ijms-logo

Journal Browser

Journal Browser

New Trends in Alzheimer's Disease Research: From Molecular Mechanisms to Therapeutics

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Neurobiology".

Deadline for manuscript submissions: closed (30 November 2023) | Viewed by 18848

Special Issue Editor

Special Issue Information

Dear Colleagues,

Alzheimer's disease (AD) is a health priority in developed societies, along with cardiovascular disease, cancer, stroke and major neuropsychiatric pathologies. The direct and indirect costs for the management of AD create a large economic burden for families, nations and health resources. The cost of worldwide dementia treatment currently exceeds USD 800 billion dollars (>1% of GDP). The average cost per patient/year ranges from USD 30,000 to USD 60,000, depending on stage of the disease, quality of medical care, social status and country. In terms of global costs (direct, indirect and social costs + costs of informal care), in 2019, the World Health Organization (WHO) estimated the total global societal cost of dementia to be about USD 1.3 trillion (>USD 2.8 trillion by 2030).

AD is the most prevalent form of dementia (50-60%). Vascular dementia (30-40%), other forms of dementia (10-15%), and mixed dementia, which is the most frequent form of dementia (>70%) in patients older than 75 years of age, are common presentations of dementia following frequent AD. AD is more frequent in women than in men; the prevalence of dementia is 30.5/1,000 in males and 48.2/1,000 in females.

The phenotype of AD is the consequence of the premature death of neurons associated with genomic, epigenomic, cerebrovascular and environmental factors. The clinical manifestation of dementia is characterized by progressive cognitive deterioration, behavioral changes and functional decline.

Conventionally, two forms of AD are differentiated: an early form (early onset AD, EOAD, <65 years) and a late-onset AD (LOAD, >65 years), within an apparent pathological continuum. EOAD is associated with familial forms of Mendelian genetics (familial AD, FAD), while LOAD shows a more complex pathogenesis, in which a multitude of polymorphic variants in over 600 genes distributed throughout the human genome converge with diverse environmental factors, which attribute the false phenotypic profile of sporadic AD to the disease.

Both forms of dementia exhibit common neuropathological hallmarks of amyloidopathy and tauopathy characterized by extracellular deposits of aggregated β-amyloid (Aβ) in senile plaques and vessels (amyloid angiopathy) and intracellular neurofibrillary tangles (NFTs), formed by the hyperphosphorylation of tau proteins in microtubules and neurofilaments, likely exerting synergistic effects on AD pathogenesis. Dendritic dystrophy and desarborization, microglia activation, astrogliosis, and neuronal loss are also typical neuropathological markers in the hippocampus and neocortex, where neurotransmitter deficits (cholinergic, monoaminergic, glutamatergic, GABAergic, neuropeptidergic), neurotrophic dysfunction, neuroinflammation, oxidative-stress-related lipid peroxidation, and cerebrovascular (hypoperfusion) damage are also present.

The scientific community, pharmaceutical industry and daily medical care are facing important challenges regarding the management of dementia. The primary causes of AD and its pathogenic mechanisms are still unclear. Reliable biomarkers for an early diagnosis are not yet available. New drugs and novel therapeutic strategies that are able to slow down or halt the course of the disease are an urgent need, assuming that present medications are inefficient and not cost-effective.  Since the disease destroys the neurons of susceptible patients for decades before showing symptoms, the golden dream of AD scientific research would be to find a preventive remedy, administered in pre-symptomatic phases, and capable of stopping the progressive destruction of the brain that leads to AD.

The objective of this Special Issue of IJMS is to offer the scientific community an open space to present new findings to (i) better understand the pathogenic mechanisms responsible for this neurodegenerative disease, (ii) identify potential predictive biomarkers that anticipate risk factors and can prophylactically intervene, and (iii) develop new preventive strategies and new forms of therapeutic intervention that slow down the course of the disease once symptoms appear. The best case scenario would be to prevent the disease from manifesting itself in those cases for which it is feasible to presymptomatically identify the risk.

