1
|
Bär J, Fanutza T, Reimann CC, Seipold L, Grohe M, Bolter JR, Delfs F, Bucher M, Gee CE, Schweizer M, Saftig P, Mikhaylova M. Non-canonical function of ADAM10 in presynaptic plasticity. Cell Mol Life Sci 2024; 81:342. [PMID: 39123091 PMCID: PMC11335265 DOI: 10.1007/s00018-024-05327-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 08/12/2024]
Abstract
A Disintegrin And Metalloproteinase 10 (ADAM10) plays a pivotal role in shaping neuronal networks by orchestrating the activity of numerous membrane proteins through the shedding of their extracellular domains. Despite its significance in the brain, the specific cellular localization of ADAM10 remains not well understood due to a lack of appropriate tools. Here, using a specific ADAM10 antibody suitable for immunostainings, we observed that ADAM10 is localized to presynapses and especially enriched at presynaptic vesicles of mossy fiber (MF)-CA3 synapses in the hippocampus. These synapses undergo pronounced frequency facilitation of neurotransmitter release, a process that play critical roles in information transfer and neural computation. We demonstrate, that in conditional ADAM10 knockout mice the ability of MF synapses to undergo this type of synaptic plasticity is greatly reduced. The loss of facilitation depends on the cytosolic domain of ADAM10 and association with the calcium sensor synaptotagmin 7 rather than ADAM10's proteolytic activity. Our findings unveil a new role of ADAM10 in the regulation of synaptic vesicle exocytosis.
Collapse
Affiliation(s)
- Julia Bär
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Tomas Fanutza
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Christopher C Reimann
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Lisa Seipold
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany
| | - Maja Grohe
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany
| | - Janike Rabea Bolter
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany
| | - Flemming Delfs
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michael Bucher
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Christine E Gee
- Department of Synaptic Physiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, ZMNH, 20251, Hamburg, Germany
| | - Paul Saftig
- Biochemisches Institut, Christian Albrechts-Universität Kiel, 24098, Kiel, Germany.
| | - Marina Mikhaylova
- AG Optobiology, Institute of Biology, Humboldt Universität Zu Berlin, 10115, Berlin, Germany.
- Guest Group, "Neuronal Protein Transport", Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
| |
Collapse
|
2
|
Alexandre-Silva V, Cominetti MR. Unraveling the dual role of ADAM10: Bridging the gap between cancer and Alzheimer's disease. Mech Ageing Dev 2024; 219:111928. [PMID: 38513842 DOI: 10.1016/j.mad.2024.111928] [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: 07/07/2023] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
An inverse association between Alzheimer's disease (AD) and cancer has been proposed. Patients with a cancer history have a decreased risk of developing AD, and AD patients have a reduced cancer incidence, which is not seen in vascular dementia patients. Given this association, common molecular and biological mechanisms that could explain this inverse relationship have been proposed before, such as Peptidylprolyl Cis/Trans Isomerase, NIMA-Interacting 1 (Pin1), Wingless and Int-1 (Wnt), and transformation-related protein 53 (p53)-mediated pathways, along with inflammation and oxidative stress-related proteins. A Disintegrin And Metalloprotease 10 (ADAM10) is a protease responsible for the cleavage of key AD- and cancer-related substrates, and it has inverse roles in those diseases: neuroprotective and disease-promoting, respectively. Thus, herein, we review the relevant literature linking AD and cancer and propose how ADAM10 activity might modulate the inverse association between the diseases. Understanding how this protease mediates those two conditions might raise some considerations in the ADAM10 pharmacological modulation for treating AD and cancer.
Collapse
|
3
|
Pelegrini LNDC, da Silva VA, Grigoli MM, Vatanabe IP, Manzine PR, Cominetti MR. Plasma ADAM10 Levels and Their Association with Alzheimer's Disease Diagnosis in Older Adults with Fewer Years of Formal Education. Dement Geriatr Cogn Disord 2024; 53:153-161. [PMID: 38583419 DOI: 10.1159/000538630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/28/2024] [Indexed: 04/09/2024] Open
Abstract
INTRODUCTION Low educational attainment is a potential risk factor for Alzheimer's disease (AD) development. Alpha-secretase ADAM10 plays a central role in AD pathology, attenuating the formation of beta-amyloid peptides and, therefore, their aggregation into senile plaques. This study seeks to investigate ADAM10 as a blood-based biomarker in mild cognitive impairment (MCI) and AD in a diverse group of community-dwelling older adults, focusing on those with limited educational attainment. METHODS Participants were recruited from public health services. Cognition was evaluated using Mini-Mental State Examination (MMSE) and Addenbrooke's Cognitive Examination - Revised (ACE-R) batteries. Blood samples were collected to analyze plasma ADAM10 levels. A logistic regression was conducted to verify the influence of plasma ADAM10 on the AD diagnosis. RESULTS Significant differences in age, years of education, prescribed medications, and cognitive test scores were found between the MCI and AD groups. Regarding cognitive performance, both ACE-R and MMSE scores displayed significant differences between groups, with post hoc analyses highlighting these distinctions, particularly between AD and cognitively unimpaired individuals. Elevated plasma ADAM10 levels were associated with a 4.5-fold increase in the likelihood of a diagnosis of MCI and a 5.9-fold increase in the likelihood of a diagnosis of AD. These findings suggest ADAM10 levels in plasma as a valuable biomarker for assessing cognitive status in older individuals with low education attainment. CONCLUSION This study underscores the potential utility of plasma ADAM10 levels as a blood-based biomarker for cognitive status, especially in individuals with low educational backgrounds, shedding light on their relevance in AD development and diagnosis.
