Using the 23K Illumina cDNA microarray to screen expression of >14,000 human genes, we identified a total of 1017 genes that are chronically up- or downregulated in AD fibroblasts, 215 of which were also differentially expressed in normal human fibroblasts within 12h after exposure to hydrogen peroxide or menadione. Pathway analysis of these 215 genes and their associated pathways revealed cellular functions that may be critically dysregulated by oxidative stress and play a critical role in the etiology and/or pathology of AD. We then examined the AD fibroblasts for the presence of oxidative DNA damage and found increased accumulation of 8-oxo-guanine. These results indicate the possible role of oxidative stress in the gene expression profile seen in AD.
Collapse
Affiliation(s)
- Mahesh Ramamoorthy
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Peter Sykora
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Morten Scheibye-Knudsen
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Christopher Dunn
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Cindy Kasmer
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Deborah L. Croteau
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Vilhelm A. Bohr
- Laboratory of Molecular Gerontology, Biomedical Research Center, 251 Bayview Boulevard, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| |
Collapse
|
32
|
Wang G, Dinkins M, He Q, Zhu G, Poirier C, Campbell A, Mayer-Proschel M, Bieberich E. Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4): potential mechanism of apoptosis induction in Alzheimer disease (AD). J Biol Chem 2012; 287:21384-95. [PMID: 22532571 DOI: 10.1074/jbc.m112.340513] [Citation(s) in RCA: 280] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Amyloid protein is well known to induce neuronal cell death, whereas only little is known about its effect on astrocytes. We found that amyloid peptides activated caspase 3 and induced apoptosis in primary cultured astrocytes, which was prevented by caspase 3 inhibition. Apoptosis was also prevented by shRNA-mediated down-regulation of PAR-4, a protein sensitizing cells to the sphingolipid ceramide. Consistent with a potentially proapoptotic effect of PAR-4 and ceramide, astrocytes surrounding amyloid plaques in brain sections of the 5xFAD mouse (and Alzheimer disease patient brain) showed caspase 3 activation and were apoptotic when co-expressing PAR-4 and ceramide. Apoptosis was not observed in astrocytes with deficient neutral sphingomyelinase 2 (nSMase2), indicating that ceramide generated by nSMase2 is critical for amyloid-induced apoptosis. Antibodies against PAR-4 and ceramide prevented amyloid-induced apoptosis in vitro and in vivo, suggesting that apoptosis was mediated by exogenous PAR-4 and ceramide, potentially associated with secreted lipid vesicles. This was confirmed by the analysis of lipid vesicles from conditioned medium showing that amyloid peptide induced the secretion of PAR-4 and C18 ceramide-enriched exosomes. Exosomes were not secreted by nSMase2-deficient astrocytes, indicating that ceramide generated by nSMase2 is critical for exosome secretion. Consistent with the ceramide composition in amyloid-induced exosomes, exogenously added C18 ceramide restored PAR-4-containing exosome secretion in nSMase2-deficient astrocytes. Moreover, isolated PAR-4/ceramide-enriched exosomes were taken up by astrocytes and induced apoptosis in the absence of amyloid peptide. Taken together, we report a novel mechanism of apoptosis induction by PAR-4/ceramide-enriched exosomes, which may critically contribute to Alzheimer disease.
Collapse
Affiliation(s)
- Guanghu Wang
- Program in Developmental Neurobiology, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia 30912, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Scheffler K, Stenzel J, Krohn M, Lange C, Hofrichter J, Schumacher T, Brüning T, Plath AS, Walker L, Pahnke J. Determination of spatial and temporal distribution of microglia by 230nm-high-resolution, high-throughput automated analysis reveals different amyloid plaque populations in an APP/PS1 mouse model of Alzheimer's disease. Curr Alzheimer Res 2012; 8:781-8. [PMID: 21244350 DOI: 10.2174/156720511797633179] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Revised: 10/24/2010] [Accepted: 11/10/2010] [Indexed: 12/17/2022]
Abstract
One early and prominent pathologic feature of Alzheimer's disease (AD) is the appearance of activated microglia in the vicinity of developing β-amyloid deposits. However, the precise role of microglia during the course of AD is still under discussion. Microglia have been reported to degrade and clear β-amyloid, but they also can exert deleterious effects due to overwhelming inflammatory reactions. Here, we demonstrate the occurrence of developing plaque populations with distinct amounts of associated microglia using time-dependent analyses of plaque morphology and the spatial distribution of microglia in an APP/PS1 mouse model. In addition to a population of larger plaques (>700µm(2)) that are occupied by a moderate contingent of microglial cells across the course of aging, a second type of small β-amyloid deposits develops (≤400µm(2)) in which the plaque core is enveloped by a relatively large number of microglia. Our analyses indicate that microglia are strongly activated early in the emergence of senile plaques, but that activation is diminished in the later stages of plaque evolution (>150 days). These findings support the view that microglia serve to restrict the growth of senile plaques, and do so in a way that minimizes local inflammatory damage to other components of the brain.