Prof. Dr. Ramón Cacabelos
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Alzheimer’s disease
  • pathogenic mechanisms
  • genomics
  • epigenetics
  • biomarkers
  • prevention
  • treatment
  • pharmacogenetics

Published Papers (7 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review, Other

18 pages, 2521 KiB  
Article
Sex-Dependent Altered Expression of Cannabinoid Signaling in Hippocampal Astrocytes of the Triple Transgenic Mouse Model of Alzheimer’s Disease: Implications for Controlling Astroglial Activity
by Beatriz Pacheco-Sánchez, Rubén Tovar, Meriem Ben Rabaa, Lourdes Sánchez-Salido, Antonio Vargas, Juan Suárez, Fernando Rodríguez de Fonseca and Patricia Rivera
Int. J. Mol. Sci. 2023, 24(16), 12598; https://doi.org/10.3390/ijms241612598 - 09 Aug 2023
Cited by 1 | Viewed by 1378
Abstract
Alzheimer’s disease (AD) is a common neurodegenerative disease. In AD-associated neuroinflammation, astrocytes play a key role, finding glial activation both in patients and in animal models. The endocannabinoid system (ECS) is a neurolipid signaling system with anti-inflammatory and neuroprotective properties implicated in AD. [...] Read more.
Alzheimer’s disease (AD) is a common neurodegenerative disease. In AD-associated neuroinflammation, astrocytes play a key role, finding glial activation both in patients and in animal models. The endocannabinoid system (ECS) is a neurolipid signaling system with anti-inflammatory and neuroprotective properties implicated in AD. Astrocytes respond to external cannabinoid signals and also have their own cannabinoid signaling. Our main objective is to describe the cannabinoid signaling machinery present in hippocampal astrocytes from 3×Tg-AD mice to determine if they are actively involved in the neurodegenerative process. Primary cultures of astrocytes from the hippocampus of 3×Tg-AD and non-Tg offspring were carried out. We analyzed the gene expression of astrogliosis markers, the main components of the ECS and Ca2+ signaling. 3×Tg-AD hippocampal astrocytes show low inflammatory activity (Il1b, Il6, and Gls) and Ca2+ flow (P2rx5 and Mcu), associated with low cannabinoid signaling (Cnr1 and Cnr2). These results were more evident in females. Our study corroborates glial involvement in AD pathology, in which cannabinoid signaling plays an important role. 3×Tg-AD mice born with hippocampal astrocytes with differential gene expression of the ECS associated with an innate attenuation of their activity. In addition, we show that there are sex differences from birth in this AD animal, which should be considered when investigating the pathogenesis of the disease. Full article
Show Figures

Graphical abstract

19 pages, 4236 KiB  
Article
Berberine Rescues D-Ribose-Induced Alzheimer‘s Pathology via Promoting Mitophagy
by Chuanling Wang, Qian Zou, Yinshuang Pu, Zhiyou Cai and Yong Tang
Int. J. Mol. Sci. 2023, 24(6), 5896; https://doi.org/10.3390/ijms24065896 - 20 Mar 2023
Cited by 7 | Viewed by 3872
Abstract
Mitochondrial dysfunction is considered an early event of Alzheimer disease (AD). D-ribose is a natural monosaccharide that exists in cells, especially in mitochondria, and can lead to cognitive dysfunction. However, the reason for this is unclear. Berberine (BBR) is an isoquinoline alkaloid that [...] Read more.
Mitochondrial dysfunction is considered an early event of Alzheimer disease (AD). D-ribose is a natural monosaccharide that exists in cells, especially in mitochondria, and can lead to cognitive dysfunction. However, the reason for this is unclear. Berberine (BBR) is an isoquinoline alkaloid that can target mitochondria and has great prospect in the treatment of AD. The methylation of PINK1 reinforces the burden of Alzheimer’s pathology. This study explores the role of BBR and D-ribose in the mitophagy and cognitive function of AD related to DNA methylation. APP/PS1 mice and N2a cells were treated with D-ribose, BBR, and mitophagy inhibitor Mdivi-1 to observe their effects on mitochondrial morphology, mitophagy, neuron histology, AD pathology, animal behavior, and PINK1 methylation. The results showed that D-ribose induced mitochondrial dysfunction, mitophagy damage, and cognitive impairment. However, BBR inhibition of PINK1 promoter methylation can reverse the above effects caused by D-ribose, improve mitochondrial function, and restore mitophagy through the PINK1–Parkin pathway, thus reducing cognitive deficits and the burden of AD pathology. This experiment puts a new light on the mechanism of action of D-ribose in cognitive impairment and reveals new insights in the use of BBR for AD treatment. Full article
Show Figures

