ijms-logo

Journal Browser

Journal Browser

Neuroinflammation in the Pathogenesis of Alzheimer's Disease and Related Dementias

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

Deadline for manuscript submissions: closed (15 February 2024) | Viewed by 23911

Special Issue Editor


E-Mail Website
Guest Editor
Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, 720 S Donahue Dr., Auburn, AL 36849, USA
Interests: Alzheimer’s disease; cerebral amyloid angiopathy; blood-brain barrier; neuroinflammation; drug development; translational therapeutics
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues, 

Alzheimer’s disease (AD) is the most common neurodegenerative disease worldwide. Besides memory loss and progressive dementia, AD is characterized by several pathological hallmarks including the deposition of amyloid-β (Aβ) plaques, neurofibrillary tangles (NFT) of hyper-phosphorylated tau, blood–brain barrier breakdown, oxidative stress, and neuronal loss. In addition, neuroinflammation has a prominent role in the pathogenesis of AD, as suggested by the increased levels of inflammatory markers in AD patients as well as findings from genome-wide association studies associating the involvement of various single-nucleotide polymorphisms of genes related to neuroinflammation with the risk of developing AD. Inflammation in the brain has an important neuroprotective role during the acute-phase response; however, in the long term, neuroinflammation becomes detrimental. The activation of microglia and astrocytes triggers an innate immune response to release various pro-inflammatory mediators and toxic products such as reactive oxygen species and cytokines, which could collectively increase Aβ production and tau hyper-phosphorylation and thus contribute to disease progression and severity. Modulating the release of neuroinflammatory molecules and targeting pathological gliosis represent a valuable therapeutic approach and may have beneficial effects on the onset, progress, and severity of AD. In addition, external factors such as systemic inflammation, diabetes, and obesity could interfere with immunological processes of the brain and further promote disease progression; hence, the modulation of risk factors could lead to preventive strategies for AD.

We are pleased to invite you to participate in this Special Issue through research articles, comprehensive reviews, or short communications about neuroinflammation in AD, treatments targeting neuroinflammation, and related topics.

Prof. Dr. Amal Kaddoumi
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

  • neuroinflammation
  • drug development
  • microglia, astrocytes, cytokines, TREM2
  • risk factors
  • biomarkers

Published Papers (9 papers)

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

Research

Jump to: Review

18 pages, 122750 KiB  
Article
Autosomal Dominant Alzheimer’s Disease Mutations in Human Microglia Are Not Sufficient to Trigger Amyloid Pathology in WT Mice but Might Affect Pathology in 5XFAD Mice
by Carmen Romero-Molina, Sarah M. Neuner, Marcelina Ryszawiec, Alice Pébay, Dominantly Inherited Alzheimer Network, Edoardo Marcora and Alison Goate
Int. J. Mol. Sci. 2024, 25(5), 2565; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25052565 - 22 Feb 2024
Viewed by 1247
Abstract
Several genetic variants that affect microglia function have been identified as risk factors for Alzheimer’s Disease (AD), supporting the importance of this cell type in disease progression. However, the effect of autosomal dominant mutations in the amyloid precursor protein (APP) or [...] Read more.
Several genetic variants that affect microglia function have been identified as risk factors for Alzheimer’s Disease (AD), supporting the importance of this cell type in disease progression. However, the effect of autosomal dominant mutations in the amyloid precursor protein (APP) or the presenilin (PSEN1/2) genes has not been addressed in microglia in vivo. We xenotransplanted human microglia derived from non-carriers and carriers of autosomal dominant AD (ADAD)-causing mutations in the brain of hCSF1 WT or 5XFAD mice. We observed that ADAD mutations in microglia are not sufficient to trigger amyloid pathology in WT mice. In 5XFAD mice, we observed a non-statistically significant increase in amyloid plaque volume and number of dystrophic neurites, coupled with a reduction in plaque-associated microglia in the brain of mice xenotransplanted with ADAD human microglia compared to mice xenotransplanted with non-ADAD microglia. In addition, we observed a non-statistically significant impairment in working and contextual memory in 5XFAD mice xenotransplanted with ADAD microglia compared to those xenotransplanted with non-ADAD-carrier microglia. We conclude that, although not sufficient to initiate amyloid pathology in the healthy brain, mutations in APP and PSEN1 in human microglia might cause mild changes in pathological and cognitive outcomes in 5XFAD mice in a manner consistent with increased AD risk. Full article
Show Figures