Collapse
Affiliation(s)
- Katja Scheffler
- University of Rostock, Department of Neurology, Neurodegeneration Research Laboratory, Gehlsheimer Str. 20, 18147 Rostock, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Neill D. Should Alzheimer's disease be equated with human brain ageing? A maladaptive interaction between brain evolution and senescence. Ageing Res Rev 2012; 11:104-22. [PMID: 21763787 DOI: 10.1016/j.arr.2011.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/26/2011] [Accepted: 06/28/2011] [Indexed: 10/18/2022]
Abstract
In this review Alzheimer's disease is seen as a maladaptive interaction between human brain evolution and senescence. It is predicted to occur in everyone although does not necessarily lead to dementia. The pathological process is initiated in relation to a senescence mediated functional down-regulation in the posteromedial cortex (Initiation Phase). This leads to a loss of glutamatergic excitatory input to layer II entorhinal cortex neurons. A human specific maladaptive neuroplastic response is initiated in these neurons leading to neuronal dysfunction, NFT formation and death. This leads to further loss of glutamatergic excitatory input and propagation of the maladaptive response along excitatory pathways linking evolutionary progressed vulnerable neurons (Propagation Phase). Eventually neurons are affected in many brain areas resulting in dementia. Possible therapeutic approaches include enhancing glutamatergic transmission. The theory may have implications with regards to how Alzheimer's disease is classified.
Collapse
|
35
|
Bi BT, Lin HB, Cheng YF, Zhou H, Lin T, Zhang MZ, Li TJ, Xu JP. Promotion of β-amyloid production by C-reactive protein and its implications in the early pathogenesis of Alzheimer's disease. Neurochem Int 2011; 60:257-66. [PMID: 22202667 DOI: 10.1016/j.neuint.2011.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/06/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
Abstract
C-reactive protein (CRP) and β-amyloid protein (Aβ) are involved in the development of Alzheimer's disease (AD). However, the relationship between CRP and Aβ production is unclear. In vitro and in vivo experiments were performed to investigate the association of CRP with Aβ production. Using the rat adrenal pheochromocytoma cell line (PC12 cells) to mimic neurons, cytotoxicity was evaluated by cell viability and supernatant lactate dehydrogenase (LDH) activity. The levels of amyloid precursor protein (APP), beta-site APP cleaving enzyme (BACE-1), and presenilins (PS-1 and PS-2) were investigated using real-time polymerase chain reaction and Western blotting analysis. Aβ1-42 was measured by enzyme-linked immunosorbent assay. The relevance of CRP and Aβ as well as potential mechanisms were studied using APP/PS1 transgenic (Tg) mice. Treatment with 0.5-4.0 μM CRP for 48 h decreased cell viability and increased LDH leakage in PC12 cells. Incubation with CRP at a sub-toxic concentration of 0.2 μM increased the mRNA levels of APP, BACE-1, PS-1, and PS-2, as well as Aβ1-42 production. CRP inhibitor reversed the CRP-induced upregulations of the mRNA levels of APP, BACE-1, PS-1, and PS-2, and the protein levels of APP, BACE-1, PS-1, and Aβ1-42, but did not reversed Aβ1-42 cytotoxicity. The cerebral levels of CRP and Aβ1-42 in APP/PS1 Tg mice were positively correlated, accompanied with the elevated mRNA expressions of serum amyloid P component (SAP), complement component 1q (C1q), and tumor necrosis factor-α (TNF-α). These results suggest that CRP cytotoxicity is associated with Aβ formation and Aβ-related markers expressions; CRP and Aβ were relevant in early-stage AD; CRP may be an important trigger in AD pathogenesis.
