1
|
Olesen MA, Villavicencio-Tejo F, Cuevas-Espinoza V, Quintanilla RA. Unknown roles of tau pathology in neurological disorders. Challenges and new perspectives. Ageing Res Rev 2025; 103:102594. [PMID: 39577774 DOI: 10.1016/j.arr.2024.102594] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Aging presents progressive changes that increase the susceptibility of the central nervous system (CNS) to suffer neurological disorders (NDs). Several studies have reported that an aged brain suffering from NDs shows the presence of pathological forms of tau protein, a microtubule accessory protein (MAP) critical for neuronal function. In this context, accumulative evidence has shown a pivotal contribution of pathological forms of tau to Alzheimer's disease (AD) and tauopathies. However, current investigations have implicated tau toxicity in other NDs that affect the central nervous system (CNS), including Parkinson's disease (PD), Huntington's disease (HD), Traumatic brain injury (TBI), Multiple sclerosis (MS), and Amyotrophic lateral sclerosis (ALS). These diseases are long-term acquired, affecting essential functions such as motor movement, cognition, hearing, and vision. Previous evidence indicated that toxic forms of tau do not have a critical contribution to the genesis or progression of these diseases. However, recent studies have shown that these tau forms contribute to neuronal dysfunction, inflammation, oxidative damage, and mitochondrial impairment events that contribute to the pathogenesis of these NDs. Recent studies have suggested that these neuropathologies could be associated with a prion-like behavior of tau, which induces a pathological dissemination of these toxic protein forms to different brain areas. Moreover, it has been suggested that this toxic propagation of tau from neurons into neighboring cells impairs the function of glial cells, oligodendrocytes, and endothelial cells by affecting metabolic function and mitochondrial health and inducing oxidative damage by tau pathology. Therefore, in this review, we will discuss current evidence demonstrating the critical role of toxic tau forms on NDs not related to AD and how its propagation and induced-bioenergetics failure may contribute to the pathogenic mechanism present in these NDs.
Collapse
Affiliation(s)
- Margrethe A Olesen
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Francisca Villavicencio-Tejo
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Víctor Cuevas-Espinoza
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile
| | - Rodrigo A Quintanilla
- Laboratory of Neurodegenerative Diseases, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Chile.
| |
Collapse
|
2
|
Marvian AT, Strauss T, Tang Q, Tuck BJ, Keeling S, Rüdiger D, Mirzazadeh Dizaji N, Mohammad-Beigi H, Nuscher B, Chakraborty P, Sutherland DS, McEwan WA, Köglsperger T, Zahler S, Zweckstetter M, Lichtenthaler SF, Wurst W, Schwarz S, Höglinger G. Distinct regulation of Tau Monomer and aggregate uptake and intracellular accumulation in human neurons. Mol Neurodegener 2024; 19:100. [PMID: 39736627 DOI: 10.1186/s13024-024-00786-w] [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: 01/23/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND The prion-like spreading of Tau pathology is the leading cause of disease progression in various tauopathies. A critical step in propagating pathologic Tau in the brain is the transport from the extracellular environment and accumulation inside naïve neurons. Current research indicates that human neurons internalize both the physiological extracellular Tau (eTau) monomers and the pathological eTau aggregates. However, similarities or differences in neuronal transport mechanisms between Tau species remain elusive. METHOD Monomers, oligomers, and fibrils of recombinant 2N4R Tau were produced and characterized by biochemical and biophysical methods. A neuronal eTau uptake and accumulation assay was developed for human induced pluripotent stem cell-derived neurons (iPSCNs) and Lund human mesencephalic cells (LUHMES)-derived neurons. Mechanisms of uptake and cellular accumulation of eTau species were studied by using small molecule inhibitors of endocytic mechanisms and siRNAs targeting Tau uptake mediators. RESULTS Extracellular Tau aggregates accumulated more than monomers in human neurons, mainly due to the higher efficiency of small fibrillar and soluble oligomeric aggregates in intraneuronal accumulation. A competition assay revealed a distinction in the neuronal accumulation between physiological eTau Monomers and pathology-relevant aggregates, suggesting differential transport mechanisms. Blocking heparan sulfate proteoglycans (HSPGs) with heparin only inhibited the accumulation of eTau aggregates, whereas monomers' uptake remained unaltered. At the molecular level, the downregulation of genes involved in HSPG synthesis exclusively blocked neuronal accumulation of eTau aggregates but not monomers, suggesting its role in the transport of pathologic Tau. Moreover, the knockdown of LRP1, as a receptor of Tau, mainly reduced the accumulation of monomeric form, confirming its involvement in Tau's physiological transport. CONCLUSION These data propose that despite the similarity in the cellular mechanism, the uptake and accumulation of eTau Monomers and aggregates in human neurons are regulated by different molecular mediators. Thus, they address the possibility of targeting the pathological spreading of Tau aggregates without disturbing the probable physiological or non-pathogenic transport of Tau Monomers.
Collapse
Affiliation(s)
- Amir T Marvian
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Tabea Strauss
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Qilin Tang
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
| | - Benjamin J Tuck
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Sophie Keeling
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel Rüdiger
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Negar Mirzazadeh Dizaji
- Faculty for Chemistry and Pharmacy, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Hossein Mohammad-Beigi
- Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kgs., Lyngby, Denmark
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - Brigitte Nuscher
- Division of Metabolic Biochemistry, Biomedical Center (BMC), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Pijush Chakraborty
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Centre (iNANO), Aarhus University, 8000, Aarhus C, Denmark
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Cambridge, UK
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Thomas Köglsperger
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany
- Department of Translational Brain Research, DZNE-German Center for Neurodegenerative Diseases, 81377, Munich, Germany
| | - Stefan Zahler
- Department of Pharmacy, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Markus Zweckstetter
- Department for NMR-based Structural Biology, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Gӧttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Gӧttingen, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Wolfgang Wurst
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences, Technical University Munich, Freising, Germany
| | - Sigrid Schwarz
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany
- Haag, Geriatric Clinic Haag, Oberbayern, Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (LMU), Klinikum, Germany.
- Department of Neurology, University Hospital, Ludwig-Maximilians-Universität (LMU), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
- Department of Neurology, Hanover Medical School, Hanover, Germany.
- Center for Systems Neuroscience, Hanover, Germany.
| |
Collapse
|
3
|
Falkon KF, Danford L, Gutierrez Kuri E, Esquinca-Moreno P, Peña Señeriz YL, Smith S, Wickline JL, Louwrier A, McPhail JA, Sayre NL, Hopp SC. Microglia internalize tau monomers and fibrils using distinct receptors but similar mechanisms. Alzheimers Dement 2024. [PMID: 39713861 DOI: 10.1002/alz.14418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/27/2024] [Accepted: 10/25/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) and other tauopathies are characterized by intracellular aggregates of microtubule-associated protein tau that are actively released and promote proteopathic spread. Microglia engulf pathological proteins, but how they endocytose tau is unknown. METHODS We measured endocytosis of different tau species by microglia after pharmacological modulation of macropinocytosis or clathrin-mediated endocytosis (CME) or antagonism/genetic depletion of known tau receptors heparan-sulfate proteoglycans (HSPGs) and low-density lipoprotein receptor-related protein 1 (LRP1). RESULTS Dynamin inhibition decreased microglial endocytosis of all tested tau species. Meanwhile, HSPG antagonism blocked only fibril uptake, and LRP1 antagonism or genetic depletion inconsistently inhibited the endocytosis of fibrils and monomers. Cre recombinase robustly enhanced tau uptake with partial selectivity for fibrils. DISCUSSION These data show that microglia take up both tau monomers and aggregates via a dynamin-dependent form of endocytosis (eg, CME) but may differ in using HSPGs for entry depending on species. HIGHLIGHTS Microglial endocytosis of tau monomers and fibrils is dynamin-dependent. HSPG antagonism blocks microglial uptake of tau fibrils but not monomers. LRP1 antagonism or knockdown inconsistently inhibits tau uptake. TAT-Cre stimulates semi-selective uptake of fibrils over monomers.
Collapse
Affiliation(s)
- Kristian F Falkon
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Liliana Danford
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Eduardo Gutierrez Kuri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Paulina Esquinca-Moreno
- Voelcker Biomedical Research Academy, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Yaren L Peña Señeriz
- Graduate School of Biomedical Sciences, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Sabrina Smith
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Jessica L Wickline
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Ariel Louwrier
- Research and Development Department, StressMarq Biosciences, Victoria, British Columbia, Canada
| | - Jacob A McPhail
- Research and Development Department, StressMarq Biosciences, Victoria, British Columbia, Canada
- Institute for Neurodegenerative Diseases, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California, USA
| | - Naomi L Sayre
- Research Division, South Texas Veteran's Health Care System, San Antonio, Texas, USA
- Department of Neurosurgery, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| |
Collapse
|
4
|
Borràs C, Canyelles M, Santos D, Rotllan N, Núñez E, Vázquez J, Maspoch D, Cano-Sarabia M, Carmona-Iragui M, Sirisi S, Lleó A, Fortea J, Alcolea D, Blanco-Vaca F, Escolà-Gil JC, Tondo M. Impaired Cerebrospinal Fluid Lipoprotein-Mediated Cholesterol Delivery to Neurons in Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-5682870. [PMID: 39764088 PMCID: PMC11703344 DOI: 10.21203/rs.3.rs-5682870/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
In the central nervous system, apolipoprotein (APO) E-containing high-density lipoprotein (HDL)-like particles mediate the transport of glial-derived cholesterol to neurons, which is essential for neuronal membrane remodeling and maintenance of the myelin sheath. Despite this, the role of HDL-like cholesterol trafficking on Alzheimer's disease (AD) pathogenesis remains poorly understood. We aimed to examine cholesterol transport via HDL-like particles in cerebrospinal fluid (CSF) of AD patients compared to control individuals. Additionally, we explored the ability of reconstituted HDL containing different APOE isoforms to regulate cholesterol transport. We evaluated the capacity of CSF HDL-like particles to facilitate radiolabeled unesterified cholesterol efflux from A172 human glioblastoma astrocytes and to deliver cholesterol to SH-SY5Y human neuronal cells. The HDL-like proteome in the AD and control groups was analyzed by liquid chromatography-mass spectrometry (LC-MS/MS). Reconstituted HDL nanoparticles were prepared by combining phospholipids and cholesterol with human APOE3 or APOE4, followed by radiolabeling with unesterified cholesterol. Our results showed that cholesterol efflux from astrocytes to CSF were similar between AD patients and controls, both under baseline conditions and after activation of ATP-binding cassette transporters A1 and G1. However, CSF HDL-like particle-mediated neuronal cholesterol uptake was significantly reduced in the AD group. LC-MS/MS analysis identified 775 proteins associated with HDL-like particles in both groups, with no major alterations in proteins linked to cholesterol metabolism. However, 27 proteins involved in non-cholesterol-related processes were differentially expressed. Notably, synthetic reconstituted HDL particles containing APOE4 exhibited reduced capacity to deliver cholesterol to neurons compared to those with APOE3. These findings indicate that CSF HDL-like particles from patients with AD demonstrate impaired cholesterol delivery to neurons. Our study highlights APOE4 as a critical contributor to abnormal neuronal cholesterol uptake in AD pathophysiology.
Collapse
Affiliation(s)
| | | | - David Santos
- Centro de Investigación Biomédica en Red Diabetes y Enfermedades Metabólicas Asociadas
| | | | - Estefanía Núñez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Jesús Vázquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III
| | - Daniel Maspoch
- Catalan Institute of Nanoscience and Nanotechnology (ICN2)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Nedelkov D, Tsokolas ZE, Rodrigues MS, Sible I, Han SD, Kerman BE, Renteln M, Mack WJ, Pascoal TA, Yassine HN. Increased cerebrospinal fluid and plasma apoE glycosylation is associated with reduced levels of Alzheimer's disease biomarkers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629619. [PMID: 39763949 PMCID: PMC11702616 DOI: 10.1101/2024.12.20.629619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
The apolipoprotein E ( APOE ) ε4 allele is the strongest genetic risk factor for Alzheimer's disease (AD). ApoE is glycosylated with an O-linked Core-1 sialylated glycan at several sites, yet the impact and function of this glycosylation on AD biomarkers remains unclear. We examined apoE glycosylation in a cohort of cerebrospinal fluid (CSF, n=181) and plasma (n= 178) samples from the Alzheimer's Disease Neuroimaging Initiative (ADNI) stratified into 4 groups: cognitively normal (CN), Mild Cognitive Impairment (MCI), progressors and non-progressors based on delayed word recall performance over 4 years. We observed decreasing glycosylation from apoE2 > apoE3 > apoE4 in CSF, and in plasma (apoE3 > apoE4). ApoE glycosylation was reduced in the MCI compared with CN groups, and in progressors compared to non-progressors. In CSF, higher apoE glycosylation associated cross-sectionally with lower total tau (t-tau), p-tau181, and with higher Aβ 1-42 . Similar associations of apoE glycosylation with higher Aβ 1-42 were observed in plasma. In CSF, greater apoE4 glycosylation was associated with lower t-tau and p-tau181. Over a 6-year period, higher baseline levels of CSF apoE glycosylation predicted lower rates of increase in CSF t-tau and p-tau181 and lower rates of decrease in CSF Aβ 1-42 . These results indicate strong associations of apoE glycosylation with biomarkers of AD pathology independent of apoE genotype, warranting a deeper understanding of the functional role of apoE glycosylation on AD tau pathology.
