1
|
Basavarajappa BS, Subbanna S. Unveiling the Potential of Phytocannabinoids: Exploring Marijuana's Lesser-Known Constituents for Neurological Disorders. Biomolecules 2024; 14:1296. [PMID: 39456229 PMCID: PMC11506053 DOI: 10.3390/biom14101296] [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: 09/25/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Cannabis sativa is known for producing over 120 distinct phytocannabinoids, with Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) being the most prominent, primarily in their acidic forms. Beyond Δ9-THC and CBD, a wide array of lesser-known phytocannabinoids, along with terpenes, flavonoids, and alkaloids, demonstrate diverse pharmacological activities, interacting with the endocannabinoid system (eCB) and other biological pathways. These compounds, characterized by phenolic structures and hydroxyl groups, possess lipophilic properties, allowing them to cross the blood-brain barrier (BBB) effectively. Notably, their antioxidant, anti-inflammatory, and neuro-modulatory effects position them as promising agents in treating neurodegenerative disorders. While research has extensively examined the neuropsychiatric and neuroprotective effects of Δ9-THC, other minor phytocannabinoids remain underexplored. Due to the well-established neuroprotective potential of CBD, there is growing interest in the therapeutic benefits of non-psychotropic minor phytocannabinoids (NMPs) in brain disorders. This review highlights the emerging research on these lesser-known compounds and their neuroprotective potential. It offers insights into their therapeutic applications across various major neurological conditions.
Collapse
Affiliation(s)
- Balapal S. Basavarajappa
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA;
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shivakumar Subbanna
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA;
| |
Collapse
|
2
|
Singh A, Ansari VA, Mahmood T, Hasan SM, Wasim R, Maheshwari S, Akhtar J, Sheikh S, Vishwakarma VK. Targeting Abnormal Tau Phosphorylation for Alzheimer's Therapeutics. Horm Metab Res 2024; 56:482-488. [PMID: 38350636 DOI: 10.1055/a-2238-1384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Alzheimer's disease (AD) is a widespread neurodegenerative disorder characterized by progressive memory and cognitive decline, posing a formidable public health challenge. This review explores the intricate interplay between two pivotal players in AD pathogenesis: β-amyloid (Aβ) and tau protein. While the amyloid cascade theory has long dominated AD research, recent developments have ignited debates about its centrality. Aβ plaques and tau NFTs are hallmark pathologies in AD. Aducanumab and lecanemab, monoclonal antibodies targeting Aβ, have been approved, albeit amidst controversy, raising questions about the therapeutic efficacy of Aβ-focused interventions. On the other hand, tau, specifically its hyperphosphorylation, disrupts microtubule stability and contributes to neuronal dysfunction. Various post-translational modifications of tau drive its aggregation into NFTs. Emerging treatments targeting tau, such as GSK-3β and CDK5 inhibitors, have shown promise in preclinical and clinical studies. Restoring the equilibrium between protein kinases and phosphatases, notably protein phosphatase-2A (PP2A), is a promising avenue for AD therapy, as tau is primarily regulated by its phosphorylation state. Activation of tau-specific phosphatases offers potential for mitigating tau pathology. The evolving landscape of AD drug development emphasizes tau-centric therapies and reevaluation of the amyloid cascade hypothesis. Additionally, exploring the role of neuroinflammation and its interaction with tau pathology present promising research directions.
Collapse
Affiliation(s)
- Aditya Singh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | | | | | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Suvaiv Sheikh
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | |
Collapse
|
3
|
Maheshwari S, Singh A, Ansari VA, Mahmood T, Wasim R, Akhtar J, Verma A. Navigating the dementia landscape: Biomarkers and emerging therapies. Ageing Res Rev 2024; 94:102193. [PMID: 38215913 DOI: 10.1016/j.arr.2024.102193] [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: 12/28/2023] [Accepted: 01/06/2024] [Indexed: 01/14/2024]
Abstract
The field of dementia research has witnessed significant developments in our understanding of neurodegenerative disorders, with a particular focus on Alzheimer's disease (AD) and Frontotemporal Dementia (FTD). Dementia, a collection of symptoms arising from the degeneration of brain cells, presents a significant healthcare challenge, especially as its prevalence escalates with age. This abstract delves into the complexities of these disorders, the role of biomarkers in their diagnosis and monitoring, as well as emerging neurophysiological insights. In the context of AD, anti-amyloid therapy has gained prominence, aiming to reduce the accumulation of amyloid-beta (Aβ) plaques in the brain, a hallmark of the disease. Notably, Leqembi recently received full FDA approval, marking a significant breakthrough in AD treatment. Additionally, ongoing phase 3 clinical trials are investigating novel therapies, including Masitinib and NE3107, focusing on cognitive and functional improvements in AD patients. In the realm of FTD, research has unveiled distinct neuropathological features, including the involvement of proteins like TDP-43 and progranulin, providing valuable insights into the diagnosis and management of this heterogeneous condition. Biomarkers, including neurofilaments and various tau fragments, have shown promise in enhancing diagnostic accuracy. Neurophysiological techniques, such as transcranial magnetic stimulation (TMS), have contributed to our understanding of AD and FTD. TMS has uncovered unique neurophysiological signatures, highlighting impaired plasticity, hyperexcitability, and altered connectivity in AD, while FTD displays differences in neurotransmitter systems, particularly GABAergic and glutamatergic circuits. Lastly, ongoing clinical trials in anti-amyloid therapy for AD, such as Simufilam, Solanezumab, Gantenerumab, and Remternetug, offer hope for individuals affected by this devastating disease, with the potential to alter the course of cognitive decline. These advancements collectively illuminate the evolving landscape of dementia research and the pursuit of effective treatments for these challenging conditions.