Collapse
Affiliation(s)
- Bing-Tian Bi
- Department of Pharmacology, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, China
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kaufmann D, Dogra AK, Wink M. Myrtenal inhibits acetylcholinesterase, a known Alzheimer target. ACTA ACUST UNITED AC 2011; 63:1368-71. [PMID: 21899553 DOI: 10.1111/j.2042-7158.2011.01344.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Inhibition of acetylcholinesterase (AChE) is a common treatment for early stages of the most general form of dementia, Alzheimer's disease. In this study selected components of essential oils, which carry a variety of important functional groups, were tested for their in-vitro anti-acetylcholinesterase activity. METHODS In-vitro anti-acetylcholinesterase activity was measured by an adapted version of Ellman's colorimetric assay. KEY FINDINGS 1,8-cineole, carvacrol, myrtenal and verbenone apparently inhibited AChE; the highest inhibitory activity was observed for myrtenal (IC50 = 0.17 mm). This is the first study showing the AChE inhibitory activity of myrtenal. CONCLUSIONS Our investigations provided evidence for the efficacy of monoterpenes as inhibitors of AChE.
Collapse
Affiliation(s)
- Dorothea Kaufmann
- Institute of Pharmacy and Molecular Biotechnology, Department of Biology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany.
| | | | | |
Collapse
|
37
|
A novel presenilin 1 mutation (Ser169del) in a Chinese family with early-onset Alzheimer's disease. Neurosci Lett 2010; 468:34-7. [DOI: 10.1016/j.neulet.2009.10.055] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Revised: 09/28/2009] [Accepted: 10/16/2009] [Indexed: 11/22/2022]
|
38
|
Variation of Monoamine Neurotransmitter in Forebrain of Presenilin-1/Presenilin-2 Double Knockout Mice*. PROG BIOCHEM BIOPHYS 2009. [DOI: 10.3724/sp.j.1206.2009.00196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Oxidative damage increased in presenilin1/presenilin2 conditional double knockout mice. Neurosci Bull 2009; 25:131-7. [PMID: 19448687 DOI: 10.1007/s12264-009-0114-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE This report aims to describe the oxidative damage profile in brain of presenilin1 and presenilin2 conditional double knockout mice (dKO) at both early and late age stages, and to discuss the correlation between oxidative stress and the Alzheimer-like phenotypes of dKO mice. METHODS The protein level of Abeta(42) in dKO cortex and free 8-OHdG level in urine were measured by ELISA. Thiobarbituric acid method and spectrophotometric DNPH assay were used to determine the lipid peroxidation and protein oxidation in cortex, respectively. SOD and GSH-PX activities were assessed by SOD Assay Kit-WST and GSH-PX assay kit, separately. RESULTS Significant decrease of Abeta(42) was verified in dKO cortex at 6 months as compared to control mice. Although lipid peroxidation (assessed by MDA) was increased only in dKO cortex at 3 months and protein oxidation (assessed by carbonyl groups) was basically unchanged in dKO cortex, ELISA analysis revealed that free 8-OHdG, which was an indicator of DNA lesion, was significantly decreased in urine of dKO mice from 3 months to 12 months. Activities of SOD and GSH-PX in dKO and control cortices showed no statistical difference except a significant increase of GSH-PX activity in dKO mice at 9 months. CONCLUSION Oxidative damage, especially DNA lesion, was correlated with the neurodegenerative symptoms that appeared in dKO mice without the deposition of Abeta(42). Triggers of oxidative damage could be the inflammatory mediators released by activated microglia and astrocytes.