Collapse
|
6
|
Kadamangudi S, Marcatti M, Zhang WR, Fracassi A, Kayed R, Limon A, Taglialatela G. Amyloid-β oligomers increase the binding and internalization of tau oligomers in human synapses. Acta Neuropathol 2024; 149:2. [PMID: 39688618 DOI: 10.1007/s00401-024-02839-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024]
Abstract
In Alzheimer's disease (AD), the propagation and spreading of CNS tau pathology closely correlates with cognitive decline, positioning tau as an attractive therapeutic target. Amyloid beta (Aβ) has been strongly implicated in driving tau spread, whereas primary tauopathies such as primary age-related tauopathy (PART)-which lack Aβ pathology-exhibit limited tau spread and minimal-to-no cognitive decline. Emerging evidence converges on a trans-synaptic mechanism of tau spread, facilitated by the transfer of misfolded tau aggregates (e.g. soluble oligomers). However, it is unclear whether Aβ oligomers modulate the binding and internalization of tau oligomers in human synapses. Our translationally focused paradigms utilize post-mortem brain specimens from Control, PART, and AD patients. Synaptosomes isolated from the temporal cortex of all three groups were incubated with preformed recombinant tauO (rtauO), ± preformed recombinant AβO (rAβO), and oligomer binding/internalization was quantified via flow cytometry following proteinase K (PK) digestion of surface-bound oligomers. TauO-synapse interactions were visualized using EM immunogold. Brain-derived tau oligomers (BDTO) from AD and PART PBS-soluble hippocampal fractions were co-immunoprecipitated and analyzed via mass spectrometry to compare synaptic tauO interactomes in primary and secondary tauopathies, thereby inferring the role of Aβ. AD synaptosomes, enriched in endogenous Aβ pathology, exhibited increased rtauO internalization compared to PART synaptosomes. This observation was mirrored in Control synaptosomes, where recombinant rAβO significantly increased rtauO binding and internalization. PK pre-treatment abolished this effect, implicating synaptic membrane proteins in AβO-mediated tauO internalization. While both PART and AD BDTO were broadly enriched in synaptic proteins, AD BDTO exhibited differential enrichment of endocytic proteins across pre- and post-synaptic compartments, whereas PART BDTO showed no significant synaptic enrichment. This study demonstrates that Aβ oligomers enhance tau oligomer binding and drive its internalization through synaptic membrane proteins. These findings offer novel mechanistic insights underlying pathological tau spreading directly within human synapses and emphasize the therapeutic potential of targeting Aβ-tau interactions.
Collapse
Affiliation(s)
- Shrinath Kadamangudi
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Michela Marcatti
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Wen-Ru Zhang
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Anna Fracassi
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Rakez Kayed
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Agenor Limon
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Giulio Taglialatela
- Department of Neurology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA.
| |
Collapse
|
7
|
Ning Y, Zhang Y, Jiang T, Feng J, Zhan J, Ou C, Wang L. LRP1-mediated p-tau propagation contributes to cognitive impairment after chronic neuropathic pain in rats. Neurosci Res 2024:S0168-0102(24)00155-X. [PMID: 39674403 DOI: 10.1016/j.neures.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Trigeminal neuralgia (TN) is a prevalent chronic neuropathic pain syndrome characterized by severe pain, often accompanied by cognitive dysfunction and cerebral degeneration. However, its mechanisms remain poorly understood. Hyperphosphorylation of tau protein (p-tau) is often seen in neurodegenerative disorders such as Alzheimer's disease (AD). LRP1 expression on brain neurons and microglial cells is believed to facilitate the propagation of p-tau. We established a TN rat model via infraorbital nerve chronic constrictive injury (ION-CCI). Once the model was established, we investigated the association between p-tau and cognitive impairment in TN rats by evaluating behavioral and degenerative markers. During the initial phase, we noted an increase in p-tau level in the prefrontal cortex and hippocampal tissues of TN rats. The accompanied impaired learning and memory abilities suggested cognitive dysfunction. Blocking p-tau synthesis by orally administering a protein phosphatase and by injecting adenoviral vectors targeting LRP1 into the lateral ventricle of rats ameliorated cognitive impairment. This suggests that cognitive decline in TN rats is linked to elevated p-tau levels. Our findings show that LRP1-mediated p-tau propagation may drive cognitive impairment associated with neuropathic pain in TN rats.
Collapse
Affiliation(s)
- Youzhi Ning
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yue Zhang
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Tao Jiang
- Department of Anesthesiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jian Zhan
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cehua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| | - Lu Wang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
8
|
Pashaei S, Shabani S, Mohammadi S, Morozova-Roche LA, Salari N, Rahimi Z, Khodarahmi R. Differential Expression of Neurodegeneration-Related Genes in SH-SY5Y Neuroblastoma Cells Under the Influence of Cyclophilin A: Could the Enzyme be a Likely Trigger and Therapeutic Target for Alzheimer's Disease? Neurochem Res 2024; 50:47. [PMID: 39636462 DOI: 10.1007/s11064-024-04253-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/08/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024]
Abstract
The function and mechanism of Cyclophilin A (CypA) in modulating gene expression associated with Alzheimer's disease (AD) remain unclear. This multifunctional protein is found to be elevated in the cerebrospinal fluid (CSF) of individuals at risk for AD. The cytotoxic effects of CypA, including both wild-type and the mutant R55A, were assessed using the MTT assay. Prior to this evaluation, the purified recombinant protein was validated through enzymatic activity assays and western blot analysis. Following treatment with CypA and transient transfection using the CypA construct, real-time PCR (qRT-PCR) and western blotting were conducted to analyze the expression of factors involved in various signaling pathways, with an emphasis on inflammation, cell death, and intercellular communication. The findings indicate that CypA has a significant impact on the gene expression of factors associated with inflammation and the progression of AD in SH-SY5Y cells. It can be concluded that CypA is capable of regulating gene expression in SH-SY5Y cells, either in a manner dependent on or independent of its enzymatic activity. Additionally, the influence of this multifunctional protein on gene expression is contingent upon the specific site of action, as well as the dosage and duration of exposure to the cells.
Collapse
Affiliation(s)
- Somayeh Pashaei
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sasan Shabani
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Mohammadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Nader Salari
- Department of Biostatics, School of Public Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Khodarahmi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
9
|
Parra Bravo C, Naguib SA, Gan L. Cellular and pathological functions of tau. Nat Rev Mol Cell Biol 2024; 25:845-864. [PMID: 39014245 DOI: 10.1038/s41580-024-00753-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 07/18/2024]
Abstract
Tau protein is involved in various cellular processes, including having a canonical role in binding and stabilization of microtubules in neurons. Tauopathies are neurodegenerative diseases marked by the abnormal accumulation of tau protein aggregates in neurons, as seen, for example, in conditions such as frontotemporal dementia and Alzheimer disease. Mutations in tau coding regions or that disrupt tau mRNA splicing, tau post-translational modifications and cellular stress factors (such as oxidative stress and inflammation) increase the tendency of tau to aggregate and interfere with its clearance. Pathological tau is strongly implicated in the progression of neurodegenerative diseases, and the propagation of tau aggregates is associated with disease severity. Recent technological advancements, including cryo-electron microscopy and disease models derived from human induced pluripotent stem cells, have increased our understanding of tau-related pathology in neurodegenerative conditions. Substantial progress has been made in deciphering tau aggregate structures and the molecular mechanisms that underlie protein aggregation and toxicity. In this Review, we discuss recent insights into the diverse cellular functions of tau and the pathology of tau inclusions and explore the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Celeste Parra Bravo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Sarah A Naguib
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
- Neuroscience Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| |
Collapse
|
10
|
Zhou F, Zhao Y, Sun Y, Chen W. Molecular Insights into Tau Pathology and its Therapeutic Strategies in Alzheimer's Disease. J Integr Neurosci 2024; 23:197. [PMID: 39613463 DOI: 10.31083/j.jin2311197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 12/01/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. The two major hallmarks of this disease are extracellular amyloid plaques and intracellular neurofibrillary tangles in the brain, accompanied by loss of neurons and synapses. The plaques and tangles mainly consist of amyloid-β (Aβ) and tau protein, respectively. Most of the therapeutic strategies for AD to date have focused on Aβ. However, there is still no effective therapy available. In recent years, the clinical therapeutic failure of targeting Aβ pathology has resulted in increased interest towards tau-based therapeutics. In the current review, we focus on the research progress regarding the pathological mechanisms of tau protein in this disease and discuss tau-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Futao Zhou
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yushi Zhao
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Yangyan Sun
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| | - Wanjiao Chen
- School of Basic Medicine, Gannan Medical University, 341000 Ganzhou, Jiangxi, China
| |
Collapse
|
11
|
Tyagi M, Chadha R, de Hoog E, Sullivan KR, Walker AC, Northrop A, Fabian B, Fuxreiter M, Hyman BT, Shepherd JD. Arc mediates intercellular tau transmission via extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619703. [PMID: 39484489 PMCID: PMC11526995 DOI: 10.1101/2024.10.22.619703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Intracellular neurofibrillary tangles that consist of misfolded tau protein1 cause neurodegeneration in Alzheimer's disease (AD) and frontotemporal dementia (FTD). Tau pathology spreads cell-to-cell2 but the exact mechanisms of tau release and intercellular transmission remain poorly defined. Tau is released from neurons as free protein or in extracellular vesicles (EVs)3-5 but the role of these different release mechanisms in intercellular tau transmission is unclear. Here, we show that the neuronal gene Arc is critical for packaging tau into EVs. Brain EVs purified from human tau (hTau) transgenic rTg4510 mice (rTgWT) contain high levels of hTau that are capable of seeding tau pathology. In contrast, EVs purified from rTgWT crossed with Arc knock-out mice (rTgArc KO) have significantly less hTau and cannot seed tau aggregation. Arc facilitates the release of hTau in EVs produced via the I-BAR protein IRSp53, but not free tau. Arc protein directly binds hTau to form a fuzzy complex that we identified in both mouse and human brain tissue. We find that pathological intracellular hTau accumulates in neurons in rTgArc KO mice, which correlates with accelerated neuron loss in the hippocampus. Finally, we find that intercellular tau transmission is significantly abrogated in Arc KO mice. We conclude that Arc-dependent release of tau in EVs plays a significant role in intracellular tau elimination and intercellular tau transmission.
Collapse
Affiliation(s)
- Mitali Tyagi
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Radhika Chadha
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Eric de Hoog
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | | | - Alicia C. Walker
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Ava Northrop
- Department of Neurobiology, University of Utah, Salt Lake City, USA
| | - Balazs Fabian
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Germany
| | - Monika Fuxreiter
- Department of Biomedical Sciences University of Padova, Padova, Italy
| | - Bradley T. Hyman
- Department of Neurology, Massachusetts Alzheimer’s Disease Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | | |
Collapse
|
12
|
Qiao L, Du X, Wang H, Wang Z, Gao S, Zhao CQ. Research Progress on the Strategies for Crossing the Blood-Brain Barrier. Mol Pharm 2024; 21:4786-4803. [PMID: 39231367 DOI: 10.1021/acs.molpharmaceut.4c00447] [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] [Indexed: 09/06/2024]
Abstract
Recently, the incidence of brain diseases, such as central nervous system degenerative diseases, brain tumors, and cerebrovascular diseases, has increased. However, the blood-brain barrier (BBB) limits the effective delivery of drugs to brain disease areas. Therefore, the mainstream direction of new drug development for these diseases is to engineer drugs that can better cross the BBB to exert their effects in the brain. This paper reviews the research progress and application of the main trans-BBB drug delivery strategies (receptor/transporter-mediated BBB crossing, focused ultrasound to open the BBB, adenosine agonist reversible opening of the BBB, aromatic resuscitation, transnasal administration, cell-mediated trans-BBB crossing, and viral vector system-mediated brain drug delivery). Meanwhile, the potential applications, advantages, and disadvantages of these strategies for crossing the BBB are analyzed. Finally, the future development prospects of strategies for crossing the BBB are also discussed. These strategies have potential value for treating brain diseases.