Figure 1

18 pages, 2425 KiB  
Article
Gene Expression Profiling as a Novel Diagnostic Tool for Neurodegenerative Disorders
by Olaia Martínez-Iglesias, Vinogran Naidoo, Juan Carlos Carril, Silvia Seoane, Natalia Cacabelos and Ramón Cacabelos
Int. J. Mol. Sci. 2023, 24(6), 5746; https://doi.org/10.3390/ijms24065746 - 17 Mar 2023
Cited by 5 | Viewed by 2054
Abstract
There is a lack of effective diagnostic biomarkers for neurodegenerative disorders (NDDs). Here, we established gene expression profiles for diagnosing Alzheimer’s disease (AD), Parkinson’s disease (PD), and vascular (VaD)/mixed dementia. Patients with AD had decreased APOE, PSEN1, and ABCA7 mRNA expression. Subjects with [...] Read more.
There is a lack of effective diagnostic biomarkers for neurodegenerative disorders (NDDs). Here, we established gene expression profiles for diagnosing Alzheimer’s disease (AD), Parkinson’s disease (PD), and vascular (VaD)/mixed dementia. Patients with AD had decreased APOE, PSEN1, and ABCA7 mRNA expression. Subjects with VaD/mixed dementia had 98% higher PICALM mRNA levels, but 75% lower ABCA7 mRNA expression than healthy individuals. Patients with PD and PD-related disorders showed increased SNCA mRNA levels. There were no differences in mRNA expression for OPRK1, NTRK2, and LRRK2 between healthy subjects and NDD patients. APOE mRNA expression had high diagnostic accuracy for AD, and moderate accuracy for PD and VaD/mixed dementia. PSEN1 mRNA expression showed promising accuracy for AD. PICALM mRNA expression was less accurate as a biomarker for AD. ABCA7 and SNCA mRNA expression showed high-to-excellent diagnostic accuracy for AD and PD, and moderate-to-high accuracy for VaD/mixed dementia. The APOE E4 allele reduced APOE expression in patients with different APOE genotypes. There was no association between PSEN1, PICALM, ABCA7, and SNCA gene polymorphisms and expression. Our study suggests that gene expression analysis has diagnostic value for NDDs and provides a liquid biopsy alternative to current diagnostic methods. Full article
Show Figures

Figure 1

21 pages, 4402 KiB  
Article
A Polyaminobiaryl-Based β-secretase Modulator Alleviates Cognitive Impairments, Amyloid Load, Astrogliosis, and Neuroinflammation in APPSwe/PSEN1ΔE9 Mice Model of Amyloid Pathology
by Marie Tautou, Florian Descamps, Paul-Emmanuel Larchanché, Luc Buée, Jamal El Bakali, Patricia Melnyk and Nicolas Sergeant
Int. J. Mol. Sci. 2023, 24(6), 5285; https://doi.org/10.3390/ijms24065285 - 09 Mar 2023
Cited by 3 | Viewed by 1443
Abstract
The progress in Alzheimer’s disease (AD) treatment suggests a combined therapeutic approach targeting the two lesional processes of AD, which include amyloid plaques made of toxic Aβ species and neurofibrillary tangles formed of aggregates of abnormally modified Tau proteins. A pharmacophoric design, novel [...] Read more.
The progress in Alzheimer’s disease (AD) treatment suggests a combined therapeutic approach targeting the two lesional processes of AD, which include amyloid plaques made of toxic Aβ species and neurofibrillary tangles formed of aggregates of abnormally modified Tau proteins. A pharmacophoric design, novel drug synthesis, and structure-activity relationship enabled the selection of a polyamino biaryl PEL24-199 compound. The pharmacologic activity consists of a non-competitive β-secretase (BACE1) modulatory activity in cells. Curative treatment of the Thy-Tau22 model of Tau pathology restores short-term spatial memory, decreases neurofibrillary degeneration, and alleviates astrogliosis and neuroinflammatory reactions. Modulatory effects of PEL24-199 towards APP catalytic byproducts are described in vitro, but whether PEL24-199 can alleviate the Aβ plaque load and associated inflammatory counterparts in vivo remains to be elucidated. We investigated short- and long-term spatial memory, Aβ plaque load, and inflammatory processes in APPSwe/PSEN1ΔE9 PEL24-199 treated transgenic model of amyloid pathology to achieve this objective. PEL24-199 curative treatment induced the recovery of spatial memory and decreased the amyloid plaque load in association with decreased astrogliosis and neuroinflammation. The present results underline the synthesis and selection of a promising polyaminobiaryl-based drug that modulates both Tau and, in this case, APP pathology in vivo via a neuroinflammatory-dependent process. Full article
Show Figures

