1
|
Di Lorenzo D. Tau Protein and Tauopathies: Exploring Tau Protein-Protein and Microtubule Interactions, Cross-Interactions and Therapeutic Strategies. ChemMedChem 2024; 19:e202400180. [PMID: 39031682 DOI: 10.1002/cmdc.202400180] [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: 03/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024]
Abstract
Tau, a microtubule-associated protein (MAP), is essential to maintaining neuronal stability and function in the healthy brain. However, aberrant modifications and pathological aggregations of Tau are implicated in various neurodegenerative disorders, collectively known as tauopathies. The most common Tauopathy is Alzheimer's Disease (AD) counting nowadays more than 60 million patients worldwide. This comprehensive review delves into the multifaceted realm of Tau protein, puzzling out its intricate involvement in both physiological and pathological roles. Emphasis is put on Tau Protein-Protein Interactions (PPIs), depicting its interaction with tubulin, microtubules and its cross-interaction with other proteins such as Aβ1-42, α-synuclein, and the chaperone machinery. In the realm of therapeutic strategies, an overview of diverse possibilities is presented with their relative clinical progresses. The focus is mostly addressed to Tau protein aggregation inhibitors including recent small molecules, short peptides and peptidomimetics with specific focus on compounds that showed a double anti aggregative activity on both Tau protein and Aβ amyloid peptide. This review amalgamates current knowledge on Tau protein and evolving therapeutic strategies, providing a comprehensive resource for researchers seeking to deepen their understanding of the Tau protein and for scientists involved in the development of new peptide-based anti-aggregative Tau compounds.
Collapse
Affiliation(s)
- Davide Di Lorenzo
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, D-33615, Bielefeld, Germany
| |
Collapse
|
2
|
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
|
3
|
Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Elewa YHA, AL‐Farga A, Aqlan F, Zahran MH, Batiha GE. Sleep disorders cause Parkinson's disease or the reverse is true: Good GABA good night. CNS Neurosci Ther 2024; 30:e14521. [PMID: 38491789 PMCID: PMC10943276 DOI: 10.1111/cns.14521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative brain disease due to degeneration of dopaminergic neurons (DNs) presented with motor and non-motor symptoms. PD symptoms are developed in response to the disturbance of diverse neurotransmitters including γ-aminobutyric acid (GABA). GABA has a neuroprotective effect against PD neuropathology by protecting DNs in the substantia nigra pars compacta (SNpc). It has been shown that the degeneration of GABAergic neurons is linked with the degeneration of DNs and the progression of motor and non-motor PD symptoms. GABA neurotransmission is a necessary pathway for normal sleep patterns, thus deregulation of GABAergic neurotransmission in PD could be the potential cause of sleep disorders in PD. AIM Sleep disorders affect GABA neurotransmission leading to memory and cognitive dysfunction in PD. For example, insomnia and short sleep duration are associated with a reduction of brain GABA levels. Moreover, PD-related disorders including rigidity and nocturia influence sleep patterns leading to fragmented sleep which may also affect PD neuropathology. However, the mechanistic role of GABA in PD neuropathology regarding motor and non-motor symptoms is not fully elucidated. Therefore, this narrative review aims to clarify the mechanistic role of GABA in PD neuropathology mainly in sleep disorders, and how good GABA improves PD. In addition, this review of published articles tries to elucidate how sleep disorders such as insomnia and REM sleep behavior disorder (RBD) affect PD neuropathology and severity. The present review has many limitations including the paucity of prospective studies and most findings are taken from observational and preclinical studies. GABA involvement in the pathogenesis of PD has been recently discussed by recent studies. Therefore, future prospective studies regarding the use of GABA agonists in the management of PD are suggested to observe their distinct effects on motor and non-motor symptoms. CONCLUSION There is a bidirectional relationship between the pathogenesis of PD and sleep disorders which might be due to GABA deregulation.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ammar AL‐Farga
- Biochemistry Department, College of SciencesUniversity of JeddahJeddahSaudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of SciencesIbb UniversityIbb GovernorateYemen
| | | | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurEgypt
| |
Collapse
|
4
|
Mahady L, Perez SE, Malek-Ahmadi M, Mufson EJ. Oligomeric, phosphorylated, and truncated tau and spliceosome pathology within the entorhinal-hippocampal connectome across stages of Alzheimer's disease. J Comp Neurol 2023; 531:2080-2108. [PMID: 36989381 PMCID: PMC10539478 DOI: 10.1002/cne.25466] [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: 11/10/2022] [Revised: 01/27/2023] [Accepted: 02/06/2023] [Indexed: 03/31/2023]
Abstract
Neurofibrillary tangles (NFTs) contain abnormally phosphorylated tau proteins, which spread within components of the medial temporal lobe (MTL) memory circuit in Alzheimer's disease (AD). Here, we used quantitative immunohistochemistry to determine the density of posttranslational oligomeric (TOC1 and TNT1), phosphorylated (AT8), and late truncated (TauC3) tau epitopes within the MTL subfields including entorhinal cortex (EC) layer II, subiculum, Cornu Ammonis (CA) subfields, and dentate gyrus (DG) in subjects who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), and AD. We also examined whether alterations of the nuclear alternative splicing protein, SRSF2, are associated with tau pathology. Although a significant increase in TOC1, TNT1, and AT8 neuron density occurred in the EC in MCI and AD, subicular, DG granule cell, and CA1 and CA3 densities were only significantly higher in AD. TauC3 counts were not different between connectome regions and clinical groups. SRSF2 intensity in AT8-positive cells decreased significantly in all regions independent of the clinical groups examined. CA1 and subicular AT8, TauC3, and oligomeric densities correlated across clinical groups. EC AT8 counts correlated with CA subfields and subicular and DG values across clinical groups. Oligomeric and AT8 CA1, EC, and subicular density correlated with Braak stage. Decreased nuclear SRSF2 in the presence of cytoplasmic phosphorylated tau suggests a dual-hit process in NFT formation within the entorhinal hippocampal connectome during the onset of AD. Although oligomeric and phosphorylated tau follow a stereotypical pattern, clinical disease stage determined density of tau deposition and not anatomic location within the entorhinal-hippocampal connectome.
