Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
1.
Int J Mol Sci ; 25(17)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39273325

ABSTRACT

Cosmic radiation experienced during space travel may increase the risk of cognitive impairment. While simulated galactic cosmic radiation (GCRsim) has led to memory deficits in wildtype (WT) mice, it has not been investigated whether GCRsim in combination with genetic risk factors for Alzheimer's disease (AD) worsens memory further in aging mice. Here, we investigated the central nervous system (CNS) effects of 0 Gy (sham) or 0.75 Gy five-ion GCRsim or 2 Gy gamma radiation (IRR) in 14-month-old female and male APPNL-F/NL-F knock-in (KI) mice bearing humanized ApoE3 or ApoE4 (APP;E3F and APP;E4F). As travel to a specialized facility was required for irradiation, both traveled sham-irradiated C57BL/6J WT and KI mice and non-traveled (NT) KI mice acted as controls for potential effects of travel. Mice underwent four behavioral tests at 20 months of age and were euthanized for pathological and biochemical analyses 1 month later. Fecal samples were collected pre- and post-irradiation at four different time points. GCRsim seemed to impair memory in male APP;E3F mice compared to their sham counterparts. Travel tended to improve cognition in male APP;E3F mice and lowered total Aß in female and male APP;E3F mice compared to their non-traveled counterparts. Sham-irradiated male APP;E4F mice accumulated more fibrillar amyloid than their APP;E3F counterparts. Radiation exposure had only modest effects on behavior and brain changes, but travel-, sex-, and genotype-specific effects were seen. Irradiated mice had immediate and long-term differences in their gut bacterial composition that correlated to Alzheimer's disease phenotypes.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Cognition , Cosmic Radiation , Mice, Transgenic , Animals , Female , Male , Cosmic Radiation/adverse effects , Mice , Cognition/radiation effects , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Gene Knock-In Techniques , Mice, Inbred C57BL , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Disease Models, Animal , Sex Factors , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Humans
2.
Life Sci Alliance ; 7(12)2024 Dec.
Article in English | MEDLINE | ID: mdl-39348937

ABSTRACT

The amyloid ß peptide (Aß), starting with pyroglutamate (pE) at position 3 and ending at position 42 (Aß3pE-42), predominantly accumulates in the brains of Alzheimer's disease. Consistently, donanemab, a therapeutic antibody raised against Aß3pE-42, has been shown to be effective in recent clinical trials. Although the primary Aß produced physiologically is Aß1-40/42, an explanation for how and why this physiological Aß is converted to the pathological form remains elusive. Here, we present experimental evidence that accounts for the aging-associated Aß3pE-42 deposition: Aß3pE-42 was metabolically more stable than other Aßx-42 variants; deficiency of neprilysin, the major Aß-degrading enzyme, induced a relatively selective deposition of Aß3pE-42 in both APP transgenic and App knock-in mouse brains; Aß3pE-42 deposition always colocalized with Pittsburgh compound B-positive cored plaques in APP transgenic mouse brains; and under aberrant conditions, such as a significant reduction in neprilysin activity, aminopeptidases, dipeptidyl peptidases, and glutaminyl-peptide cyclotransferase-like were up-regulated in the progression of aging, and a proportion of Aß1-42 may be processed to Aß3pE-42. Our findings suggest that anti-Aß therapies are more effective if given before Aß3pE-42 deposition.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Brain , Epitopes , Mice, Transgenic , Neprilysin , Amyloid beta-Peptides/metabolism , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Mice , Humans , Brain/metabolism , Neprilysin/metabolism , Epitopes/immunology , Epitopes/metabolism , Peptide Fragments/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Amyloid beta-Protein Precursor/metabolism , Antibodies, Monoclonal, Humanized
3.
J Alzheimers Dis ; 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39331103
4.
J Biochem ; 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259503