Collapse
Affiliation(s)
| | | | | | - Izabela Pereira Vatanabe
- Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil
- Pharmaceutical Sciences Faculty, University of São Paulo, São Paulo, Brazil
| | | | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos, Brazil,
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland,
| |
Collapse
|
4
|
Roselli S, Satir TM, Camacho R, Fruhwürth S, Bergström P, Zetterberg H, Agholme L. APP-BACE1 Interaction and Intracellular Localization Regulate Aβ Production in iPSC-Derived Cortical Neurons. Cell Mol Neurobiol 2023; 43:3653-3668. [PMID: 37355492 PMCID: PMC10477112 DOI: 10.1007/s10571-023-01374-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/09/2023] [Indexed: 06/26/2023]
Abstract
Alzheimer's disease (AD) is characterized pathologically by amyloid β (Aβ)-containing plaques. Generation of Aβ from amyloid precursor protein (APP) by two enzymes, β- and γ-secretase, has therefore been in the AD research spotlight for decades. Despite this, how the physical interaction of APP with the secretases influences APP processing is not fully understood. Herein, we compared two genetically identical human iPSC-derived neuronal cell types: low Aβ-secreting neuroprogenitor cells (NPCs) and high Aβ-secreting mature neurons, as models of low versus high Aβ production. We investigated levels of substrate, enzymes and products of APP amyloidogenic processing and correlated them with the proximity of APP to β- and γ-secretase in endo-lysosomal organelles. In mature neurons, increased colocalization of full-length APP with the β-secretase BACE1 correlated with increased β-cleavage product sAPPβ. Increased flAPP/BACE1 colocalization was mainly found in early endosomes. In the same way, increased colocalization of APP-derived C-terminal fragment (CTF) with presenilin-1 (PSEN1), the catalytic subunit of γ-secretase, was seen in neurons as compared to NPCs. Furthermore, most of the interaction of APP with BACE1 in low Aβ-secreting NPCs seemed to derive from CTF, the remaining APP part after BACE1 cleavage, indicating a possible novel product-enzyme inhibition. In conclusion, our results suggest that interaction of APP and APP cleavage products with their secretases can regulate Aβ production both positively and negatively. β- and γ-Secretases are difficult targets for AD treatment due to their ubiquitous nature and wide range of substrates. Therefore, targeting APP-secretase interactions could be a novel treatment strategy for AD. Colocalization of APP species with BACE1 in a novel model of low- versus high-Aβ secretion-Two genetically identical human iPSC-derived neuronal cell types: low Aβ-secreting neuroprogenitor cells (NPCs) and high Aβ secreting mature neurons, were compared. Increased full-length APP (flAPP)/BACE1 colocalization in early endosomes was seen in neurons, while APP-CTF/BACE1 colocalization was much higher than flAPP/BACE1 colocalization in NPCs, although the cellular location was not determined.
Collapse
Affiliation(s)
- Sandra Roselli
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden.
| | - Tugce Munise Satir
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden
| | - Rafael Camacho
- Centre for Cellular Imaging, Core Facilities, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 7A, 405 30, Gothenburg, Sweden
| | - Stefanie Fruhwürth
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden
| | - Petra Bergström
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden
| | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Building V3, Mölndal Hospital, 431 80, Mölndal, Sweden
- Department of Neurodegenerative Disease, Institute of Neurology, University College London Queen Square, Queen Square, London, WC1N 3BG, UK
- UK Dementia Research Institute at UCL, Cruciform Building, Gower Street, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Units 1501-1502, 1512-1518, 15/F, Building 17W, Hong Kong Science Park, Shatin, N.T., Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, 600 Highland Avenue, Madison, WI, 53792, USA
| | - Lotta Agholme
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Blå Stråket 15, Sahlgrenska Hospital, 405 30, Gothenburg, Sweden
| |
Collapse
|
5
|
Romero-Fernandez W, Carvajal-Tapia C, Prusky A, Katdare KA, Wang E, Shostak A, Ventura-Antunes L, Harmsen HJ, Lippmann ES, Fuxe K, MacGurn JA, Borroto-Escuela DO, Schrag MS. Detection, visualization and quantification of protein complexes in human Alzheimer's disease brains using proximity ligation assay. Sci Rep 2023; 13:11948. [PMID: 37488165 PMCID: PMC10366145 DOI: 10.1038/s41598-023-38000-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 06/30/2023] [Indexed: 07/26/2023] Open
Abstract
Examination of healthy and diseased human brain is essential to translational neuroscience. Protein-protein interactions play a pivotal role in physiological and pathological processes, but their detection is difficult, especially in aged and fixed human brain tissue. We used the in-situ proximity ligation assay (PLA) to broaden the range of molecular interactions assessable in-situ in the human neuropathology. We adapted fluorescent in-situ PLA to detect ubiquitin-modified proteins in human brains with Alzheimer's disease (AD), including approaches for the management of autofluorescence and quantification using a high-content image analysis system. We confirmed that phosphorylated microtubule-associated protein tau (Serine202, Threonine205) aggregates were modified by ubiquitin and that phospho-tau-ubiquitin complexes were increased in hippocampal and frontal cortex regions in AD compared to non-AD brains. Overall, we refined PLA for use in human neuropathology, which has revealed a profound change in the distribution of ubiquitin in AD brain and its association with characteristic tau pathologies.