Collapse
Affiliation(s)
- Shubhrat Maheshwari
- Faculty of Pharmaceutical Sciences Rama University Mandhana, Bithoor Road, Kanpur, Uttar Pradesh 209217, India; Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| | - Aditya Singh
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Vaseem Ahamad Ansari
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Tarique Mahmood
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Rufaida Wasim
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Juber Akhtar
- Department of Pharmaceutics, Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| | - Amita Verma
- Sam Higginbottom University of Agriculture, Technology and Sciences, Prayagraj, 21107, U.P., India.
| |
Collapse
|
4
|
Zilioli A, Misirocchi F, Pancaldi B, Mutti C, Ganazzoli C, Morelli N, Pellegrini FF, Messa G, Scarlattei M, Mohanty R, Ruffini L, Westman E, Spallazzi M. Predicting amyloid-PET status in a memory clinic: The role of the novel antero-posterior index and visual rating scales. J Neurol Sci 2023; 455:122806. [PMID: 38006829 DOI: 10.1016/j.jns.2023.122806] [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: 09/18/2023] [Revised: 10/27/2023] [Accepted: 11/16/2023] [Indexed: 11/27/2023]
Abstract
INTRODUCTION Visual rating scales are increasingly utilized in clinical practice to assess atrophy in crucial brain regions among patients with cognitive disorders. However, their capacity to predict Alzheimer's disease (AD)-related pathology remains unexplored, particularly within a heterogeneous memory clinic population. This study aims to assess the accuracy of a novel visual rating assessment, the antero-posterior index (API) scale, in predicting amyloid-PET status. Furthermore, the study seeks to determine the optimal cohort-based cutoffs for the medial temporal atrophy (MTA) and parietal atrophy (PA) scales and to integrate the main visual rating scores into a predictive model. METHODS We conducted a retrospective analysis of brain MRI and high-resolution TC scans from 153 patients with cognitive disorders who had undergone amyloid-PET assessments due to suspected AD pathology in a real-world memory clinic setting. RESULTS The API scale (cutoff ≥1) exhibited the highest accuracy (AUC = 0.721) among the visual rating scales. The combination of the cohort-based MTA and PA threshold with the API yielded favorable accuracy (AUC = 0.787). Analyzing a cohort of MCI/Mild dementia patients below 75 years of age, the API scale and the predictive model improved their accuracy (AUC = 0.741 and 0.813, respectively), achieving excellent results in the early-onset population (AUC = 0.857 and 0.949, respectively). CONCLUSION Our study emphasizes the significance of visual rating scales in predicting amyloid-PET positivity within a real-world memory clinic. Implementing the novel API scale, alongside our cohort-based MTA and PA thresholds, has the potential to substantially enhance diagnostic accuracy.
Collapse
Affiliation(s)
- Alessandro Zilioli
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Francesco Misirocchi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy.
| | - Beatrice Pancaldi
- Department of Medicine and Surgery, Unit of Neurology, University of Parma, Parma, Italy
| | - Carlotta Mutti
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy; Sleep Disorders Center, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Nicola Morelli
- Department of Neurology, G. da Saliceto Hospital, Piacenza, Italy
| | | | - Giovanni Messa
- Center for Cognitive Disorders, AUSL Parma, Parma, Italy
| | - Maura Scarlattei
- Nuclear Medicine Unit, University Hospital of Parma, Parma, Italy
| | - Rosaleena Mohanty
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden
| | - Livia Ruffini
- Nuclear Medicine Unit, University Hospital of Parma, Parma, Italy
| | - Eric Westman
- Division of Clinical Geriatrics; Center for Alzheimer Research; Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Blickagången 16 (NEO building, floor 7th), 14152, Huddinge, Stockholm, Sweden; Department of Neuroimaging, Center for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Marco Spallazzi
- Department of Medicine and Surgery, Unit of Neurology, University-Hospital of Parma, Parma, Italy
| |
Collapse
|
5
|
Gaitsch H, Franklin RJM, Reich DS. Cell-free DNA-based liquid biopsies in neurology. Brain 2023; 146:1758-1774. [PMID: 36408894 PMCID: PMC10151188 DOI: 10.1093/brain/awac438] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/22/2022] Open
Abstract
This article reviews recent developments in the application of cell-free DNA-based liquid biopsies to neurological diseases. Over the past few decades, an explosion of interest in the use of accessible biofluids to identify and track molecular disease has revolutionized the fields of oncology, prenatal medicine and others. More recently, technological advances in signal detection have allowed for informative analysis of biofluids that are typically sparse in cells and other circulating components, such as CSF. In parallel, advancements in epigenetic profiling have allowed for novel applications of liquid biopsies to diseases without characteristic mutational profiles, including many degenerative, autoimmune, inflammatory, ischaemic and infectious disorders. These events have paved the way for a wide array of neurological conditions to benefit from enhanced diagnostic, prognostic, and treatment abilities through the use of liquid biomarkers: a 'liquid biopsy' approach. This review includes an overview of types of liquid biopsy targets with a focus on circulating cell-free DNA, methods used to identify and probe potential liquid biomarkers, and recent applications of such biomarkers to a variety of complex neurological conditions including CNS tumours, stroke, traumatic brain injury, Alzheimer's disease, epilepsy, multiple sclerosis and neuroinfectious disease. Finally, the challenges of translating liquid biopsies to use in clinical neurology settings-and the opportunities for improvement in disease management that such translation may provide-are discussed.
Collapse
Affiliation(s)
- Hallie Gaitsch
- NIH-Oxford-Cambridge Scholars Program, Wellcome-MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | | | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Milano C, Hoxhaj D, Del Chicca M, Pascazio A, Paoli D, Tommasini L, Vergallo A, Pizzanelli C, Tognoni G, Nuti A, Ceravolo R, Siciliano G, Hampel H, Baldacci F. Alzheimer's Disease and Neurosyphilis: Meaningful Commonalities and Differences of Clinical Phenotype and Pathophysiological Biomarkers. J Alzheimers Dis 2023; 94:611-625. [PMID: 37334599 DOI: 10.3233/jad-230170] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Neurosyphilis-associated cognitive and behavioral impairment- historically coined as "general paralysis of the insane"- share clinical and neuroradiological features with the neurodegenerative disease spectrum, in particular Alzheimer's disease (AD). Anatomopathological similarities have been extensively documented, i.e., neuronal loss, fibrillary alterations, and local amyloid-β deposition. Consequently, accurate classification and timely differential diagnosis may be challenging. OBJECTIVE To describe clinical, bio-humoral, brain MRI, FDG-PET, and amyloid-PET features in cases of neurosyphilis with an AD-like phenotypical presentation, as well as clinical outcome in terms of response to antibiotic therapy. METHODS We selected the studies comparing patients with AD and with neurosyphilis associated cognitive impairment, to investigate candidate biomarkers classifying the two neurological diseases. RESULTS The neuropsychological phenotype of general paralysis, characterized by episodic memory impairment and executive disfunction, substantially mimics clinical AD features. Neuroimaging often shows diffuse or medial temporal cortical atrophy, thus contributing to a high rate of misdiagnosis. Cerebrospinal fluid (CSF)-based analysis may provide supportive diagnostic value, since increased proteins or cells are often found in neurosyphilis, while published data on pathophysiological AD candidate biomarkers are controversial. Finally, psychometric testing using cross-domain cognitive tests, may highlight a wider range of compromised functions in neurosyphilis, involving language, attention, executive function, and spatial ability, which are atypical for AD. CONCLUSION Neurosyphilis should be considered a potential etiological differential diagnosis of cognitive impairment whenever imaging, neuropsychological or CSF features are atypical for AD, in order to promptly start antibiotic therapy and delay or halt cognitive decline and disease progression.
Collapse
Affiliation(s)
- Chiara Milano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Domeniko Hoxhaj
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marta Del Chicca
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Alessia Pascazio
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Davide Paoli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Tommasini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Chiara Pizzanelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gloria Tognoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Angelo Nuti
- Division of Neurology, Versilia Hospital, Lido di Camaiore, Italy
| | - Roberto Ceravolo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
7
|
Rasheed A, Zaheer AB, Munawwar A, Sarfraz Z, Sarfraz A, Robles-Velasco K, Cherrez-Ojeda I. The Allosteric Antagonist of the Sigma-2 Receptors-Elayta (CT1812) as a Therapeutic Candidate for Mild to Moderate Alzheimer's Disease: A Scoping Systematic Review. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010001. [PMID: 36675950 PMCID: PMC9866790 DOI: 10.3390/life13010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/10/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Nearly 35 million people worldwide live with Alzheimer's disease (AD). The prevalence of the disease is expected to rise two-fold by 2050. With only symptomatic treatment options available, it is essential to understand the developments and existing evidence that aims to target brain pathology and dementia outcomes. This scoping systematic review aimed to collate existing evidence of CT1812 for use in patients with AD and summarize the methodologies of ongoing trials. Adhering to PRISMA Statement 2020 guidelines, PubMed/MEDLINE, Embase, Cochrane, and ClinicalTrials.gov were systematically searched through up to 15 November 2022 by applying the following keywords: CT1812, Alzheimer's disease, dementia, and/or sigma-2 receptor. Three completed clinical trials were included along with three ongoing records of clinical trials. The three completed trials were in Phases I and II of testing. The sample size across all three trials was 135. CT1812 reached endpoints across the trials and obtained a maximum concentration in the cerebrospinal fluid with 97-98% receptor occupancy. The findings of this systematic review must be used with caution as the results, while mostly favorable so far, must be replicated in higher-powered, placebo-controlled Phase II-III trials.
Collapse
Affiliation(s)
- Anum Rasheed
- Department of Research, Services Institute of Medical Sciences, Lahore 54000, Pakistan
| | - Ahmad Bin Zaheer
- Department of Research, Al Nafees Medical College and Hospital, Isra University, Islamabad 44000, Pakistan
| | - Aqsa Munawwar
- Department of Research, Services Institute of Medical Sciences, Lahore 54000, Pakistan
| | - Zouina Sarfraz
- Department of Research and Publications, Fatima Jinnah Medical University, Lahore 54000, Pakistan
- Correspondence: (Z.S.); (I.C.-O.)
| | - Azza Sarfraz
- Department of Pediatrics and Child Health, The Aga Khan University, Karachi 74000, Pakistan
| | - Karla Robles-Velasco
- Department of Allergy, Immunology & Pulmonary Medicine, Universidad Espíritu Santo, Samborondón 092301, Ecuador
| | - Ivan Cherrez-Ojeda
- Department of Allergy, Immunology & Pulmonary Medicine, Universidad Espíritu Santo, Samborondón 092301, Ecuador
- Correspondence: (Z.S.); (I.C.-O.)
| |
Collapse
|
8
|
Chin E, Jaqua E, Safaeipour M, Ladue T. Conventional Versus New Treatment: Comparing the Effects of Acetylcholinesterase Inhibitors and N-Methyl-D-Aspartate Receptor Antagonist With Aducanumab. Cureus 2022; 14:e31065. [DOI: 10.7759/cureus.31065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2022] [Indexed: 11/06/2022] Open
|
9
|
Yuyama K, Sun H, Igarashi Y, Monde K, Hirase T, Nakayama M, Makino Y. Immuno-digital invasive cleavage assay for analyzing Alzheimer's amyloid ß-bound extracellular vesicles. Alzheimers Res Ther 2022; 14:140. [PMID: 36184615 PMCID: PMC9528138 DOI: 10.1186/s13195-022-01073-w] [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: 01/31/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The protracted preclinical stage of Alzheimer's disease (AD) provides the opportunity for early intervention to prevent the disease; however, the lack of minimally invasive and easily detectable biomarkers and their measurement technologies remain unresolved. Extracellular vesicles (EVs) are nanosized membrane vesicles released from a variety of cells and play important roles in cell-cell communication. Neuron-derived and ganglioside-enriched EVs capture amyloid-ß protein, a major AD agent, and transport it into glial cells for degradation; this suggests that EVs influence Aß accumulation in the brain. EV heterogeneity, however, requires the use of a highly sensitive technique for measuring specific EVs in biofluid. In this study, immuno-digital invasive cleavage assay (idICA) was developed for quantitating target-intact EVs. METHODS EVs were captured onto ganglioside GM1-specific cholera toxin B subunit (CTB)-conjugated magnetic beads and detected with a DNA oligonucleotide-labeled Aß antibody. Fluorescence signals for individual EVs were then counted using an invasive cleavage assay (ICA). This idICA examines the Aß-bound and GM1-containing EVs isolated from the culture supernatant of human APP-overexpressing N2a (APP-N2a) cells and APP transgenic mice sera. RESULTS The idICA quantitatively detected Aß-bound and GM1-containing EVs isolated from culture supernatants of APP-N2a cells and sera of AD model mice. The idICA levels of Aß-associated EVs in blood gradually increased from 3- to 12-month-old mice, corresponding to the progression of Aß accumulations in the brain of AD model mice. CONCLUSIONS The present findings suggest that peripheral EVs harboring Aß and GM1 reflect Aß burden in mice. The idICA is a valuable tool for easy quantitative detection of EVs as an accessible biomarker for preclinical AD diagnosis.
Collapse
Affiliation(s)
- Kohei Yuyama
- Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, 001-0021, Japan.
| | - Hui Sun
- Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, 001-0021, Japan
| | - Yasuyuki Igarashi
- Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, 001-0021, Japan
| | - Kenji Monde
- Faculty of Advanced Life Science, Hokkaido University, Kita-21, Nishi-11, Kita-ku, Sapporo, 001-0021, Japan
| | - Takumi Hirase
- Technical Research Institute, TOPPAN INC., 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| | - Masato Nakayama
- Technical Research Institute, TOPPAN INC., 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| | - Yoichi Makino
- Technical Research Institute, TOPPAN INC., 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| |
Collapse
|
10
|
Shi B, Zhao J, Xu Z, Chen C, Xu L, Xu C, Sun M, Kuang H. Chiral Nanoparticles Force Neural Stem Cell Differentiation to Alleviate Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202475. [PMID: 36008133 PMCID: PMC9561871 DOI: 10.1002/advs.202202475] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/08/2022] [Indexed: 06/04/2023]
Abstract
The differentiation of neural stem cells via nanomaterials has attracted attention and has become a potential tool. However, the chirality effect in neural stem cell differentiation has not been investigated. Here, this study shows that chiral nanoparticles (NPs) with strong chirality can efficiently accelerate the differentiation of mouse neural stem cells (NSCs) into neurons under near-infrared (NIR) light illumination. L-type NPs are 1.95 times greater than D-type NPs in promoting NSCs differentiation due to their 1.47-fold endocytosis efficiency. Whole gene expression map analysis reveals that circularly polarized light illumination and chiral NPs irradiation significantly upregulate Map2, Yap1, and Taz genes, resulting in mechanical force, cytoskeleton protein action, and accelerated NSCs differentiation. In vivo experiments show that successful differentiation can further alleviate symptoms in Alzheimer's disease mice. Moreover, the clearance of L-type NPs on amyloid and hyperphosphorylated p-tau protein reachs 68.24% and 66.43%, respectively, under the synergy of NIR irradiation. The findings suggest that strong chiral nanomaterials may have advantages in guiding cell development and can be used in biomedicine.
Collapse
Affiliation(s)
- Baimei Shi
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Jing Zhao
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Zhuojia Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chen Chen
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and BiodetectionJiangnan UniversityWuxiJiangsu214122China
- State Key Laboratory of Food Science and TechnologyJiangnan UniversityWuxiJiangsu214122China
| |
Collapse
|
11
|
Menardi A, Dotti L, Ambrosini E, Vallesi A. Transcranial magnetic stimulation treatment in Alzheimer's disease: a meta-analysis of its efficacy as a function of protocol characteristics and degree of personalization. J Neurol 2022; 269:5283-5301. [PMID: 35781536 PMCID: PMC9468063 DOI: 10.1007/s00415-022-11236-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/06/2022]
Abstract
Alzheimer's disease (AD) represents the most common type of neurodegenerative disorder. Although our knowledge on the causes of AD remains limited and no curative treatments are available, several interventions have been proposed in trying to improve patients' symptomatology. Among those, transcranial magnetic stimulation (TMS) has been shown a promising, safe and noninvasive intervention to improve global cognitive functioning. Nevertheless, we currently lack agreement between research studies on the optimal stimulation protocol yielding the highest efficacy in these patients. To answer this query, we conducted a systematic literature search in PubMed, PsycINFO and Scopus databases and meta-analysis of studies published in the last 10 years (2010-2021) according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Differently from prior published meta-analytic work, we investigated whether protocols that considered participants-specific neuroimaging scans for the selection of individualized stimulation targets held more successful outcomes compared to those relying on a generalized targeting selection criteria. We then compared the effect sizes of subsets of studies based on additional protocol characteristics (frequency, duration of intervention, number of stimulation sites, use of concomitant cognitive training and patients' educational level). Our results confirm TMS efficacy in improving global cognitive functioning in mild-to-moderate AD patients, but also highlight the flaws of current protocols characteristics, including a possible lack of sufficient personalization in stimulation protocols.
Collapse
Affiliation(s)
- Arianna Menardi
- Department of Neuroscience, University of Padova, 35121, Padua, Italy.
- Padova Neuroscience Center, University of Padova, Padua, Italy.
| | - Lisa Dotti
- Department of General Psychology, University of Padova, Padua, Italy
| | - Ettore Ambrosini
- Department of Neuroscience, University of Padova, 35121, Padua, Italy
- Padova Neuroscience Center, University of Padova, Padua, Italy
- Department of General Psychology, University of Padova, Padua, Italy
| | - Antonino Vallesi
- Department of Neuroscience, University of Padova, 35121, Padua, Italy
- Padova Neuroscience Center, University of Padova, Padua, Italy
| |
Collapse
|
12
|
Gong X, Zhang H, Liu X, Liu Y, Liu J, Fapohunda FO, Lü P, Wang K, Tang M. Is liquid biopsy mature enough for the diagnosis of Alzheimer's disease? Front Aging Neurosci 2022; 14:977999. [PMID: 35992602 PMCID: PMC9389010 DOI: 10.3389/fnagi.2022.977999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
Abstract
The preclinical diagnosis and clinical practice for Alzheimer's disease (AD) based on liquid biopsy have made great progress in recent years. As liquid biopsy is a fast, low-cost, and easy way to get the phase of AD, continual efforts from intense multidisciplinary studies have been made to move the research tools to routine clinical diagnostics. On one hand, technological breakthroughs have brought new detection methods to the outputs of liquid biopsy to stratify AD cases, resulting in higher accuracy and efficiency of diagnosis. On the other hand, diversiform biofluid biomarkers derived from cerebrospinal fluid (CSF), blood, urine, Saliva, and exosome were screened out and biologically verified. As a result, more detailed knowledge about the molecular pathogenesis of AD was discovered and elucidated. However, to date, how to weigh the reports derived from liquid biopsy for preclinical AD diagnosis is an ongoing question. In this review, we briefly introduce liquid biopsy and the role it plays in research and clinical practice. Then, we summarize the established fluid-based assays of the current state for AD diagnostic such as ELISA, single-molecule array (Simoa), Immunoprecipitation-Mass Spectrometry (IP-MS), liquid chromatography-MS, immunomagnetic reduction (IMR), multimer detection system (MDS). In addition, we give an updated list of fluid biomarkers in the AD research field. Lastly, the current outstanding challenges and the feasibility to use a stand-alone biomarker in the joint diagnostic strategy are discussed.
Collapse
Affiliation(s)
- Xun Gong
- Department of Rheumatology and Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hantao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Xiaoyan Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Yi Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
- Institute of Animal Husbandry, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Junlin Liu
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | | | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Kun Wang
- Children’s Center, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Min Tang
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
13
|
Recombinant Integrin β1 Signal Peptide Blocks Gliosis Induced by Aβ Oligomers. Int J Mol Sci 2022; 23:ijms23105747. [PMID: 35628557 PMCID: PMC9146559 DOI: 10.3390/ijms23105747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/27/2023] Open
Abstract
Glial cells participate actively in the early cognitive decline in Alzheimer’s disease (AD) pathology. In fact, recent studies have found molecular and functional abnormalities in astrocytes and microglia in both animal models and brains of patients suffering from this pathology. In this regard, reactive gliosis intimately associated with amyloid plaques has become a pathological hallmark of AD. A recent study from our laboratory reports that astrocyte reactivity is caused by a direct interaction between amyloid beta (Aβ) oligomers and integrin β1. Here, we have generated four recombinant peptides including the extracellular domain of integrin β1, and evaluated their capacity both to bind in vitro to Aβ oligomers and to prevent in vivo Aβ oligomer-induced gliosis and endoplasmic reticulum stress. We have identified the minimal region of integrin β1 that binds to Aβ oligomers. This region is called signal peptide and corresponds to the first 20 amino acids of the integrin β1 N-terminal domain. This recombinant integrin β1 signal peptide prevented Aβ oligomer-induced ROS generation in primary astrocyte cultures. Furthermore, we carried out intrahippocampal injection in adult mice of recombinant integrin β1 signal peptide combined with or without Aβ oligomers and we evaluated by immunohistochemistry both astrogliosis and microgliosis as well as endoplasmic reticulum stress. The results show that recombinant integrin β1 signal peptide precluded both astrogliosis and microgliosis and endoplasmic reticulum stress mediated by Aβ oligomers in vivo. We have developed a molecular tool that blocks the activation of the molecular cascade that mediates gliosis via Aβ oligomer/integrin β1 signaling.
Collapse
|
14
|
Valverde A, Gordón Pidal JM, Montero-Calle A, Arévalo B, Serafín V, Calero M, Moreno-Guzmán M, López MÁ, Escarpa A, Yáñez-Sedeño P, Barderas R, Campuzano S, Pingarrón JM. Paving the way for reliable Alzheimer's disease blood diagnosis by quadruple electrochemical immunosensing. ChemElectroChem 2022. [DOI: 10.1002/celc.202200055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Alejandro Valverde
- Universidad Complutense de Madrid Facultad de Ciencias Quimicas Analytical Chemistry SPAIN
| | - José M. Gordón Pidal
- Universidad de Alcala Analytical Chemistry, Physical Chemistry and Chemical Engineering SPAIN
| | - Ana Montero-Calle
- Instituto de Salud Carlos III Chronic Disease Programme, UFIEC SPAIN
| | - Beatriz Arévalo
- Universidad Complutense de Madrid Facultad de Ciencias Quimicas Analytical Chemistry SPAIN
| | - Verónica Serafín
- Universidad Complutense de Madrid Facultad de Ciencias Quimicas Analytical Chemistry SPAIN
| | | | | | - Miguel Ángel López
- Universidad de Alcala Analytical Chemsitry, Physical Chemistry and Chemical Engineering SPAIN
| | - Alberto Escarpa
- Universidad de Alcala Analytical Chemistry, Physical Chemistry and Chemical Engineering SPAIN
| | - Paloma Yáñez-Sedeño
- Universidad Complutense de Madrid Facultad de Ciencias Quimicas Analytical Chemistry SPAIN
| | - Rodrigo Barderas
- Instituto de Salud Carlos III Chronic Disease Programme, UFIEC SPAIN
| | - Susana Campuzano
- Universidad Complutense de Madrid Facultad de Ciencias Quimicas Analytical Chemistry SPAIN
| | - José Manuel Pingarrón
- Universidad Complutense de Madrid Química Analítica Av. Complutense s/n 28040 Madrid SPAIN
| |
Collapse
|
15
|
OUP accepted manuscript. Brain 2022; 145:2250-2275. [DOI: 10.1093/brain/awac096] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
|
16
|
Milošević M, Arsić A, Cvetković Z, Vučić V. Memorable Food: Fighting Age-Related Neurodegeneration by Precision Nutrition. Front Nutr 2021; 8:688086. [PMID: 34422879 PMCID: PMC8374314 DOI: 10.3389/fnut.2021.688086] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022] Open
Abstract
Healthcare systems worldwide are seriously challenged by a rising prevalence of neurodegenerative diseases (NDDs), which mostly, but not exclusively, affect the ever-growing population of the elderly. The most known neurodegenerative diseases are Alzheimer's (AD) and Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis, but some viral infections of the brain and traumatic brain injury may also cause NDD. Typical for NDD are the malfunctioning of neurons and their irreversible loss, which often progress irreversibly to dementia and ultimately to death. Numerous factors are involved in the pathogenesis of NDD: genetic variability, epigenetic changes, extent of oxidative/nitrosative stress, mitochondrial dysfunction, and DNA damage. The complex interplay of all the above-mentioned factors may be a fingerprint of neurodegeneration, with different diseases being affected to different extents by particular factors. There is a voluminous body of evidence showing the benefits of regular exercise to brain health and cognitive functions. Moreover, the importance of a healthy diet, balanced in macro- and micro-nutrients, in preventing neurodegeneration and slowing down a progression to full-blown disease is evident. Individuals affected by NDD almost inevitably have low-grade inflammation and anomalies in lipid metabolism. Metabolic and lipid profiles in NDD can be improved by the Mediterranean diet. Many studies have associated the Mediterranean diet with a decreased risk of dementia and AD, but a cause-and-effect relationship has not been deduced. Studies with caloric restriction showed neuroprotective effects in animal models, but the results in humans are inconsistent. The pathologies of NDD are complex and there is a great inter-individual (epi)genetic variance within any population. Furthermore, the gut microbiome, being deeply involved in nutrient uptake and lipid metabolism, also represents a pillar of the gut microbiome-brain axis and is linked with the pathogenesis of NDD. Numerous studies on the role of different micronutrients (omega-3 fatty acids, bioactive polyphenols from fruit and medicinal plants) in the prevention, prediction, and treatment of NDD have been conducted, but we are still far away from a personalized diet plan for individual NDD patients. For this to be realized, large-scale cohorts that would include the precise monitoring of food intake, mapping of genetic variants, epigenetic data, microbiome studies, and metabolome, lipidome, and transcriptome data are needed.
Collapse
Affiliation(s)
- Maja Milošević
- Department of Neuroendocrinology, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Arsić
- Department of Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Zorica Cvetković
- Department of Hematology, Clinical Hospital Center Zemun, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vesna Vučić
- Department of Nutritional Biochemistry and Dietology, Centre of Research Excellence in Nutrition and Metabolism, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
17
|
Stoiljkovic M, Horvath TL, Hajós M. Therapy for Alzheimer's disease: Missing targets and functional markers? Ageing Res Rev 2021; 68:101318. [PMID: 33711510 PMCID: PMC8131215 DOI: 10.1016/j.arr.2021.101318] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
The development of the next generation therapy for Alzheimer's disease (AD) presents a huge challenge given the number of promising treatment candidates that failed in trials, despite recent advancements in understanding of genetic, pathophysiologic and clinical characteristics of the disease. This review reflects some of the most current concepts and controversies in developing disease-modifying and new symptomatic treatments. It elaborates on recent changes in the AD research strategy for broadening drug targets, and potentials of emerging non-pharmacological treatment interventions. Established and novel biomarkers are discussed, including emerging cerebrospinal fluid and plasma biomarkers reflecting tau pathology, neuroinflammation and neurodegeneration. These fluid biomarkers together with neuroimaging findings can provide innovative objective assessments of subtle changes in brain reflecting disease progression. A particular emphasis is given to neurophysiological biomarkers which are well-suited for evaluating the brain overall neural network integrity and function. Combination of multiple biomarkers, including target engagement and outcome biomarkers will empower translational studies and facilitate successful development of effective therapies.
Collapse
Affiliation(s)
- Milan Stoiljkovic
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA; Department of Pharmacology, University of Nis School of Medicine, Nis, Serbia.
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Mihály Hajós
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, 06520, USA; Cognito Therapeutics, Cambridge, MA, 02138, USA
| |
Collapse
|
18
|
Small GTPases of the Rab and Arf Families: Key Regulators of Intracellular Trafficking in Neurodegeneration. Int J Mol Sci 2021; 22:ijms22094425. [PMID: 33922618 PMCID: PMC8122874 DOI: 10.3390/ijms22094425] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Small guanosine triphosphatases (GTPases) of the Rab and Arf families are key regulators of vesicle formation and membrane trafficking. Membrane transport plays an important role in the central nervous system. In this regard, neurons require a constant flow of membranes for the correct distribution of receptors, for the precise composition of proteins and organelles in dendrites and axons, for the continuous exocytosis/endocytosis of synaptic vesicles and for the elimination of dysfunctional proteins. Thus, it is not surprising that Rab and Arf GTPases have been associated with neurodegenerative diseases such as Alzheimer’s and Parkinson’s. Both pathologies share characteristics such as the presence of protein aggregates and/or the fragmentation of the Golgi apparatus, hallmarks that have been related to both Rab and Arf GTPases functions. Despite their relationship with neurodegenerative disorders, very few studies have focused on the role of these GTPases in the pathogenesis of neurodegeneration. In this review, we summarize their importance in the onset and progression of Alzheimer’s and Parkinson’s diseases, as well as their emergence as potential therapeutical targets for neurodegeneration.
Collapse
|
19
|
Hugon J, Paquet C. The PKR/P38/RIPK1 Signaling Pathway as a Therapeutic Target in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22063136. [PMID: 33808629 PMCID: PMC8003462 DOI: 10.3390/ijms22063136] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/15/2021] [Accepted: 03/16/2021] [Indexed: 12/23/2022] Open
Abstract
Neuropathological lesions in Alzheimer’s disease (AD) include amyloid plaques formed by the accumulation of amyloid peptides, neurofibrillary tangles made of hyperphosphorylated tau protein, synaptic and neuronal degenerations, and neuroinflammation. The cause of AD is unknown, but according to the amyloid hypothesis, amyloid oligomers could lead to the activation of kinases such as eukaryotic translation initiation factor 2-alpha kinase 2 (PKR), p38, and receptor-interacting serine/threonine-protein kinase 1 (RIPK1), which all belong to the same stress-activated pathway. Many toxic kinase activations have been described in AD patients and in experimental models. A p38 mitogen-activated protein kinase inhibitor was recently tested in clinical trials but with unsuccessful results. The complex PKR/P38/RIPK1 (PKR/dual specificity mitogen-activated protein kinase kinase 6 (MKK6)/P38/MAP kinase-activated protein kinase 2 (MK2)/RIPK1) is highly activated in AD brains and in the brains of AD transgenic animals. To delineate the implication of this pathway in AD, we carried out a search on PubMed including PKR/MKK6/p38/MK2/RIPK1, Alzheimer, and therapeutics. The involvement of this signaling pathway in the genesis of AD lesions, including Aβ accumulations and tau phosphorylation as well as cognitive decline, is demonstrated by the reports described in this review. A future combination strategy with kinase inhibitors should be envisaged to modulate the consequences for neurons and other brain cells linked to the abnormal activation of this pathway.
Collapse
Affiliation(s)
- Jacques Hugon
- Correspondence: ; Tel.: +33-140-054-313; Fax: +33-140-054-339
| | | |
Collapse
|
20
|
Imbimbo BP, Ippati S, Watling M, Balducci C. Accelerating Alzheimer's disease drug discovery and development: what's the way forward? Expert Opin Drug Discov 2021; 16:727-735. [PMID: 33653187 DOI: 10.1080/17460441.2021.1887132] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: As the global burden of Alzheimer's disease (AD) grows, an effective disease-modifying therapy remains a distant prospect following the repeated failure of multiple therapeutics targeting β-amyloid and (it seems) tau over many years of costly effort. The repeated failure of single-target therapies to meaningfully modify disease progression raises major questions about the validity of many aspects of drug development in this area, especially target selection.Area covered: The authors explore the critical questions raised by a review of the collective experience to date, relating to why findings with non-clinical models and clinical biomarkers so frequently fail to translate to positive outcomes in clinical trials, which alternatives should be considered, and how we can design and conduct clinical trials that can successfully identify and quantify meaningful benefits in the future.Expert opinion: It is our opinion that we must recognize and accept the need to consider less specific, more multimodal approaches to addressing neurodegeneration in AD if we are to make progress - and we must avoid repeating the well intentioned, but ultimately erroneous, assumptions of the past.
Collapse
Affiliation(s)
- Bruno P Imbimbo
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | - Stefania Ippati
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Mark Watling
- CNS & Pain Department, TranScrip Partners, Reading, UK
| | - Claudia Balducci
- Department of Neuroscience, IRCCS, Istituto Di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| |
Collapse
|
21
|
Valverde A, Montero-Calle A, Barderas R, Calero M, Yáñez-Sedeño P, Campuzano S, Pingarrón J. Electrochemical immunoplatform to unravel neurodegeneration and Alzheimer's disease through the determination of neurofilament light protein. Electrochim Acta 2021. [DOI: 10.1016/j.electacta.2021.137815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
22
|
Zhao Y, Jaber V, Alexandrov PN, Vergallo A, Lista S, Hampel H, Lukiw WJ. microRNA-Based Biomarkers in Alzheimer's Disease (AD). Front Neurosci 2020; 14:585432. [PMID: 33192270 PMCID: PMC7664832 DOI: 10.3389/fnins.2020.585432] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is a multifactorial, age-related neurological disease characterized by complex pathophysiological dynamics taking place at multiple biological levels, including molecular, genetic, epigenetic, cellular and large-scale brain networks. These alterations account for multiple pathophysiological mechanisms such as brain protein accumulation, neuroinflammatory/neuro-immune processes, synaptic dysfunction, and neurodegeneration that eventually lead to cognitive and behavioral decline. Alterations in microRNA (miRNA) signaling have been implicated in the epigenetics and molecular genetics of all neurobiological processes associated with AD pathophysiology. These changes encompass altered miRNA abundance, speciation and complexity in anatomical regions of the CNS targeted by the disease, including modified miRNA expression patterns in brain tissues, the systemic circulation, the extracellular fluid (ECF) and the cerebrospinal fluid (CSF). miRNAs have been investigated as candidate biomarkers for AD diagnosis, disease prediction, prognosis and therapeutic purposes because of their involvement in multiple brain signaling pathways in both health and disease. In this review we will: (i) highlight the significantly heterogeneous nature of miRNA expression and complexity in AD tissues and biofluids; (ii) address how information may be extracted from these data to be used as a diagnostic, prognostic and/or screening tools across the entire continuum of AD, from the preclinical stage, through the prodromal, i.e., mild cognitive impairment (MCI) phase all the way to clinically overt dementia; and (iii) consider how specific miRNA expression patterns could be categorized using miRNA reporters that span AD pathophysiological initiation and disease progression.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Cell Biology and Anatomy, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Vivian Jaber
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | | | - Andrea Vergallo
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Simone Lista
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’Hôpital, Paris, France
- Institute of Memory and Alzheimer’s Disease (IM2A), Department of Neurology, Pitié-Salpêtrière Hospital, AP-HP, Boulevard de l’hôpital, Paris, France
| | - Harald Hampel
- Sorbonne University, GRC n° 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Russian Academy of Medical Sciences, Moscow, Russia
- Department of Ophthalmology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, LA, United States
- Department of Neurology, LSU Neuroscience Center Louisiana State University Health Science Center, New Orleans, LA, United States
| |
Collapse
|
23
|
Barbagallo C, Di Martino MT, Grasso M, Salluzzo MG, Scionti F, Cosentino FII, Caruso G, Barbagallo D, Di Pietro C, Ferri R, Caraci F, Purrello M, Ragusa M. Uncharacterized RNAs in Plasma of Alzheimer's Patients Are Associated with Cognitive Impairment and Show a Potential Diagnostic Power. Int J Mol Sci 2020; 21:ijms21207644. [PMID: 33076555 PMCID: PMC7588983 DOI: 10.3390/ijms21207644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) diagnosis is actually based on clinical evaluation and brain-imaging tests, and it can often be confirmed only post-mortem. Therefore, new non-invasive molecular biomarkers are necessary to improve AD diagnosis. As circulating microRNA biomarkers have been proposed for many diseases, including AD, we aimed to identify new diagnostic non-small RNAs in AD. Whole transcriptome analysis was performed on plasma samples of five AD and five unaffected individuals (CTRL) using the Clariom D Pico Assay, followed by validation in real-time PCR on 37 AD patients and 37 CTRL. Six differentially expressed (DE) transcripts were identified: GS1-304P7.3 (upregulated), NONHSAT090268, TC0100011037, TC0400008478, TC1400008125, and UBE2V1 (downregulated). Peripheral blood mononuclear cells (PBMCs) may influence the expression of circulating RNAs and their analysis has been proposed to improve AD clinical management. Accordingly, DE transcript expression was also evaluated in PBMCs, showing no difference between AD and CTRL. ROC (receiver operating characteristic) curve analysis was performed to evaluate the diagnostic accuracy of each DE transcript and a signature including all of them. A correlation between cognitive impairment and GS1-304P7.3, NONHSAT090268, TC0100011037, and TC0400008478 was detected, suggesting a potential association between their extracellular abundance and AD clinical phenotype. Finally, this study identified six transcripts showing altered expression in the plasma of AD patients. Given the need for new, accurate blood biomarkers for AD diagnosis, these transcripts may be considered for further analyses in larger cohorts, also in combination with other biomarkers, aiming to identify specific RNA-based biomarkers to be eventually applied to clinical practice.
Collapse
Affiliation(s)
- Cristina Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.D.M.); (F.S.)
| | - Margherita Grasso
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Maria Grazia Salluzzo
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (M.T.D.M.); (F.S.)
| | | | - Giuseppe Caruso
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
| | - Davide Barbagallo
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Raffaele Ferri
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy
- Correspondence:
| | - Michele Purrello
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
| | - Marco Ragusa
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics G. Sichel, University of Catania, 95123 Catania, Italy; (C.B.); (D.B.); (C.D.P.); (M.P.); (M.R.)
- Oasi Research Institute—IRCCS, 94018 Troina, Italy; (M.G.); (M.G.S.); (F.I.I.C.); (G.C.); (R.F.)
| |
Collapse
|
24
|
Lukiw WJ, Vergallo A, Lista S, Hampel H, Zhao Y. Biomarkers for Alzheimer's Disease (AD) and the Application of Precision Medicine. J Pers Med 2020; 10:E138. [PMID: 32967128 PMCID: PMC7565758 DOI: 10.3390/jpm10030138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/08/2020] [Accepted: 09/15/2020] [Indexed: 02/08/2023] Open
Abstract
An accurate diagnosis of Alzheimer's disease (AD) currently stands as one of the most difficult and challenging in all of clinical neurology. AD is typically diagnosed using an integrated knowledge and assessment of multiple biomarkers and interrelated factors. These include the patient's age, gender and lifestyle, medical and genetic history (both clinical- and family-derived), cognitive, physical, behavioral and geriatric assessment, laboratory examination of multiple AD patient biofluids, especially within the systemic circulation (blood serum) and cerebrospinal fluid (CSF), multiple neuroimaging-modalities of the brain's limbic system and/or retina, followed up in many cases by post-mortem neuropathological examination to finally corroborate the diagnosis. More often than not, prospective AD cases are accompanied by other progressive, age-related dementing neuropathologies including, predominantly, a neurovascular and/or cardiovascular component, multiple-infarct dementia (MID), frontotemporal dementia (FTD) and/or strokes or 'mini-strokes' often integrated with other age-related neurological and non-neurological disorders including cardiovascular disease and cancer. Especially over the last 40 years, enormous research efforts have been undertaken to discover, characterize, and quantify more effectual and reliable biological markers for AD, especially during the pre-clinical or prodromal stages of AD so that pre-emptive therapeutic treatment strategies may be initiated. While a wealth of genetic, neurobiological, neurochemical, neuropathological, neuroimaging and other diagnostic information obtainable for a single AD patient can be immense: (i) it is currently challenging to integrate and formulate a definitive diagnosis for AD from this multifaceted and multidimensional information; and (ii) these data are unfortunately not directly comparable with the etiopathological patterns of other AD patients even when carefully matched for age, gender, familial genetics, and drug history. Four decades of AD research have repeatedly indicated that diagnostic profiles for AD are reflective of an extremely heterogeneous neurological disorder. This commentary will illuminate the heterogeneity of biomarkers for AD, comment on emerging investigative approaches and discuss why 'precision medicine' is emerging as our best paradigm yet for the most accurate and definitive prediction, diagnosis, and prognosis of this insidious and lethal brain disorder.
Collapse
Affiliation(s)
- Walter J. Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA;
- Department of Cell Biology and Anatomy, LSU-HSC, New Orleans, LA 70112, USA
- Department of Ophthalmology, LSU Neuroscience Center, LSU-HSC, New Orleans, LA 70112, USA
- Department Neurology, LSU Neuroscience Center, LSU-HSC, New Orleans, LA 70112, USA
| | - Andrea Vergallo
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière hospital, F-75013 Paris, France; (A.V.); (S.L.); (H.H.)
| | - Simone Lista
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière hospital, F-75013 Paris, France; (A.V.); (S.L.); (H.H.)
- Brain & Spine Institute (ICM), INSERM U 1127, CNRS UMR 7225, Boulevard de l’Hôpital, F-75013 Paris, France
- Department of Neurology, Institute of Memory and Alzheimer’s Disease (IM2A), Pitié-Salpêtrière Hospital, AP-HP, F-75013 Paris, France
| | - Harald Hampel
- Sorbonne University, GRC no 21, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière hospital, F-75013 Paris, France; (A.V.); (S.L.); (H.H.)
| | - Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science Center, New Orleans, LA 70112, USA;
- Department of Cell Biology and Anatomy, LSU-HSC, New Orleans, LA 70112, USA
| |
Collapse
|
25
|
Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases. Int J Mol Sci 2020. [DOI: 10.3390/ijms21176312
expr 858053618 + 832508766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases.
Collapse
|
26
|
Arrazola Sastre A, Luque Montoro M, Gálvez-Martín P, Lacerda HM, Lucia A, Llavero F, Zugaza JL. Small GTPases of the Ras and Rho Families Switch on/off Signaling Pathways in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E6312. [PMID: 32878220 PMCID: PMC7504559 DOI: 10.3390/ijms21176312] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/16/2022] Open
Abstract
Small guanosine triphosphatases (GTPases) of the Ras superfamily are key regulators of many key cellular events such as proliferation, differentiation, cell cycle regulation, migration, or apoptosis. To control these biological responses, GTPases activity is regulated by guanine nucleotide exchange factors (GEFs), GTPase activating proteins (GAPs), and in some small GTPases also guanine nucleotide dissociation inhibitors (GDIs). Moreover, small GTPases transduce signals by their downstream effector molecules. Many studies demonstrate that small GTPases of the Ras family are involved in neurodegeneration processes. Here, in this review, we focus on the signaling pathways controlled by these small protein superfamilies that culminate in neurodegenerative pathologies, such as Alzheimer's disease (AD) and Parkinson's disease (PD). Specifically, we concentrate on the two most studied families of the Ras superfamily: the Ras and Rho families. We summarize the latest findings of small GTPases of the Ras and Rho families in neurodegeneration in order to highlight these small proteins as potential therapeutic targets capable of slowing down different neurodegenerative diseases.
Collapse
Affiliation(s)
- Alazne Arrazola Sastre
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain
| | - Miriam Luque Montoro
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
| | - Patricia Gálvez-Martín
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, 180041 Granada, Spain;
- R&D Human Health, Bioibérica S.A.U., 08950 Barcelona, Spain
| | | | - Alejandro Lucia
- Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain;
- Research Institute of the Hospital 12 de Octubre (i+12), 28041 Madrid, Spain
| | - Francisco Llavero
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Faculty of Sport Science, European University of Madrid, 28670 Madrid, Spain;
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Science Park of the Universidad del País Vasco/Euskal Herriko Unibertsitatea (UPV/EHU), 48940 Leioa, Spain; (A.A.S.); (M.L.M.)
- Department of Genetics, Physical Anthropology, and Animal Physiology, Faculty of Science and Technology, UPV/EHU, 48940 Leioa, Spain
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|