Collapse
Affiliation(s)
- Laura Mahady
- Dept. of Translational Neuroscience, Phoenix, AZ
| | | | | | - Elliott J. Mufson
- Dept. of Translational Neuroscience, Phoenix, AZ
- Dept. of Neurology, Barrow Neurological Institute, Phoenix, AZ 85013
| |
Collapse
|
5
|
Vande Vyver M, Daeninck L, De Smet G, Aourz N, Sahu S, Engelborghs S, Pauwels K, De Bundel D, Smolders I. The intracerebral injection of Aβ 1-42 oligomers does not invariably alter seizure susceptibility in mice. Front Aging Neurosci 2023; 15:1239140. [PMID: 37744393 PMCID: PMC10512828 DOI: 10.3389/fnagi.2023.1239140] [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: 06/12/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Objectives Epileptiform activity and seizures are present in patients with Alzheimer's disease (AD) and genetic animal models of AD. Amyloid beta 1-42 (Aβ1-42) oligomers are thought to be crucial in AD and can cause neuronal hyperexcitability in vitro. However, it is unclear whether these Aβ1-42 oligomers cause the increased seizure susceptibility in vivo in people with AD and in AD animal models, nor via which mechanisms it would do so. We investigated this question by injecting Aβ1-42 oligomers intracerebrally in mice and assessed its impact on seizure susceptibility. Materials and methods We performed a single intracerebral injection of synthetic Aβ1-42 oligomers or scrambled Aβ1-42 in NMRI mice in three different cohorts and subjected them to an i.v. infusion of a chemoconvulsant. We evoked the seizures 1.5 h, 1 week, or 3 weeks after the intracerebral injection of Aβ1-42 oligomers, covering also the timepoints and injection locations that were used by others in similar experimental set-ups. Results With a thioflavine T assay and transmission electron microscopy we confirmed that Aβ1-42 monomers spontaneously aggregated to oligomers. We did not find an effect of Aβ1-42 oligomers on susceptibility to seizures - evoked 1.5 h, 1 week or 3 weeks - after their intracerebral injection. Significance The lack of effect of Aβ1-42 oligomers on seizure susceptibility in our experiments contrasts with recent findings in similar experimental set-ups. Contradicting conclusions are frequent in experiments with Aβ1-42 and they are often attributed to subtle differences in the various aggregation forms of the Aβ1-42 used in different experiments. We confirmed the presence of Aβ1-42 oligomers with state-of-the-art methods but cannot ascertain that the protein aggregates we used are identical to those used by others. Whether our findings or those previously published best represent the role of Aβ1-42 oligomers on seizures in AD remains unclear.
Collapse
Affiliation(s)
- Maxime Vande Vyver
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel, Brussels, Belgium
- NEUR Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Louise Daeninck
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Gino De Smet
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Najat Aourz
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Surajit Sahu
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Sebastiaan Engelborghs
- Department of Neurology and Bru-BRAIN, Universitair Ziekenhuis Brussel, Brussels, Belgium
- NEUR Research Group, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedical Sciences, Reference Center for Biological Markers of Dementia (BIODEM), University of Antwerp, Antwerp, Belgium
| | - Kris Pauwels
- RESEARCH Department, Vrije Universiteit Brussel, Brussels, Belgium
| | - Dimitri De Bundel
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry, Drug Analysis and Drug Information, Research Group Experimental Pharmacology (EFAR), Center for Neurosciences (C4N), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
6
|
Wang Q, Yao S, Yang ZX, Zhou C, Zhang Y, Zhang Y, Zhang L, Li JT, Xu ZJ, Zhu WL, Zhang NX, Ye Y, Feng LY. Pharmacological characterization of the small molecule 03A10 as an inhibitor of α-synuclein aggregation for Parkinson's disease treatment. Acta Pharmacol Sin 2023; 44:1122-1134. [PMID: 36627343 PMCID: PMC10203352 DOI: 10.1038/s41401-022-01039-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/05/2022] [Indexed: 01/11/2023] Open
Abstract
Aggregation of α-synuclein, a component of Lewy bodies (LBs) or Lewy neurites in Parkinson's disease (PD), is strongly linked with disease development, making it an attractive therapeutic target. Inhibiting aggregation can slow or prevent the neurodegenerative process. However, the bottleneck towards achieving this goal is the lack of such inhibitors. In the current study, we established a high-throughput screening platform to identify candidate compounds for preventing the aggregation of α-synuclein among the natural products in our in-house compound library. We found that a small molecule, 03A10, i.e., (+)-desdimethylpinoresinol, which is present in the fruits of Vernicia fordii (Euphorbiaceae), modulated aggregated α-synuclein, but not monomeric α-synuclein, to prevent further elongation of α-synuclein fibrils. In α-synuclein-overexpressing cell lines, 03A10 (10 μM) efficiently prevented α-synuclein aggregation and markedly ameliorated the cellular toxicity of α-synuclein fibril seeds. In the MPTP/probenecid (MPTP/p) mouse model, oral administration of 03A10 (0.3 mg· kg-1 ·d-1, 1 mg ·kg-1 ·d-1, for 35 days) significantly alleviated behavioral deficits, tyrosine hydroxylase (TH) neuron degeneration and p-α-synuclein aggregation in the substantia nigra (SN). As the Braak hypothesis postulates that the prevailing site of early PD pathology is the gastrointestinal tract, we inoculated α-synuclein preformed fibrils (PFFs) into the mouse colon. We demonstrated that α-synuclein PFF inoculation promoted α-synuclein pathology and neuroinflammation in the gut and brain; oral administration of 03A10 (5 mg· kg-1 ·d-1, for 4 months) significantly attenuated olfactory deficits, α-synuclein accumulation and neuroinflammation in the olfactory bulb and SN. We conclude that 03A10 might be a promising drug candidate for the treatment of PD. 03A10 might be a novel drug candidate for PD treatment, as it inhibits α-synuclein aggregation by modulating aggregated α-synuclein rather than monomeric α-synuclein to prevent further elongation of α-synuclein fibrils and prevent α-synuclein toxicity in vitro, in an MPTP/p mouse model, and PFF-inoculated mice.