ABSTRACT

Alzheimer's disease (AD) is the most common etiology of dementia. The transcription factor NF-E2-related factor 2 (NRF2) induces the expression of genes encoding phase II detoxification and antioxidant genes. NRF2 is regulated by Kelch-like ECH-associated protein 1 (KEAP1), and the KEAP1-NRF2 system is the key regulatory system involved in cytoprotection. To examine whether pharmacological induction of NRF2 expression alleviates AD phenotypes in vivo, we employed two AD mouse models, i.e., App  NL-G-F/NL-G-F (AppNLGF) and APPV717I::TAUP301L (APP/TAU) mice. As the synthetic oleanane triterpenoid 1-[2-cyano-3,12-dioxooleana-1,9(11-dien-28-oyl)] (CDDO)-4(-pyridin-2-yl)-imidazole (CDDO-2P-Im) exhibits strong NRF2-inducing activity, we treated AD model mice with CDDO-2P-Im. We found that Aß42 levels were markedly greater in the brains of AppNLGF mice than in those of APP/TAU mice. CDDO-2P-Im treatment significantly decreased Aß42 levels, but not Aß40 levels, in APP/TAU mice. Consequently, CDDO-2P-Im also decreased the ratio of Aß42/Aß40, a vital marker of amyloid plaque formation. LC-MS/MS analyses revealed that CDDO-2P-Im was delivered to the brains of the APP/TAU mice. CDDO-2P-Im induced the expression of detoxification and antioxidant gene targets of NRF2 and elevated reduced glutathione (GSH) levels in the mouse brain. These results support the notion that CDDO-2P-Im ameliorates AD-related pathologic changes.

5.
Sci Rep ; 14(1): 18471, 2024 08 09.
Article in English | MEDLINE | ID: mdl-39122814

ABSTRACT

Generation and accumulation of amyloid-ß (Aß) protein in the brain are the primary causes of Alzheimer's disease (AD). Alcadeins (Alcs composed of Alcα, Alcß and Alcγ family) are a neuronal membrane protein that is subject to proteolytic processing, as is Aß protein precursor (APP), by APP secretases. Previous observations suggest that Alcs are involved in the pathophysiology of Alzheimer's disease (AD). Here, we generated new mouse AppNL-F (APP-KI) lines with either Alcα- or Alcß-deficient background and analyzed APP processing and Aß accumulation through the aging process. The Alcα-deficient APP-KI (APP-KI/Alcα-KO) mice enhanced brain Aß accumulation along with increased amyloidogenic ß-site cleavage of APP through the aging process whereas Alcß-deficient APP-KI (APP-KI/Alcß-KO) mice neither affected APP metabolism nor Aß accumulation at any age. More colocalization of APP and BACE1 was observed in the endolysosomal pathway in neurons of APP-KI/Alcα-KO mice compared to APP-KI and APP-KI/Alcß-KO mice. These results indicate that Alcα plays an important role in the neuroprotective function by suppressing the amyloidogenic cleavage of APP by BACE1 in the brain, which is distinct from the neuroprotective function of Alcß, in which p3-Alcß peptides derived from Alcß restores the viability in neurons impaired by toxic Aß.


Subject(s)
Aging , Amyloid Precursor Protein Secretases , Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Brain , Animals , Mice , Aging/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/genetics , Brain/metabolism , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Mice, Knockout , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Neurons/metabolism
6.
Lab Anim (NY) ; 53(9): 244-251, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39215182

ABSTRACT

Nonhuman primates (NHPs), which are closely related to humans, are useful in biomedical research, and an increasing number of NHP disease models have been reported using gene editing. However, many disease-related genes cause perinatal death when manipulated homozygously by gene editing. In addition, NHP resources, which are limited, should be efficiently used. Here, to address these issues, we developed a method of introducing heterozygous genetic modifications into common marmosets by combining Platinum transcription activator-like effector nuclease (TALEN) and a gene-editing strategy in oocytes. We succeeded in introducing the heterozygous exon 9 deletion mutation in the presenilin 1 gene, which causes familial Alzheimer's disease in humans, using this technology. As a result, we obtained animals with the expected genotypes and confirmed several Alzheimer's disease-related biochemical changes. This study suggests that highly efficient heterozygosity-oriented gene editing is possible using TALEN and oocytes and is an effective method for producing genetically modified animals.