Figure 1

15 pages, 923 KiB  
Article
The Synergic Effect of AT(N) Profiles and Depression on the Risk of Conversion to Dementia in Patients with Mild Cognitive Impairment
by Marta Marquié, Fernando García-Gutiérrez, Adelina Orellana, Laura Montrreal, Itziar de Rojas, Pablo García-González, Raquel Puerta, Clàudia Olivé, Amanda Cano, Isabel Hernández, Maitée Rosende-Roca, Liliana Vargas, Juan Pablo Tartari, Ester Esteban-De Antonio, Urszula Bojaryn, Mario Ricciardi, Diana M. Ariton, Vanesa Pytel, Montserrat Alegret, Gemma Ortega, Ana Espinosa, Alba Pérez-Cordón, Ángela Sanabria, Nathalia Muñoz, Núria Lleonart, Núria Aguilera, Ainhoa García-Sánchez, Emilio Alarcón-Martín, Lluís Tárraga, Agustín Ruiz, Mercè Boada and Sergi Valeroadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2023, 24(2), 1371; https://doi.org/10.3390/ijms24021371 - 10 Jan 2023
Cited by 4 | Viewed by 2016
Abstract
Few studies have addressed the impact of the association between Alzheimer’s disease (AD) biomarkers and NPSs in the conversion to dementia in patients with mild cognitive impairment (MCI), and no studies have been conducted on the interaction effect of these two risk factors. [...] Read more.
Few studies have addressed the impact of the association between Alzheimer’s disease (AD) biomarkers and NPSs in the conversion to dementia in patients with mild cognitive impairment (MCI), and no studies have been conducted on the interaction effect of these two risk factors. AT(N) profiles were created using AD-core biomarkers quantified in cerebrospinal fluid (CSF) (normal, brain amyloidosis, suspected non-Alzheimer pathology (SNAP) and prodromal AD). NPSs were assessed using the Neuropsychiatric Inventory Questionnaire (NPI-Q). A total of 500 individuals with MCI were followed-up yearly in a memory unit. Cox regression analysis was used to determine risk of conversion, considering additive and multiplicative interactions between AT(N) profile and NPSs on the conversion to dementia. A total of 224 participants (44.8%) converted to dementia during the 2-year follow-up study. Pathologic AT(N) groups (brain amyloidosis, prodromal AD and SNAP) and the presence of depression and apathy were associated with a higher risk of conversion to dementia. The additive combination of the AT(N) profile with depression exacerbates the risk of conversion to dementia. A synergic effect of prodromal AD profile with depressive symptoms is evidenced, identifying the most exposed individuals to conversion among MCI patients. Full article
Show Figures