Figure 1

11 pages, 1660 KiB  
Communication
Long-Term Pioglitazone Treatment Has No Significant Impact on Microglial Activation and Tau Pathology in P301S Mice
by Lea Helena Kunze, François Ruch, Gloria Biechele, Florian Eckenweber, Karin Wind-Mark, Lina Dinkel, Paul Feyen, Peter Bartenstein, Sibylle Ziegler, Lars Paeger, Sabina Tahirovic, Jochen Herms and Matthias Brendel
Int. J. Mol. Sci. 2023, 24(12), 10106; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms241210106 - 14 Jun 2023
Cited by 2 | Viewed by 1147
Abstract
Neuroinflammation is one disease hallmark on the road to neurodegeneration in primary tauopathies. Thus, immunomodulation might be a suitable treatment strategy to delay or even prevent the occurrence of symptoms and thus relieve the burden for patients and caregivers. In recent years, the [...] Read more.
Neuroinflammation is one disease hallmark on the road to neurodegeneration in primary tauopathies. Thus, immunomodulation might be a suitable treatment strategy to delay or even prevent the occurrence of symptoms and thus relieve the burden for patients and caregivers. In recent years, the peroxisome proliferator-activated receptor γ (PPARγ) has received increasing attention as it is immediately involved in the regulation of the immune system and can be targeted by the anti-diabetic drug pioglitazone. Previous studies have shown significant immunomodulation in amyloid-β (Aβ) mouse models by pioglitazone. In this study, we performed long-term treatment over six months in P301S mice as a tauopathy model with either pioglitazone or placebo. We performed serial 18 kDa translocator protein positron-emission-tomography (TSPO-PET) imaging and terminal immunohistochemistry to assess microglial activation during treatment. Tau pathology was quantified via immunohistochemistry at the end of the study. Long-term pioglitazone treatment had no significant effect on TSPO-PET, immunohistochemistry read-outs of microglial activation, or tau pathology levels in P301S mice. Thus, we conclude that pioglitazone modifies the time course of Aβ-dependent microglial activation, but does not significantly modulate microglial activation in response to tau pathology. Full article
Show Figures

Figure 1

26 pages, 26907 KiB  
Article
L-Type Ca2+ Channel Inhibition Rescues the LPS-Induced Neuroinflammatory Response and Impairments in Spatial Memory and Dendritic Spine Formation
by Jieun Kim, Seong Gak Jeon, Ha-Ram Jeong, HyunHee Park, Jae-Ick Kim and Hyang-Sook Hoe
Int. J. Mol. Sci. 2022, 23(21), 13606; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms232113606 - 6 Nov 2022
Cited by 6 | Viewed by 2545
Abstract
Ca2+ signaling is implicated in the transition between microglial surveillance and activation. Several L-type Ca2+ channel blockers (CCBs) have been shown to ameliorate neuroinflammation by modulating microglial activity. In this study, we examined the effects of the L-type CCB felodipine on [...] Read more.
Ca2+ signaling is implicated in the transition between microglial surveillance and activation. Several L-type Ca2+ channel blockers (CCBs) have been shown to ameliorate neuroinflammation by modulating microglial activity. In this study, we examined the effects of the L-type CCB felodipine on LPS-mediated proinflammatory responses. We found that felodipine treatment significantly diminished LPS-evoked proinflammatory cytokine levels in BV2 microglial cells in an L-type Ca2+ channel-dependent manner. In addition, felodipine leads to the inhibition of TLR4/AKT/STAT3 signaling in BV2 microglial cells. We further examined the effects of felodipine on LPS-stimulated neuroinflammation in vivo and found that daily administration (3 or 7 days, i.p.) significantly reduced LPS-mediated gliosis and COX-2 and IL-1β levels in C57BL/6 (wild-type) mice. Moreover, felodipine administration significantly reduced chronic neuroinflammation-induced spatial memory impairment, dendritic spine number, and microgliosis in C57BL/6 mice. Taken together, our results suggest that the L-type CCB felodipine could be repurposed for the treatment of neuroinflammation/cognitive function-associated diseases. Full article
Show Figures

Figure 1

25 pages, 3103 KiB  
Article
Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Microglia from APOE Targeted Replacement Mice
by Isha Mhatre-Winters, Aseel Eid, Yoonhee Han, Kim Tieu and Jason R. Richardson
Int. J. Mol. Sci. 2022, 23(17), 9829; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23179829 - 29 Aug 2022
Cited by 6 | Viewed by 2695
Abstract
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide [...] Read more.
The sex and APOE4 genotype are significant risk factors for Alzheimer’s disease (AD); however, the mechanism(s) responsible for this interaction are still a matter of debate. Here, we assess the responses of mixed-sex and sex-specific APOE3 and APOE4 primary microglia (PMG) to lipopolysaccharide and interferon-gamma. In our investigation, inflammatory cytokine profiles were assessed by qPCR and multiplex ELISA assays. Mixed-sex APOE4 PMG exhibited higher basal mRNA expression and secreted levels of TNFa and IL1b. In sex-specific cultures, basal expression and secreted levels of IL1b, TNFa, IL6, and NOS2 were 2–3 fold higher in APOE4 female PMG compared to APOE4 males, with both higher than APOE3 cells. Following an inflammatory stimulus, the expression of pro-inflammatory cytokines and the secreted cytokine level were upregulated in the order E4 female > E4 male > E3 female > E3 male in sex-specific cultures. These data indicate that the APOE4 genotype and female sex together contribute to a greater inflammatory response in PMG isolated from targeted replacement humanized APOE mice. These data are consistent with clinical data and indicate that sex-specific PMG may provide a platform for exploring mechanisms of genotype and sex differences in AD related to neuroinflammation and neurodegeneration. Full article
Show Figures