Collapse
Affiliation(s)
- Li Qiao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Xiuwei Du
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Hua Wang
- College of Intelligence and Information Engineering, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Zhiyi Wang
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Shijie Gao
- Experimental Centre, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| | - Chun-Qin Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, P. R. China
| |
Collapse
|
13
|
Min JH, Sarlus H, Harris RA. MAD-microbial (origin of) Alzheimer's disease hypothesis: from infection and the antimicrobial response to disruption of key copper-based systems. Front Neurosci 2024; 18:1467333. [PMID: 39416952 PMCID: PMC11480022 DOI: 10.3389/fnins.2024.1467333] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Microbes have been suspected to cause Alzheimer's disease since at least 1908, but this has generally remained unpopular in comparison to the amyloid hypothesis and the dominance of Aβ and Tau. However, evidence has been accumulating to suggest that these earlier theories are but a manifestation of a common cause that can trigger and interact with all the major molecular players recognized in AD. Aβ, Tau and ApoE, in particular appear to be molecules with normal homeostatic functions but also with alternative antimicrobial functions. Their alternative functions confer the non-immune specialized neuron with some innate intracellular defenses that appear to be re-appropriated from their normal functions in times of need. Indeed, signs of infection of the neurons by biofilm-forming microbial colonies, in synergy with herpes viruses, are evident from the clinical and preclinical studies we discuss. Furthermore, we attempt to provide a mechanistic understanding of the AD landscape by discussing the antimicrobial effect of Aβ, Tau and ApoE and Lactoferrin in AD, and a possible mechanistic link with deficiency of vital copper-based systems. In particular, we focus on mitochondrial oxidative respiration via complex 4 and ceruloplasmin for iron homeostasis, and how this is similar and possibly central to neurodegenerative diseases in general. In the case of AD, we provide evidence for the microbial Alzheimer's disease (MAD) theory, namely that AD could in fact be caused by a long-term microbial exposure or even long-term infection of the neurons themselves that results in a costly prolonged antimicrobial response that disrupts copper-based systems that govern neurotransmission, iron homeostasis and respiration. Finally, we discuss potential treatment modalities based on this holistic understanding of AD that incorporates the many separate and seemingly conflicting theories. If the MAD theory is correct, then the reduction of microbial exposure through use of broad antimicrobial and anti-inflammatory treatments could potentially alleviate AD although this requires further clinical investigation.
Collapse
Affiliation(s)
- Jin-Hong Min
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital at Solna, Stockholm, Sweden
| | | | | |
Collapse
|
14
|
Chen Y, Holtzman DM. New insights into innate immunity in Alzheimer's disease: from APOE protective variants to therapies. Trends Immunol 2024; 45:768-782. [PMID: 39278789 DOI: 10.1016/j.it.2024.08.001] [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: 06/28/2024] [Revised: 08/16/2024] [Accepted: 08/21/2024] [Indexed: 09/18/2024]
Abstract
Recent discoveries of rare variants of human APOE may shed light on novel therapeutic strategies for Alzheimer's disease (AD). Here, we highlight the newly identified protective variant [APOE3 Christchurch (APOE3ch, R136S)] as an example. We summarize human AD and mouse amyloidosis and tauopathy studies from the past 5 years that have been associated with this R136S variant. We also propose a potential mechanism for how this point mutation might lead to protection against AD pathology, from the molecular level, to cells, to mouse models, and potentially, to humans. Lastly, we extend our discussion of the recent insights gained regarding different APOE variants to putative therapeutic approaches in AD.
Collapse
Affiliation(s)
- Yun Chen
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St Louis, St Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University in St Louis, St Louis, MO 63110, USA.
| |
Collapse
|
15
|
Liu E, Zhang Y, Wang JZ. Updates in Alzheimer's disease: from basic research to diagnosis and therapies. Transl Neurodegener 2024; 13:45. [PMID: 39232848 PMCID: PMC11373277 DOI: 10.1186/s40035-024-00432-x] [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: 03/12/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder, characterized pathologically by extracellular deposition of β-amyloid (Aβ) into senile plaques and intracellular accumulation of hyperphosphorylated tau (pTau) as neurofibrillary tangles. Clinically, AD patients show memory deterioration with varying cognitive dysfunctions. The exact molecular mechanisms underlying AD are still not fully understood, and there are no efficient drugs to stop or reverse the disease progression. In this review, we first provide an update on how the risk factors, including APOE variants, infections and inflammation, contribute to AD; how Aβ and tau become abnormally accumulated and how this accumulation plays a role in AD neurodegeneration. Then we summarize the commonly used experimental models, diagnostic and prediction strategies, and advances in periphery biomarkers from high-risk populations for AD. Finally, we introduce current status of development of disease-modifying drugs, including the newly officially approved Aβ vaccines, as well as novel and promising strategies to target the abnormal pTau. Together, this paper was aimed to update AD research progress from fundamental mechanisms to the clinical diagnosis and therapies.
Collapse
Affiliation(s)
- Enjie Liu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yao Zhang
- Department of Endocrine, Liyuan Hospital, Key Laboratory of Ministry of Education for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430077, China
| | - Jian-Zhi Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China.
| |
Collapse
|
16
|
Chen F, Zhao J, Meng F, He F, Ni J, Fu Y. The vascular contribution of apolipoprotein E to Alzheimer's disease. Brain 2024; 147:2946-2965. [PMID: 38748848 DOI: 10.1093/brain/awae156] [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: 12/24/2023] [Revised: 03/23/2024] [Accepted: 04/21/2024] [Indexed: 09/04/2024] Open
Abstract
Alzheimer's disease, the most prevalent form of dementia, imposes a substantial societal burden. The persistent inadequacy of disease-modifying drugs targeting amyloid plaques and neurofibrillary tangles suggests the contribution of alternative pathogenic mechanisms. A frequently overlooked aspect is cerebrovascular dysfunction, which may manifest early in the progression of Alzheimer's disease pathology. Mounting evidence underscores the pivotal role of the apolipoprotein E gene, particularly the apolipoprotein ε4 allele as the strongest genetic risk factor for late-onset Alzheimer's disease, in the cerebrovascular pathology associated with Alzheimer's disease. In this review, we examine the evidence elucidating the cerebrovascular impact of both central and peripheral apolipoprotein E on the pathogenesis of Alzheimer's disease. We present a novel three-hit hypothesis, outlining potential mechanisms that shed light on the intricate relationship among different pathogenic events. Finally, we discuss prospective therapeutics targeting the cerebrovascular pathology associated with apolipoprotein E and explore their implications for future research endeavours.
Collapse
Affiliation(s)
- Feng Chen
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jing Zhao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou 215123, China
| | - Fanxia Meng
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Fangping He
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jie Ni
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuan Fu
- Department of Neurology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
17
|
Matsuo K, Nshihara H. Rebuilding insight into the pathophysiology of Alzheimer's disease through new blood-brain barrier models. Neural Regen Res 2024; 19:1954-1960. [PMID: 38227521 DOI: 10.4103/1673-5374.390978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/01/2023] [Indexed: 01/17/2024] Open
Abstract
The blood-brain barrier is a unique function of the microvasculature in the brain parenchyma that maintains homeostasis in the central nervous system. Blood-brain barrier breakdown is a common pathology in various neurological diseases, such as Alzheimer's disease, stroke, multiple sclerosis, and Parkinson's disease. Traditionally, it has been considered a consequence of neuroinflammation or neurodegeneration, but recent advanced imaging techniques and detailed studies in animal models show that blood-brain barrier breakdown occurs early in the disease process and may precede neuronal loss. Thus, the blood-brain barrier is attractive as a potential therapeutic target for neurological diseases that lack effective therapeutics. To elucidate the molecular mechanism underlying blood-brain barrier breakdown and translate them into therapeutic strategies for neurological diseases, there is a growing demand for experimental models of human origin that allow for functional assessments. Recently, several human induced pluripotent stem cell-derived blood-brain barrier models have been established and various in vitro blood-brain barrier models using microdevices have been proposed. Especially in the Alzheimer's disease field, the human evidence for blood-brain barrier dysfunction has been demonstrated and human induced pluripotent stem cell-derived blood-brain barrier models have suggested the putative molecular mechanisms of pathological blood-brain barrier. In this review, we summarize recent evidence of blood-brain barrier dysfunction in Alzheimer's disease from pathological analyses, imaging studies, animal models, and stem cell sources. Additionally, we discuss the potential future directions for blood-brain barrier research.
Collapse
Affiliation(s)
- Kinya Matsuo
- Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Hideaki Nshihara
- Department of Neurotherapeutics, Yamaguchi University, Ube, Japan
| |
Collapse
|
18
|
Jackson RJ, Hyman BT, Serrano-Pozo A. Multifaceted roles of APOE in Alzheimer disease. Nat Rev Neurol 2024; 20:457-474. [PMID: 38906999 DOI: 10.1038/s41582-024-00988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
For the past three decades, apolipoprotein E (APOE) has been known as the single greatest genetic modulator of sporadic Alzheimer disease (AD) risk, influencing both the average age of onset and the lifetime risk of developing AD. The APOEε4 allele significantly increases AD risk, whereas the ε2 allele is protective relative to the most common ε3 allele. However, large differences in effect size exist across ethnoracial groups that are likely to depend on both global genetic ancestry and local genetic ancestry, as well as gene-environment interactions. Although early studies linked APOE to amyloid-β - one of the two culprit aggregation-prone proteins that define AD - in the past decade, mounting work has associated APOE with other neurodegenerative proteinopathies and broader ageing-related brain changes, such as neuroinflammation, energy metabolism failure, loss of myelin integrity and increased blood-brain barrier permeability, with potential implications for longevity and resilience to pathological protein aggregates. Novel mouse models and other technological advances have also enabled a number of therapeutic approaches aimed at either attenuating the APOEε4-linked increased AD risk or enhancing the APOEε2-linked AD protection. This Review summarizes this progress and highlights areas for future research towards the development of APOE-directed therapeutics.
Collapse
Affiliation(s)
- Rosemary J Jackson
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| | - Alberto Serrano-Pozo
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Massachusetts Alzheimer's Disease Research Center, Charlestown, MA, USA.
| |
Collapse
|
19
|
Yoo CJ, Choi Y, Bok E, Lin Y, Cheon M, Lee YH, Kim J. Complement receptor 4 mediates the clearance of extracellular tau fibrils by microglia. FEBS J 2024; 291:3499-3520. [PMID: 38715400 DOI: 10.1111/febs.17150] [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: 11/09/2023] [Revised: 03/08/2024] [Accepted: 04/18/2024] [Indexed: 08/03/2024]
Abstract
Tauopathies exhibit a characteristic accumulation of misfolded tau aggregates in the brain. Tau pathology shows disease-specific spatiotemporal propagation through intercellular transmission, which is closely correlated with the progression of clinical manifestations. Therefore, identifying molecular mechanisms that prevent tau propagation is critical for developing therapeutic strategies for tauopathies. The various innate immune receptors, such as complement receptor 3 (CR3) and complement receptor 4 (CR4), have been reported to play a critical role in the clearance of various extracellular toxic molecules by microglia. However, their role in tau clearance has not been studied yet. In the present study, we investigated the role of CR3 and CR4 in regulating extracellular tau clearance. We found that CR4 selectively binds to tau fibrils but not to tau monomers, whereas CR3 does not bind to either of them. Inhibiting CR4, but not CR3, significantly reduces the uptake of tau fibrils by BV2 cells and primary microglia. By contrast, inhibiting CR4 has no effect on the uptake of tau monomers by BV2 cells. Furthermore, inhibiting CR4 suppresses the clearance of extracellular tau fibrils, leading to more seed-competent tau fibrils remaining in the extracellular space relative to control samples. We also provide evidence that the expression of CR4 is upregulated in the brains of human Alzheimer's disease patients and the PS19 mouse model of tauopathy. Taken together, our data strongly support that CR4 is a previously undescribed receptor for the clearance of tau fibrils in microglia and may represent a novel therapeutic target for tauopathy.
Collapse
Affiliation(s)
- Chang Jae Yoo
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), South Korea
| | - Youngtae Choi
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Eugene Bok
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Yuxi Lin
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang, South Korea
| | - Mookyung Cheon
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang, South Korea
- Bio-Analytical Science, University of Science and Technology, Daejeon, South Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon, South Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, South Korea
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Jaekwang Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu, South Korea
| |
Collapse
|
20
|
Yamamoto K, Scilabra SD, Bonelli S, Jensen A, Scavenius C, Enghild JJ, Strickland DK. Novel insights into the multifaceted and tissue-specific roles of the endocytic receptor LRP1. J Biol Chem 2024; 300:107521. [PMID: 38950861 PMCID: PMC11325810 DOI: 10.1016/j.jbc.2024.107521] [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: 03/13/2024] [Revised: 06/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
Receptor-mediated endocytosis provides a mechanism for the selective uptake of specific molecules thereby controlling the composition of the extracellular environment and biological processes. The low-density lipoprotein receptor-related protein 1 (LRP1) is a widely expressed endocytic receptor that regulates cellular events by modulating the levels of numerous extracellular molecules via rapid endocytic removal. LRP1 also participates in signalling pathways through this modulation as well as in the interaction with membrane receptors and cytoplasmic adaptor proteins. LRP1 SNPs are associated with several diseases and conditions such as migraines, aortic aneurysms, cardiopulmonary dysfunction, corneal clouding, and bone dysmorphology and mineral density. Studies using Lrp1 KO mice revealed a critical, nonredundant and tissue-specific role of LRP1 in regulating various physiological events. However, exactly how LRP1 functions to regulate so many distinct and specific processes is still not fully clear. Our recent proteomics studies have identified more than 300 secreted proteins that either directly interact with LRP1 or are modulated by LRP1 in various tissues. This review will highlight the remarkable ability of this receptor to regulate secreted molecules in a tissue-specific manner and discuss potential mechanisms underpinning such specificity. Uncovering the depth of these "hidden" specific interactions modulated by LRP1 will provide novel insights into a dynamic and complex extracellular environment that is involved in diverse biological and pathological processes.