Collapse
|
40
|
Tang BL. Neuronal protein trafficking associated with Alzheimer disease: from APP and BACE1 to glutamate receptors. Cell Adh Migr 2009; 3:118-28. [PMID: 19372755 DOI: 10.4161/cam.3.1.7254] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Aberrant and/or cumulative amyloid-beta (Abeta) production, resulting from proteolytic processing of the amyloid precursor protein (APP) by beta and gamma-secretases, have been postulated to be a main etiological basis of Alzheimer disease (AD). A number of proteins influence the subcellular trafficking itinerary of APP and the beta-site APP-cleaving enzyme (BACE1) between the cell surface, endosomes and the trans-Golgi network (TGN). Available evidence suggests that co-residence of APP and BACE1 in the endosomal compartments promotes amyloidogenesis. Retrograde transport of APP out of the endosome to the TGN reduces Abeta production, while APP routed to and kept at the cell surface enhances its non-amyloidogenic, alpha-secretase-mediated processing. Changes in post-Golgi membrane trafficking in aging neurons that may influence APP processing is particularly relevant to late-onset, idiopathic AD. Dystrophic axons are key features of AD pathology, and impaired axonal transport could play crucial roles in the pathogenesis of idiopathic AD. Recent evidence has also indicated that Abeta-induced synaptic defects and memory impairment could be explained by a loss of both AMPA and NMDA receptors through endocytosis. Detail understanding of factors that influence these neuronal trafficking processes will open up novel therapeutic avenues for preventing or delaying the onset of symptomatic AD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
41
|
Woodhouse A, Shepherd CE, Sokolova A, Carroll VL, King AE, Halliday GM, Dickson TC, Vickers JC. Cytoskeletal alterations differentiate presenilin-1 and sporadic Alzheimer's disease. Acta Neuropathol 2009; 117:19-29. [PMID: 19015863 DOI: 10.1007/s00401-008-0458-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2008] [Revised: 11/06/2008] [Accepted: 11/07/2008] [Indexed: 12/25/2022]
Abstract
Most cases of Alzheimer's disease (AD) are sporadic in nature, although rarer familial AD (FAD) cases have provided important insights into major pathological disease mechanisms. Mutations in the presenilin 1 gene (PS1) are responsible for the majority of FAD cases, causing an earlier age of onset and more rapid progression to end-stage disease than seen in sporadic AD. We have investigated the cytoskeletal alterations in neuritic AD pathology in a cohort of FAD cases in comparison to sporadic AD and pathologically aged cases. Tau-immunoreactive neurofibrillary tangle (NFT) loads were similar between PS1 FAD and sporadic AD cases. Similarly, plaque loads, both beta-amyloid (Abeta) and thioflavine S, in PS1 FAD and sporadic AD cases were not significantly different; however, in pathologically aged cases, they were significantly lower than those in PS1 cases, but were not different from sporadic AD cases. The 'cotton wool' plaque characteristic of PS1 cases did not demonstrate a high density of dystrophic neurites compared to other Abeta plaque types, but did demonstrate a localised mass effect on the neuropil. Despite minimal differences in plaque and NFT loads, immunolabelling demonstrated clear phenotypic differences in the NFTs and dystrophic neurites in PS1 FAD cases. Presenilin-1 cases exhibited significantly (P < 0.05) more tau-positive NFTs that were immunolabelled by the antibody SMI312 (anti-phosphorylated NF protein and phosphorylated tau) than sporadic AD cases. Presenilin-1 cases also exhibited numerous ring-like NF-positive and elongated tau-labelled dystrophic neurites, whereas these dystrophic neurite types were only abundant at the very early (pathologically aged cases) or very late stages of sporadic AD progression, respectively. These differences in cytoskeletal pathology in PS1 cases suggest an accelerated rate of neuritic pathology development, potentially due to mutant PS1 influencing multiple pathogenic pathways.
Collapse
Affiliation(s)
- Adele Woodhouse
- Wicking Dementia Research and Education Centre and NeuroRepair Group, Menzies Research Institute, Private Bag 29, Hobart, TAS, 7001, Australia
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhu M, Gu F, Shi J, Hu J, Hu Y, Zhao Z. Increased oxidative stress and astrogliosis responses in conditional double-knockout mice of Alzheimer-like presenilin-1 and presenilin-2. Free Radic Biol Med 2008; 45:1493-9. [PMID: 18822370 DOI: 10.1016/j.freeradbiomed.2008.08.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 08/28/2008] [Accepted: 08/29/2008] [Indexed: 10/21/2022]
Abstract
Conditional presenilin 1 and presenilin 2 double knockout causes memory dysfunction and reproduces neurodegenerative phenotypes of Alzheimer disease (AD) in mice. Oxidative stress has been long implicated predominantly in amyloidosis-mediated AD pathologies; however, its role in response to the loss-of-function pathogenic mechanism of AD remains unclear. In this study, we examined the oxidative stress status in PS1 and PS2 double-knockout (PS cDKO) mice using F(2)-isoprostanes (iPF(2alpha)-III) as the marker of lipid peroxidation. Lipid peroxidation was enhanced in a gender- and age-related manner in the PS cDKO mice independent of brain Abeta deposition. Such oxidative abnormalities predominantly in cerebral cortex at 2-4 months of age preceded the onset of many pronounced AD neuropathologies, suggesting that increased lipid peroxidation is not only an early pathophysiological response to PS inactivation, but also a potential culprit responsible for the AD-like neurodegenerative pathologies in the PS cDKO mice. Western blot analysis of cortical glial fibrillary acidic protein demonstrated an increased astrogliosis response to PS inactivation, in particular in the PS cDKO mice at as young as 2 months of age, suggesting that lipid peroxidation and neuronal injury may be closely associated with the loss-of-function neuropathogenic mechanism of AD.
Collapse
Affiliation(s)
- Manjie Zhu
- Key Laboratory of Brain Functional Genomics, Ministry of Education of China and the Science and Technology Commission of Shanghai Municipality, Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai 200062, China
| | | | | | | | | | | |
Collapse
|
43
|
Bellucci C, Lilli C, Baroni T, Parnetti L, Sorbi S, Emiliani C, Lumare E, Calabresi P, Balloni S, Bodo M. Differences in extracellular matrix production and basic fibroblast growth factor response in skin fibroblasts from sporadic and familial Alzheimer's disease. Mol Med 2007; 13:542-50. [PMID: 17660861 PMCID: PMC1933258 DOI: 10.2119/2007-00034.bellucci] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2007] [Accepted: 07/09/2007] [Indexed: 11/06/2022] Open
Abstract
Extracellular matrix (ECM) molecules and growth factors, such as fibroblast growth factor (FGF), play a crucial role in Alzheimer's disease (AD). The purpose of this investigation was to determine whether phenotypic alterations in ECM production are present in non-neuronal AD cells associated with different FGF expression and response. Synthesis of glycosaminoglycans (GAG) and collagen were measured in skin fibroblasts from patients with familial, sporadic AD (FAD and SAD respectively), and from age-matched controls by radiolabeled precursors. Proteoglycans (PG), metalloprotease (MMP)-1, and FGF gene expressions were measured by reverse transcription-polymerase chain reaction. The results showed different ECM neosynthesis and mRNA levels in the two AD fibroblast populations. FAD accumulated more collagen and secreted less GAG than SAD. Biglycan PG was upregulated in FAD while betaglycan, syndecan, and decorin were markedly downregulated in SAD fibroblasts. We found a significant decrease of MMP1, more marked in FAD than in SAD fibroblasts. Constitutive FGF expression was greatly reduced in both pathological conditions (SAD>FAD). Moreover, an inverse high affinity/low affinity FGF receptor ratio between SAD and FAD fibroblasts was observed. FGF treatment differently modulated ECM molecule production and gene expression in the two cell populations. These observations in association with the changes in FGF gene expression and in the FGF receptor number, suggest that cellular mechanisms downstream from FGF receptor binding are involved in the two different forms of AD.
Collapse
Affiliation(s)
- Catia Bellucci
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Cinzia Lilli
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Tiziano Baroni
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Lucilla Parnetti
- Department of Specialistic Medicine and Public Health, Neuroscience Clinical section, University of Perugia, Italy
| | - Sandro Sorbi
- Department of Neurologic and Psychiatric Sciences, University of Firenze, Italy
| | - Carla Emiliani
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | | | - Paolo Calabresi
- Department of Specialistic Medicine and Public Health, Neuroscience Clinical section, University of Perugia, Italy
| | - Stefania Balloni
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Maria Bodo
- Department of Specialistic Medicine and Public Health, Neuroscience Clinical section, University of Perugia, Italy
- Address correspondence and reprint requests to Maria Bodo, Department of Specialistic Medicine and Public Health, Neuroscience Clinical section, University of Perugia, Italy. Phone/Fax: 075-5857432; E-mail:
| |
Collapse
|
44
|
Abstract
2,4-dinitrophenol (DNP) has long been known to be toxic at high concentrations,
an effect related to uncoupling of mitochondrial oxidative phosphorylation. Five
years ago, however, we reported that low concentrations of DNP protect neurons
against the toxicity of the amyloid-β peptide. Since then, a number of
other studies have provided evidence of beneficial actions of DNP (at low
concentrations), including neuroprotection against different types of insult,
blockade of amyloid aggregation, stimulation of neurite outgrowth and neuronal
differentiation, and even extension of lifespan in certain organisms. Some of
these effects appear due to mild mitochondrial uncoupling and prevention of
oxidative stress, whereas other actions are related to activation of additional
intracellular signaling pathways. This study discusses the evidence supporting
beneficial neuroprotective actions of DNP. DNP and other compounds with similar
biological activities may be of interest in the development of novel therapeutic
approaches for neurodegenerative diseases and other neurological disorders.
Collapse
Affiliation(s)
- Sérgio Teixeira Ferreira
- Instituto de Bioquímica Médica, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda Guarino De Felice
- Instituto de Bioquímica Médica, Programa de Bioquímica e Biofísica Celular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
45
|
Woodhouse A, Vickers JC, Adlard PA, Dickson TC. Dystrophic neurites in TgCRND8 and Tg2576 mice mimic human pathological brain aging. Neurobiol Aging 2007; 30:864-74. [PMID: 17950493 DOI: 10.1016/j.neurobiolaging.2007.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2007] [Revised: 08/29/2007] [Accepted: 09/05/2007] [Indexed: 10/22/2022]
Abstract
The morphology and neurochemistry of beta-amyloid (A beta) plaque-associated dystrophic neurites present in TgCRND8 and Tg2576 mice was demonstrated to be strikingly similar to that observed in pathologically aged human cases, but not in Alzheimer's disease (AD) cases. Specifically, pathologically aged cases and both transgenic mouse lines exhibited alpha-internexin- and neurofilament-triplet-labelled ring- and bulb-like dystrophic neurites, but no classical hyperphosphorylated-tau dystrophic neurite pathology. In contrast, AD cases demonstrated abundant classical hyperphosphorylated-tau-labelled dystrophic neurites, but no neurofilament-triplet-labelled ring-like dystrophic neurites. Importantly, quantitation demonstrated that the A beta plaques in TgCRND8 mice were highly axonopathic, and localised displacement or clipping of apical dendrite segments was also associated with A beta plaques in both transgenic mouse models. These results suggest that neuronal pathology in these mice represent an accurate and valuable model for understanding, and developing treatments for, the early brain changes of AD.
Collapse
Affiliation(s)
- Adele Woodhouse
- NeuroRepair Group, Menzies Research Institute, 43 Collins Street, Hobart, 7001 Tasmania, Australia.
| | | | | | | |
Collapse
|
46
|
Hatchett CS, Tyler S, Armstrong D, Dawbarn D, Allen SJ. Familial Alzheimer's disease presenilin 1 mutation M146V increases gamma secretase cutting of p75NTR in vitro. Brain Res 2007; 1147:248-55. [PMID: 17349981 DOI: 10.1016/j.brainres.2007.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 02/01/2007] [Accepted: 02/02/2007] [Indexed: 01/22/2023]
Abstract
The cholinergic neurons of the basal forebrain are amongst the first to degenerate in Alzheimer's disease. These neurons are unique in the brain, expressing the tyrosine kinase receptor TrkA, together with the common neurotrophin receptor p75NTR; both of which bind nerve growth factor. Activation of the TrkA receptor is important in the maintenance of cell viability, whereas the p75NTR receptor has been implicated in apoptosis. Mutations in the gene for presenilin 1, a multi-transmembrane aspartyl protease, are known to cause familial Alzheimer's disease. This is thought to be due to their effect on gamma-secretase-dependent processing of amyloid precursor protein and subsequent formation of amyloid. Since p75NTR was recently shown to undergo gamma-secretase regulated intramembrane proteolysis, this study examines the effect of familial Alzheimer mutations on processing of p75NTR. PC12 cells were stably transfected with familial mutations M146V, A246E and deltaE9 and wild-type presenilin 1 and were examined here for gamma-secretase-dependent proteolysis of p75NTR. Overexpression of wild-type presenilin 1 did not increase gamma-secretase-mediated cleavage of p75NTR. However, by contrast, the presence of the M146V mutation was shown to significantly increase cleavage of p75NTR compared with the other mutations. Survival of cholinergic neurons will depend on the balance between the receptors TrkA and p75NTR, and their respective signalling pathways. Thus alterations in p75NTR proteolysis may influence this equilibrium. The novel finding that a mutation may increase processing of p75NTR may have implications for the pathogenic outcome in Alzheimer's disease.
Collapse
Affiliation(s)
- Caroline Sara Hatchett
- Molecular Neurobiology Unit, University of Bristol, CSSB, Dorothy Hodgkin Building, Bristol BS1 3NY, UK
| | | | | | | | | |
Collapse
|
47
|
Abstract
The process of L1 specification early in plant embryogenesis, and subsequent maintenance and elaboration of epidermal organization, are fundamental to plant growth and fitness. To occur in a co-ordinated fashion, these processes require considerable cell–cell cross-talk. It is perhaps then unsurprising that several classes of plant RLKs (receptor-like kinases), as well as other membrane-localized signalling components, have been implicated both in epidermal specification and in patterning events governing the distribution of epidermal cell types. However, despite our growing knowledge of the roles of these signalling molecules, remarkably little is understood regarding their function at the cellular level. In particular the potential role of regulated proteolytic cleavage in controlling the activity of signalling molecules at the plant plasma membrane has remained largely unaddressed despite its massive importance in signalling in animal systems. Because of the relative physical accessibility of their expression domains, molecules involved in epidermal development present opportunities for investigating mechanisms of cell–cell signalling in planta. Advances in understanding the potential regulatory processing of membrane-localized signalling molecules during epidermal development will be examined using parallels with animal systems to highlight potential future directions for this field of research.
Collapse
Affiliation(s)
- G C Ingram
- Institute for Molecular Plant Science, Rutherford Building, Kings Buildings, University of Edinburgh, Edinburgh EH9 3JR, UK.
| |
Collapse
|
48
|
Francis YI, Diss JKJ, Kariti M, Stephanou A, Latchman DS. p300 activation by Presenilin 1 but not by its M146L mutant. Neurosci Lett 2007; 413:137-40. [PMID: 17197080 DOI: 10.1016/j.neulet.2006.11.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 10/25/2006] [Accepted: 11/17/2006] [Indexed: 11/26/2022]
Abstract
The transcriptional co-activator p300 plays an important role in regulating gene expression in a number of different cell types. We have shown that wild type (WT) Presenilin 1 (PS1) stimulates the transcriptional activity ability of CREB Binding Protein (CBP), a close homolog of p300, whereas the Alzheimer's disease (AD) associated mutant of PS1 does not have this effect. A recent report has suggested that mutant PS1 can also disrupt the TCF/beta-catenin/CBP interaction but has no effect on the TCF/beta-catenin/p300 interaction. This suggests that the malregulation of CBP, but not of p300, caused by mutation in PS1 may be involved in the disease process. Here we show that wild type PS1 stimulates the transcriptional activity ability of p300 whereas an Alzheimer's disease-associated mutant of PS1 did not produce this effect. To our knowledge, this is the first report that shows regulation of p300 activity by WT PS1 and not by mutant PS1, indicating that like CBP, p300 can be differentially regulated by WT PS1 compared to its AD-associated mutant.
Collapse
Affiliation(s)
- Yitshak I Francis
- Medical Molecular Biology Unit, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | | | | | | | | |
Collapse
|
49
|
Li D, Parks SB, Kushner JD, Nauman D, Burgess D, Ludwigsen S, Partain J, Nixon RR, Allen CN, Irwin RP, Jakobs PM, Litt M, Hershberger RE. Mutations of presenilin genes in dilated cardiomyopathy and heart failure. Am J Hum Genet 2006; 79:1030-9. [PMID: 17186461 PMCID: PMC1698711 DOI: 10.1086/509900] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Accepted: 09/27/2006] [Indexed: 12/31/2022] Open
Abstract
Two common disorders of the elderly are heart failure and Alzheimer disease (AD). Heart failure usually results from dilated cardiomyopathy (DCM). DCM of unknown cause in families has recently been shown to result from genetic disease, highlighting newly discovered disease mechanisms. AD is the most frequent neurodegenerative disease of older Americans. Familial AD is caused most commonly by presenilin 1 (PSEN1) or presenilin 2 (PSEN2) mutations, a discovery that has greatly advanced the field. The presenilins are also expressed in the heart and are critical to cardiac development. We hypothesized that mutations in presenilins may also be associated with DCM and that their discovery could provide new insight into the pathogenesis of DCM and heart failure. A total of 315 index patients with DCM were evaluated for sequence variation in PSEN1 and PSEN2. Families positive for mutations underwent additional clinical, genetic, and functional studies. A novel PSEN1 missense mutation (Asp333Gly) was identified in one family, and a single PSEN2 missense mutation (Ser130Leu) was found in two other families. Both mutations segregated with DCM and heart failure. The PSEN1 mutation was associated with complete penetrance and progressive disease that resulted in the necessity of cardiac transplantation or in death. The PSEN2 mutation showed partial penetrance, milder disease, and a more favorable prognosis. Calcium signaling was altered in cultured skin fibroblasts from PSEN1 and PSEN2 mutation carriers. These data indicate that PSEN1 and PSEN2 mutations are associated with DCM and heart failure and implicate novel mechanisms of myocardial disease.
Collapse
Affiliation(s)
- Duanxiang Li
- Familial Dilated Cardiomyopathy Research Program, Division of Cardiovascular Medicine and the Department of Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Mondadori CRA, Buchmann A, Mustovic H, Schmidt CF, Boesiger P, Nitsch RM, Hock C, Streffer J, Henke K. Enhanced brain activity may precede the diagnosis of Alzheimer's disease by 30 years. Brain 2006; 129:2908-22. [PMID: 17012294 DOI: 10.1093/brain/awl266] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Presenilin 1 (PSEN1) mutations cause autosomal dominant familial Alzheimer's disease (FAD). PSEN1 mutation carriers undergo the course of cognitive deterioration, which is typical for sporadic Alzheimer's disease but disease onset is earlier and disease progression is faster. Here, we sought to detect signs of FAD in presymptomatic carriers of the PSEN1 mutation (C410Y) by use of a neuropsychological examination, functional MRI during learning and memory tasks and MRI volumetry. We examined five non-demented members of a FAD family and 21 non-related controls. Two of the five family members were carrying the mutation; one was 20 years old and the other 45 years old. The age of clinical manifestation of FAD in the family studied here is approximately 48 years. Neuropsychological assessments suggested subtle problems with episodic memory in the 20-year-old mutation carrier. The middle-aged mutation carrier fulfilled criteria for amnestic mild cognitive impairment. The 20-year-old mutation carrier exhibited increased, while the middle-aged mutation carrier exhibited decreased brain activity compared to controls within memory-related neural networks during episodic learning and retrieval, but not during a working-memory task. The increased memory-related brain activity in the young mutation carrier might reflect a compensatory effort to overcome preclinical neural dysfunction caused by first pathological changes. The activity reductions in the middle-aged mutation carrier might reflect gross neural dysfunction in a more advanced stage of neuropathology. These data suggest that functional neuroimaging along with tasks that challenge specifically those brain areas which are initial targets of Alzheimer's disease pathology may reveal activity alterations on a single-subject level decades before the clinical manifestation of Alzheimer's disease.
Collapse
|