Figure 1

Review

Jump to: Research, Other

26 pages, 1724 KiB  
Review
Alcohol as a Modifiable Risk Factor for Alzheimer’s Disease—Evidence from Experimental Studies
by Devaraj V. Chandrashekar, Ross A. Steinberg, Derick Han and Rachita K. Sumbria
Int. J. Mol. Sci. 2023, 24(11), 9492; https://doi.org/10.3390/ijms24119492 - 30 May 2023
Cited by 2 | Viewed by 5145
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment and memory loss. Epidemiological evidence suggests that heavy alcohol consumption aggravates AD pathology, whereas low alcohol intake may be protective. However, these observations have been inconsistent, and because of methodological discrepancies, [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by cognitive impairment and memory loss. Epidemiological evidence suggests that heavy alcohol consumption aggravates AD pathology, whereas low alcohol intake may be protective. However, these observations have been inconsistent, and because of methodological discrepancies, the findings remain controversial. Alcohol-feeding studies in AD mice support the notion that high alcohol intake promotes AD, while also hinting that low alcohol doses may be protective against AD. Chronic alcohol feeding to AD mice that delivers alcohol doses sufficient to cause liver injury largely promotes and accelerates AD pathology. The mechanisms by which alcohol can modulate cerebral AD pathology include Toll-like receptors, protein kinase-B (Akt)/mammalian target of rapamycin (mTOR) pathway, cyclic adenosine monophosphate (cAMP) response element-binding protein phosphorylation pathway, glycogen synthase kinase 3-β, cyclin-dependent kinase-5, insulin-like growth factor type-1 receptor, modulation of β-amyloid (Aβ) synthesis and clearance, microglial mediated, and brain endothelial alterations. Besides these brain-centric pathways, alcohol-mediated liver injury may significantly affect brain Aβ levels through alterations in the peripheral-to-central Aβ homeostasis. This article reviews published experimental studies (cell culture and AD rodent models) to summarize the scientific evidence and probable mechanisms (both cerebral and hepatic) by which alcohol promotes or protects against AD progression. Full article
Show Figures

Figure 1

Other

Jump to: Research, Review

8 pages, 1511 KiB  
Opinion
Calmodulin and Amyloid Beta as Coregulators of Critical Events during the Onset and Progression of Alzheimer’s Disease
by Danton H. O’Day
Int. J. Mol. Sci. 2023, 24(2), 1393; https://doi.org/10.3390/ijms24021393 - 11 Jan 2023
Cited by 6 | Viewed by 1587
Abstract
Calmodulin (CaM) and a diversity of CaM-binding proteins (CaMBPs) are involved in the onset and progression of Alzheimer’s disease (AD). In the amyloidogenic pathway, AβPP1, BACE1 and PSEN-1 are all calcium-dependent CaMBPs as are the risk factor proteins BIN1 and TREM2. Ca2+ [...] Read more.
Calmodulin (CaM) and a diversity of CaM-binding proteins (CaMBPs) are involved in the onset and progression of Alzheimer’s disease (AD). In the amyloidogenic pathway, AβPP1, BACE1 and PSEN-1 are all calcium-dependent CaMBPs as are the risk factor proteins BIN1 and TREM2. Ca2+/CaM-dependent protein kinase II (CaMKII) and calcineurin (CaN) are classic CaMBPs involved in memory and plasticity, two events impacted by AD. Coupled with these events is the production of amyloid beta monomers (Aβ) and oligomers (Aβo). The recent revelations that Aβ and Aβo each bind to both CaM and to a host of Aβ receptors that are also CaMBPs adds a new level of complexity to our understanding of the onset and progression of AD. Multiple Aβ receptors that are proven CaMBPs (e.g., NMDAR, PMCA) are involved in calcium homeostasis an early event in AD and other neurodegenerative diseases. Other CaMBPs that are Aβ receptors are AD risk factors while still others are involved in the amyloidogenic pathway. Aβ binding to receptors not only serves to control CaM’s ability to regulate critical proteins, but it is also implicated in Aβ turnover. The complexity of the Aβ/CaM/CaMBP interactions is analyzed using two events: Aβ generation and NMDAR function. The interactions between Aβ, CaM and CaMBPs reveals a new level of complexity to critical events associated with the onset and progression of AD and may help to explain the failure to develop successful therapeutic treatments for the disease. Full article
Show Figures

Figure 1

Back to TopTop