Collapse
Affiliation(s)
- Qing Wang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Sheng Yao
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, China
| | - Ze-Xian Yang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Chen Zhou
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Yu Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Ye Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
| | - Lei Zhang
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China
| | - Jin-Tian Li
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhi-Jian Xu
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Wei-Liang Zhu
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Nai-Xia Zhang
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
- Analytical Research Center for Organic and Biological Molecules, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
| | - Yang Ye
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
- State Key Laboratory of Drug Research and Natural Products Chemistry Department, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
- School of Life Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Shanghai, 201203, China.
| | - Lin-Yin Feng
- CAS Key Laboratory of Receptor Research, Center for Neurological and Psychiatric Research and Drug Discovery (CNPRDD), Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, No.19 A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
7
|
Alrouji M, Al-Kuraishy HM, Al-Gareeb AI, Zaafar D, Batiha GES. Orexin pathway in Parkinson's disease: a review. Mol Biol Rep 2023:10.1007/s11033-023-08459-5. [PMID: 37155018 DOI: 10.1007/s11033-023-08459-5] [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: 02/23/2023] [Accepted: 04/13/2023] [Indexed: 05/10/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease (NDD) caused by dopaminergic neuron degeneration in the substantia nigra (SN). Orexin is a neuropeptide that plays a role in the pathogenesis of PD. Orexin has neuroprotective properties in dopaminergic neurons. In PD neuropathology, there is also degeneration of orexinergic neurons in the hypothalamus, in addition to dopaminergic neurons. However, the loss of orexinergic neurons in PD began after the degeneration of dopaminergic neurons. Reduced activity of orexinergic neurons has been linked to developing and progressing motor and non-motor symptoms in PD. In addition, the dysregulation of the orexin pathway is linked to the development of sleep disorders. The hypothalamic orexin pathway regulates various aspects of PD neuropathology at the cellular, subcellular, and molecular levels. Finally, non-motor symptoms, particularly insomnia and disturbed sleep, promote neuroinflammation and the accumulation of neurotoxic proteins as a result of defects in autophagy, endoplasmic reticulum (ER) stress, and the glymphatic system. As a result, this review aimed to highlight the potential role of orexin in PD neuropathology.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of clinical pharmacology and therapeutic medicine, college of medicine, Mustansiriyah University, Baghdad, Iraq
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Al Beheira, Egypt.
| |
Collapse
|
8
|
Kim JR. Oligomerization by co-assembly of β-amyloid and α-synuclein. Front Mol Biosci 2023; 10:1153839. [PMID: 37021111 PMCID: PMC10067735 DOI: 10.3389/fmolb.2023.1153839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/07/2023] [Indexed: 04/07/2023] Open
Abstract
Aberrant self-assembly of an intrinsically disordered protein is a pathological hallmark of protein misfolding diseases, such as Alzheimer's and Parkinson's diseases (AD and PD, respectively). In AD, the 40-42 amino acid-long extracellular peptide, β-amyloid (Aβ), self-assembles into oligomers, which eventually aggregate into fibrils. A similar self-association of the 140 amino acid-long intracellular protein, α-synuclein (αS), is responsible for the onset of PD pathology. While Aβ and αS are primarily extracellular and intracellular polypeptides, respectively, there is evidence of their colocalization and pathological overlaps of AD and PD. This evidence has raised the likelihood of synergistic, toxic protein-protein interactions between Aβ and αS. This mini review summarizes the findings of studies on Aβ-αS interactions related to enhanced oligomerization via co-assembly, aiming to provide a better understanding of the complex biology behind AD and PD and common pathological mechanisms among the major neurodegenerative diseases.
Collapse
|
9
|
Gomes P, Tzouanou F, Skolariki K, Vamvaka-Iakovou A, Noguera-Ortiz C, Tsirtsaki K, Waites CL, Vlamos P, Sousa N, Costa-Silva B, Kapogiannis D, Sotiropoulos I. Extracellular vesicles and Alzheimer's disease in the novel era of Precision Medicine: implications for disease progression, diagnosis and treatment. Exp Neurol 2022; 358:114183. [PMID: 35952764 PMCID: PMC9985072 DOI: 10.1016/j.expneurol.2022.114183] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/17/2022] [Accepted: 07/21/2022] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs), secreted membranous nano-sized particles, are critical intercellular messengers participating in nervous system homeostasis, while recent evidence implicates EVs in Alzheimer's disease (AD) pathogenesis. Specifically, small EVs have been shown to spread toxic proteins, induce neuronal loss, and contribute to neuroinflammation and AD progression. On the other hand, EVs can reduce amyloid-beta deposition and transfer neuroprotective substances between cells, mitigating disease mechanisms. In addition to their roles in AD pathogenesis, EVs also exhibit great potential for the diagnosis and treatment of other brain disorders, representing an advantageous tool for Precision Medicine. Herein, we summarize the contribution of small EVs to AD-related mechanisms and disease progression, as well as their potential as diagnostic and therapeutic agents for AD.
Collapse
Affiliation(s)
- Patrícia Gomes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Foteini Tzouanou
- Institute of Biosciences & Applications NCSR "Demokritos", Athens, Greece
| | | | - Anastasia Vamvaka-Iakovou
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; Institute of Biosciences & Applications NCSR "Demokritos", Athens, Greece
| | - Carlos Noguera-Ortiz
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Katerina Tsirtsaki
- Institute of Biosciences & Applications NCSR "Demokritos", Athens, Greece
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Medical Center, New York, NY, USA
| | | | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bruno Costa-Silva
- Systems Oncology Group, Champalimaud Research, Champalimaud Centre for the Unknown, Av. Brasília, 1400-038 Lisbon, Portugal
| | - Dimitrios Kapogiannis
- Laboratory of Clinical Investigation, Intramural Research Program, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; Institute of Biosciences & Applications NCSR "Demokritos", Athens, Greece.
| |
Collapse
|
10
|
Lacoursiere SG, Safar J, Westaway D, Mohajerani MH, Sutherland RJ. The effect of Aβ seeding is dependent on the presence of knock-in genes in the App NL-G-F mice. FRONTIERS IN DEMENTIA 2022; 1:941879. [PMID: 39081481 PMCID: PMC11285652 DOI: 10.3389/frdem.2022.941879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/08/2022] [Indexed: 08/02/2024]
Abstract
Alzheimer's disease (AD) is characterized by the prion-like propagation of amyloid-β (Aβ). However, the role of Aβ in cognitive impairment is still unclear. To determine the causal role of Aβ in AD, we intracerebrally seeded the entorhinal cortex of a 2-month-old App NL-G-F mouse model with an Aβ peptide derived from patients who died from rapidly progressing AD. When the mice were 3 months of age or 1 month following seeding, spatial learning and memory were tested using the Morris water task. Immunohistochemical labeling showed seeding with the Aβ was found accelerate Aβ plaque deposition and microgliosis in the App NL-G-F mice, but this was dependent on the presence of the knocked-in genes. However, we found no correlation between pathology and spatial performance. The results of the present study show the seeding effects in the App NL-G-F knock-in model, and how these are dependent on the presence of a humanized App gene. But these pathological changes were not initially causal in memory impairment.