Subject(s)
Callithrix , Exons , Gene Editing , Heterozygote , Presenilin-1 , Transcription Activator-Like Effector Nucleases , Animals , Callithrix/genetics , Gene Editing/methods , Transcription Activator-Like Effector Nucleases/genetics , Presenilin-1/genetics , Female , Disease Models, Animal , Alzheimer Disease/genetics , Animals, Genetically Modified/genetics , Oocytes/metabolism
7.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125789

ABSTRACT

In Alzheimer's disease (AD), transgenic mouse models have established links between abnormalities in the retina and those in the brain. APPNL-F/NL-F is a murine, humanized AD model that replicates several pathological features observed in patients with AD. Research has focused on obtaining quantitative parameters from optical coherence tomography (OCT) in AD. The aim of this study was to analyze, in a transversal case-control study using manual retinal segmentation via SD-OCT, the changes occurring in the retinal layers of the APPNL/F-NF/L AD model in comparison to C57BL/6J mice (WT) at 6, 9, 12, 15, 17, and 20 months of age. The analysis focused on retinal thickness in RNFL-GCL, IPL, INL, OPL, and ONL based on the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Both APPNL-F/NL-F-model and WT animals exhibited thickness changes at the time points studied. While WT showed significant changes in INL, OPL, and ONL, the AD model showed changes in all retinal layers analyzed. The APPNL-F/NL-F displayed significant thickness variations in the analyzed layers except for the IPL compared to related WT. These thickness changes closely resembled those found in humans during preclinical stages, as well as during mild and moderate AD stages, making this AD model behave more similarly to the disease in humans.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Mice, Transgenic , Retina , Tomography, Optical Coherence , Animals , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Tomography, Optical Coherence/methods , Retina/pathology , Retina/diagnostic imaging , Mice , Mice, Inbred C57BL , Humans , Aging/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Male , Female , Case-Control Studies
8.
Sci Signal ; 17(848): eadk1822, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39106321

ABSTRACT

Deposition of amyloid-ß (Aß) in the brain can impair neuronal function and contribute to cognitive decline in Alzheimer's disease (AD). Here, we found that dopamine and the dopamine precursor levodopa (also called l-DOPA) induced Aß degradation in the brain. Chemogenetic approaches in mice revealed that the activation of dopamine release from ventral tegmental area (VTA) neurons increased the abundance and activity of the Aß-degrading enzyme neprilysin and reduced the amount of Aß deposits in the prefrontal cortex in a neprilysin-dependent manner. Aged mice had less dopamine and neprilysin in the anterior cortex, a decrease that was accentuated in AD model mice. Treating AD model mice with levodopa reduced Aß deposition and improved cognitive function. These observations demonstrate that dopamine promotes brain region-specific, neprilysin-dependent degradation of Aß, suggesting that dopamine-associated strategies have the potential to treat this aspect of AD pathology.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Dopamine , Neprilysin , Ventral Tegmental Area , Neprilysin/metabolism , Neprilysin/genetics , Animals , Dopamine/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/drug effects , Levodopa/pharmacology , Brain/metabolism , Mice, Transgenic , Disease Models, Animal , Humans , Proteolysis/drug effects , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Male
9.
Biomolecules ; 14(7)2024 Jul 10.
Article in English | MEDLINE | ID: mdl-39062542

ABSTRACT

Alzheimer's disease (AD) may manifest retinal changes preceding brain pathology. A transversal case-control study utilized spectral-domain OCT angiography (SD-OCTA) and Angio-Tool software 0.6a to assess retinal vascular structures and OCT for inner and outer retina thickness in the APPNL-F/NL-F AD model at 6, 9, 12, 15, 17, and 20 months old. Comparisons to age-matched wild type (WT) were performed. The analysis focused on the three vascular plexuses using AngiooTool and on retinal thickness, which was represented with the Early Treatment Diabetic Retinopathy Study (ETDRS) sectors. Compared to WT, the APPNL-F/NL-F group exhibited both vascular and structural changes as early as 6 months persisting and evolving at 15, 17, and 20 months. Significant vascular alterations, principally in the superficial vascular complex (SVC), were observed. There was a significant decrease in the vessel area and the total vessel length in SVC, intermediate, and deep capillary plexus. The inner retina in the APPNL-F/NL-F group predominantly decreased in thickness while the outer retina showed increased thickness in most analyzed time points compared to the control group. There are early vascular and structural retinal changes that precede the cognitive changes, which appear at later stages. Therefore, the natural history of the APPNL-F/NL-F model may be more similar to human AD than other transgenic models.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Retinal Vessels , Tomography, Optical Coherence , Alzheimer Disease/pathology , Alzheimer Disease/diagnostic imaging , Animals , Retinal Vessels/diagnostic imaging , Retinal Vessels/pathology , Mice , Mice, Transgenic , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Retina/pathology , Retina/diagnostic imaging , Humans , Case-Control Studies , Male , Female
10.
bioRxiv ; 2024 Jun 09.
Article in English | MEDLINE | ID: mdl-38895278