Graphical abstract

Review

Jump to: Research

18 pages, 1527 KiB  
Review
Periodontitis: A Plausible Modifiable Risk Factor for Neurodegenerative Diseases? A Comprehensive Review
by Adelina S. Plachokova, Jolijn Gjaltema, Eliza R. C. Hagens, Zahra Hashemi, Tim B. A. Knüppe, Thomas J. M. Kootstra, Anita Visser and Bastiaan R. Bloem
Int. J. Mol. Sci. 2024, 25(8), 4504; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25084504 - 19 Apr 2024
Viewed by 561
Abstract
The aim of this comprehensive review is to summarize recent literature on associations between periodontitis and neurodegenerative diseases, explore the bidirectionality and provide insights into the plausible pathogenesis. For this purpose, systematic reviews and meta-analyses from PubMed, Medline and EMBASE were considered. Out [...] Read more.
The aim of this comprehensive review is to summarize recent literature on associations between periodontitis and neurodegenerative diseases, explore the bidirectionality and provide insights into the plausible pathogenesis. For this purpose, systematic reviews and meta-analyses from PubMed, Medline and EMBASE were considered. Out of 33 retrieved papers, 6 articles complying with the inclusion criteria were selected and discussed. Additional relevant papers for bidirectionality and pathogenesis were included. Results show an association between periodontitis and Alzheimer’s disease, with odds ratios of 3 to 5. A bidirectional relationship is suspected. For Parkinson’s disease (PD), current evidence for an association appears to be weak, although poor oral health and PD seem to be correlated. A huge knowledge gap was identified. The plausible mechanistic link for the association between periodontitis and neurodegenerative diseases is the interplay between periodontal inflammation and neuroinflammation. Three pathways are hypothesized in the literature, i.e., humoral, neuronal and cellular, with a clear role of periodontal pathogens, such as Porphyromonas gingivalis. Age, gender, race, smoking, alcohol intake, nutrition, physical activity, socioeconomic status, stress, medical comorbidities and genetics were identified as common risk factors for periodontitis and neurodegenerative diseases. Future research with main emphasis on the collaboration between neurologists and dentists is encouraged. Full article
Show Figures

Figure 1

21 pages, 3667 KiB  
Review
Necrosis Links Neurodegeneration and Neuroinflammation in Neurodegenerative Disease
by Hidenori Homma, Hikari Tanaka, Kyota Fujita and Hitoshi Okazawa
Int. J. Mol. Sci. 2024, 25(7), 3636; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25073636 - 24 Mar 2024
Viewed by 1024
Abstract
The mechanisms of neuronal cell death in neurodegenerative disease remain incompletely understood, although recent studies have made significant advances. Apoptosis was previously considered to be the only mechanism of neuronal cell death in neurodegenerative diseases. However, recent findings have challenged this dogma, identifying [...] Read more.
The mechanisms of neuronal cell death in neurodegenerative disease remain incompletely understood, although recent studies have made significant advances. Apoptosis was previously considered to be the only mechanism of neuronal cell death in neurodegenerative diseases. However, recent findings have challenged this dogma, identifying new subtypes of necrotic neuronal cell death. The present review provides an updated summary of necrosis subtypes and discusses their potential roles in neurodegenerative cell death. Among numerous necrosis subtypes, including necroptosis, paraptosis, ferroptosis, and pyroptosis, transcriptional repression-induced atypical cell death (TRIAD) has been identified as a potential mechanism of neuronal cell death. TRIAD is induced by functional deficiency of TEAD-YAP and self-amplifies via the release of HMGB1. TRIAD is a feasible potential mechanism of neuronal cell death in Alzheimer’s disease and other neurodegenerative diseases. In addition to induction of cell death, HMGB1 released during TRIAD activates brain inflammatory responses, which is a potential link between neurodegeneration and neuroinflammation. Full article
Show Figures

Figure 1

15 pages, 929 KiB  
Review
The Influence of Kynurenine Metabolites on Neurodegenerative Pathologies
by Suhrud Pathak, Rishi Nadar, Shannon Kim, Keyi Liu, Manoj Govindarajulu, Preston Cook, Courtney S. Watts Alexander, Muralikrishnan Dhanasekaran and Timothy Moore
Int. J. Mol. Sci. 2024, 25(2), 853; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms25020853 - 10 Jan 2024
Cited by 2 | Viewed by 1448
Abstract
As the kynurenine pathway’s links to inflammation, the immune system, and neurological disorders became more apparent, it attracted more and more attention. It is the main pathway through which the liver breaks down Tryptophan and the initial step in the creation of nicotinamide [...] Read more.
As the kynurenine pathway’s links to inflammation, the immune system, and neurological disorders became more apparent, it attracted more and more attention. It is the main pathway through which the liver breaks down Tryptophan and the initial step in the creation of nicotinamide adenine dinucleotide (NAD+) in mammals. Immune system activation and the buildup of potentially neurotoxic substances can result from the dysregulation or overactivation of this pathway. Therefore, it is not shocking that kynurenines have been linked to neurological conditions (Depression, Parkinson’s, Alzheimer’s, Huntington’s Disease, Schizophrenia, and cognitive deficits) in relation to inflammation. Nevertheless, preclinical research has demonstrated that kynurenines are essential components of the behavioral analogs of depression and schizophrenia-like cognitive deficits in addition to mediators associated with neurological pathologies due to their neuromodulatory qualities. Neurodegenerative diseases have been extensively associated with neuroactive metabolites of the kynurenine pathway (KP) of tryptophan breakdown. In addition to being a necessary amino acid for protein synthesis, Tryptophan is also transformed into the important neurotransmitters tryptamine and serotonin in higher eukaryotes. In this article, a summary of the KP, its function in neurodegeneration, and the approaches being used currently to target the route therapeutically are discussed. Full article
Show Figures

Figure 1

16 pages, 2488 KiB  
Review
Role of cGAS–Sting Signaling in Alzheimer’s Disease
by Manoj Govindarajulu, Sindhu Ramesh, McNeil Beasley, Graham Lynn, Caleigh Wallace, Sammie Labeau, Suhrud Pathak, Rishi Nadar, Timothy Moore and Muralikrishnan Dhanasekaran
Int. J. Mol. Sci. 2023, 24(9), 8151; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24098151 - 2 May 2023
Cited by 4 | Viewed by 3463
Abstract
There is mounting evidence that the development of Alzheimer’s disease (AD) interacts extensively with immunological processes in the brain and extends beyond the neuronal compartment. Accumulation of misfolded proteins can activate an innate immune response that releases inflammatory mediators and increases the severity [...] Read more.
There is mounting evidence that the development of Alzheimer’s disease (AD) interacts extensively with immunological processes in the brain and extends beyond the neuronal compartment. Accumulation of misfolded proteins can activate an innate immune response that releases inflammatory mediators and increases the severity and course of the disease. It is widely known that type-I interferon-driven neuroinflammation in the central nervous system (CNS) accelerates the development of numerous acute and chronic CNS diseases. It is becoming better understood how the cyclic GMP–AMP synthase (cGAS) and its adaptor protein Stimulator of Interferon Genes (STING) triggers type-I IFN-mediated neuroinflammation. We discuss the principal elements of the cGAS–STING signaling pathway and the mechanisms underlying the association between cGAS–STING activity and various AD pathologies. The current understanding of beneficial and harmful cGAS–STING activity in AD and the current treatment pathways being explored will be discussed in this review. The cGAS–STING regulation offers a novel therapeutic opportunity to modulate inflammation in the CNS because it is an upstream regulator of type-I IFNs Full article
Show Figures

Figure 1

29 pages, 1453 KiB  
Review
Glial Cell-Mediated Neuroinflammation in Alzheimer’s Disease
by Nour F. Al-Ghraiybah, Junwei Wang, Amer E. Alkhalifa, Andrew B. Roberts, Ruchika Raj, Euitaek Yang and Amal Kaddoumi
Int. J. Mol. Sci. 2022, 23(18), 10572; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231810572 - 12 Sep 2022
Cited by 32 | Viewed by 8464
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder; it is the most common cause of dementia and has no treatment. It is characterized by two pathological hallmarks, the extracellular deposits of amyloid beta (Aβ) and the intraneuronal deposits of Neurofibrillary tangles (NFTs). Yet, those two hallmarks do not explain the full pathology seen with AD, suggesting the involvement of other mechanisms. Neuroinflammation could offer another explanation for the progression of the disease. This review provides an overview of recent advances on the role of the immune cells’ microglia and astrocytes in neuroinflammation. In AD, microglia and astrocytes become reactive by several mechanisms leading to the release of proinflammatory cytokines that cause further neuronal damage. We then provide updates on neuroinflammation diagnostic markers and investigational therapeutics currently in clinical trials to target neuroinflammation. Full article
Show Figures

Graphical abstract

Back to TopTop