Collapse
Affiliation(s)
- Sean G. Lacoursiere
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Jiri Safar
- Departments of Pathology, Neurology, Psychiatry, and National Prion Disease Pathology Surveillance Center, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - David Westaway
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - Majid H. Mohajerani
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| | - Robert J. Sutherland
- Canadian Centre for Behavioural Neuroscience, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
11
|
β-Amyloid activates reactive astrocytes by enhancing glycolysis of astrocytes. Mol Biol Rep 2022; 49:4699-4707. [PMID: 35534584 DOI: 10.1007/s11033-022-07319-y] [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: 10/13/2021] [Revised: 02/16/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND The aberrant accumulation of β-amyloid peptides (Aβ), reactive astrocytes and glucose metabolism deficit are typical features in the early Alzheimer's disease (AD) pathology. Previous studies have demonstrated that astrocytes process glucose mainly by glycolysis to generate lactate. However, the changes of glycolytic metabolism of reactive astrocytes in AD are still unknown. The present study aims to explore the effect of Aβ on the astrocytic activation and glycolytic metabolism, as well as the role of glycolysis in the activation of astrocytes. METHODS AND RESULTS The primary astrocytes were cultured and treated with Aβ oligomers, Aβ-activated microglia conditioned medium (aMCM) or the glycogen phosphorylase inhibitor (DAB) for 12 h. Then ECAR was used to detect the glycolysis function of reactive astrocytes. The phenotypes of reactive astrocytes were evaluated by detecting the mRNA expression of Gfap (pan-reactive marker), and Ugt1a, Ggta1 (A1-phenotypes markers), and S100a10, Emp1 (A2-phenotypes markers) using qRT-PCR. The levels of GFAP, the marker protein of pan-reactive astrocytes, was also quantified by immunofluorescence and western-blot in Aβ, aMCM or DAB-treated astrocytes. In this study, we found that Aβ oligomers could not directly activate astrocytes or promote the glycolysis. However, Aβ oligomers could induce the activation of neurotoxic A1 astrocytes and up-regulate the glycolysis function via aMCM. Reactivity of A1-astrocytes were inhibited when the glycolytic metabolism was blocked by DAB. CONCLUSIONS The results revealed that Aβ could indirectly activate A1 astrocytes by Aβ-activated microglia, which depended on the up-regulation of the glycolysis of astrocytes. The glycolysis was crucial for the activation of the neurotoxic A1 astrocytes and inhibiting the glycolysis of neurotoxic A1 astrocytes might be a new therapeutic strategy for AD.
Collapse
|
12
|
Interactions of intrinsically disordered proteins with the unconventional chaperone human serum albumin: From mechanisms of amyloid inhibition to therapeutic opportunities. Biophys Chem 2022; 282:106743. [PMID: 35093643 DOI: 10.1016/j.bpc.2021.106743] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/29/2022]
Abstract
Human Serum Albumin (HSA), the most abundant protein in plasma, serves a diverse repertoire of biological functions including regulation of oncotic pressure and redox potential, transport of serum solutes, but also chaperoning of misfolded proteins. Here we review how HSA interacts with a wide spectrum of client proteins including intrinsically disordered proteins (IDPs) such as Aβ, the islet amyloid peptide (IAPP), alpha synuclein and stressed globular proteins such as insulin. The comparative analysis of the HSA chaperone - client interactions reveals that the amyloid-inhibitory function of HSA arises from at least four emerging mechanisms. Two mechanisms (the monomer stabilizer model and the monomer competitor model) involve the direct binding of HSA to either IDP monomers or oligomers, while other mechanisms (metal chelation and membrane protection) rely on the indirect modulation by HSA of other factors that drive IDP aggregation. While HSA is not the only extracellular chaperone, given its abundance, HSA is likely to account for a significant fraction of the chaperoning effects in plasma, thus opening new therapeutic opportunities in the context of the peripheral sink hypothesis.
Collapse
|
13
|
Rimal S, Li Y, Vartak R, Geng J, Tantray I, Li S, Huh S, Vogel H, Glabe C, Grinberg LT, Spina S, Seeley WW, Guo S, Lu B. Inefficient quality control of ribosome stalling during APP synthesis generates CAT-tailed species that precipitate hallmarks of Alzheimer's disease. Acta Neuropathol Commun 2021; 9:169. [PMID: 34663454 PMCID: PMC8522249 DOI: 10.1186/s40478-021-01268-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/26/2021] [Indexed: 11/25/2022] Open
Abstract
Amyloid precursor protein (APP) metabolism is central to Alzheimer's disease (AD) pathogenesis, but the key etiological driver remains elusive. Recent failures of clinical trials targeting amyloid-β (Aβ) peptides, the proteolytic fragments of amyloid precursor protein (APP) that are the main component of amyloid plaques, suggest that the proteostasis-disrupting, key pathogenic species remain to be identified. Previous studies suggest that APP C-terminal fragment (APP.C99) can cause disease in an Aβ-independent manner. The mechanism of APP.C99 pathogenesis is incompletely understood. We used Drosophila models expressing APP.C99 with the native ER-targeting signal of human APP, expressing full-length human APP only, or co-expressing full-length human APP and β-secretase (BACE), to investigate mechanisms of APP.C99 pathogenesis. Key findings are validated in mammalian cell culture models, mouse 5xFAD model, and postmortem AD patient brain materials. We find that ribosomes stall at the ER membrane during co-translational translocation of APP.C99, activating ribosome-associated quality control (RQC) to resolve ribosome collision and stalled translation. Stalled APP.C99 species with C-terminal extensions (CAT-tails) resulting from inadequate RQC are prone to aggregation, causing endolysosomal and autophagy defects and seeding the aggregation of amyloid β peptides, the main component of amyloid plaques. Genetically removing stalled and CAT-tailed APP.C99 rescued proteostasis failure, endolysosomal/autophagy dysfunction, neuromuscular degeneration, and cognitive deficits in AD models. Our finding of RQC factor deposition at the core of amyloid plaques from AD brains further supports the central role of defective RQC of ribosome collision and stalled translation in AD pathogenesis. These findings demonstrate that amyloid plaque formation is the consequence and manifestation of a deeper level proteostasis failure caused by inadequate RQC of translational stalling and the resultant aberrantly modified APP.C99 species, previously unrecognized etiological drivers of AD and newly discovered therapeutic targets.
Collapse
Affiliation(s)
- Suman Rimal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yu Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rasika Vartak
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ji Geng
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ishaq Tantray
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Shuangxi Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sungun Huh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Charles Glabe
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Lea T Grinberg
- Memory and Aging Center, Department of Neurology and Department of Pathology, University of California, San Francisco, CA, 94158, USA
| | - Salvatore Spina
- Memory and Aging Center, Department of Neurology and Department of Pathology, University of California, San Francisco, CA, 94158, USA
| | - William W Seeley
- Memory and Aging Center, Department of Neurology and Department of Pathology, University of California, San Francisco, CA, 94158, USA
| | - Su Guo
- Department of Bioengineering and Therapeutic Sciences, Programs in Human Genetics and Biological Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
14
|
Hu Y, Zhang Y, Ren R, Dammer EB, Xie X, Chen S, Huang Q, Huang W, Zhang R, Chen H, Wang H, Wang G. microRNA-425 loss mediates amyloid plaque microenvironment heterogeneity and promotes neurodegenerative pathologies. Aging Cell 2021; 20:e13454. [PMID: 34510683 PMCID: PMC8520725 DOI: 10.1111/acel.13454] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/19/2021] [Accepted: 08/05/2021] [Indexed: 01/09/2023] Open
Abstract
Different cellular and molecular changes underlie the pathogenesis of Alzheimer's disease (AD). Among these, neuron‐specific dysregulation is a necessary event for accumulation of classic pathologies including amyloid plaques. Here, we show that AD‐associated pathophysiology including neuronal cell death, inflammatory signaling, and endolysosomal dysfunction is spatially colocalized to amyloid plaques in regions with abnormal microRNA‐425 (miR‐425) levels and this change leads to focal brain microenvironment heterogeneity, that is, an amyloid plaque‐associated microenvironment (APAM). APAM consists of multiple specific neurodegenerative signature pathologies associated with senile plaques that contribute to the heterogeneity and complexity of AD. Remarkably, miR‐425, a neuronal‐specific regulator decreased in AD brain, maintains a normal spatial transcriptome within brain neurons. We tested the hypothesis that miR‐425 loss correlates with enhanced levels of mRNA targets downstream, supporting APAM and AD progression. A miR‐425‐deficient mouse model has enhanced APP amyloidogenic processing, neuroinflammation, neuron loss, and cognitive impairment. In the APP/PS1 mouse model, intervening with miR‐425 supplementation ameliorated APAM changes and memory deficits. This study reveals a novel mechanism of dysregulation of spatial transcriptomic changes in AD brain, identifying a probable neuronal‐specific microRNA regulator capable of staving off amyloid pathogenesis. Moreover, our findings provide new insights for developing AD treatment strategies with miRNA oligonucleotide(s).
Collapse
Affiliation(s)
- Yong‐Bo Hu
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Neurology,Shanghai East Hospital School of Medicine,Tongji University Shanghai China
| | - Yong‐Fang Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ru‐Jing Ren
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Eric B. Dammer
- Department of Biochemistry and Center for Neurodegenerative Disease Emory University School of Medicine Atlanta Georgia USA
| | - Xin‐Yi Xie
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Shi‐Wu Chen
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiang Huang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Wan‐Ying Huang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hong‐Zhuan Chen
- Institute of Interdisciplinary Science Shuguang Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gang Wang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
15
|
Yang X, Li X, Liu L, Chen YH, You Y, Gao Y, Liu YY, Yang L, Tong K, Chen DS, Hao JR, Sun N, Zhao ZM, Gao C. Transferrin-Pep63-liposomes accelerate the clearance of Aβ and rescue impaired synaptic plasticity in early Alzheimer's disease models. Cell Death Discov 2021; 7:256. [PMID: 34548476 PMCID: PMC8455582 DOI: 10.1038/s41420-021-00639-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/19/2021] [Accepted: 08/27/2021] [Indexed: 11/11/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by aberrant accumulation of extracellular β-amyloid (Aβ) peptides in the brain. Soluble Aβ oligomers are thought to be the most neurotoxic species and are correlated with cognitive dysfunction in early AD. However, there is still no effective treatment so far. We determined that Pep63, a small peptide, had a neuroprotective effect on synaptic plasticity and memory in our previous study. Here, we developed novel and multifunctional liposomes targeting both Aβ oligomers and fibrils based on a liposome delivery system. Transferrin-Pep63-liposomes (Tf-Pep63-Lip), possessing the ability for blood-brain barrier targeting, were also incorporated with phosphatidic acid (PA) and loaded with neuroprotective Pep63. We discovered that administration of Tf-Pep63-Lip could significantly reduce the Aβ burden in the hippocampus, and improve cognitive deficits in 6-month-old APP/PS1 mice in the Morris-Water maze task and fear-conditioning test with the combined effects of PA and Pep63. Tf-Pep63-Lip could capture Aβ oligomers or fibrils and then facilitated microglial chemotaxis nearby for clearance. Simultaneously, Tf-Pep63-Lip hindered Aβ1-42 aggregation and disaggregated Aβ1-42 assembly due to multivalent PA-Aβ. Pep63 effectively inhibited the binding between EphB2 and Aβ oligomers after release from liposomes and rescued NMDA receptors trafficking, the basis of synaptic plasticity. No side effects were observed in either APP/PS1 or wild-type mice, indicating that Tf-Pep63-Lip might be safe under the dosing regimen used in our experiment. Taken together, our results suggested that Tf-Pep63-Lip may serve as a safe and efficient agent for AD combination therapy.
Collapse
Affiliation(s)
- Xiu Yang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Xu Li
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Le Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Yuan-Hao Chen
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Yue You
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Yin Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Yue-Ying Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Li Yang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Kun Tong
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Di-Shi Chen
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Jing-Ru Hao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Nan Sun
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China
| | - Zi-Ming Zhao
- Jiangsu Province Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China.
| | - Can Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, Xuzhou Medical University, 221004, Xuzhou, Jiangsu, China.
| |
Collapse
|
16
|
Cornblath EJ, Li HL, Changolkar L, Zhang B, Brown HJ, Gathagan RJ, Olufemi MF, Trojanowski JQ, Bassett DS, Lee VMY, Henderson MX. Computational modeling of tau pathology spread reveals patterns of regional vulnerability and the impact of a genetic risk factor. SCIENCE ADVANCES 2021; 7:eabg6677. [PMID: 34108219 PMCID: PMC8189700 DOI: 10.1126/sciadv.abg6677] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/21/2021] [Indexed: 05/09/2023]
Abstract
Neuropathological staging studies have suggested that tau pathology spreads through the brain in Alzheimer's disease (AD) and other tauopathies, but it is unclear how neuroanatomical connections, spatial proximity, and regional vulnerability contribute. In this study, we seed tau pathology in the brains of nontransgenic mice with AD tau and quantify pathology development over 9 months in 134 brain regions. Network modeling of pathology progression shows that diffusion through the connectome is the best predictor of tau pathology patterns. Further, deviations from pure neuroanatomical spread are used to estimate regional vulnerability to tau pathology and identify related gene expression patterns. Last, we show that pathology spread is altered in mice harboring a mutation in leucine-rich repeat kinase 2. While tau pathology spread is still constrained by anatomical connectivity in these mice, it spreads preferentially in a retrograde direction. This study provides a framework for understanding neuropathological progression in tauopathies.
Collapse
Affiliation(s)
- Eli J Cornblath
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Howard L Li
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lakshmi Changolkar
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bin Zhang
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hannah J Brown
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ronald J Gathagan
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Modupe F Olufemi
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Q Trojanowski
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danielle S Bassett
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Electrical and Systems Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Santa Fe Institute, Santa Fe, NM 87501, USA
| | - Virginia M Y Lee
- Institute on Aging and Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael X Henderson
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
17
|
López-Cervantes M, Quintanar-Stephano A, Alcauter-Solórzano S, Hernández-Pando R, Aguilar-Roblero R, Gasca-Martínez D, Ortíz JJ, Vázquez-Martínez O, Ximénez-Camilli C, Díaz-Muñoz M. Cerebellar spongiform degeneration is accompanied by metabolic, cellular, and motor disruption in male rats with portacaval anastomosis. J Neurosci Res 2021; 99:2287-2304. [PMID: 34061383 DOI: 10.1002/jnr.24853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
The episodes of cerebral dysfunction, known as encephalopathy, are usually coincident with liver failure. The primary metabolic marker of liver diseases is the increase in blood ammonium, which promotes neuronal damage. In the present project, we used an experimental model of hepatic encephalopathy in male rats by portacaval anastomosis (PCA) surgery. Sham rats had a false operation. After 13 weeks of surgery, the most distinctive finding was vacuolar/spongiform neurodegeneration exclusively in the molecular layer of the cerebellum. This cerebellar damage was further characterized by metabolic, histopathological, and behavioral approaches. The results were as follows: (a) Cellular alterations, namely loss of Purkinje cells, morphological changes, such as swelling of astrocytes and Bergmann glia, and activation of microglia; (b) Cytotoxic edema, shown by an increase in aquaporin-4 and N-acetylaspartate and a reduction in taurine and choline-derivate osmolytes; (c) Metabolic adjustments, noted by the elevation of circulating ammonium, enhanced presence of glutamine synthetase, and increase in glutamine and creatine/phosphocreatine; (d) Inflammasome activation, detected by the elevation of the marker NLRP3 and microglial activation; (e) Locomotor deficits in PCA rats as assessed by the Rotarod and open field tests. These results lead us to suggest that metabolic disturbances associated with PCA can generate the cerebellar damage that is similar to morphophysiological modifications observed in amyloidogenic disorders. In conclusion, we have characterized a distinctive cerebellar multi-disruption accompanied by high levels of ammonium and associated with spongiform neurodegeneration in a model of hepatic hypofunctioning.
Collapse
Affiliation(s)
- Mayra López-Cervantes
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Andrés Quintanar-Stephano
- Departmento de Fisiología y Farmacología, Centro de Ciencia Básica, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Sarael Alcauter-Solórzano
- Laboratorio Nacional de Imagenología por Resonancia Magnética, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Rogelio Hernández-Pando
- Seccion de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición "Salvador Zubirán", Ciudad de México, Mexico
| | - Raúl Aguilar-Roblero
- División de Neurociencias, Instituto de Fisiología Celular, UNAM, Ciudad de México, Mexico
| | - Deisy Gasca-Martínez
- Unidad de Análisis Conductual, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Juan J Ortíz
- Laboratorio Nacional de Imagenología por Resonancia Magnética, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Olivia Vázquez-Martínez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Cecilia Ximénez-Camilli
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| | - Mauricio Díaz-Muñoz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Campus UNAM-Juriquilla, Querétaro, Mexico
| |
Collapse
|
18
|
Bachstetter AD, Garrett FG, Jicha GA, Nelson PT. Space-occupying brain lesions, trauma-related tau astrogliopathy, and ARTAG: a report of two cases and a literature review. Acta Neuropathol Commun 2021; 9:49. [PMID: 33757579 PMCID: PMC7986305 DOI: 10.1186/s40478-021-01152-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Astrocytes with intracellular accumulations of misfolded phosphorylated tau protein have been observed in advanced-stage chronic traumatic encephalopathy (CTE) and in other neurodegenerative conditions. There is a growing awareness that astrocytic tau inclusions are also relatively common in the brains of persons over 70 years of age-affecting approximately one-third of autopsied individuals. The pathologic hallmarks of aging-related tau astrogliopathy (ARTAG) include phosphorylated tau protein within thorn-shaped astrocytes (TSA) in subpial, subependymal, perivascular, and white matter regions, whereas granular-fuzzy astrocytes are often seen in gray matter. CTE and ARTAG share molecular and histopathologic characteristics, suggesting that trauma-related mechanism(s) may predispose to the development of tau astrogliopathy. There are presently few experimental systems to study the pathobiology of astrocytic-tau aggregation, but human studies have made recent progress. For example, leucotomy (also referred to as lobotomy) is associated with a localized ARTAG-like neuropathology decades after the surgical brain injury, suggesting that chronic brain injury of any type may predispose to later life ARTAG. To examine this idea in a different context, we report clinical and pathologic features of two middle-aged men who came to autopsy with large (> 6 cm in greatest dimension) arachnoid cysts that had physically displaced and injured the subjects' left temporal lobes through chronic mechanical stress. Despite the similarity of the size and location of the arachnoid cysts, these individuals had dissimilar neurologic outcomes and neuropathologic findings. We review the evidence for ARTAG in response to brain injury, and discuss how the location and molecular properties of astroglial tau inclusions might alter the physiology of resident astrocytes. These cases and literature review point toward possible mechanism(s) of tau aggregation in astrocytes in response to chronic brain trauma.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone St., Lexington, KY, 40536, USA.
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.
| | - Filip G Garrett
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
19
|
Moelgg K, Jummun F, Humpel C. Spreading of Beta-Amyloid in Organotypic Mouse Brain Slices and Microglial Elimination and Effects on Cholinergic Neurons. Biomolecules 2021; 11:434. [PMID: 33804246 PMCID: PMC7999593 DOI: 10.3390/biom11030434] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 01/29/2023] Open
Abstract
The extracellular deposition of β-amyloid (Aβ) is one of the major characteristics in Alzheimer´s disease (AD). The "spreading hypothesis" suggests that a pathological protein (similar to prions) spreads over the entire brain. The aim of the present study was to use organotypic brain slices of postnatal day 8-10 mice. Using collagen hydrogels, we applied different Aβ peptides onto brain slices and analyzed spreading as well as glial reactions after eight weeks of incubation. Our data showed that from all tested Aβ peptides, human Aβ42 had the most potent activity to spread over into adjacent "target" areas. This effect was potentiated when brain slices from transgenic AD mice (APP_SweDI) were cultured. When different brain areas were connected to the "target slice" the spreading activity was more intense, originating from ventral striatum and brain stem. Reactive glial-fibrillary acidic protein (GFAP) astrogliosis increased over time, but Aβ depositions co-localized only with Iba1+ microglia but not with astrocytes. Application of human Aβ42 did not cause a degeneration of cholinergic neurons. We concluded that human Aβ42 spreads over into other "target areas", causing activation of glial cells. Most of the spread Aβ42 was taken up by microglia, and thus toxic free Aβ could not damage cholinergic neurons.
Collapse
Affiliation(s)
| | | | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer’s Research, Department Psychiatry I, Medical University of Innsbruck, Anichstr 35, A-6020 Innsbruck, Austria; (K.M.); (F.J.)
| |
Collapse
|
20
|
Ulm BS, Borchelt DR, Moore BD. Remodeling Alzheimer-amyloidosis models by seeding. Mol Neurodegener 2021; 16:8. [PMID: 33588898 PMCID: PMC7885558 DOI: 10.1186/s13024-021-00429-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/01/2021] [Indexed: 11/27/2022] Open
Abstract
Alzheimer’s disease (AD) is among the most prevalent neurodegenerative diseases, with brain pathology defined by extracellular amyloid beta deposits and intracellular tau aggregates. To aid in research efforts to improve understanding of this disease, transgenic murine models have been developed that replicate aspects of AD pathology. Familial AD is associated with mutations in the amyloid precursor protein and in the presenilins (associated with amyloidosis); transgenic amyloid models feature one or more of these mutant genes. Recent advances in seeding methods provide a means to alter the morphology of resultant amyloid deposits and the age that pathology develops. In this review, we discuss the variety of factors that influence the seeding of amyloid beta pathology, including the source of seed, the time interval after seeding, the nature of the transgenic host, and the preparation of the seeding inoculum.
Collapse
Affiliation(s)
- Brittany S Ulm
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - David R Borchelt
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Brenda D Moore
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
21
|
Ritchie DL, Barria MA. Prion Diseases: A Unique Transmissible Agent or a Model for Neurodegenerative Diseases? Biomolecules 2021; 11:biom11020207. [PMID: 33540845 PMCID: PMC7912988 DOI: 10.3390/biom11020207] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
The accumulation and propagation in the brain of misfolded proteins is a pathological hallmark shared by many neurodegenerative diseases such as Alzheimer's disease (Aβ and tau), Parkinson's disease (α-synuclein), and prion disease (prion protein). Currently, there is no epidemiological evidence to suggest that neurodegenerative disorders are infectious, apart from prion diseases. However, there is an increasing body of evidence from experimental models to suggest that other pathogenic proteins such as Aβ and tau can propagate in vivo and in vitro in a prion-like mechanism, inducing the formation of misfolded protein aggregates such as amyloid plaques and neurofibrillary tangles. Such similarities have raised concerns that misfolded proteins, other than the prion protein, could potentially transmit from person-to-person as rare events after lengthy incubation periods. Such concerns have been heightened following a number of recent reports of the possible inadvertent transmission of Aβ pathology via medical and surgical procedures. This review will provide a historical perspective on the unique transmissible nature of prion diseases, examining their impact on public health and the ongoing concerns raised by this rare group of disorders. Additionally, this review will provide an insight into current evidence supporting the potential transmissibility of other pathogenic proteins associated with more common neurodegenerative disorders and the potential implications for public health.
Collapse
|
22
|
Vandendriessche C, Bruggeman A, Van Cauwenberghe C, Vandenbroucke RE. Extracellular Vesicles in Alzheimer's and Parkinson's Disease: Small Entities with Large Consequences. Cells 2020; 9:cells9112485. [PMID: 33203181 PMCID: PMC7696752 DOI: 10.3390/cells9112485] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are incurable, devastating neurodegenerative disorders characterized by the formation and spreading of protein aggregates throughout the brain. Although the exact spreading mechanism is not completely understood, extracellular vesicles (EVs) have been proposed as potential contributors. Indeed, EVs have emerged as potential carriers of disease-associated proteins and are therefore thought to play an important role in disease progression, although some beneficial functions have also been attributed to them. EVs can be isolated from a variety of sources, including biofluids, and the analysis of their content can provide a snapshot of ongoing pathological changes in the brain. This underlines their potential as biomarker candidates which is of specific relevance in AD and PD where symptoms only arise after considerable and irreversible neuronal damage has already occurred. In this review, we discuss the known beneficial and detrimental functions of EVs in AD and PD and we highlight their promising potential to be used as biomarkers in both diseases.
Collapse
Affiliation(s)
- Charysse Vandendriessche
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Arnout Bruggeman
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Department of Neurology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Caroline Van Cauwenberghe
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; (C.V.); (A.B.); (C.V.C.)
- Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
- Correspondence: ; Tel.: +32-9-3313730
| |
Collapse
|
23
|
Martinez Pomier K, Ahmed R, Melacini G. Catechins as Tools to Understand the Molecular Basis of Neurodegeneration. Molecules 2020; 25:E3571. [PMID: 32781559 PMCID: PMC7465241 DOI: 10.3390/molecules25163571] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/25/2020] [Accepted: 08/03/2020] [Indexed: 12/12/2022] Open
Abstract
Protein misfolding as well as the subsequent self-association and deposition of amyloid aggregates is implicated in the progression of several neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Modulators of amyloidogenic aggregation serve as essential tools to dissect the underlying molecular mechanisms and may offer insight on potential therapeutic solutions. These modulators include green tea catechins, which are potent inhibitors of amyloid aggregation. Although catechins often exhibit poor pharmacokinetic properties and bioavailability, they are still essential tools for identifying the drivers of amyloid aggregation and for developing other aggregation modulators through structural mimicry. As an illustration of such strategies, here we review how catechins have been used to map the toxic surfaces of oligomeric amyloid-like species and develop catechin-based phenolic compounds with enhanced anti-amyloid activity.
Collapse
Affiliation(s)
- Karla Martinez Pomier
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada;
| | - Rashik Ahmed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4M1, Canada;
| | - Giuseppe Melacini
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON L8S 4M1, Canada;
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4M1, Canada;
| |
Collapse
|
24
|
Anti-aggregation Effects of Phenolic Compounds on α-synuclein. Molecules 2020; 25:molecules25102444. [PMID: 32456274 PMCID: PMC7288075 DOI: 10.3390/molecules25102444] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/13/2022] Open
Abstract
The aggregation and deposition of α-synuclein (αS) are major pathologic features of Parkinson’s disease, dementia with Lewy bodies, and other α-synucleinopathies. The propagation of αS pathology in the brain plays a key role in the onset and progression of clinical phenotypes. Thus, there is increasing interest in developing strategies that attenuate αS aggregation and propagation. Based on cumulative evidence that αS oligomers are neurotoxic and critical species in the pathogenesis of α-synucleinopathies, we and other groups reported that phenolic compounds inhibit αS aggregation including oligomerization, thereby ameliorating αS oligomer-induced cellular and synaptic toxicities. Heterogeneity in gut microbiota may influence the efficacy of dietary polyphenol metabolism. Our recent studies on the brain-penetrating polyphenolic acids 3-hydroxybenzoic acid (3-HBA), 3,4-dihydroxybenzoic acid (3,4-diHBA), and 3-hydroxyphenylacetic acid (3-HPPA), which are derived from gut microbiota-based metabolism of dietary polyphenols, demonstrated an in vitro ability to inhibit αS oligomerization and mediate aggregated αS-induced neurotoxicity. Additionally, 3-HPPA, 3,4-diHBA, 3-HBA, and 4-hydroxybenzoic acid significantly attenuated intracellular αS seeding aggregation in a cell-based system. This review focuses on recent research developments regarding neuroprotective properties, especially anti-αS aggregation effects, of phenolic compounds and their metabolites by the gut microbiome, including our findings in the pathogenesis of α-synucleinopathies.
Collapse
|
25
|
Ciccone L, Shi C, di Lorenzo D, Van Baelen AC, Tonali N. The Positive Side of the Alzheimer's Disease Amyloid Cross-Interactions: The Case of the Aβ 1-42 Peptide with Tau, TTR, CysC, and ApoA1. Molecules 2020; 25:E2439. [PMID: 32456156 PMCID: PMC7288020 DOI: 10.3390/molecules25102439] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 05/18/2020] [Accepted: 05/22/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) represents a progressive amyloidogenic disorder whose advancement is widely recognized to be connected to amyloid-β peptides and Tau aggregation. However, several other processes likely contribute to the development of AD and some of them might be related to protein-protein interactions. Amyloid aggregates usually contain not only single type of amyloid protein, but also other type of proteins and this phenomenon can be rationally explained by the process of protein cross-seeding and co-assembly. Amyloid cross-interaction is ubiquitous in amyloid fibril formation and so a better knowledge of the amyloid interactome could help to further understand the mechanisms of amyloid related diseases. In this review, we discuss about the cross-interactions of amyloid-β peptides, and in particular Aβ1-42, with other amyloids, which have been presented either as integrated part of Aβ neurotoxicity process (such as Tau) or conversely with a preventive role in AD pathogenesis by directly binding to Aβ (such as transthyretin, cystatin C and apolipoprotein A1). Particularly, we will focus on all the possible therapeutic strategies aiming to rescue the Aβ toxicity by taking inspiration from these protein-protein interactions.
Collapse
Affiliation(s)
- Lidia Ciccone
- Department of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Chenghui Shi
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| | - Davide di Lorenzo
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| | - Anne-Cécile Van Baelen
- Département Médicaments et Technologies pour la Santé (DMTS), CEA, INRAE, Université Paris Saclay, SIMoS, 91191 Gif-sur-Yvette, France;
| | - Nicolo Tonali
- CNRS, BioCIS, Université Paris-Saclay, rue Jean-Baptiste Clément 5, 92290 Châtenay-Malabry, France; (C.S.); (D.d.L.)
| |
Collapse
|
26
|
Limanaqi F, Biagioni F, Gambardella S, Familiari P, Frati A, Fornai F. Promiscuous Roles of Autophagy and Proteasome in Neurodegenerative Proteinopathies. Int J Mol Sci 2020; 21:E3028. [PMID: 32344772 PMCID: PMC7215558 DOI: 10.3390/ijms21083028] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022] Open
Abstract
Alterations in autophagy and the ubiquitin proteasome system (UPS) are commonly implicated in protein aggregation and toxicity which manifest in a number of neurological disorders. In fact, both UPS and autophagy alterations are bound to the aggregation, spreading and toxicity of the so-called prionoid proteins, including alpha synuclein (α-syn), amyloid-beta (Aβ), tau, huntingtin, superoxide dismutase-1 (SOD-1), TAR-DNA-binding protein of 43 kDa (TDP-43) and fused in sarcoma (FUS). Recent biochemical and morphological studies add to this scenario, focusing on the coordinated, either synergistic or compensatory, interplay that occurs between autophagy and the UPS. In fact, a number of biochemical pathways such as mammalian target of rapamycin (mTOR), transcription factor EB (TFEB), Bcl2-associated athanogene 1/3 (BAG3/1) and glycogen synthase kinase beta (GSk3β), which are widely explored as potential targets in neurodegenerative proteinopathies, operate at the crossroad between autophagy and UPS. These biochemical steps are key in orchestrating the specificity and magnitude of the two degradation systems for effective protein homeostasis, while intermingling with intracellular secretory/trafficking and inflammatory pathways. The findings discussed in the present manuscript are supposed to add novel viewpoints which may further enrich our insight on the complex interactions occurring between cell-clearing systems, protein misfolding and propagation. Discovering novel mechanisms enabling a cross-talk between the UPS and autophagy is expected to provide novel potential molecular targets in proteinopathies.
Collapse
Affiliation(s)
- Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
| | - Francesca Biagioni
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Stefano Gambardella
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Pietro Familiari
- Department of Human Neurosciences, Division of Neurosurgery, Sapienza University of Rome, 00185 Roma, Italy;
| | - Alessandro Frati
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy; (F.B.); (S.G.); (A.F.)
| |
Collapse
|