ABSTRACT

Gene-editing technologies promise to create a new class of therapeutics that can achieve permanent correction with a single intervention. Besides eliminating mutant alleles in familial disease, gene-editing can also be used to favorably manipulate upstream pathophysiologic events and alter disease-course in wider patient populations, but few such feasible therapeutic avenues have been reported. Here we use CRISPR-Cas9 to edit the last exon of amyloid precursor protein (App), relevant for Alzheimer's disease (AD). Our strategy effectively eliminates an endocytic (YENPTY) motif at APP C-terminus, while preserving the N-terminus and compensatory APP-homologues. This manipulation favorably alters events along the amyloid-pathway - inhibiting toxic APP-ß-cleavage fragments (including Aß) and upregulating neuroprotective APP-α-cleavage products. AAV-driven editing ameliorates neuropathologic, electrophysiologic, and behavioral deficits in an AD knockin mouse model. Effects persist for many months, and no abnormalities are seen in WT mice even after germline App-editing; underlining overall efficacy and safety. Pathologic alterations in the glial-transcriptome of App-KI mice, as seen by single nuclei RNA-sequencing (sNuc-Seq), are also normalized by App C-terminus editing. Our strategy takes advantage of innate transcriptional rules that render terminal exons insensitive to nonsense-decay, and the upstream manipulation is expected to be effective for all forms of AD. These studies offer a path for a one-time disease-modifying treatment for AD.

11.
bioRxiv ; 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38712028

ABSTRACT

The disease's trajectory of Alzheimer's disease (AD) is associated with and worsened by hippocampal hyperexcitability. Here we show that during the asymptomatic stage in a knock in mouse model of Alzheimer's disease (APPNL-G-F/NL-G-F; APPKI), hippocampal hyperactivity occurs at the synaptic compartment, propagates to the soma and is manifesting at low frequencies of stimulation. We show that this aberrant excitability is associated with a deficient adenosine tone, an inhibitory neuromodulator, driven by reduced levels of CD39/73 enzymes, responsible for the extracellular ATP-to-adenosine conversion. Both pharmacologic (adenosine kinase inhibitor) and non-pharmacologic (ketogenic diet) restorations of the adenosine tone successfully normalize hippocampal neuronal activity. Our results demonstrated that neuronal hyperexcitability during the asymptomatic stage of a KI model of Alzheimer's disease originated at the synaptic compartment and is associated with adenosine deficient tone. These results extend our comprehension of the hippocampal vulnerability associated with the asymptomatic stage of Alzheimer's disease.

12.
PLoS One ; 19(5): e0303375, 2024.
Article in English | MEDLINE | ID: mdl-38728348

ABSTRACT

Hearing loss is a pivotal risk factor for dementia. It has recently emerged that a disruption in the intercommunication between the cochlea and brain is a key process in the initiation and progression of this disease. However, whether the cochlear properties can be influenced by pathological signals associated with dementia remains unclear. In this study, using a mouse model of Alzheimer's disease (AD), we investigated the impacts of the AD-like amyloid ß (Aß) pathology in the brain on the cochlea. Despite little detectable change in the age-related shift of the hearing threshold, we observed quantitative and qualitative alterations in the protein profile in perilymph, an extracellular fluid that fills the path of sound waves in the cochlea. Our findings highlight the potential contribution of Aß pathology in the brain to the disturbance of cochlear homeostasis.


Subject(s)
Alzheimer Disease , Cochlea , Disease Models, Animal , Perilymph , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Perilymph/metabolism , Cochlea/metabolism , Cochlea/pathology , Amyloid beta-Peptides/metabolism , Mice, Transgenic , Hearing Loss/metabolism , Hearing Loss/pathology
13.
FEBS Lett ; 598(13): 1576-1590, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38789405