Collapse
Affiliation(s)
- Kazuhiro Yamamoto
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Simone D Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy
| | - Simone Bonelli
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT, Palermo, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Anders Jensen
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Carsten Scavenius
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Dudley K Strickland
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Petralla S, Panayotova M, Franchina E, Fricker G, Puris E. Low-Density Lipoprotein Receptor-Related Protein 1 as a Potential Therapeutic Target in Alzheimer's Disease. Pharmaceutics 2024; 16:948. [PMID: 39065645 PMCID: PMC11279518 DOI: 10.3390/pharmaceutics16070948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease impacting the lives of millions of people worldwide. The formation of amyloid β (Aβ) plagues in the brain is the main pathological hallmark of AD. The Aβ deposits are formed due to the imbalance between the production and Aβ clearance in the brain and across the blood-brain barrier (BBB). In this respect, low-density lipoprotein receptor-related protein 1 (LRP1) plays a significant role by mediating both brain Aβ production and clearance. Due to its important role in AD pathogenesis, LRP1 is considered an attractive drug target for AD therapies. In the present review, we summarize the current knowledge about the role of LRP1 in AD pathogenesis as well as recent findings on changes in LRP1 expression and function in AD. Finally, we discuss the advances in utilizing LRP1 as a drug target for AD treatments as well as future perspectives on LRP1 research.
Collapse
Affiliation(s)
| | | | | | | | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany; (S.P.); (M.P.); (E.F.); (G.F.)
| |
Collapse
|
22
|
Glynn C, Rodriguez JA, Hyman BT. The structural line between prion and "prion-like": Insights from prion protein and tau. Curr Opin Neurobiol 2024; 86:102857. [PMID: 38489865 PMCID: PMC11162956 DOI: 10.1016/j.conb.2024.102857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/15/2024] [Accepted: 02/26/2024] [Indexed: 03/17/2024]
Abstract
The concept of 'prion-like' behavior has emerged in the study of diseases involving protein misfolding where fibrillar structures, called amyloids, self-propagate and induce disease in a fashion similar to prions. From a biological standpoint, in order to be considered 'prion-like,' a protein must traverse cells and tissues and further propagate via a templated conformational change. Since 2017, cryo-electron microscopy structures from patient-derived 'prion-like' amyloids, in particular tau, have been presented and revealed structural similarities shared across amyloids. Since 2021, cryo-EM structures from prions of known infectivity have been added to the ex vivo amyloid structure family. In this review, we discuss current proposals for the 'prion-like' mechanisms of spread for tau and prion protein as well as discuss different influencers on structures of aggregates from tauopathies and prion diseases. Lastly, we discuss some of the current hypotheses for what may distinguish structures that are 'prion-like' from transmissible prion structures.
Collapse
Affiliation(s)
- Calina Glynn
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA
| | - Jose A Rodriguez
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics, STROBE, NSF Science and Technology Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Cambridge, MA, USA.
| |
Collapse
|
23
|
Cooper JM, Lathuiliere A, Su EJ, Song Y, Torrente D, Jo Y, Weinrich N, Sales JD, Migliorini M, Sisson TH, Lawrence DA, Hyman BT, Strickland DK. SORL1 is a receptor for tau that promotes tau seeding. J Biol Chem 2024; 300:107313. [PMID: 38657864 PMCID: PMC11145553 DOI: 10.1016/j.jbc.2024.107313] [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: 01/05/2024] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.
Collapse
Affiliation(s)
- Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aurelien Lathuiliere
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA; Department of Rehabilitation and Geriatrics, Memory Center, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Enming J Su
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yuyu Song
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Daniel Torrente
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York, USA
| | - Youhwa Jo
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Nicholas Weinrich
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Diaz Sales
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mary Migliorini
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas H Sisson
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
24
|
Rabanal-Ruiz Y, Pedrero-Prieto CM, Sanchez-Rodriguez L, Flores-Cuadrado A, Saiz-Sanchez D, Frontinan-Rubio J, Ubeda-Banon I, Duran Prado M, Martinez-Marcos A, Peinado JR. Differential accumulation of human β-amyloid and tau from enriched extracts in neuronal and endothelial cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167204. [PMID: 38679217 DOI: 10.1016/j.bbadis.2024.167204] [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: 10/30/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
While Aβ and Tau cellular distribution has been largely studied, the comparative internalization and subcellular accumulation of Tau and Aβ isolated from human brain extracts in endothelial and neuronal cells has not yet been unveiled. We have previously demonstrated that controlled enrichment of Aβ from human brain extracts constitutes a valuable tool to monitor cellular internalization in vitro and in vivo. Herein, we establish an alternative method to strongly enrich Aβ and Tau aggregates from human AD brains, which has allowed us to study and compare the cellular internalization, distribution and toxicity of both proteins within brain barrier endothelial (bEnd.3) and neuronal (Neuro2A) cells. Our findings demonstrate the suitability of human enriched brain extracts to monitor the intracellular distribution of human Aβ and Tau, which, once internalized, show dissimilar sorting to different organelles within the cell and differential toxicity, exhibiting higher toxic effects on neuronal cells than on endothelial cells. While tau is strongly concentrated preferentially in mitochondria, Aβ is distributed predominantly within the endolysosomal system in endothelial cells, whereas the endoplasmic reticulum was its preferential location in neurons. Altogether, our findings display a picture of the interactions that human Aβ and Tau might establish in these cells.
Collapse
Affiliation(s)
- Y Rabanal-Ruiz
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - C M Pedrero-Prieto
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - L Sanchez-Rodriguez
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Flores-Cuadrado
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - D Saiz-Sanchez
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - J Frontinan-Rubio
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - I Ubeda-Banon
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - M Duran Prado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain
| | - A Martinez-Marcos
- Department of Medical Sciences, Ciudad Real Medical School, Neuroplasticity and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| | - Juan R Peinado
- Department of Medical Sciences, Ciudad Real Medical School, Oxidative Stress and Neurodegeneration Group, Regional Center for Biomedical Research, University of Castilla-La Mancha, Ciudad Real, Spain.
| |
Collapse
|
25
|
Balczon R, Lin MT, Voth S, Nelson AR, Schupp JC, Wagener BM, Pittet JF, Stevens T. Lung endothelium, tau, and amyloids in health and disease. Physiol Rev 2024; 104:533-587. [PMID: 37561137 PMCID: PMC11281824 DOI: 10.1152/physrev.00006.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 06/26/2023] [Accepted: 08/04/2023] [Indexed: 08/11/2023] Open
Abstract
Lung endothelia in the arteries, capillaries, and veins are heterogeneous in structure and function. Lung capillaries in particular represent a unique vascular niche, with a thin yet highly restrictive alveolar-capillary barrier that optimizes gas exchange. Capillary endothelium surveys the blood while simultaneously interpreting cues initiated within the alveolus and communicated via immediately adjacent type I and type II epithelial cells, fibroblasts, and pericytes. This cell-cell communication is necessary to coordinate the immune response to lower respiratory tract infection. Recent discoveries identify an important role for the microtubule-associated protein tau that is expressed in lung capillary endothelia in the host-pathogen interaction. This endothelial tau stabilizes microtubules necessary for barrier integrity, yet infection drives production of cytotoxic tau variants that are released into the airways and circulation, where they contribute to end-organ dysfunction. Similarly, beta-amyloid is produced during infection. Beta-amyloid has antimicrobial activity, but during infection it can acquire cytotoxic activity that is deleterious to the host. The production and function of these cytotoxic tau and amyloid variants are the subject of this review. Lung-derived cytotoxic tau and amyloid variants are a recently discovered mechanism of end-organ dysfunction, including neurocognitive dysfunction, during and in the aftermath of infection.
Collapse
Affiliation(s)
- Ron Balczon
- Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Mike T Lin
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Sarah Voth
- Department of Cell Biology and Physiology, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States
| | - Amy R Nelson
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| | - Jonas C Schupp
- Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yale University, New Haven, Connecticut, United States
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | - Brant M Wagener
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Jean-Francois Pittet
- Department of Anesthesiology and Perioperative Medicine, University of Alabama-Birmingham, Birmingham, Alabama, United States
| | - Troy Stevens
- Department of Physiology and Cell Biology, University of South Alabama, Mobile, Alabama, United States
- Department of Internal Medicine, University of South Alabama, Mobile, Alabama, United States
- Center for Lung Biology, University of South Alabama, Mobile, Alabama, United States
| |
Collapse
|
26
|
Eisenbaum M, Pearson A, Ortiz C, Koprivica M, Cembran A, Mullan M, Crawford F, Ojo J, Bachmeier C. Repetitive head trauma and apoE4 induce chronic cerebrovascular alterations that impair tau elimination from the brain. Exp Neurol 2024; 374:114702. [PMID: 38301863 PMCID: PMC10922621 DOI: 10.1016/j.expneurol.2024.114702] [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: 12/04/2023] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
Repetitive mild traumatic brain injuries (r-mTBI) sustained in the military or contact sports have been associated with the accumulation of extracellular tau in the brain, which may contribute to the pathogenesis of neurodegenerative tauopathies. The expression of the apolipoprotein E4 (apoE4) isoform has been associated with higher levels of tau in the brain, and worse clinical outcomes after r-mTBI, though the influence of apoE genotype on extracellular tau dynamics in the brain is poorly understood. We recently demonstrated that extracellular tau can be eliminated across blood-brain barrier (BBB), which is progressively impaired following r-mTBI. The current studies investigated the influence of repetitive mild TBI (r-mTBI) and apoE genotype on the elimination of extracellular solutes from the brain. Following intracortical injection of biotin-labeled tau into humanized apoE-Tr mice, the levels of exogenous tau residing in the brain of apoE4 mice were elevated compared to other isoforms, indicating reduced tau elimination. Additionally, we found exposure to r-mTBI increased tau residence in apoE2 mice, similar to our observations in E2FAD animals. Each of these findings may be the result of diminished tau efflux via LRP1 at the BBB, as LRP1 inhibition significantly reduced tau uptake in endothelial cells and decreased tau transit across an in vitro model of the BBB (basolateral-to-apical). Notably, we showed that injury and apoE status, (particularly apoE4) resulted in chronic alterations in BBB integrity, pericyte coverage, and AQP4 polarization. These aberrations coincided with an atypical reactive astrocytic gene signature indicative of diminished CSF-ISF exchange. Our work found that CSF movement was reduced in the chronic phase following r-mTBI (>18 months post injury) across all apoE genotypes. In summary, we show that apoE genotype strongly influences cerebrovascular homeostasis, which can lead to age-dependent deficiencies in the elimination of toxic proteins from the brain, like tau, particularly in the aftermath of head trauma.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Joseph Ojo
- The Roskamp Institute, Sarasota, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA; Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
27
|
Xu H, Qiu Q, Hu P, Hoxha K, Jang E, O'Reilly M, Kim C, He Z, Marotta N, Changolkar L, Zhang B, Wu H, Schellenberg GD, Kraemer B, Luk KC, Lee EB, Trojanowski JQ, Brunden KR, Lee VMY. MSUT2 regulates tau spreading via adenosinergic signaling mediated ASAP1 pathway in neurons. Acta Neuropathol 2024; 147:55. [PMID: 38472475 PMCID: PMC10933148 DOI: 10.1007/s00401-024-02703-3] [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: 12/06/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Inclusions comprised of microtubule-associated protein tau (tau) are implicated in a group of neurodegenerative diseases, collectively known as tauopathies, that include Alzheimer's disease (AD). The spreading of misfolded tau "seeds" along neuronal networks is thought to play a crucial role in the progression of tau pathology. Consequently, restricting the release or uptake of tau seeds may inhibit the spread of tau pathology and potentially halt the advancement of the disease. Previous studies have demonstrated that the Mammalian Suppressor of Tauopathy 2 (MSUT2), an RNA binding protein, modulates tau pathogenesis in a transgenic mouse model. In this study, we investigated the impact of MSUT2 on tau pathogenesis using tau seeding models. Our findings indicate that the loss of MSUT2 mitigates human tau seed-induced pathology in neuron cultures and mouse models. In addition, MSUT2 regulates many gene transcripts, including the Adenosine Receptor 1 (A1AR), and we show that down regulation or inhibition of A1AR modulates the activity of the "ArfGAP with SH3 Domain, Ankyrin Repeat, and PH Domain 1 protein" (ASAP1), thereby influencing the internalization of pathogenic tau seeds into neurons resulting in reduction of tau pathology.
Collapse
Affiliation(s)
- Hong Xu
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Qi Qiu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Ministry of Education), Shanghai Ocean University, Shanghai, China
| | - Kevt'her Hoxha
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elliot Jang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mia O'Reilly
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher Kim
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhuohao He
- Interdisciplinary Research Center On Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Nicholas Marotta
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lakshmi Changolkar
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bin Zhang
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, Penn Epigenetics Institute, Institute of Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerard D Schellenberg
- Department of Pathology and Laboratory Medicine, Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian Kraemer
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, 98108, USA
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, 98195, USA
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, 98104, USA
| | - Kelvin C Luk
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Edward B Lee
- Translational Neuropathology Research Laboratory, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kurt R Brunden
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Virginia M-Y Lee
- Department of Pathology and Laboratory Medicine, Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
28
|
Carreras Mascaro A, Grochowska MM, Boumeester V, Dits NFJ, Bilgiҫ EN, Breedveld GJ, Vergouw L, de Jong FJ, van Royen ME, Bonifati V, Mandemakers W. LRP10 and α-synuclein transmission in Lewy body diseases. Cell Mol Life Sci 2024; 81:75. [PMID: 38315424 PMCID: PMC10844361 DOI: 10.1007/s00018-024-05135-0] [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: 06/27/2023] [Revised: 01/13/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024]
Abstract
Autosomal dominant variants in LRP10 have been identified in patients with Lewy body diseases (LBDs), including Parkinson's disease (PD), Parkinson's disease-dementia (PDD), and dementia with Lewy bodies (DLB). Nevertheless, there is little mechanistic insight into the role of LRP10 in disease pathogenesis. In the brains of control individuals, LRP10 is typically expressed in non-neuronal cells like astrocytes and neurovasculature, but in idiopathic and genetic cases of PD, PDD, and DLB, it is also present in α-synuclein-positive neuronal Lewy bodies. These observations raise the questions of what leads to the accumulation of LRP10 in Lewy bodies and whether a possible interaction between LRP10 and α-synuclein plays a role in disease pathogenesis. Here, we demonstrate that wild-type LRP10 is secreted via extracellular vesicles (EVs) and can be internalised via clathrin-dependent endocytosis. Additionally, we show that LRP10 secretion is highly sensitive to autophagy inhibition, which induces the formation of atypical LRP10 vesicular structures in neurons in human-induced pluripotent stem cells (iPSC)-derived brain organoids. Furthermore, we show that LRP10 overexpression leads to a strong induction of monomeric α-synuclein secretion, together with time-dependent, stress-sensitive changes in intracellular α-synuclein levels. Interestingly, patient-derived astrocytes carrying the c.1424 + 5G > A LRP10 variant secrete aberrant high-molecular-weight species of LRP10 in EV-free media fractions. Finally, we show that this truncated patient-derived LRP10 protein species (LRP10splice) binds to wild-type LRP10, reduces LRP10 wild-type levels, and antagonises the effect of LRP10 on α-synuclein levels and distribution. Together, this work provides initial evidence for a possible functional role of LRP10 in LBDs by modulating intra- and extracellular α-synuclein levels, and pathogenic mechanisms linked to the disease-associated c.1424 + 5G > A LRP10 variant, pointing towards potentially important disease mechanisms in LBDs.
Collapse
Affiliation(s)
- Ana Carreras Mascaro
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martyna M Grochowska
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Valerie Boumeester
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Natasja F J Dits
- Department of Urology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ece Naz Bilgiҫ
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Guido J Breedveld
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Leonie Vergouw
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frank Jan de Jong
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Vincenzo Bonifati
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim Mandemakers
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
29
|
Asiamah EA, Feng B, Guo R, Yaxing X, Du X, Liu X, Zhang J, Cui H, Ma J. The Contributions of the Endolysosomal Compartment and Autophagy to APOEɛ4 Allele-Mediated Increase in Alzheimer's Disease Risk. J Alzheimers Dis 2024; 97:1007-1031. [PMID: 38306054 DOI: 10.3233/jad-230658] [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] [Indexed: 02/03/2024]
Abstract
Apolipoprotein E4 (APOE4), although yet-to-be fully understood, increases the risk and lowers the age of onset of Alzheimer's disease (AD), which is the major cause of dementia among elderly individuals. The endosome-lysosome and autophagy pathways, which are necessary for homeostasis in both neurons and glia, are dysregulated even in early AD. Nonetheless, the contributory roles of these pathways to developing AD-related pathologies in APOE4 individuals and models are unclear. Therefore, this review summarizes the dysregulations in the endosome-lysosome and autophagy pathways in APOE4 individuals and non-human models, and how these anomalies contribute to developing AD-relevant pathologies. The available literature suggests that APOE4 causes endosomal enlargement, increases endosomal acidification, impairs endosomal recycling, and downregulates exosome production. APOE4 impairs autophagy initiation and inhibits basal autophagy and autophagy flux. APOE4 promotes lysosome formation and trafficking and causes ApoE to accumulate in lysosomes. APOE4-mediated changes in the endosome, autophagosome and lysosome could promote AD-related features including Aβ accumulation, tau hyperphosphorylation, glial dysfunction, lipid dyshomeostasis, and synaptic defects. ApoE4 protein could mediate APOE4-mediated endosome-lysosome-autophagy changes. ApoE4 impairs vesicle recycling and endosome trafficking, impairs the synthesis of autophagy genes, resists being dissociated from its receptors and degradation, and forms a stable folding intermediate that could disrupt lysosome structure. Drugs such as molecular correctors that target ApoE4 molecular structure and enhance autophagy may ameliorate the endosome-lysosome-autophagy-mediated increase in AD risk in APOE4 individuals.
Collapse
Affiliation(s)
- Ernest Amponsah Asiamah
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, PMB UCC, Cape Coast, Ghana
| | - Baofeng Feng
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| | - Ruiyun Guo
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xu Yaxing
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xiaofeng Du
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Xin Liu
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Jinyu Zhang
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
| | - Huixian Cui
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| | - Jun Ma
- Hebei Medical University-Galway University of Ireland Stem Cell Research Center, Hebei Medical University, Hebei, China
- Hebei Research Center for Stem Cell Medical Translational Engineering, Hebei, China
- Hebei Technology Innovation Center for Stem Cell and Regenerative Medicine, Hebei, China
| |
Collapse
|
30
|
Eisenbaum M, Pearson A, Ortiz C, Mullan M, Crawford F, Ojo J, Bachmeier C. ApoE4 expression disrupts tau uptake, trafficking, and clearance in astrocytes. Glia 2024; 72:184-205. [PMID: 37668005 DOI: 10.1002/glia.24469] [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: 02/03/2023] [Revised: 08/07/2023] [Accepted: 08/26/2023] [Indexed: 09/06/2023]
Abstract
Tauopathies are a collection of neurodegenerative diseases characterized by the accumulation of pathogenic aggregates of the microtubule-associated protein tau. Despite the prevalence and diversity of tau astrogliopathy in tauopathies, the interactions between astrocytes and tau in the brain, and the influence of neurodegenerative genetic risk factors like the apolipoprotein E4 (apoE4) isoform, are largely unknown. Here, we leveraged primary and immortalized astrocytes expressing humanized apoE isoforms to characterize the mechanisms by which astrocytes interact with and eliminate extracellular tau, and the influence of apoE genotype on these processes. Our work indicates that astrocytes rapidly internalize, process, and release tau via an exosomal secretory mechanism under physiological conditions. However, we found that apoE4 disrupted these processes in comparison to apoE3, resulting in an astrocytic phenotype prone to intracellular tau accumulation. Furthermore, exposure to repetitive mild traumatic brain injuries exacerbated the apoE4-induced impairments in tau processing and elimination by astrocytes in apoE4 targeted-replacement mice. The diminished ability of apoE4 astrocytes to eliminate extracellular tau can lead to an accumulation of pathogenic tau, which induces mitochondrial dysfunction, as demonstrated by our studies. In total, our findings suggest that the apoE4 isoform lowers the threshold of astrocytic resilience to pathogenic tau, rendering them susceptible to bioenergetic deficits in the early stages of neurodegenerative diseases such as traumatic brain injury, potentially contributing to neurological decline.
Collapse
Affiliation(s)
| | | | | | | | - Fiona Crawford
- The Roskamp Institute, Sarasota, Florida, USA
- James A. Haley Veterans' Hospital, Tampa, Florida, USA
| | - Joseph Ojo
- The Roskamp Institute, Sarasota, Florida, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, Florida, USA
- Bay Pines VA Healthcare System, Bay Pines, Florida, USA
| |
Collapse
|
31
|
Pluta R, Kocki J, Bogucki J, Bogucka-Kocka A, Czuczwar SJ. LRP1 and RAGE Genes Transporting Amyloid and Tau Protein in the Hippocampal CA3 Area in an Ischemic Model of Alzheimer's Disease with 2-Year Survival. Cells 2023; 12:2763. [PMID: 38067191 PMCID: PMC10706460 DOI: 10.3390/cells12232763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/21/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Explaining changes at the gene level that occur during neurodegeneration in the CA3 area is crucial from the point of view of memory impairment and the development of post-ischemic dementia. An ischemic model of Alzheimer's disease was used to evaluate changes in the expression of genes related to amyloid transport in the CA3 region of the hippocampus after 10 min of brain ischemia with survival of 2, 7 and 30 days and 12, 18 and 24 months. The quantitative reverse transcriptase PCR assay revealed that the expression of the LRP1 and RAGE genes involved in amyloid transport was dysregulated from 2 days to 24 months post-ischemia in the CA3 area of the hippocampus. LRP1 gene expression 2 and 7 days after ischemia was below control values. However, its expression from day 30 to 24 months, survival after an ischemic episode was above control values. RAGE gene expression 2 days after ischemia was below control values, reaching a maximum increase 7 and 30 days post-ischemia. Then, after 12, 18 and 24 months, it was again below the control values. The data indicate that in the CA3 area of the hippocampus, an episode of brain ischemia causes the increased expression of the RAGE gene for 7-30 days during the acute phase and that of LRP1 from 1 to 24 months after ischemia during the chronic stage. In other words, in the early post-ischemic stage, the expression of the gene that transport amyloid to the brain increases (7-30 days). Conversely, in the late post-ischemic stage, amyloid scavenging/cleaning gene activity increases, reducing and/or preventing further neuronal damage or facilitating the healing of damaged sites. This is how the new phenomenon of pyramidal neuronal damage in the CA3 area after ischemia is defined. In summary, post-ischemic modification of the LRP1 and RAGE genes is useful in the study of the ischemic pathways and molecular factors involved in the development of Alzheimer's disease.
Collapse
Affiliation(s)
- Ryszard Pluta
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Jacek Bogucki
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Anna Bogucka-Kocka
- Department of Biology and Genetics, Medical University of Lublin, 20-093 Lublin, Poland;
| | | |
Collapse
|
32
|
Salem HA, Abu-Elfotuh K, Alzahrani S, Rizk NI, Ali HS, Elsherbiny N, Aljohani A, Hamdan AME, Chellasamy P, Abdou NS, Gowifel AMH, Darwish A, Ibrahim OM, Abd Elmageed ZY. Punicalagin's Protective Effects on Parkinson's Progression in Socially Isolated and Socialized Rats: Insights into Multifaceted Pathway. Pharmaceutics 2023; 15:2420. [PMID: 37896179 PMCID: PMC10610313 DOI: 10.3390/pharmaceutics15102420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Parkinson's disease (PD) is a gradual deterioration of dopaminergic neurons, leading to motor impairments. Social isolation (SI), a recognized stressor, has recently gained attention as a potential influencing factor in the progress of neurodegenerative illnesses. We aimed to investigate the intricate relationship between SI and PD progression, both independently and in the presence of manganese chloride (MnCl2), while evaluating the punicalagin (PUN) therapeutic effects, a natural compound established for its cytoprotective, anti-inflammatory, and anti-apoptotic activities. In this five-week experiment, seven groups of male albino rats were organized: G1 (normal control), G2 (SI), G3 (MnCl2), G4 (SI + MnCl2), G5 (SI + PUN), G6 (MnCl2 + PUN), and G7 (SI + PUN + MnCl2). The results revealed significant changes in behavior, biochemistry, and histopathology in rats exposed to SI and/or MnCl2, with the most pronounced effects detected in the SI rats concurrently exposed to MnCl2. These effects were associated with augmented oxidative stress biomarkers and reduced antioxidant activity of the Nrf2/HO-1 pathway. Additionally, inflammatory pathways (HMGB1/RAGE/TLR4/NF-ᴋB/NLRP3/Caspase-1 and JAK-2/STAT-3) were upregulated, while dysregulation of signaling pathways (PI3K/AKT/GSK-3β/CREB), sustained endoplasmic reticulum stress by activation PERK/CHOP/Bcl-2, and impaired autophagy (AMPK/SIRT-1/Beclin-1 axis) were observed. Apoptosis induction and a decrease in monoamine levels were also noted. Remarkably, treatment with PUN effectively alleviated behaviour, histopathological changes, and biochemical alterations induced by SI and/or MnCl2. These findings emphasize the role of SI in PD progress and propose PUN as a potential therapeutic intervention to mitigate PD. PUN's mechanisms of action involve modulation of pathways such as HMGB1/RAGE/TLR4/NF-ᴋB/NLRP3/Caspase-1, JAK-2/STAT-3, PI3K/AKT/GSK-3β/CREB, AMPK/SIRT-1, Nrf2/HO-1, and PERK/CHOP/Bcl-2.
Collapse
Affiliation(s)
- Hoda A. Salem
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt;
| | - Karema Abu-Elfotuh
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt;
| | - Sharifa Alzahrani
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.S.A.); (S.A.)
| | - Nermin I. Rizk
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Menouf 32952, Egypt;
| | - Howaida S. Ali
- Department of Pharmacology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia; (H.S.A.); (S.A.)
- Department of Pharmacology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Nehal Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Alhanouf Aljohani
- Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Ahmed M. E. Hamdan
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | | | - Nada S. Abdou
- Faculty of Medicine, Misr University for Science and Technology (MUST), Giza 11556, Egypt;
| | - Ayah M. H. Gowifel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 11571, Egypt;
| | - Alshaymaa Darwish
- Biochemistry Department, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt;
| | - Osama Mohamed Ibrahim
- Clinical Pharmacy Department, Faculty of Pharmacy, University of Tanta, Tanta 31527, Egypt;
| | - Zakaria Y. Abd Elmageed
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of Louisiana at Monroe, Monroe, LA 71203, USA;
| |
Collapse
|
33
|
Zheng W, Zhou T, Zhang Y, Ding J, Xie J, Wang S, Wang Z, Wang K, Shen L, Zhu Y, Gao C. Simplified α 2-macroglobulin as a TNF-α inhibitor for inflammation alleviation in osteoarthritis and myocardial infarction therapy. Biomaterials 2023; 301:122247. [PMID: 37487780 DOI: 10.1016/j.biomaterials.2023.122247] [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: 10/14/2022] [Revised: 06/25/2023] [Accepted: 07/16/2023] [Indexed: 07/26/2023]
Abstract
Tumor necrosis factor α (TNF-α) is a leading proinflammatory cytokine as the master regulator of inflammation in chronic inflammation diseases. Although TNF-α antagonists such as small molecules and peptides are in development, comparable effectiveness in TNF-α neutralization is hardly achieved only with TNF-α capture. In this study, simplified α2-macroglobulin (SM) as a novel TNF-α inhibitor was fabricated to relieve inflammation response by TNF-α capture and internalization with lysosomal degradation. SM was prepared by conjugating a TNF-α-targeting peptide with a receptor binding domain (RBD) derived from α2-macroglobulin through a synthetic biology strategy. SM exhibited effective capture and bioactivity inhibition of TNF-α. Improved endocytosis of TNF-α into lysosomes was observed with SM in macrophages. Even challenged with LPS/IFNγ, the macrophages showed relieved inflammation response with SM treatment. When administrated in chronic inflammation injury in vivo, SM achieved comparable therapeutic efficacy with Infliximab, showing ameliorated cartilage degeneration with relieved inflammation in osteoarthritis (OA) and preserved cardiac function with mitigated myocardium injury in myocardial infarction (MI). These results suggest that SM functioning in TNF-α capture-internalization mechanism might be promising therapeutic alternatives of TNF-α antibodies.
Collapse
Affiliation(s)
- Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Tong Zhou
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yuxiang Zhang
- Department of Plastic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang Province, PR China; Key Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, 310009, China; Dr. Li Dak Sum Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jie Ding
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Jieqi Xie
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Shuqin Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Zhaoyi Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Kai Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Liyin Shen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yang Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Dr. Li Dak Sum Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
| |
Collapse
|
34
|
Hromadkova L, Kim C, Haldiman T, Peng L, Zhu X, Cohen M, de Silva R, Safar JG. Evolving prion-like tau conformers differentially alter postsynaptic proteins in neurons inoculated with distinct isolates of Alzheimer's disease tau. Cell Biosci 2023; 13:174. [PMID: 37723591 PMCID: PMC10507869 DOI: 10.1186/s13578-023-01133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023] Open
Abstract
OBJECTIVES Although accumulation of misfolded tau species has been shown to predict cognitive decline in patients with Alzheimer's disease (AD) and other tauopathies but with the remarkable diversity of clinical manifestations, neuropathology profiles, and time courses of disease progression remaining unexplained by current genetic data. We considered the diversity of misfolded tau conformers present in individual AD cases as an underlying driver of the phenotypic variations of AD and progressive loss of synapses. METHODS To model the mechanism of tau propagation and synaptic toxicity of distinct tau conformers, we inoculated wild-type primary mouse neurons with structurally characterized Sarkosyl-insoluble tau isolates from the frontal cortex of six AD cases and monitored the impact for fourteen days. We analyzed the accumulation rate, tau isoform ratio, and conformational characteristics of de novo-induced tau aggregates with conformationally sensitive immunoassays, and the dynamics of synapse formation, maintenance, and their loss using a panel of pre-and post-synaptic markers. RESULTS At the same concentrations of tau, the different AD tau isolates induced accumulation of misfolded predominantly 4-repeat tau aggregates at different rates in mature neurons, and demonstrated distinct conformational characteristics corresponding to the original AD brain tau. The time-course of the formation of misfolded tau aggregates and colocalization correlated with significant loss of synapses in tau-inoculated cell cultures and the reduction of synaptic connections implicated the disruption of postsynaptic compartment as an early event. CONCLUSIONS The data obtained with mature neurons expressing physiological levels and adult isoforms of tau protein demonstrate markedly different time courses of endogenous tau misfolding and differential patterns of post-synaptic alterations. These and previous biophysical data argue for an ensemble of various misfolded tau aggregates in individual AD brains and template propagation of their homologous conformations in neurons with different rates and primarily postsynaptic interactors. Modeling tau aggregation in mature differentiated neurons provides a platform for investigating divergent molecular mechanisms of tau strain propagation and for identifying common structural features of misfolded tau and critical interactors for new therapeutic targets and approaches in AD.
Collapse
Affiliation(s)
- Lenka Hromadkova
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Chae Kim
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Tracy Haldiman
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Lihua Peng
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
| | - Xiongwei Zhu
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
- Departments of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Mark Cohen
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA
- National Prion Disease Pathology Surveillance Center, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Rohan de Silva
- Reta Lila Weston Institute, UCL Queen Square Institute of Neurology, London, WC1N 1PJ, UK
| | - Jiri G Safar
- Departments of Pathology, Case Western Reserve University School of Medicine, 2085 Adelbert Rd, Cleveland, OH, 44106, USA.
- Departments of Neurology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
- Departments of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
35
|
Geerts H, Bergeler S, Walker M, van der Graaf PH, Courade JP. Analysis of clinical failure of anti-tau and anti-synuclein antibodies in neurodegeneration using a quantitative systems pharmacology model. Sci Rep 2023; 13:14342. [PMID: 37658103 PMCID: PMC10474108 DOI: 10.1038/s41598-023-41382-0] [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: 04/10/2023] [Accepted: 08/25/2023] [Indexed: 09/03/2023] Open
Abstract
Misfolded proteins in Alzheimer's disease and Parkinson's disease follow a well-defined connectomics-based spatial progression. Several anti-tau and anti-alpha synuclein (aSyn) antibodies have failed to provide clinical benefit in clinical trials despite substantial target engagement in the experimentally accessible cerebrospinal fluid (CSF). The proposed mechanism of action is reducing neuronal uptake of oligomeric protein from the synaptic cleft. We built a quantitative systems pharmacology (QSP) model to quantitatively simulate intrasynaptic secretion, diffusion and antibody capture in the synaptic cleft, postsynaptic membrane binding and internalization of monomeric and oligomeric tau and aSyn proteins. Integration with a physiologically based pharmacokinetic (PBPK) model allowed us to simulate clinical trials of anti-tau antibodies gosuranemab, tilavonemab, semorinemab, and anti-aSyn antibodies cinpanemab and prasineuzumab. Maximal target engagement for monomeric tau was simulated as 45% (semorinemab) to 99% (gosuranemab) in CSF, 30% to 99% in ISF but only 1% to 3% in the synaptic cleft, leading to a reduction of less than 1% in uptake of oligomeric tau. Simulations for prasineuzumab and cinpanemab suggest target engagement of free monomeric aSyn of only 6-8% in CSF, 4-6% and 1-2% in the ISF and synaptic cleft, while maximal target engagement of aggregated aSyn was predicted to reach 99% and 80% in the synaptic cleft with similar effects on neuronal uptake. The study generates optimal values of selectivity, sensitivity and PK profiles for antibodies. The study identifies a gradient of decreasing target engagement from CSF to the synaptic cleft as a key driver of efficacy, quantitatively identifies various improvements for drug design and emphasizes the need for QSP modelling to support the development of tau and aSyn antibodies.
Collapse
Affiliation(s)
- Hugo Geerts
- Certara US, 100 Overlook Centre, Suite 101, Princeton, NJ, 08540, USA.
| | - Silke Bergeler
- Certara US, 100 Overlook Centre, Suite 101, Princeton, NJ, 08540, USA
- Bristol-Meyers-Squibb, Lawrenceville, NJ, 08648, USA
| | - Mike Walker
- Certara UK, Canterbury Innovation Centre, University Road, Canterbury, CT2 7FG, Kent, UK
| | - Piet H van der Graaf
- Certara UK, Canterbury Innovation Centre, University Road, Canterbury, CT2 7FG, Kent, UK
| | | |
Collapse
|
36
|
Lathuiliere A, Jo Y, Perbet R, Donahue C, Commins C, Quittot N, Fan Z, Bennett RE, Hyman BT. Specific detection of tau seeding activity in Alzheimer's disease using rationally designed biosensor cells. Mol Neurodegener 2023; 18:53. [PMID: 37553663 PMCID: PMC10408046 DOI: 10.1186/s13024-023-00643-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND The prion-like propagation of tau in neurodegenerative disorders implies that misfolded pathological tau can recruit the normal protein and template its aggregation. Here, we report the methods for the development of sensitive biosensor cell lines for the detection of tau seeding activity. RESULTS We performed the rational design of novel tau probes based on the current structural knowledge of pathological tau aggregates in Alzheimer's disease. We generated Förster resonance energy transfer (FRET)-based biosensor stable cell lines and characterized their sensitivity, specificity, and overall ability to detect bioactive tau in human samples. As compared to the reference biosensor line, the optimized probe design resulted in an increased efficiency in the detection of tau seeding. The increased sensitivity allowed for the detection of lower amount of tau seeding competency in human brain samples, while preserving specificity for tau seeds found in Alzheimer's disease. CONCLUSIONS This next generation of FRET-based biosensor cells is a novel tool to study tau seeding activity in Alzheimer's disease human samples, especially in samples with low levels of seeding activity, which may help studying early tau-related pathological events.
Collapse
Affiliation(s)
- Aurelien Lathuiliere
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
- Memory Center, Department of Rehabilitation and Geriatrics, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Youhwa Jo
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
| | - Romain Perbet
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
| | - Cameron Donahue
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
| | - Caitlin Commins
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
| | - Noé Quittot
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
| | - Zhanyun Fan
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
| | - Rachel E Bennett
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA
- Harvard Medical School, Boston, MA, USA
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital/Harvard Medical School, 114 16Th Street, Charlestown, MA, 02129, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Raut S, Bhalerao A, Powers M, Gonzalez M, Mancuso S, Cucullo L. Hypometabolism, Alzheimer's Disease, and Possible Therapeutic Targets: An Overview. Cells 2023; 12:2019. [PMID: 37626828 PMCID: PMC10453773 DOI: 10.3390/cells12162019] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/19/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
The brain is a highly dynamic organ that requires a constant energy source to function normally. This energy is mostly supplied by glucose, a simple sugar that serves as the brain's principal fuel source. Glucose transport across the blood-brain barrier (BBB) is primarily controlled via sodium-independent facilitated glucose transport, such as by glucose transporter 1 (GLUT1) and 3 (GLUT3). However, other glucose transporters, including GLUT4 and the sodium-dependent transporters SGLT1 and SGLT6, have been reported in vitro and in vivo. When the BBB endothelial layer is crossed, neurons and astrocytes can absorb the glucose using their GLUT1 and GLUT3 transporters. Glucose then enters the glycolytic pathway and is metabolized into adenosine triphosphate (ATP), which supplies the energy to support cellular functions. The transport and metabolism of glucose in the brain are impacted by several medical conditions, which can cause neurological and neuropsychiatric symptoms. Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, traumatic brain injury (TBI), schizophrenia, etc., are a few of the most prevalent disorders, characterized by a decline in brain metabolism or hypometabolism early in the course of the disease. Indeed, AD is considered a metabolic disorder related to decreased brain glucose metabolism, involving brain insulin resistance and age-dependent mitochondrial dysfunction. Although the conventional view is that reduced cerebral metabolism is an effect of neuronal loss and consequent brain atrophy, a growing body of evidence points to the opposite, where hypometabolism is prodromal or at least precedes the onset of brain atrophy and the manifestation of clinical symptoms. The underlying processes responsible for these glucose transport and metabolic abnormalities are complicated and remain poorly understood. This review article provides a comprehensive overview of the current understanding of hypometabolism in AD and potential therapeutic targets.
Collapse
Affiliation(s)
- Snehal Raut
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Aditya Bhalerao
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Michael Powers
- Department of Biological and Biomedical Sciences, Oakland University, Rochester, MI 48309, USA;
| | - Minelly Gonzalez
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Salvatore Mancuso
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| | - Luca Cucullo
- Department of Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI 48309, USA; (S.R.); (A.B.); (M.G.); (S.M.)
| |
Collapse
|
38
|
Mah D, Zhu Y, Su G, Zhao J, Canning A, Gibson J, Song X, Stancanelli E, Xu Y, Zhang F, Linhardt RJ, Liu J, Wang L, Wang C. Apolipoprotein E Recognizes Alzheimer's Disease Associated 3-O Sulfation of Heparan Sulfate. Angew Chem Int Ed Engl 2023; 62:e202212636. [PMID: 37014788 PMCID: PMC10430763 DOI: 10.1002/anie.202212636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/05/2023]
Abstract
Apolipoprotein E (ApoE)'s ϵ4 alle is the most important genetic risk factor for late onset Alzheimer's Disease (AD). Cell-surface heparan sulfate (HS) is a cofactor for ApoE/LRP1 interaction and the prion-like spread of tau pathology between cells. 3-O-sulfo (3-O-S) modification of HS has been linked to AD through its interaction with tau, and enhanced levels of 3-O-sulfated HS and 3-O-sulfotransferases in the AD brain. In this study, we characterized ApoE/HS interactions in wildtype ApoE3, AD-linked ApoE4, and AD-protective ApoE2 and ApoE3-Christchurch. Glycan microarray and SPR assays revealed that all ApoE isoforms recognized 3-O-S. NMR titration localized ApoE/3-O-S binding to the vicinity of the canonical HS binding motif. In cells, the knockout of HS3ST1-a major 3-O sulfotransferase-reduced cell surface binding and uptake of ApoE. 3-O-S is thus recognized by both tau and ApoE, suggesting that the interplay between 3-O-sulfated HS, tau and ApoE isoforms may modulate AD risk.
Collapse
Affiliation(s)
- Dylan Mah
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Yanan Zhu
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani School of Medicine, Tampa, FL 33620, USA
| | - Guowei Su
- Glycan Therapeutics, Raleigh, NC 27606, USA
| | - Jing Zhao
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
- China Agricultural University, Beijing, 100083, China
| | - Ashely Canning
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - James Gibson
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Xuehong Song
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani School of Medicine, Tampa, FL 33620, USA
| | - Eduardo Stancanelli
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Fuming Zhang
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Robert J Linhardt
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Jian Liu
- Glycan Therapeutics, Raleigh, NC 27606, USA
- Division of Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Morsani School of Medicine, Tampa, FL 33620, USA
| | - Chunyu Wang
- Department of Chemistry and Chemical Biology, Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
39
|
Sarafanov AG. Plasma Clearance of Coagulation Factor VIII and Extension of Its Half-Life for the Therapy of Hemophilia A: A Critical Review of the Current State of Research and Practice. Int J Mol Sci 2023; 24:ijms24108584. [PMID: 37239930 DOI: 10.3390/ijms24108584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Factor VIII (FVIII) is an important component of blood coagulation as its congenital deficiency results in life-threatening bleeding. Current prophylactic therapy of the disease (hemophilia A) is based on 3-4 intravenous infusions of therapeutic FVIII per week. This poses a burden on patients, demanding reduction of infusion frequency by using FVIII with extended plasma half-life (EHL). Development of these products requires understanding FVIII plasma clearance mechanisms. This paper overviews (i) an up-to-date state of the research in this field and (ii) current EHL FVIII products, including recently approved efanesoctocog alfa, for which the plasma half-life exceeds a biochemical barrier posed by von Willebrand factor, complexed with FVIII in plasma, which results in ~1 per week infusion frequency. We focus on the EHL FVIII products' structure and function, in particular related to the known discrepancy in results of one-stage clotting (OC) and chromogenic substrate (CS) assays used to assign the products' potency, dosing, and for clinical monitoring in plasma. We suggest a possible root cause of these assays' discrepancy that is also pertinent to EHL factor IX variants used to treat hemophilia B. Finally, we discuss approaches in designing future EHL FVIII variants, including those to be used for hemophilia A gene therapy.
Collapse
Affiliation(s)
- Andrey G Sarafanov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
40
|
Chen J, Fan A, Li S, Xiao Y, Fu Y, Chen JS, Zi D, Zeng LH, Tan J. APP mediates tau uptake and its overexpression leads to the exacerbated tau pathology. Cell Mol Life Sci 2023; 80:123. [PMID: 37071198 PMCID: PMC11071805 DOI: 10.1007/s00018-023-04774-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 03/11/2023] [Accepted: 03/16/2023] [Indexed: 04/19/2023]
Abstract
Alzheimer's disease (AD), as the most common type of dementia, has two pathological hallmarks, extracellular senile plaques composed of β-amyloid peptides and intracellular neurofibrillary tangles containing phosphorylated-tau protein. Amyloid precursor protein (APP) and tau each play central roles in AD, although how APP and tau interact and synergize in the disease process is largely unknown. Here, we showed that soluble tau interacts with the N-terminal of APP in vitro in cell-free and cell culture systems, which can be further confirmed in vivo in the brain of 3XTg-AD mouse. In addition, APP is involved in the cellular uptake of tau through endocytosis. APP knockdown or N-terminal APP-specific antagonist 6KApoEp can prevent tau uptake in vitro, resulting in an extracellular tau accumulation in cultured neuronal cells. Interestingly, in APP/PS1 transgenic mouse brain, the overexpression of APP exacerbated tau propagation. Moreover, in the human tau transgenic mouse brain, overexpression of APP promotes tau phosphorylation, which is significantly remediated by 6KapoEp. All these results demonstrate the important role of APP in the tauopathy of AD. Targeting the pathological interaction of N-terminal APP with tau may provide an important therapeutic strategy for AD.
Collapse
Affiliation(s)
- Jiang Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Anran Fan
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Song Li
- First Affiliated Hospital of Dalian Medical University, Dalian, 116021, Liaoning, China
| | - Yan Xiao
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yanlin Fu
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Jun-Sheng Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Dan Zi
- Department of Gynecology, Guizhou Provincial People's Hospital, Guiyang, 550025, Guizhou, China
| | - Ling-Hui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Jun Tan
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
41
|
Mazura AD, Pietrzik CU. Endocrine Regulation of Microvascular Receptor-Mediated Transcytosis and Its Therapeutic Opportunities: Insights by PCSK9-Mediated Regulation. Pharmaceutics 2023; 15:pharmaceutics15041268. [PMID: 37111752 PMCID: PMC10144601 DOI: 10.3390/pharmaceutics15041268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Currently, many neurological disorders lack effective treatment options due to biological barriers that effectively separate the central nervous system (CNS) from the periphery. CNS homeostasis is maintained by a highly selective exchange of molecules, with tightly controlled ligand-specific transport systems at the blood-brain barrier (BBB) playing a key role. Exploiting or modifying these endogenous transport systems could provide a valuable tool for targeting insufficient drug delivery into the CNS or pathological changes in the microvasculature. However, little is known about how BBB transcytosis is continuously regulated to respond to temporal or chronic changes in the environment. The aim of this mini-review is to draw attention to the sensitivity of the BBB to circulating molecules derived from peripheral tissues, which may indicate a fundamental endocrine-operating regulatory system of receptor-mediated transcytosis at the BBB. We present our thoughts in the context of the recent observation that low-density lipoprotein receptor-related protein 1 (LRP1)-mediated clearance of brain amyloid-β (Aβ) across the BBB is negatively regulated by peripheral proprotein convertase subtilisin/kexin type 9 (PCSK9). We hope that our conclusions will inspire future investigations of the BBB as dynamic communication interface between the CNS and periphery, whose peripheral regulatory mechanisms could be easily exploited for therapeutic purposes.
Collapse
Affiliation(s)
- Alexander D Mazura
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| | - Claus U Pietrzik
- Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg, University Mainz, Duesbergweg 6, 55128 Mainz, Germany
| |
Collapse
|
42
|
Mukadam AS, Miller LVC, Smith AE, Vaysburd M, Sakya SA, Sanford S, Keeling S, Tuck BJ, Katsinelos T, Green C, Skov L, Kaalund SS, Foss S, Mayes K, O’Connell K, Wing M, Knox C, Banbury J, Avezov E, Rowe JB, Goedert M, Andersen JT, James LC, McEwan WA. Cytosolic antibody receptor TRIM21 is required for effective tau immunotherapy in mouse models. Science 2023; 379:1336-1341. [PMID: 36996217 PMCID: PMC7614512 DOI: 10.1126/science.abn1366] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/09/2023] [Indexed: 04/01/2023]
Abstract
Aggregates of the protein tau are proposed to drive pathogenesis in neurodegenerative diseases. Tau can be targeted by using passively transferred antibodies (Abs), but the mechanisms of Ab protection are incompletely understood. In this work, we used a variety of cell and animal model systems and showed that the cytosolic Ab receptor and E3 ligase TRIM21 (T21) could play a role in Ab protection against tau pathology. Tau-Ab complexes were internalized to the cytosol of neurons, which enabled T21 engagement and protection against seeded aggregation. Ab-mediated protection against tau pathology was lost in mice that lacked T21. Thus, the cytosolic compartment provides a site of immunotherapeutic protection, which may help in the design of Ab-based therapies in neurodegenerative disease.
Collapse
Affiliation(s)
- Aamir S Mukadam
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Lauren VC Miller
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Annabel E Smith
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Marina Vaysburd
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Siri A Sakya
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Sophie Sanford
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Sophie Keeling
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Benjamin J Tuck
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Taxiarchis Katsinelos
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Chris Green
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Lise Skov
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Sanne S Kaalund
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - Stian Foss
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Keith Mayes
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Kevin O’Connell
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Mark Wing
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Claire Knox
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Jessica Banbury
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Edward Avezov
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
- Cambridge University Hospitals NHS Trust, Cambridge, CB2 0SZ
| | - Michel Goedert
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Jan Terje Andersen
- Department of Immunology, University of Oslo and Oslo University Hospital Rikshospitalet, N-0424 Oslo, Norway
- Institute of Clinical Medicine and Department of Pharmacology, University of Oslo and Oslo University Hospital, N-0372 Oslo, Norway
| | - Leo C James
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - William A McEwan
- UK Dementia Research Institute at the University of Cambridge, Hills Road, Cambridge CB2 0AH, UK
- Department of Clinical Neurosciences, University of Cambridge, CB2 0AH
| |
Collapse
|
43
|
I F. The unique neuropathological vulnerability of the human brain to aging. Ageing Res Rev 2023; 87:101916. [PMID: 36990284 DOI: 10.1016/j.arr.2023.101916] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Alzheimer's disease (AD)-related neurofibrillary tangles (NFT), argyrophilic grain disease (AGD), aging-related tau astrogliopathy (ARTAG), limbic predominant TDP-43 proteinopathy (LATE), and amygdala-predominant Lewy body disease (LBD) are proteinopathies that, together with hippocampal sclerosis, progressively appear in the elderly affecting from 50% to 99% of individuals aged 80 years, depending on the disease. These disorders usually converge on the same subject and associate with additive cognitive impairment. Abnormal Tau, TDP-43, and α-synuclein pathologies progress following a pattern consistent with an active cell-to-cell transmission and abnormal protein processing in the host cell. However, cell vulnerability and transmission pathways are specific for each disorder, albeit abnormal proteins may co-localize in particular neurons. All these alterations are unique or highly prevalent in humans. They all affect, at first, the archicortex and paleocortex to extend at later stages to the neocortex and other regions of the telencephalon. These observations show that the phylogenetically oldest areas of the human cerebral cortex and amygdala are not designed to cope with the lifespan of actual humans. New strategies aimed at reducing the functional overload of the human telencephalon, including optimization of dream repair mechanisms and implementation of artificial circuit devices to surrogate specific brain functions, appear promising.
Collapse
Affiliation(s)
- Ferrer I
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain; Emeritus Researcher of the Bellvitge Institute of Biomedical Research (IDIBELL), Barcelona, Spain; Biomedical Research Network of Neurodegenerative Diseases (CIBERNED), Barcelona, Spain; Institute of Neurosciences, University of Barcelona, Barcelona, Spain; Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
44
|
Beenken A, Cerutti G, Brasch J, Guo Y, Sheng Z, Erdjument-Bromage H, Aziz Z, Robbins-Juarez SY, Chavez EY, Ahlsen G, Katsamba PS, Neubert TA, Fitzpatrick AWP, Barasch J, Shapiro L. Structures of LRP2 reveal a molecular machine for endocytosis. Cell 2023; 186:821-836.e13. [PMID: 36750096 PMCID: PMC9993842 DOI: 10.1016/j.cell.2023.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/29/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023]
Abstract
The low-density lipoprotein (LDL) receptor-related protein 2 (LRP2 or megalin) is representative of the phylogenetically conserved subfamily of giant LDL receptor-related proteins, which function in endocytosis and are implicated in diseases of the kidney and brain. Here, we report high-resolution cryoelectron microscopy structures of LRP2 isolated from mouse kidney, at extracellular and endosomal pH. The structures reveal LRP2 to be a molecular machine that adopts a conformation for ligand binding at the cell surface and for ligand shedding in the endosome. LRP2 forms a homodimer, the conformational transformation of which is governed by pH-sensitive sites at both homodimer and intra-protomer interfaces. A subset of LRP2 deleterious missense variants in humans appears to impair homodimer assembly. These observations lay the foundation for further understanding the function and mechanism of LDL receptors and implicate homodimerization as a conserved feature of the LRP receptor subfamily.
Collapse
Affiliation(s)
- Andrew Beenken
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Gabriele Cerutti
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Julia Brasch
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA
| | - Yicheng Guo
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA
| | - Hediye Erdjument-Bromage
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Zainab Aziz
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | | | - Estefania Y Chavez
- Department of Pediatrics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Goran Ahlsen
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Phinikoula S Katsamba
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Thomas A Neubert
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anthony W P Fitzpatrick
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Jonathan Barasch
- Division of Nephrology, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Columbia University George M. O'Brien Urology Center, New York, NY 10032, USA.
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Aaron Diamond AIDS Research Center, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA.
| |
Collapse
|
45
|
Abstract
Alzheimer's disease (AD) was described in 1906 as a dementing disease marked by the presence of two types of fibrillar aggregates in the brain: neurofibrillary tangles and senile plaques. The process of aggregation and formation of the aggregates has been a major focus of investigation ever since the discoveries that the tau protein is the predominant protein in tangles and amyloid β is the predominant protein in plaques. The idea that smaller, oligomeric species of amyloid may also be bioactive has now been clearly established. This review examines the possibility that soluble, nonfibrillar, bioactive forms of tau-the "tau we cannot see"-comprise a dominant driver of neurodegeneration in AD.
Collapse
Affiliation(s)
- Bradley Hyman
- Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, USA;
| |
Collapse
|
46
|
Prediction Model for Sensory Perception Abnormality in Autism Spectrum Disorder. Int J Mol Sci 2023; 24:ijms24032367. [PMID: 36768688 PMCID: PMC9916460 DOI: 10.3390/ijms24032367] [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: 12/04/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by heterogeneous clinical phenotypes. Patients often experience abnormal sensory perception, which may further affect the ASD core phenotype, significantly and adversely affecting their quality of life. However, biomarkers for the diagnosis of ASD sensory perception abnormality are currently elusive. We sought to identify potential biomarkers related to ASD sensory perception abnormality to construct a prediction model that could facilitate the early identification of and screening for ASD. Differentially expressed genes in ASD were obtained from the Gene Expression Omnibus database and were screened for genes related to sensory perception abnormality. After enrichment analysis, the random forest method was used to identify disease-characteristic genes. A prediction model was constructed with an artificial neural network. Finally, the results were validated using data from the dorsal root ganglion, cerebral cortex, and striatum of the BTBR T+ Itpr3tf/J (BTBR) ASD mouse model. A total of 1869 differentially expressed genes in ASD were screened, among which 16 genes related to sensory perception abnormality were identified. According to enrichment analysis, these 16 genes were mainly related to actin, cholesterol metabolism, and tight junctions. Using random forest, 15 disease-characteristic genes were screened for model construction. The area under the curve of the training set validation result was 0.999, and for the model function validation, the result was 0.711, indicating high accuracy. The validation of BTBR mice confirmed the reliability of using these disease-characteristic genes for prediction of ASD. In conclusion, we developed a highly accurate model for predicting ASD sensory perception abnormality from 15 disease-characteristic genes. This model provides a new method for the early identification and diagnosis of ASD sensory perception abnormality.
Collapse
|
47
|
Common and Specific Marks of Different Tau Strains Following Intra-Hippocampal Injection of AD, PiD, and GGT Inoculum in hTau Transgenic Mice. Int J Mol Sci 2022; 23:ijms232415940. [PMID: 36555581 PMCID: PMC9787745 DOI: 10.3390/ijms232415940] [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: 10/27/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Heterozygous hTau mice were used for the study of tau seeding. These mice express the six human tau isoforms, with a high predominance of 3Rtau over 4Rtau. The following groups were assessed: (i) non-inoculated mice aged 9 months (n = 4); (ii) Alzheimer's Disease (AD)-inoculated mice (n = 4); (iii) Globular Glial Tauopathy (GGT)-inoculated mice (n = 4); (iv) Pick's disease (PiD)-inoculated mice (n = 4); (v) control-inoculated mice (n = 4); and (vi) inoculated with vehicle alone (n = 2). AD-inoculated mice showed AT8-immunoreactive neuronal pre-tangles, granular aggregates, and dots in the CA1 region of the hippocampus, dentate gyrus (DG), and hilus, and threads and dots in the ipsilateral corpus callosum. GGT-inoculated mice showed unique or multiple AT8-immunoreactive globular deposits in neurons, occasionally extended to the proximal dendrites. PiD-inoculated mice showed a few loose pre-tangles in the CA1 region, DG, and cerebral cortex near the injection site. Coiled bodies were formed in the corpus callosum in AD-inoculated mice, but GGT-inoculated mice lacked globular glial inclusions. Tau deposits in inoculated mice co-localized active kinases p38-P and SAPK/JNK-P, thus suggesting active phosphorylation of the host tau. Tau deposits were absent in hTau mice inoculated with control homogenates and vehicle alone. Deposits in AD-inoculated hTau mice contained 3Rtau and 4Rtau; those in GGT-inoculated mice were mainly stained with anti-4Rtau antibodies, but a small number of deposits contained 3Rtau. Deposits in PiD-inoculated mice were stained with anti-3Rtau antibodies, but rare neuronal, thread-like, and dot-like deposits showed 4Rtau immunoreactivity. These findings show that tau strains produce different patterns of active neuronal seeding, which also depend on the host tau. Unexpected 3Rtau and 4Rtau deposits after inoculation of homogenates from 4R and 3R tauopathies, respectively, suggests the regulation of exon 10 splicing of the host tau during the process of seeding, thus modulating the plasticity of the cytoskeleton.
Collapse
|
48
|
Ando K, Nagaraj S, Küçükali F, de Fisenne MA, Kosa AC, Doeraene E, Lopez Gutierrez L, Brion JP, Leroy K. PICALM and Alzheimer's Disease: An Update and Perspectives. Cells 2022; 11:3994. [PMID: 36552756 PMCID: PMC9776874 DOI: 10.3390/cells11243994] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified the PICALM (Phosphatidylinositol binding clathrin-assembly protein) gene as the most significant genetic susceptibility locus after APOE and BIN1. PICALM is a clathrin-adaptor protein that plays a critical role in clathrin-mediated endocytosis and autophagy. Since the effects of genetic variants of PICALM as AD-susceptibility loci have been confirmed by independent genetic studies in several distinct cohorts, there has been a number of in vitro and in vivo studies attempting to elucidate the underlying mechanism by which PICALM modulates AD risk. While differential modulation of APP processing and Aβ transcytosis by PICALM has been reported, significant effects of PICALM modulation of tau pathology progression have also been evidenced in Alzheimer's disease models. In this review, we summarize the current knowledge about PICALM, its physiological functions, genetic variants, post-translational modifications and relevance to AD pathogenesis.
Collapse
Affiliation(s)
- Kunie Ando
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Siranjeevi Nagaraj
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Fahri Küçükali
- Complex Genetics of Alzheimer’s Disease Group, VIB Center for Molecular Neurology, VIB Antwerp, Department of Biomedical Sciences, University of Antwerp, 2000 Antwerp, Belgium
| | - Marie-Ange de Fisenne
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Andreea-Claudia Kosa
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Emilie Doeraene
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Lidia Lopez Gutierrez
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuropathology and Neuroanatomy, Faculty of Medicine, Université Libre de Bruxelles, ULB Neuroscience Institute, 808 Route de Lennik, 1070 Brussels, Belgium
| |
Collapse
|
49
|
Liu S, Chen T, Chen B, Liu Y, Lu X, Li J. Lrpap1 deficiency leads to myopia through TGF-β-induced apoptosis in zebrafish. Cell Commun Signal 2022; 20:162. [PMID: 36261846 PMCID: PMC9580148 DOI: 10.1186/s12964-022-00970-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/03/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Frameshift mutations in LRPAP1 are responsible for autosomal recessive high myopia in human beings but its underlying mechanism remains elusive. This study aims to investigate the effect of LRPAP1 defect on ocular refractive development and its involved mechanism. METHODS A lrpap1 mutant zebrafish line with homozygous frameshift mutation was generated by CRISPR/Cas9 technology and confirmed by Sanger sequencing. The ocular refractive phenotype was analyzed by calculating the relative refractive error (RRE) with vivo photography and histological analysis at different development stages, together with examining ocular structure change via transmission electron microscopy. Further, RNA sequencing and bioinformatics analysis were performed. The potentially involved signaling pathway as well as the interacted protein were investigated in vivo. RESULTS The lrpap1 homozygous mutant zebrafish line showed myopic phenotype. Specifically, the mutant lines showed larger eye axial length-to-body length in one-month old individuals and a myopic shift with an RRE that changed after two months. Collagen fibers became thinning and disordered in the sclera. Further, RNA sequencing and bioinformatics analysis indicated that apoptosis signaling was activated in mutant line; this was further confirmed by acridine orange and TUNEL staining. Moreover, the expression of TGF-β protein was elevated in the mutant lines. Finally, the treatment of wild-type embryos with a TGF-β agonist aggravated the degree of eyeball apoptosis; conversely, the use of a TGF-β inhibitor mitigated apoptosis in mutant embryos. CONCLUSION The study provides functional evidence of a link between lrpap1 and myopia, suggesting that lrpap1 deficiency could lead to myopia through TGF-β-induced apoptosis signaling. Video abstract.
Collapse
Affiliation(s)
- Shanshan Liu
- grid.284723.80000 0000 8877 7471Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Chen
- grid.284723.80000 0000 8877 7471Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Binghao Chen
- grid.459579.30000 0004 0625 057XDepartment of Orthopedics, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yijun Liu
- grid.413107.0Department of Foot and Ankle Surgery, Center for Orthopedic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiaohe Lu
- grid.284723.80000 0000 8877 7471Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiali Li
- grid.284723.80000 0000 8877 7471Department of Ophthalmology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
50
|
Chun H, Kurasawa JH, Olivares P, Marakasova ES, Shestopal SA, Hassink GU, Karnaukhova E, Migliorini M, Obi JO, Smith AK, Wintrode PL, Durai P, Park K, Deredge D, Strickland DK, Sarafanov AG. Characterization of interaction between blood coagulation factor VIII and LRP1 suggests dynamic binding by alternating complex contacts. J Thromb Haemost 2022; 20:2255-2269. [PMID: 35810466 PMCID: PMC9804390 DOI: 10.1111/jth.15817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/15/2022] [Accepted: 07/01/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Deficiency in blood coagulation factor VIII (FVIII) results in life-threating bleeding (hemophilia A) treated by infusions of FVIII concentrates. To improve disease treatment, FVIII has been modified to increase its plasma half-life, which requires understanding mechanisms of FVIII catabolism. An important catabolic actor is hepatic low density lipoprotein receptor-related protein 1 (LRP1), which also regulates many other clinically significant processes. Previous studies showed complexity of FVIII site for binding LRP1. OBJECTIVES To characterize binding sites between FVIII and LRP1 and suggest a model of the interaction. METHODS A series of recombinant ligand-binding complement-type repeat (CR) fragments of LRP1 including mutated variants was generated in a baculovirus system and tested for FVIII interaction using surface plasmon resonance, tissue culture model, hydrogen-deuterium exchange mass spectrometry, and in silico. RESULTS Multiple CR doublets within LRP1 clusters II and IV were identified as alternative FVIII-binding sites. These interactions follow the canonical binding mode providing major binding energy, and additional weak interactions are contributed by adjacent CR domains. A representative CR doublet was shown to have multiple contact sites on FVIII. CONCLUSIONS FVIII and LRP1 interact via formation of multiple complex contacts involving both canonical and non-canonical binding combinations. We propose that FVIII-LRP1 interaction occurs via switching such alternative binding combinations in a dynamic mode, and that this mechanism is relevant to other ligand interactions of the low-density lipoprotein receptor family members including LRP1.
Collapse
Affiliation(s)
- Haarin Chun
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - James H. Kurasawa
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
Biologics Engineering, R&D, AstraZeneca, GaithersburgMarylandUSA
| | - Philip Olivares
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Ekaterina S. Marakasova
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
(1) Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver SpringMarylandUSA
- Present address:
George Mason University, School of Systems Biology, FairfaxVirginiaUSA
| | - Svetlana A. Shestopal
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Gabriela U. Hassink
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
- Present address:
GSK‐Rockville Center for Vaccines Research, RockvilleMarylandUSA
| | - Elena Karnaukhova
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| | - Mary Migliorini
- Center for Vascular and Inflammatory DiseasesDepartments of Surgery and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Juliet O. Obi
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Ally K. Smith
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Patrick L. Wintrode
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Prasannavenkatesh Durai
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Keunwan Park
- Natural Product Informatics Research CenterKorea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Daniel Deredge
- Department of Pharmaceutical SciencesUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Dudley K. Strickland
- Center for Vascular and Inflammatory DiseasesDepartments of Surgery and PhysiologyUniversity of Maryland School of MedicineBaltimoreMarylandUSA
| | - Andrey G. Sarafanov
- Center for Biologics Evaluation and ResearchU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|