Collapse
Affiliation(s)
- Wilber Romero-Fernandez
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA.
| | - Cristian Carvajal-Tapia
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
| | - Alex Prusky
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA
| | - Emmeline Wang
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
| | - Alena Shostak
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
| | - Lissa Ventura-Antunes
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
| | - Hannah J Harmsen
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37240, USA
| | - Ethan S Lippmann
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, 37235, USA
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, 37235, USA
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institute, Solna, 17177, Stockholm, Sweden
| | - Jason A MacGurn
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, 37240, USA
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institute, Solna, 17177, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Department of Human Physiology, Sport and Exercise, Faculty of Medicine, University of Malaga, Edificio Lopez-Penalver, Jimenez Fraud 10, 29071, Málaga, Spain
| | - Matthew S Schrag
- Department of Neurology, Vanderbilt University Medical Center, MRBIII 465 21St Avenue S, Suite 6158, Nashville, TN, 37240, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
6
|
de la Fuente AG, Pelucchi S, Mertens J, Di Luca M, Mauceri D, Marcello E. Novel therapeutic approaches to target neurodegeneration. Br J Pharmacol 2023; 180:1651-1673. [PMID: 36965025 PMCID: PMC10952850 DOI: 10.1111/bph.16078] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 03/27/2023] Open
Abstract
Ageing is the main risk factor common to most primary neurodegenerative disorders. Indeed, age-related brain alterations have been long considered to predispose to neurodegeneration. Although protein misfolding and the accumulation of toxic protein aggregates have been considered as causative events in neurodegeneration, several other biological pathways affected by brain ageing also contribute to pathogenesis. Here, we discuss the evidence showing the involvement of the mechanisms controlling neuronal structure, gene expression, autophagy, cell metabolism and neuroinflammation in the onset and progression of neurodegenerative disorders. Furthermore, we review the therapeutic strategies currently under development or as future approaches designed to normalize these pathways, which may then increase brain resilience to cope with toxic protein species. In addition to therapies targeting the insoluble protein aggregates specifically associated with each neurodegenerative disorder, these novel pharmacological approaches may be part of combined therapies designed to rescue brain function.
Collapse
Affiliation(s)
- Alerie G. de la Fuente
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL)AlicanteSpain
- Instituto de Neurociencias CSIC‐UMHAlicanteSpain
- Wellcome‐Wolfson Institute for Experimental MedicineQueen's University BelfastBelfastUK
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
| | - Jerome Mertens
- Institute of Molecular BiologyLeopold‐Franzens‐Universität InnsbruckInnsbruckAustria
- Department of NeurosciencesUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| | - Daniela Mauceri
- Institute of Anatomy and Cell BiologyDepartment of Molecular and Cellular Neuroscience, University of MarburgMarburgGermany
- Department of NeurobiologyInterdisciplinary Centre for Neurosciences (IZN), Heidelberg UniversityHeidelbergGermany
| | - Elena Marcello
- Department of Pharmacological and Biomolecular SciencesUniversity of MilanMilanItaly
| |
Collapse
|
7
|
Romero-Fernandez W, Carvajal-Tapia C, Prusky A, Katdare K, Wang E, Shostak A, Ventura-Antunes L, Harmsen H, Lippmann E, Borroto-Escuela D, MacGurn J, Fuxe K, Schrag M. Detection, Visualization and Quantification of Protein Complexes in Human Alzheimer's Disease Brains using Proximity Ligation Assay. RESEARCH SQUARE 2023:rs.3.rs-2570335. [PMID: 36824944 PMCID: PMC9949263 DOI: 10.21203/rs.3.rs-2570335/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Examination of healthy and diseased human brain is essential to translational neuroscience. Protein-protein interactions play a pivotal role in physiological and pathological processes, but their detection is difficult, especially in aged and fixed human brain tissue. We used the proximity ligation assay (PLA) to broaden the range of molecular interactions assessable in-situ in human neuropathology. We adapted fluorescent in-situ PLA to detect ubiquitin-modified proteins in human brains with Alzheimer's disease (AD), including approaches for the management of autofluorescence and quantification using a high-content image analysis system. We confirmed that hyperphosphorylated microtubule-associated protein tau (Serine202, Threonine205) aggregates were modified by ubiquitin and that phospho-tau-ubiquitin complexes were increased in hippocampal and frontal cortex regions in AD compared to non-AD brains. Overall, we refined PLA for use in human neuropathology, which has revealed a profound change in the distribution of ubiquitin in AD brain and its association with characteristic tau pathologies.
Collapse
|
8
|
A Disintegrin and Metalloproteinase 10 (ADAM10) Is Essential for Oligodendrocyte Precursor Development and Myelination in the Mouse Brain. Mol Neurobiol 2023; 60:1675-1689. [PMID: 36550333 PMCID: PMC9899191 DOI: 10.1007/s12035-022-03163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A disintegrin and metalloproteinase 10 (ADAM10) plays an essential role in the regulation of survival, proliferation, migration, and differentiation of various neural cells. Nevertheless, the role of ADAM10 in oligodendrocyte precursors (OPCs) and myelination in the central nervous system (CNS) of developing and adult mouse brains is still unknown. We generated ADAM10 conditional knockout (ADAM10 cKO) mice lacking the ADAM10 gene primarily in OPCs by crossing NG2-Cre mice with ADAM10 loxp/loxp mice. We found that OPCs expressed ADAM10 in the mouse corpus callosum and the hippocampus. ADAM10 cKO mice showed significant loss of back hair and reduction in weight and length on postnatal (30 ± 2.1) day, died at (65 ± 5) days after birth, and exhibited the "anxiety and depression-like" performances. Conditional knockout of ADAM10 in OPCs resulted in a prominent increase in myelination and a decrease in the number of OPCs in the corpus callosum at P30 owing to premyelination and lack of proliferation of OPCs. Moreover, the number of proliferating OPCs and mature oligodendrocytes (OLs) also decreased with age in the corpus callosum of ADAM10 cKO mice from P30 to P60. Western blot and RT-PCR results showed that the activation of Notch-1 and its four target genes, Hes1, Hes5, Hey1, and Hey2, was inhibited in the corpus callosum tissue of ADAM10 knockout mice. In our study, we provided experimental evidence to demonstrate that ADAM10 is essential for modulating CNS myelination and OPC development by activating Notch-1 signaling in the developing and adult mouse brain.
Collapse
|
9
|
Piscopo P, Crestini A, Carbone E, Rivabene R, Ancidoni A, Lo Giudice M, Corbo M, Vanacore N, Lacorte E. A systematic review on drugs for synaptic plasticity in the treatment of dementia. Ageing Res Rev 2022; 81:101726. [PMID: 36031056 DOI: 10.1016/j.arr.2022.101726] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 01/31/2023]
Abstract
The aim of the present systematic review (SR) was to provide an overview of all published and unpublished clinical trials investigating the safety and efficacy of disease-modifying drugs targeting synaptic plasticity in dementia. Searches on CT.gov and EuCT identified 27 trials (4 phase-1, 1 phase-1/2, 18 phase-2, 1 phase-2/3, 1 phase-3, 1 phase-4, and 1 not reported). Twenty of them completed, and seven are currently active or enrolling. The structured bibliographic searches yielded 3585 records. A total of 12 studies were selected on Levetiracetam, Masitinib, Saracatinib, BI 40930, Bryostatin 1, PF-04447943 and Edonerpic drugs. We used RoB tool for quality analysis of randomized studies. Efficacy was assessed as a primary outcome in all studies except one and the main scale used was ADAS-Cog (7 studies), MMSE and CDR (4 studies). Safety and tolerability were reported in eleven studies. The incidence of SAEs was similar between treatment and placebo. At the moment, only one molecule reached phase-3. This could suggest that research on these drugs is still preliminary. Of all, three studies reported promising results on Levetiracetam, Bryostatin 1 and Masitinib.
Collapse
Affiliation(s)
- P Piscopo
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy.
| | - A Crestini
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - E Carbone
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - R Rivabene
- Department of Neuroscience, Italian National Institute of Health, Rome, Italy
| | - A Ancidoni
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - M Lo Giudice
- Need Institute, Foundation for Cure and Rehabilitation of Neurological Diseases, Milan, Italy
| | - M Corbo
- Department of Neurorehabilitation Sciences, Casa Cura Policlinico, Milan, Italy.
| | - N Vanacore
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| | - E Lacorte
- National Center for Disease Prevention ad Heath Promotion, Italian National Institute of Health, Rome, Italy
| |
Collapse
|
10
|
Zhai X, Shan S, Wan J, Tian H, Wang J, Xin L. Silver Nanoparticles Induce a Size-dependent Neurotoxicity to SH-SY5Y Neuroblastoma Cells via Ferritinophagy-mediated Oxidative Stress. Neurotox Res 2022; 40:1369-1379. [PMID: 36040578 DOI: 10.1007/s12640-022-00570-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/21/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022]
Abstract
Silver nanoparticles (AgNPs) are widely used in a variety of consumer products because of their antibacterial and antifungal characteristics, but little is known about their toxicity to the brain. In this study, we investigated AgNP-induced neurotoxicity using the human neuroblastoma cancer (SH-SY5Y) cell line. After a 24 h treatment of AgNPs with two primary sizes (5 and 50 nm labeled as Ag-5 and Ag-50, respectively), a series of toxicological endpoints including cell viability, expression of proteins and genes in amyloid precursor protein (APP) amyloid hydrolysis process and ferritinophagy signaling pathways, oxidative stress, intracellular iron levels, and molecular regulators of iron metabolism were evaluated. Our results showed that both Ag-5 and Ag-50 induced notable neurotoxic effects on SH-SY5Y cells indicated by cell proliferation inhibition, increased BACE1 protein expression, and decreased APP and ADAM10 gene expression. Activation of nuclear receptor coactivator 4-mediated ferritinophagy and blockade of autophagic flux were induced by AgNPs, accompanied by intracellular iron accumulation and overexpression of divalent metal-ion transporter-1 and ferroportin1 in SH-SY5Y cells. In addition, AgNPs significantly decreased glutathione peroxidase 4 protein expression but increased malondialdehyde concentration, suggesting that AgNP-induced iron accumulation may trigger oxidative stress by disruption of the intracellular oxidant and antioxidant systems. In addition, compared with Ag-50, Ag-5 with higher cellular uptake efficiency caused more detrimental effects on SH-SY5Y cells. In conclusion, our findings demonstrated a size-dependent neurotoxicity in SH-SY5Y cells by AgNPs via ferritinophagy-mediated oxidative stress.
Collapse
Affiliation(s)
- Xuedi Zhai
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Shan Shan
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Jianmei Wan
- Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Hailin Tian
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China
| | - Jianshu Wang
- Suzhou Center for Disease Prevention and Control, 72 Sanxiang Road, Suzhou, Jiangsu, 215004, China.
| | - Lili Xin
- School of Public Health, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
11
|
Molecular Pathophysiological Mechanisms in Huntington's Disease. Biomedicines 2022; 10:biomedicines10061432. [PMID: 35740453 PMCID: PMC9219859 DOI: 10.3390/biomedicines10061432] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022] Open
Abstract
Huntington’s disease is an inherited neurodegenerative disease described 150 years ago by George Huntington. The genetic defect was identified in 1993 to be an expanded CAG repeat on exon 1 of the huntingtin gene located on chromosome 4. In the following almost 30 years, a considerable amount of research, using mainly animal models or in vitro experiments, has tried to unravel the complex molecular cascades through which the transcription of the mutant protein leads to neuronal loss, especially in the medium spiny neurons of the striatum, and identified excitotoxicity, transcriptional dysregulation, mitochondrial dysfunction, oxidative stress, impaired proteostasis, altered axonal trafficking and reduced availability of trophic factors to be crucial contributors. This review discusses the pathogenic cascades described in the literature through which mutant huntingtin leads to neuronal demise. However, due to the ubiquitous presence of huntingtin, astrocytes are also dysfunctional, and neuroinflammation may additionally contribute to Huntington’s disease pathology. The quest for therapies to delay the onset and reduce the rate of Huntington’s disease progression is ongoing, but is based on findings from basic research.
Collapse
|
12
|
Pelucchi S, Gardoni F, Di Luca M, Marcello E. Synaptic dysfunction in early phases of Alzheimer's Disease. HANDBOOK OF CLINICAL NEUROLOGY 2022; 184:417-438. [PMID: 35034752 DOI: 10.1016/b978-0-12-819410-2.00022-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The synapse is the locus of plasticity where short-term alterations in synaptic strength are converted to long-lasting memories. In addition to the presynaptic terminal and the postsynaptic compartment, a more holistic view of the synapse includes the astrocytes and the extracellular matrix to form a tetrapartite synapse. All these four elements contribute to synapse health and are crucial for synaptic plasticity events and, thereby, for learning and memory processes. Synaptic dysfunction is a common pathogenic trait of several brain disorders. In Alzheimer's Disease, the degeneration of synapses can be detected at the early stages of pathology progression before neuronal degeneration, supporting the hypothesis that synaptic failure is a major determinant of the disease. The synapse is the place where amyloid-β peptides are generated and is the target of the toxic amyloid-β oligomers. All the elements constituting the tetrapartite synapse are altered in Alzheimer's Disease and can synergistically contribute to synaptic dysfunction. Moreover, the two main hallmarks of Alzheimer's Disease, i.e., amyloid-β and tau, act in concert to cause synaptic deficits. Deciphering the mechanisms underlying synaptic dysfunction is relevant for the development of the next-generation therapeutic strategies aimed at modifying the disease progression.
Collapse
Affiliation(s)
- Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
13
|
Preeti K, Sood A, Fernandes V. Metabolic Regulation of Glia and Their Neuroinflammatory Role in Alzheimer's Disease. Cell Mol Neurobiol 2021; 42:2527-2551. [PMID: 34515874 DOI: 10.1007/s10571-021-01147-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disorder. It is characterized clinically by progressive memory loss and impaired cognitive function. Its progression occurs from neuronal synapse loss to amyloid pathology and Tau deposit which eventually leads to the compromised neuronal function. Neurons in central nervous tissue work in a composite and intricate network with the glia and vascular cells. Microglia and astrocytes are becoming the prime focus due to their involvement in various aspects of neurophysiology, such as trophic support to neurons, synaptic modulation, and brain surveillance. AD is also often considered as the sequela of prolonged metabolic dyshomeostasis. The neuron and glia have different metabolic profiles as cytosolic glycolysis and mitochondrial-dependent oxidative phosphorylation (OXPHOS), especially under dyshomeostasis or with aging pertaining to their unique genetic built-up. Various efforts are being put in to decipher the role of mitochondrial dynamics regarding their trafficking, fission/fusion imbalance, and mitophagy spanning over both neurons and glia to improve aging-related brain health. The mitochondrial dysfunction may lead to activation in various signaling mechanisms causing metabolic reprogramming in glia cells, further accelerating AD-related pathogenic events. The glycolytic-dominant astrocytes switch to the neurotoxic phenotype, i.e., disease-associated astrocyte under metabolic stress. The microglia also transform from resting to reactive phenotype, i.e., disease-associated microglia. It may also exist in otherwise a misconception an M1, glycolytic, or M2, an OXPHOS-dependent phenotype. Further, glial transformation plays a vital role in regulating hallmarks of AD pathologies like synapse maintenance, amyloid, and Tau clearance. In this updated review, we have tried to emphasize the metabolic regulation of glial reactivity, mitochondrial quality control mechanisms, and their neuroinflammatory response in Alzheimer's progression.
Collapse
Affiliation(s)
- Kumari Preeti
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| | - Anika Sood
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Valencia Fernandes
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
14
|
Mendsaikhan A, Tooyama I, Serrano GE, Beach TG, Walker DG. Loss of Lysosomal Proteins Progranulin and Prosaposin Associated with Increased Neurofibrillary Tangle Development in Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:741-753. [PMID: 34374777 PMCID: PMC8433593 DOI: 10.1093/jnen/nlab056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disease causing cognitive decline in the aging population. To develop disease-modifying treatments, understanding the mechanisms behind the pathology is important, which should include observations using human brain samples. We reported previously on the association of lysosomal proteins progranulin (PGRN) and prosaposin (PSAP) with amyloid plaques in non-demented aged control and AD brains. In this study, we investigated the possible involvement of PGRN and PSAP in tangle formation using human brain tissue sections of non-demented aged control subjects and AD cases and compared with cases of frontotemporal dementia with granulin (GRN) mutations. The study revealed that decreased amounts of PGRN and PSAP proteins were detected even in immature neurofibrillary tangles, while colocalization was still evident in adjacent neurons in all cases. Results suggest that neuronal loss of PGRN preceded loss of PSAP as tangles developed and matured. The GRN mutation cases exhibited almost complete absence of PGRN in most neurons, while PSAP signal was preserved. Although based on correlative data, we suggest that reduced levels of PGRN and PSAP and their interaction in neurons might predispose to accumulation of p-Tau protein.
Collapse
Affiliation(s)
- Anarmaa Mendsaikhan
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona (GES, TGB)
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, Arizona (GES, TGB)
| | - Douglas G Walker
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Shiga, Japan (AM, IT, DGW)
- School of Life Sciences and Neurodegenerative Disease Research Center, Arizona State University, Tempe, Arizona (DGW)
| |
Collapse
|
15
|
Hsia HE, Tüshaus J, Feng X, Hofmann LI, Wefers B, Marciano DK, Wurst W, Lichtenthaler SF. Endoglycan (PODXL2) is proteolytically processed by ADAM10 (a disintegrin and metalloprotease 10) and controls neurite branching in primary neurons. FASEB J 2021; 35:e21813. [PMID: 34390512 DOI: 10.1096/fj.202100475r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/22/2021] [Accepted: 07/07/2021] [Indexed: 01/24/2023]
Abstract
Cell adhesion is tightly controlled in multicellular organisms, for example, through proteolytic ectodomain shedding of the adhesion-mediating cell surface transmembrane proteins. In the brain, shedding of cell adhesion proteins is required for nervous system development and function, but the shedding of only a few adhesion proteins has been studied in detail in the mammalian brain. One such adhesion protein is the transmembrane protein endoglycan (PODXL2), which belongs to the CD34-family of highly glycosylated sialomucins. Here, we demonstrate that endoglycan is broadly expressed in the developing mouse brains and is proteolytically shed in vitro in mouse neurons and in vivo in mouse brains. Endoglycan shedding in primary neurons was mediated by the transmembrane protease a disintegrin and metalloprotease 10 (ADAM10), but not by its homolog ADAM17. Functionally, endoglycan deficiency reduced the branching of neurites extending from primary neurons in vitro, whereas deletion of ADAM10 had the opposite effect and increased neurite branching. Taken together, our study discovers a function for endoglycan in neurite branching, establishes endoglycan as an ADAM10 substrate and suggests that ADAM10 cleavage of endoglycan may contribute to neurite branching.
Collapse
Affiliation(s)
- Hung-En Hsia
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Johanna Tüshaus
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Xiao Feng
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Laura I Hofmann
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Benedikt Wefers
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany
| | - Denise K Marciano
- Departments of Cell Biology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Institute of Developmental Genetics, Helmholtz Center Munich, Neuherberg/Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Technical University of Munich-Weihenstephan, Neuherberg/Munich, Neuherberg, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
16
|
Barron JC, Hurley EP, Parsons MP. Huntingtin and the Synapse. Front Cell Neurosci 2021; 15:689332. [PMID: 34211373 PMCID: PMC8239291 DOI: 10.3389/fncel.2021.689332] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Huntington disease (HD) is a monogenic disease that results in a combination of motor, psychiatric and cognitive symptoms. HD is caused by a CAG trinucleotide repeat expansion in the huntingtin (HTT) gene, which results in the production of a pathogenic mutant HTT protein (mHTT). Although there is no cure at present for HD, a number of RNA-targeting therapies have recently entered clinical trials which aim to lower mHTT production through the use of antisense oligonucleotides (ASOs) and RNAi. However, many of these treatment strategies are non-selective in that they cannot differentiate between non-pathogenic wild type HTT (wtHTT) and the mHTT variant. As HD patients are already born with decreased levels of wtHTT, these genetic therapies may result in critically low levels of wtHTT. The consequence of wtHTT reduction in the adult brain is currently under debate, and here we argue that wtHTT loss is not well-tolerated at the synaptic level. Synaptic dysfunction is an extremely sensitive measure of subsequent cell death, and is known to precede neurodegeneration in numerous brain diseases including HD. The present review focuses on the prominent role of wtHTT at the synapse and considers the consequences of wtHTT loss on both pre- and postsynaptic function. We discuss how wtHTT is implicated in virtually all major facets of synaptic neurotransmission including anterograde and retrograde transport of proteins to/from terminal buttons and dendrites, neurotransmitter release, endocytic vesicle recycling, and postsynaptic receptor localization and recycling. We conclude that wtHTT presence is essential for proper synaptic function.
Collapse
Affiliation(s)
- Jessica C Barron
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Emily P Hurley
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, St. John's, NL, Canada
| |
Collapse
|
17
|
Dore K, Carrico Z, Alfonso S, Marino M, Koymans K, Kessels HW, Malinow R. PSD-95 protects synapses from β-amyloid. Cell Rep 2021; 35:109194. [PMID: 34077732 PMCID: PMC8237704 DOI: 10.1016/j.celrep.2021.109194] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/16/2021] [Accepted: 05/10/2021] [Indexed: 01/22/2023] Open
Abstract
Beta-amyloid (Aβ) depresses excitatory synapses by a poorly understood mechanism requiring NMDA receptor (NMDAR) function. Here, we show that increased PSD-95, a major synaptic scaffolding molecule, blocks the effects of Aβ on synapses. The protective effect persists in tissue lacking the AMPA receptor subunit GluA1, which prevents the confounding synaptic potentiation by increased PSD-95. Aβ modifies the conformation of the NMDAR C-terminal domain (CTD) and its interaction with protein phosphatase 1 (PP1), producing synaptic weakening. Higher endogenous levels or overexpression of PSD-95 block Aβ-induced effects on the NMDAR CTD conformation, its interaction with PP1, and synaptic weakening. Our results indicate that increased PSD-95 protects synapses from Aβ toxicity, suggesting that low levels of synaptic PSD-95 may be a molecular sign indicating synapse vulnerability to Aβ. Importantly, pharmacological inhibition of its depalmitoylation increases PSD-95 at synapses and rescues deficits caused by Aβ, possibly opening a therapeutic avenue against Alzheimer's disease.
Collapse
Affiliation(s)
- Kim Dore
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA.
| | - Zachary Carrico
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Stephanie Alfonso
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Marc Marino
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| | - Karin Koymans
- Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Helmut W Kessels
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA; Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, the Netherlands
| | - Roberto Malinow
- Center for Neural Circuits and Behavior, Department of Neuroscience and Section for Neurobiology, Division of Biology, University of California, San Diego, San Diego, CA 92093, USA
| |
Collapse
|
18
|
Pereira Vatanabe I, Peron R, Mantellatto Grigoli M, Pelucchi S, De Cesare G, Magalhães T, Manzine PR, Figueredo Balthazar ML, Di Luca M, Marcello E, Cominetti MR. ADAM10 Plasma and CSF Levels Are Increased in Mild Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22052416. [PMID: 33670873 PMCID: PMC7957802 DOI: 10.3390/ijms22052416] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 01/03/2023] Open
Abstract
ADAM10 is the main α-secretase that participates in the non-amyloidogenic cleavage of amyloid precursor protein (APP) in neurons, inhibiting the production of β-amyloid peptide (Aβ) in Alzheimer’s disease (AD). Strong recent evidence indicates the importance of the localization of ADAM10 for its activity as a protease. In this study, we investigated ADAM10 activity in plasma and CSF samples of patients with amnestic mild cognitive impairment (aMCI) and mild AD compared with cognitively healthy controls. Our results indicated that plasma levels of soluble ADAM10 were significantly increased in the mild AD group, and that in these samples the protease was inactive, as determined by activity assays. The same results were observed in CSF samples, indicating that the increased plasma ADAM10 levels reflect the levels found in the central nervous system. In SH-SY5Y neuroblastoma cells, ADAM10 achieves its major protease activity in the fraction obtained from plasma membrane lysis, where the mature form of the enzyme is detected, confirming the importance of ADAM10 localization for its activity. Taken together, our results demonstrate the potential of plasma ADAM10 to act as a biomarker for AD, highlighting its advantages as a less invasive, easier, faster, and lower-cost processing procedure, compared to existing biomarkers.
Collapse
Affiliation(s)
- Izabela Pereira Vatanabe
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Rafaela Peron
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Marina Mantellatto Grigoli
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | - Silvia Pelucchi
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Giulia De Cesare
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Thamires Magalhães
- Department of Neurology, University of Campinas, Campinas 13083-887, Brazil; (T.M.); (M.L.F.B.)
| | - Patricia Regina Manzine
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
| | | | - Monica Di Luca
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
| | - Elena Marcello
- Department of Pharmacological and Biomolecular Sciences, Universitá Degli Studi di Milano, 20133 Milan, Italy; (S.P.); (G.D.C.); (M.D.L.)
- Correspondence: (E.M.); (M.R.C.); Tel.: +39-02-5031-8314 (E.M.); +55-16-3306-6663 (M.R.C.)
| | - Marcia Regina Cominetti
- Department of Gerontology, Federal University of São Carlos, São Carlos 13565-905, Brazil; (I.P.V.); (R.P.); (M.M.G.); (P.R.M.)
- Correspondence: (E.M.); (M.R.C.); Tel.: +39-02-5031-8314 (E.M.); +55-16-3306-6663 (M.R.C.)
| |
Collapse
|
19
|
Oliveira Monteiro MPA, Salheb Oliveira DSM, Manzine PR, Crispim Nascimento CM, Dos Santos Orlandi AA, de Oliveira Gomes GA, Dos Santos Orlandi F, Zazzetta MS, Pott-Junior H, Cominetti MR. ADAM10 plasma levels predict worsening in cognition of older adults: a 3-year follow-up study. ALZHEIMERS RESEARCH & THERAPY 2021; 13:18. [PMID: 33419480 PMCID: PMC7792035 DOI: 10.1186/s13195-020-00750-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Blood-based biomarkers for Alzheimer's disease (AD) are highly needed in clinic practice. So far, the gold standards for AD diagnosis are brain neuroimaging and beta-amyloid peptide, total tau, and phosphorylated tau in cerebrospinal fluid (CSF); however, they are not attractive for large-scale screening. Blood-based biomarkers allow an initial large-scale screening of patients under suspicion that could later be tested for the already established CSF biomarkers. To this regard, in this study, we evaluated whether plasma ADAM10 levels would be predictors of declines in cognition in community-dwelling older adults after a 3-year period follow-up. METHODS This was a 3-year longitudinal cohort study that included 219 community-dwelling older adults. Sociodemographic, clinical, lifestyle, depressive symptoms (GDS), and cognitive data (Mini-Mental State Examination, MMSE; Clock Drawing test, CDT) were gathered. The measurement of ADAM10 plasma levels was performed using a sandwich ELISA kit. Bivariate comparisons between groups were performed using Wilcoxon-Mann-Whitney for continuous data and Pearson's chi-square tests with Yates continuity correction for categorical data. Longitudinal analyzes of changes in the MMSE scores were performed using linear mixed-effects modeling. RESULTS Baseline MMSE scores and ADAM10 levels were significantly associated with MMSE scores on the follow-up assessment. When analyzing the interaction with time, normal MMSE scores and the ADAM10 plasma levels at baseline presented a significant and independent negative association with MMSE score values on the follow-up assessment. The analyses also showed that the predictive effect of ADAM10 plasma levels on decreasing MMSE scores on follow-up seems to be more pronounced in participants with normal MMSE, when compared with those with altered MMSE scores at baseline. CONCLUSIONS Considering that ADAM10 increase in plasma is detected as soon as in mild cognitive impairment (MCI) patients, the results presented here may support the complementary clinical use of this biomarker, in addition to the classical AD biomarkers. Taken together, these results provide the first direct evidence that changes in ADAM10 plasma levels are predictors of cognitive worsening in older adults. Moreover, this work can shed light on the study of blood biomarkers for AD and contribute to the advancement of the area.
Collapse
Affiliation(s)
- Maria Patrícia A Oliveira Monteiro
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Danielle S M Salheb Oliveira
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Patrícia R Manzine
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Carla M Crispim Nascimento
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | | | - Grace A de Oliveira Gomes
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Fabiana Dos Santos Orlandi
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Marisa S Zazzetta
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil
| | - Henrique Pott-Junior
- Department of Medicine, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Marcia R Cominetti
- Department of Gerontology, Federal University of São Carlos (UFSCar), Rodovia Washington Luís, km 235, São Carlos, SP, 13565-905, Brazil.
| |
Collapse
|