ABSTRACT

Alzheimer's disease (AD) involves reduced glutathione levels, causing oxidative stress and contributing to neuronal cell death. Our prior research identified diminished glutamate-cysteine ligase catalytic subunit (GCLC) as linked to cell death. However, the effect of GCLC on AD features such as amyloid and tau pathology remained unclear. To address this, we investigated amyloid pathology and tau pathology in mice by combining neuron-specific conditional GCLC knockout mice with amyloid precursor protein (App) knockin (KI) or microtubule-associated protein tau (MAPT) KI mice. Intriguingly, GCLC knockout resulted in an increased Aß42/40 ratio. Additionally, GCLC deficiency in MAPT KI mice accelerated the oligomerization of tau through intermolecular disulfide bonds. These findings suggest that the decline in glutathione levels, due to aging or AD pathology, may contribute to the progression of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Glutathione , Neurons , Peptide Fragments , tau Proteins , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , tau Proteins/metabolism , tau Proteins/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Glutathione/metabolism , Mice , Neurons/metabolism , Neurons/pathology , Peptide Fragments/metabolism , Peptide Fragments/genetics , Mice, Knockout , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Disease Models, Animal , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics
14.
Front Neurosci ; 18: 1372297, 2024.
Article in English | MEDLINE | ID: mdl-38572146

ABSTRACT

Introduction: The study of the pathophysiology study of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. Methods: The humanized APPNL-G-F knock-in mouse line was crossed to the PS19 MAPTP301S, over-expression mouse line to create the dual APPNL-G-F/PS19 MAPTP301S line. The resulting pathologies were characterized by immunochemical methods and PCR. Results: We now report on a double transgenic APPNL-G-F/PS19 MAPTP301S mouse that at 6 months of age exhibits robust A plaque accumulation, intense MAPT pathology, strong inflammation and extensive neurodegeneration. The presence of A pathology potentiated the other major pathologies, including MAPT pathology, inflammation and neurodegeneration. MAPT pathology neither changed levels of amyloid precursor protein nor potentiated A accumulation. Interestingly, study of immunofluorescence in cleared brains indicates that microglial inflammation was generally stronger in the hippocampus, dentate gyrus and entorhinal cortex, which are regions with predominant MAPT pathology. The APPNL-G-F/MAPTP301S mouse model also showed strong accumulation of N6-methyladenosine (m6A), which was recently shown to be elevated in the AD brain. m6A primarily accumulated in neuronal soma, but also co-localized with a subset of astrocytes and microglia. The accumulation of m6A corresponded with increases in METTL3 and decreases in ALKBH5, which are enzymes that add or remove m6A from mRNA, respectively. Discussion: Our understanding of the pathophysiology of Alzheimer's disease (AD) has been hampered by lack animal models that recapitulate the major AD pathologies, including extracellular -amyloid (A) deposition, intracellular aggregation of microtubule associated protein tau (MAPT), inflammation and neurodegeneration. The APPNL-G-F/MAPTP301S mouse recapitulates many features of AD pathology beginning at 6 months of aging, and thus represents a useful new mouse model for the field.

15.
Neurobiol Aging ; 139: 20-29, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38583392

ABSTRACT

Brazilian green propolis (propolis) is a chemically complex resinous substance that is a potentially viable therapeutic agent for Alzheimer's disease. Herein, propolis induced a transient increase in intracellular Ca2+ concentration ([Ca2+]i) in Neuro-2A cells; moreover, propolis-induced [Ca2+]i elevations were suppressed prior to 24-h pretreatment with amyloid-ß. To reveal the effect of [Ca2+]i elevation on impaired cognition, we performed memory-related behavioral tasks in APP-KI mice relative to WT mice at 4 and 12 months of age. Propolis, at 300-1000 mg/kg/d for 8 wk, significantly ameliorated cognitive deficits in APP-KI mice at 4 months, but not at 12 months of age. Consistent with behavioral observations, injured hippocampal long-term potentiation was markedly ameliorated in APP-KI mice at 4 months of age following repeated propolis administration. In addition, repeated administration of propolis significantly activated intracellular calcium signaling pathway in the CA1 region of APP-KI mice. These results suggest a preventive effect of propolis on cognitive decline through the activation of intracellular calcium signaling pathways in CA1 region of AD mice model.


Subject(s)
Alzheimer Disease , Calcium , Cognitive Dysfunction , Disease Models, Animal , Propolis , Animals , Propolis/therapeutic use , Propolis/administration & dosage , Propolis/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Alzheimer Disease/psychology , Alzheimer Disease/etiology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/drug therapy , Calcium/metabolism , Mice, Transgenic , Calcium Signaling/drug effects , Long-Term Potentiation/drug effects , Male , Amyloid beta-Peptides/metabolism , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Mice
16.
ACS Chem Neurosci ; 15(10): 2058-2069, 2024 05 15.
Article in English | MEDLINE | ID: mdl-38652895

ABSTRACT

Amyloid plaques composed of fibrils of misfolded Aß peptides are pathological hallmarks of Alzheimer's disease (AD). Aß fibrils are polymorphic in their tertiary and quaternary molecular structures. This structural polymorphism may carry different pathologic potencies and can putatively contribute to clinical phenotypes of AD. Therefore, mapping of structural polymorphism of Aß fibrils and structural evolution over time is valuable to understanding disease mechanisms. Here, we investigated how Aß fibril structures in situ differ in Aß plaque of different mouse models expressing familial mutations in the AßPP gene. We imaged frozen brains with a combination of conformation-sensitive luminescent conjugated oligothiophene (LCO) ligands and Aß-specific antibodies. LCO fluorescence mapping revealed that mouse models APP23, APPPS1, and AppNL-F have different fibril structures within Aß-amyloid plaques depending on the AßPP-processing genotype. Co-staining with Aß-specific antibodies showed that individual plaques from APP23 mice expressing AßPP Swedish mutation have two distinct fibril polymorph regions of core and corona. The plaque core is predominantly composed of compact Aß40 fibrils, and the corona region is dominated by diffusely packed Aß40 fibrils. Conversely, the AßPP knock-in mouse AppNL-F, expressing the AßPP Iberian mutation along with Swedish mutation has tiny, cored plaques consisting mainly of compact Aß42 fibrils, vastly different from APP23 even at elevated age up to 21 months. Age-dependent polymorph rearrangement of plaque cores observed for APP23 and APPPS1 mice >12 months, appears strongly promoted by Aß40 and was hence minuscule in AppNL-F. These structural studies of amyloid plaques in situ can map disease-relevant fibril polymorph distributions to guide the design of diagnostic and therapeutic molecules.


Subject(s)
Amyloid beta-Peptides , Amyloid beta-Protein Precursor , Plaque, Amyloid , Animals , Humans , Mice , Aging/metabolism , Aging/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Mice, Transgenic , Mutation , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Conformation
17.
Front Aging Neurosci ; 16: 1361847, 2024.
Article in English | MEDLINE | ID: mdl-38469162

ABSTRACT

Introduction: Alzheimer's disease (AD), the most common neurodegenerative disease, is characterized by accumulated amyloid-ß (Aß) plaques, aggregated phosphorylated tau protein, gliosis-associated neuroinflammation, synaptic dysfunction, and cognitive impairment. Many cohort studies indicate that tooth loss is a risk factor for AD. The detailed mechanisms underlying the association between AD and tooth loss, however, are not yet fully understood. Methods: We explored the involvement of early tooth loss in the neuropathogenesis of the adult AppNL-G-F mouse AD model. The maxillary molars were extracted bilaterally in 1-month-old male mice soon after tooth eruption. Results: Plasma corticosterone levels were increased and spatial learning memory was impaired in these mice at 6 months of age. The cerebral cortex and hippocampus of AD mice with extracted teeth showed an increased accumulation of Aß plaques and phosphorylated tau proteins, and increased secretion of the proinflammatory cytokines, including interleukin 1ß (IL-1ß) and tumor necrosis factor α (TNF-α), accompanied by an increased number of microglia and astrocytes, and decreased synaptophysin expression. AD mice with extracted teeth also had a shorter lifespan than the control mice. Discussion: These findings revealed that long-term tooth loss is a chronic stressor, activating the recruitment of microglia and astrocytes; exacerbating neuroinflammation, Aß deposition, phosphorylated tau accumulation, and synaptic dysfunction; and leading to spatial learning and memory impairments in AD model mice.

18.
J Oral Biosci ; 66(2): 329-338, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38521152

ABSTRACT

OBJECTIVES: Many patients with Alzheimer's disease (AD) experience behavioral and psychological symptoms of dementia (BPSD), which significantly affect their quality of life. It is known that 5-Hydroxytryptamine (5-HT) plays a crucial role in the development of BPSD. While the relationship between tooth loss and AD symptoms has been acknowledged, the aspect of aggression has not been focused on until now. Despite the established importance of 5-HT in BPSD, how tooth loss is related to the exacerbation of AD symptoms, especially in terms of aggression, remains largely unexplored. Although nutritional status is known to influence the progression of dementia, the specific effect of tooth loss on peripheral symptoms, notably aggression, is not well understood. METHODS: In our study, we conducted maxillary molar extractions in aged C57BL/6J and AppNL-G-F mice and observed their condition over a 3-month period. During this time, we documented significant behavioral and genetic differences between mice in the control groups and mice that underwent tooth extraction. Notably, mice that underwent tooth extraction exhibited a considerable decline in cognitive function and increased in aggression 3 months after tooth extraction compared with the control groups (C57BL/6J and AppNL-G-Fmice). RESULTS: Our findings suggest that molar loss may lead to reduced 5-HT levels in the hippocampus, possibly mediated by the trigeminal nerve, contributing to the development of aggression and BPSD in AD. CONCLUSION: This study sheds light on the intricate relationships between oral health, 5-HT, and AD symptoms, offering valuable insights into potential therapeutic avenues for managing BPSD in patients with dementia.


Subject(s)
Aggression , Mice, Inbred C57BL , Tooth Loss , Animals , Mice , Tooth Loss/genetics , Tooth Loss/psychology , Aggression/psychology , Aggression/physiology , Behavior, Animal , Disease Models, Animal , Dementia/genetics , Dementia/psychology , Mice, Transgenic , Male , Alzheimer Disease/genetics , Alzheimer Disease/psychology , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism
19.
Neurosci Res ; 205: 40-46, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38508957

ABSTRACT

Sleep apnea is regarded as an important risk factor in the pathogenesis of Alzheimer disease (AD). Chronic intermittent hypoxia treatment (IHT) given during the sleep period of the circadian cycle in experimental animals is a well-established sleep apnea model. Here we report that transient IHT for 4 days on AD model mice causes Aß overproduction 2 months after IHT presumably via upregulation of synaptic BACE1, side-by-side with tau hyperphosphorylation. These results suggest that even transient IHT may be sufficient to cause long-lasting changes in the molecules measured as AD biomarkers in the brain.


Subject(s)
Amyloid beta-Peptides , Disease Models, Animal , Sleep Apnea Syndromes , tau Proteins , Animals , tau Proteins/metabolism , Phosphorylation , Amyloid beta-Peptides/metabolism , Sleep Apnea Syndromes/metabolism , Sleep Apnea Syndromes/physiopathology , Male , Mice , Mice, Inbred C57BL , Hypoxia/metabolism , Alzheimer Disease/metabolism , Brain/metabolism , Amyloid Precursor Protein Secretases/metabolism
20.
J Neuroinflammation ; 21(1): 55, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383481

ABSTRACT

BACKGROUND: Neuroinflammation substantially contributes to the pathology of Alzheimer's disease (AD), the most common form of dementia. Studies have reported that nuclear factor erythroid 2-related factor 2 (Nrf2) attenuates neuroinflammation in the mouse models of neurodegenerative diseases, however, the detailed mechanism remains unclear. METHODS: The effects of dimethyl fumarate (DMF), a clinically used drug to activate the Nrf2 pathway, on neuroinflammation were analyzed in primary astrocytes and AppNL-G-F (App-KI) mice. The cognitive function and behavior of DMF-administrated App-KI mice were evaluated. For the gene expression analysis, microglia and astrocytes were directly isolated from the mouse cerebral cortex by magnetic-activated cell sorting, followed by quantitative PCR. RESULTS: DMF treatment activated some Nrf2 target genes and inhibited the expression of proinflammatory markers in primary astrocytes. Moreover, chronic oral administration of DMF attenuated neuroinflammation, particularly in astrocytes, and reversed cognitive dysfunction presumably by activating the Nrf2-dependent pathway in App-KI mice. Furthermore, DMF administration inhibited the expression of STAT3/C3 and C3 receptor in astrocytes and microglia isolated from App-KI mice, respectively, suggesting that the astrocyte-microglia crosstalk is involved in neuroinflammation in mice with AD. CONCLUSION: The activation of astrocytic Nrf2 signaling confers neuroprotection in mice with AD by controlling neuroinflammation, particularly by regulating astrocytic C3-STAT3 signaling. Furthermore, our study has implications for the repositioning of DMF as a drug for AD treatment.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Mice , Animals , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Mice, Transgenic , Neuroinflammatory Diseases , NF-E2-Related Factor 2/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Disease Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL