Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.240
Filter
1.
JCI Insight ; 9(14)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39133647

ABSTRACT

The aggregation and prion-like propagation of tau are the hallmarks of Alzheimer's disease (AD) and other tauopathies. However, the molecular mechanisms underlying the assembly and spread of tau pathology remain elusive. Epidemiological data show that exposure to fine particulate matter (PM2.5) is associated with an increased risk of AD. However, the molecular mechanisms remain unknown. Here, we showed that PM2.5 triggered the aggregation of tau and promoted the formation of tau fibrils. Injection of PM2.5-induced tau preformed fibrils (PFFs) into the hippocampus of tau P301S transgenic mice promoted the aggregation of tau and induced cognitive deficits and synaptic dysfunction. Furthermore, intranasal administration of PM2.5 exacerbated tau pathology and induced cognitive impairment in tau P301S mice. In conclusion, our results indicated that PM2.5 exposure promoted tau pathology and induced cognitive impairments. These results provide mechanistic insight into how PM2.5 increases the risk of AD.


Subject(s)
Disease Models, Animal , Hippocampus , Mice, Transgenic , Particulate Matter , Tauopathies , tau Proteins , Animals , Particulate Matter/toxicity , tau Proteins/metabolism , Mice , Tauopathies/metabolism , Tauopathies/pathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/drug effects , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/chemically induced , Alzheimer Disease/etiology , Protein Aggregation, Pathological/metabolism , Humans , Male
2.
PLoS One ; 19(8): e0303008, 2024.
Article in English | MEDLINE | ID: mdl-39146256

ABSTRACT

The nascent polypeptide-associate complex (NAC) is a heterodimeric chaperone complex that binds near the ribosome exit tunnel and is the first point of chaperone contact for newly synthesized proteins. Deletion of the NAC induces embryonic lethality in many multi-cellular organisms. Previous work has shown that the deletion of the NAC rescues cells from prion-induced cytotoxicity. This counterintuitive result led us to hypothesize that NAC disruption would improve viability in cells expressing human misfolding proteins. Here, we show that NAC disruption improves viability in cells expressing expanded polyglutamine and also leads to delayed and reduced aggregation of expanded polyglutamine and changes in polyglutamine aggregate morphology. Moreover, we show that NAC disruption leads to changes in de novo yeast prion induction. These results indicate that the NAC plays a critical role in aggregate organization as a potential therapeutic target in neurodegenerative disorders.


Subject(s)
Molecular Chaperones , Peptides , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Peptides/metabolism , Humans , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Prions/metabolism , Protein Aggregates , Protein Aggregation, Pathological/metabolism
3.
ACS Chem Neurosci ; 15(14): 2545-2564, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38979773

ABSTRACT

Alzheimer's disease (AD) is a progressive multifaceted neurodegenerative disease and remains a formidable global health challenge. The current medication for AD gives symptomatic relief and, thus, urges us to look for alternative disease-modifying therapies based on a multitarget directed approach. Looking at the remarkable progress made in peptide drug development in the last decade and the benefits associated with peptides, they offer valuable chemotypes [multitarget directed ligands (MTDLs)] as AD therapeutics. This review recapitulates the current developments made in harnessing peptides as MTDLs in combating AD by targeting multiple key pathways involved in the disease's progression. The peptides hold immense potential and represent a convincing avenue in the pursuit of novel AD therapeutics. While hurdles remain, ongoing research offers hope that peptides may eventually provide a multifaceted approach to combat AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Oxidative Stress , tau Proteins , Animals , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Cholinesterases/metabolism , Oxidative Stress/drug effects , Peptides/pharmacology , Peptides/therapeutic use , Protein Aggregates/drug effects , Protein Aggregates/physiology , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , tau Proteins/metabolism
4.
NPJ Syst Biol Appl ; 10(1): 80, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080352

ABSTRACT

A complex interplay between various processes underlies the neuropathology of Alzheimer's disease (AD) and its progressive course. Several lines of evidence point to the coupling between Aß aggregation and neuroinflammation and its role in maintaining brain homeostasis during the long prodromal phase of AD. Little is however known about how this protective mechanism fails and as a result, an irreversible and progressive transition to clinical AD occurs. Here, we introduce a minimal model of a coupled system of Aß aggregation and inflammation, numerically simulate its dynamical behavior, and analyze its bifurcation properties. The introduced model represents the following events: generation of Aß monomers, aggregation of Aß monomers into oligomers and fibrils, induction of inflammation by Aß aggregates, and clearance of various Aß species. Crucially, the rates of Aß generation and clearance are modulated by inflammation level following a Hill-type response function. Despite its relative simplicity, the model exhibits enormously rich dynamics ranging from overdamped kinetics to sustained oscillations. We then specify the region of inflammation- and coupling-related parameters space where a transition to oscillatory dynamics occurs and demonstrate how changes in Aß aggregation parameters could shift this oscillatory region in parameter space. Our results reveal the propensity of coupled Aß aggregation-inflammation systems to oscillatory dynamics and propose prolonged sustained oscillations and their consequent immune system exhaustion as a potential mechanism underlying the transition to a more progressive phase of amyloid pathology in AD. The implications of our results in regard to early diagnosis of AD and anti-AD drug development are discussed.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Inflammation , Amyloid beta-Peptides/metabolism , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Inflammation/metabolism , Brain/metabolism , Brain/pathology , Models, Biological , Protein Aggregation, Pathological/metabolism , Protein Aggregates
5.
Basic Clin Pharmacol Toxicol ; 135(3): 237-249, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39020526

ABSTRACT

Alzheimer's disease is characterized by progressive cognitive decline, and behavioural and psychological symptoms of dementia are common. The APOE ε4 allele, a genetic risk factor, significantly increases susceptibility to the disease. Despite efforts to effectively treat the disease, only seven drugs are approved for its treatment, and only two of these prevent its progression. This highlights the need to identify new pharmacological options. This review focuses on mimetic peptides, small molecule correctors and HAE-4 antibodies that target ApoE. These drugs reduce ß-amyloid-induced neurodegeneration in preclinical models. In addition, loop diuretics such as bumetanide and furosemide show the potential to reduce the prevalence of Alzheimer's disease in humans, and antidepressants such as imipramine improve cognitive function in individuals diagnosed with Alzheimer's disease. Consistent with this, both classes of drugs have been shown to exert neuroprotective effects by inhibiting ApoE4-catalysed Aß aggregation in preclinical models. Moreover, peroxisome proliferator-activated receptor ligands, particularly pioglitazone and rosiglitazone, reduce ApoE4-induced neurodegeneration in animal models. However, they do not prevent the cognitive decline in APOE ε4 allele carriers. Finally, ApoE4 impairs the integrity of the blood-brain barrier and haemostasis. On this basis, ApoE4 modulation is a promising avenue for the treatment of late-onset Alzheimer's disease.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apolipoprotein E4 , Brain , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Apolipoprotein E4/metabolism , Apolipoprotein E4/genetics , Animals , Amyloid beta-Peptides/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Disease Models, Animal
6.
ACS Chem Neurosci ; 15(16): 3044-3052, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39082221

ABSTRACT

The intrinsically disordered protein α-Synuclein is identified as a major toxic aggregate in Parkinson's as well as several other neurodegenerative diseases. Recent work on this protein has focused on the effects of posttranslational modifications on aggregation kinetics. Among them, O-GlcNAcylation of α-Synuclein has been observed to inhibit the aggregation propensity of the protein. Here, we investigate the monomer dynamics of two O-GlcNAcylated α-Synucleins, α-Syn(gT72), and α-Syn(gS87) and correlate them with the aggregation kinetics. We find that, compared to the unmodified protein, glycosylation at T72 makes the protein less compact and more diffusive, while glycosylation at S87 makes the protein more compact and less diffusive. Based on a model of the earliest steps in aggregation, we predict that T72 should aggregate slower than unmodified protein, which is confirmed by ThT fluorescence measurements. In contrast, S87 should aggregate faster, which is not mirrored in ThT kinetics of later fibril formation but does not rule out a higher rate of formation of small oligomers. Together, these results show that posttranslational modifications do not uniformly affect aggregation propensity.


Subject(s)
Protein Processing, Post-Translational , alpha-Synuclein , alpha-Synuclein/metabolism , Kinetics , Protein Processing, Post-Translational/physiology , Humans , Glycosylation , Protein Aggregates/physiology , Protein Aggregates/drug effects , Acetylglucosamine/metabolism , Protein Aggregation, Pathological/metabolism
7.
J Theor Biol ; 593: 111900, 2024 Oct 07.
Article in English | MEDLINE | ID: mdl-38992461

ABSTRACT

The formation of amyloid beta (Aß) deposits (senile plaques) is one of the hallmarks of Alzheimer's disease (AD). This study investigates what processes are primarily responsible for their formation. A model is developed to simulate the diffusion of amyloid beta (Aß) monomers, the production of free Aß aggregates through nucleation and autocatalytic processes, and the deposition of these aggregates into senile plaques. The model suggests that efficient degradation of Aß monomers alone may suffice to prevent the growth of senile plaques, even without degrading Aß aggregates and existing plaques. This is because the degradation of Aß monomers interrupts the supply of reactants needed for plaque formation. The impact of Aß monomer diffusivity is demonstrated to be small, enabling the application of the lumped capacitance approximation and the derivation of approximate analytical solutions for limiting cases with both small and large rates of Aß aggregate deposition into plaques. It is found that the rate of plaque growth is governed by two competing processes. One is the deposition rate of free Aß aggregates into senile plaques. If this rate is small, the plaque grows slowly. However, if the rate of deposition of Aß aggregates into senile plaques is very large, the free Aß aggregates are removed from the intracellular fluid by deposition into the plaques, leaving insufficient free Aß aggregates to catalyze the production of new aggregates. This suggests that under certain conditions, Aß plaques may offer neuroprotection and impede their own growth. Additionally, it indicates that there exists an optimal rate of deposition of free Aß aggregates into the plaques, at which the plaques attain their maximum size.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Plaque, Amyloid , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Models, Biological , Protein Aggregates , Protein Aggregation, Pathological/metabolism
8.
ACS Chem Neurosci ; 15(15): 2925-2935, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39009034

ABSTRACT

Polyglutamine (polyQ) diseases are devastating neurodegenerative disorders characterized by abnormal expansion of glutamine repeats within specific proteins. The aggregation of polyQ proteins is a critical pathological hallmark of these diseases. Arginine was identified as a promising inhibitory compound because it prevents polyQ-protein monomers from forming intra- and intermolecular ß-sheet structures and hinders polyQ proteins from aggregating to form oligomers. Such an aggregation inhibitory effect was not observed in other amino acids. However, the underlying molecular mechanism of the aggregation inhibition and the factors that differentiate arginine from other amino acids, in terms of the inhibition of the polyQ-protein aggregation, remain poorly understood. Here, we performed replica-permutation molecular dynamics simulations to elucidate the molecular mechanism by which arginine inhibits the formation of the intramolecular ß-sheet structure of a polyQ monomer. We found that the intramolecular ß-sheet structure with more than four ß-bridges of the polyQ monomer with arginine is more unstable than without any ligand and with lysine. We also found that arginine has 1.6-2.1 times more contact with polyQ than lysine. In addition, we revealed that arginine forms more hydrogen bonds with the main chain of the polyQ monomer than lysine. More hydrogen bonds formed between arginine and polyQ inhibit polyQ from forming the long intramolecular ß-sheet structure. It is known that intramolecular ß-sheet structure enhances intermolecular ß-sheet structure between proteins. These effects are thought to be the reason for the inhibition of polyQ aggregation. This study provides insights into the molecular events underlying arginine's inhibition of polyQ-protein aggregation.


Subject(s)
Arginine , Molecular Dynamics Simulation , Peptides , Peptides/chemistry , Peptides/pharmacology , Arginine/chemistry , Hydrogen Bonding , Protein Aggregates/drug effects , Humans , Protein Aggregation, Pathological/metabolism
9.
Mol Cells ; 47(8): 100089, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38971320

ABSTRACT

The accumulation of aggregation-prone proteins in a specific neuronal population is a common feature of neurodegenerative diseases, which is correlated with the development of pathological lesions in diseased brains. The formation and progression of pathological protein aggregates in susceptible neurons induce cellular dysfunction, resulting in progressive degeneration. Moreover, recent evidence supports the notion that the cell-to-cell transmission of pathological protein aggregates may be involved in the onset and progression of many neurodegenerative diseases. Indeed, several studies have identified different pathological aggregate strains. Although how these different aggregate strains form remains unclear, a variety of biomolecular compositions or cross-seeding events promoted by the presence of other protein aggregates in the cellular environment may affect the formation of different strains of pathological aggregates, which in turn can influence complex pathologies in diseased brains. In this review, we summarize the recent results regarding cell-to-cell transmission and the molecular heterogeneity of pathological aggregate strains, raising key questions for future research directions.


Subject(s)
Neurodegenerative Diseases , Protein Aggregation, Pathological , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Humans , Protein Aggregation, Pathological/metabolism , Animals , Protein Aggregates
10.
J Membr Biol ; 257(3-4): 151-164, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38888760

ABSTRACT

Protein aggregation plays a crucial role in the development of several neurodegenerative diseases. It is important to understand the aggregation process for the detection of the onset of these diseases. Alzheimer's Disease (AD) is one of the most prevalent neurodegenerative diseases caused by the aggregation of Aß-40 and Aß-42 peptides. The smaller oligomers lead to the formation of protein plaque at the neural membranes leading to memory loss and other disorders. Interestingly, aggregation takes place at the neural membranes, therefore the membrane composition seems to play an important role in the aggregation process. Despite a large number of literatures on the effect of lipid composition on protein aggregation, there are very few concise reviews that highlight the role of membrane composition in protein aggregation. In this review, we have discussed the implication of membrane composition on the aggregation of amyloid beta peptide with a special emphasis on cholesterol. We have further discussed the role of the degree of unsaturation of fatty acids and the participation of apolipoprotein E4 (ApoE4) in the onset of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Membrane Lipids , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/chemistry , Humans , Membrane Lipids/metabolism , Membrane Lipids/chemistry , Alzheimer Disease/metabolism , Protein Aggregates , Cell Membrane/metabolism , Cell Membrane/chemistry , Cholesterol/metabolism , Cholesterol/chemistry , Animals , Protein Aggregation, Pathological/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism
11.
Open Biol ; 14(6): 230418, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835240

ABSTRACT

Mutations in the protein superoxide dismutase-1 (SOD1) promote its misfolding and aggregation, ultimately causing familial forms of the debilitating neurodegenerative disease amyotrophic lateral sclerosis (ALS). Currently, over 220 (mostly missense) ALS-causing mutations in the SOD1 protein have been identified, indicating that common structural features are responsible for aggregation and toxicity. Using in silico tools, we predicted amyloidogenic regions in the ALS-associated SOD1-G85R mutant, finding seven regions throughout the structure. Introduction of proline residues into ß-strands II (I18P) or III (I35P) reduced the aggregation propensity and toxicity of SOD1-G85R in cells, significantly more so than proline mutations in other amyloidogenic regions. The I18P and I35P mutations also reduced the capability of SOD1-G85R to template onto previously formed non-proline mutant SOD1 aggregates as measured by fluorescence recovery after photobleaching. Finally, we found that, while the I18P and I35P mutants are less structurally stable than SOD1-G85R, the proline mutants are less aggregation-prone during proteasome inhibition, and less toxic to cells overall. Our research highlights the importance of a previously underappreciated SOD1 amyloidogenic region in ß-strand II (15QGIINF20) to the aggregation and toxicity of SOD1 in ALS mutants, and suggests that ß-strands II and III may be good targets for the development of SOD1-associated ALS therapies.


Subject(s)
Amyotrophic Lateral Sclerosis , Protein Aggregates , Superoxide Dismutase-1 , Superoxide Dismutase-1/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/chemistry , Humans , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Mutation , Protein Conformation, beta-Strand , Models, Molecular , Proline/metabolism , Amyloid/metabolism , Amyloid/chemistry , Protein Folding
12.
Commun Biol ; 7(1): 776, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38937578

ABSTRACT

Aggregation of the human islet amyloid polypeptide (hIAPP) contributes to the development and progression of Type 2 Diabetes (T2D). hIAPP aggregates within a few hours at few micromolar concentration in vitro but exists at millimolar concentrations in vivo. Natively occurring inhibitors of hIAPP aggregation might therefore provide a model for drug design against amyloid formation associated with T2D. Here, we describe the combined ability of low pH, zinc, and insulin to inhibit hIAPP fibrillation. Insulin dose-dependently slows hIAPP aggregation near neutral pH but had less effect on the aggregation kinetics at acidic pH. We determine that insulin alters hIAPP aggregation in two manners. First, insulin diverts the aggregation pathway to large nonfibrillar aggregates with ThT-positive molecular structure, rather than to amyloid fibrils. Second, soluble insulin suppresses hIAPP dimer formation, which is an important early aggregation event. Further, we observe that zinc significantly modulates the inhibition of hIAPP aggregation by insulin. We hypothesize that this effect arose from controlling the oligomeric state of insulin and show that hIAPP interacts more strongly with monomeric than oligomeric insulin.


Subject(s)
Insulin , Islet Amyloid Polypeptide , Protein Aggregates , Zinc , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Hydrogen-Ion Concentration , Humans , Zinc/pharmacology , Zinc/metabolism , Zinc/chemistry , Insulin/metabolism , Protein Aggregates/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Kinetics , Amyloid/metabolism , Amyloid/chemistry , Protein Aggregation, Pathological/metabolism
13.
Acta Neuropathol ; 147(1): 104, 2024 06 19.
Article in English | MEDLINE | ID: mdl-38896345

ABSTRACT

TAR DNA-binding protein 43 (TDP-43) is an RNA binding protein found within ribonucleoprotein granules tethered to lysosomes via annexin A11. TDP-43 protein forms inclusions in many neurodegenerative diseases including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration with TDP-43 inclusions (FTLD-TDP) and limbic predominant age-related TDP-43 encephalopathy neuropathologic change (LATE-NC). Annexin A11 is also known to form aggregates in ALS cases with pathogenic variants in ANXA11. Annexin A11 aggregation has not been described in sporadic ALS, FTLD-TDP or LATE-NC cases. To explore the relationship between TDP-43 and annexin A11, genetic analysis of 822 autopsy cases was performed to identify rare ANXA11 variants. In addition, an immunohistochemical study of 368 autopsy cases was performed to identify annexin A11 aggregates. Insoluble annexin A11 aggregates which colocalize with TDP-43 inclusions were present in all FTLD-TDP Type C cases. Annexin A11 inclusions were also seen in a small proportion (3-6%) of sporadic and genetic forms of FTLD-TDP types A and B, ALS, and LATE-NC. In addition, we confirm the comingling of annexin A11 and TDP-43 aggregates in an ALS case with the pathogenic ANXA11 p.G38R variant. Finally, we found abundant annexin A11 inclusions as the primary pathologic finding in a case of progressive supranuclear palsy-like frontotemporal dementia with prominent striatal vacuolization due to a novel variant, ANXA11 p.P75S. By immunoblot, FTLD-TDP with annexinopathy and ANXA11 variant cases show accumulation of insoluble ANXA11 including a truncated fragment. These results indicate that annexin A11 forms a diverse and heterogeneous range of aggregates in both sporadic and genetic forms of TDP-43 proteinopathies. In addition, the finding of a primary vacuolar annexinopathy due to ANXA11 p.P75S suggests that annexin A11 aggregation is sufficient to cause neurodegeneration.


Subject(s)
Annexins , DNA-Binding Proteins , Frontotemporal Lobar Degeneration , Humans , Aged , Annexins/genetics , Annexins/metabolism , Female , Male , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Frontotemporal Lobar Degeneration/genetics , Frontotemporal Lobar Degeneration/pathology , Frontotemporal Lobar Degeneration/metabolism , Middle Aged , Aged, 80 and over , TDP-43 Proteinopathies/pathology , TDP-43 Proteinopathies/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Inclusion Bodies/pathology , Inclusion Bodies/metabolism , Brain/pathology , Brain/metabolism , Protein Aggregation, Pathological/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism
14.
BMB Rep ; 57(6): 263-272, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835114

ABSTRACT

Amyloid-ß (Aß) is one of the amyloidogenic intrinsically disordered proteins (IDPs) that self-assemble to protein aggregates, incurring cell malfunction and cytotoxicity. While Aß has been known to regulate multiple physiological functions, such as enhancing synaptic functions, aiding in the recovery of the blood-brain barrier/brain injury, and exhibiting tumor suppression/antimicrobial activities, the hydrophobicity of the primary structure promotes pathological aggregations that are closely associated with the onset of Alzheimer's disease (AD). Aß proteins consist of multiple isoforms with 37-43 amino acid residues that are produced by the cleavage of amyloid-ß precursor protein (APP). The hydrolytic products of APP are secreted to the extracellular regions of neuronal cells. Aß 1-42 (Aß42) and Aß 1-40 (Aß40) are dominant isoforms whose significance in AD pathogenesis has been highlighted in numerous studies to understand the molecular mechanism and develop AD diagnosis and therapeutic strategies. In this review, we focus on the differences between Aß42 and Aß40 in the molecular mechanism of amyloid aggregations mediated by the two additional residues (Ile41 and Ala42) of Aß42. The current comprehension of Aß42 and Aß40 in AD progression is outlined, together with the structural features of Aß42/Aß40 amyloid fibrils, and the aggregation mechanisms of Aß42/Aß40. Furthermore, the impact of the heterogeneous distribution of Aß isoforms during amyloid aggregations is discussed in the system mimicking the coexistence of Aß42 and Aß40 in human cerebrospinal fluid (CSF) and plasma. [BMB Reports 2024; 57(6): 263-272].


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Protein Isoforms , Animals , Humans , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Peptide Fragments/metabolism , Peptide Fragments/chemistry , Protein Aggregates/physiology , Protein Aggregation, Pathological/metabolism , Protein Isoforms/metabolism
15.
Acta Neuropathol Commun ; 12(1): 84, 2024 05 31.
Article in English | MEDLINE | ID: mdl-38822421

ABSTRACT

Alpha-synuclein (αsyn) is an intrinsically disordered protein that aggregates in the brain in several neurodegenerative diseases collectively called synucleinopathies. Phosphorylation of αsyn at serine 129 (PSER129) was considered rare in the healthy human brain but is enriched in pathological αsyn aggregates and is used as a specific marker for disease inclusions. However, recent observations challenge this assumption by demonstrating that PSER129 results from neuronal activity and can be readily detected in the non-diseased mammalian brain. Here, we investigated experimental conditions under which two distinct PSER129 pools, namely endogenous-PSER129 and aggregated-PSER129, could be detected and differentiated in the mammalian brain. Results showed that in the wild-type (WT) mouse brain, perfusion fixation conditions greatly influenced the detection of endogenous-PSER129, with endogenous-PSER129 being nearly undetectable after delayed perfusion fixation (30-min and 1-h postmortem interval). Exposure to anesthetics (e.g., Ketamine or xylazine) before perfusion did not significantly influence endogenous-PSER129 detection or levels. In situ, non-specific phosphatase calf alkaline phosphatase (CIAP) selectively dephosphorylated endogenous-PSER129 while αsyn preformed fibril (PFF)-seeded aggregates and genuine disease aggregates (Lewy pathology and Papp-Lantos bodies in Parkinson's disease and multiple systems atrophy brain, respectively) were resistant to CIAP-mediated dephosphorylation. The phosphatase resistance of aggregates was abolished by sample denaturation, and CIAP-resistant PSER129 was closely associated with proteinase K (PK)-resistant αsyn (i.e., a marker of aggregation). CIAP pretreatment allowed for highly specific detection of seeded αsyn aggregates in a mouse model that accumulates non-aggregated-PSER129. We conclude that αsyn aggregates are impervious to phosphatases, and CIAP pretreatment increases detection specificity for aggregated-PSER129, particularly in well-preserved biological samples (e.g., perfusion fixed or flash-frozen mammalian tissues) where there is a high probability of interference from endogenous-PSER129. Our findings have important implications for the mechanism of PSER129-accumulation in the synucleinopathy brain and provide a simple experimental method to differentiate endogenous-from aggregated PSER129.


Subject(s)
Brain , Mice, Inbred C57BL , alpha-Synuclein , Animals , Humans , Male , Mice , Alkaline Phosphatase/metabolism , alpha-Synuclein/metabolism , Brain/metabolism , Brain/pathology , Mice, Transgenic , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Protein Aggregates/physiology , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Synucleinopathies/metabolism , Synucleinopathies/pathology
16.
Nat Commun ; 15(1): 4695, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824138

ABSTRACT

Which isoforms of apolipoprotein E (apoE) we inherit determine our risk of developing late-onset Alzheimer's Disease (AD), but the mechanism underlying this link is poorly understood. In particular, the relevance of direct interactions between apoE and amyloid-ß (Aß) remains controversial. Here, single-molecule imaging shows that all isoforms of apoE associate with Aß in the early stages of aggregation and then fall away as fibrillation happens. ApoE-Aß co-aggregates account for ~50% of the mass of diffusible Aß aggregates detected in the frontal cortices of homozygotes with the higher-risk APOE4 gene. We show how dynamic interactions between apoE and Aß tune disease-related functions of Aß aggregates throughout the course of aggregation. Our results connect inherited APOE genotype with the risk of developing AD by demonstrating how, in an isoform- and lipidation-specific way, apoE modulates the aggregation, clearance and toxicity of Aß. Selectively removing non-lipidated apoE4-Aß co-aggregates enhances clearance of toxic Aß by glial cells, and reduces secretion of inflammatory markers and membrane damage, demonstrating a clear path to AD therapeutics.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides , Apolipoprotein E4 , Apolipoproteins E , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Humans , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Animals , Apolipoprotein E4/metabolism , Apolipoprotein E4/genetics , Protein Isoforms/metabolism , Protein Isoforms/genetics , Mice , Female , Protein Aggregates , Male , Protein Aggregation, Pathological/metabolism , Mice, Transgenic , Neuroglia/metabolism
17.
Mol Neurodegener ; 19(1): 46, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862967

ABSTRACT

RNA binding proteins have emerged as central players in the mechanisms of many neurodegenerative diseases. In particular, a proteinopathy of fused in sarcoma (FUS) is present in some instances of familial Amyotrophic lateral sclerosis (ALS) and about 10% of sporadic Frontotemporal lobar degeneration (FTLD). Here we establish that focal injection of sonicated human FUS fibrils into brains of mice in which ALS-linked mutant or wild-type human FUS replaces endogenous mouse FUS is sufficient to induce focal cytoplasmic mislocalization and aggregation of mutant and wild-type FUS which with time spreads to distal regions of the brain. Human FUS fibril-induced FUS aggregation in the mouse brain of humanized FUS mice is accelerated by an ALS-causing FUS mutant relative to wild-type human FUS. Injection of sonicated human FUS fibrils does not induce FUS aggregation and subsequent spreading after injection into naïve mouse brains containing only mouse FUS, indicating a species barrier to human FUS aggregation and its prion-like spread. Fibril-induced human FUS aggregates recapitulate pathological features of FTLD including increased detergent insolubility of FUS and TAF15 and amyloid-like, cytoplasmic deposits of FUS that accumulate ubiquitin and p62, but not TDP-43. Finally, injection of sonicated FUS fibrils is shown to exacerbate age-dependent cognitive and behavioral deficits from mutant human FUS expression. Thus, focal seeded aggregation of FUS and further propagation through prion-like spread elicits FUS-proteinopathy and FTLD-like disease progression.


Subject(s)
Disease Progression , Frontotemporal Dementia , Mice, Transgenic , RNA-Binding Protein FUS , Animals , Humans , Mice , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/metabolism , Brain/metabolism , Brain/pathology , Disease Models, Animal , Frontotemporal Dementia/pathology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/genetics , Protein Aggregation, Pathological/metabolism , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/genetics
18.
Molecules ; 29(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38930877

ABSTRACT

Tau protein is a microtubule-associated protein that is widely distributed in the central nervous system and maintains and regulates neuronal morphology and function. Tau protein aggregates abnormally and forms neurofibrillary tangles in neurodegenerative diseases, disrupting the structure and function of neurons and leading to neuronal death, which triggers the initiation and progression of neurological disorders. The aggregation of tau protein in neurodegenerative diseases is associated with post-translational modifications, which may affect the hydrophilicity, spatial conformation, and stability of tau protein, promoting tau protein aggregation and the formation of neurofibrillary tangles. Therefore, studying the role of tau protein in neurodegenerative diseases and the mechanism of aberrant aggregation is important for understanding the mechanism of neurodegenerative diseases and finding therapeutic approaches. This review describes the possible mechanisms by which tau protein promotes neurodegenerative diseases, the post-translational modifications of tau protein and associated influencing factors, and the current status of drug discovery and development related to tau protein, which may contribute to the development of new therapeutic approaches to alleviate or treat neurodegenerative diseases.


Subject(s)
Neurodegenerative Diseases , Protein Processing, Post-Translational , tau Proteins , tau Proteins/metabolism , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Animals , Drug Development , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/drug therapy , Neurofibrillary Tangles/metabolism , Molecular Targeted Therapy
19.
J Neurol ; 271(8): 4826-4851, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38869826

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal disease. As its pathological mechanisms are not well understood, there are no efficient therapeutics for it at present. While it is highly heterogenous both etiologically and clinically, it has a common salient hallmark, i.e., aberrant protein aggregation (APA). The upstream pathogenesis and the downstream effects of APA in ALS are sophisticated and the investigation of this pathology would be of consequence for understanding ALS. In this paper, the pathomechanism of APA in ALS and the candidate treatment strategies for it are discussed.


Subject(s)
Amyotrophic Lateral Sclerosis , Protein Aggregation, Pathological , Amyotrophic Lateral Sclerosis/metabolism , Humans , Protein Aggregation, Pathological/metabolism , Protein Aggregates/physiology , Animals
20.
Sci Adv ; 10(18): eadl2991, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38691615

ABSTRACT

Amyloid fibrils of tau are increasingly accepted as a cause of neuronal death and brain atrophy in Alzheimer's disease (AD). Diminishing tau aggregation is a promising strategy in the search for efficacious AD therapeutics. Previously, our laboratory designed a six-residue, nonnatural amino acid inhibitor D-TLKIVW peptide (6-DP), which can prevent tau aggregation in vitro. However, it cannot block cell-to-cell transmission of tau aggregation. Here, we find D-TLKIVWC (7-DP), a d-cysteine extension of 6-DP, not only prevents tau aggregation but also fragments tau fibrils extracted from AD brains to neutralize their seeding ability and protect neuronal cells from tau-induced toxicity. To facilitate the transport of 7-DP across the blood-brain barrier, we conjugated it to magnetic nanoparticles (MNPs). The MNPs-DP complex retains the inhibition and fragmentation properties of 7-DP alone. Ten weeks of MNPs-DP treatment appear to reverse neurological deficits in the PS19 mouse model of AD. This work offers a direction for development of therapies to target tau fibrils.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Magnetite Nanoparticles , tau Proteins , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , tau Proteins/metabolism , tau Proteins/chemistry , Mice , Humans , Magnetite Nanoparticles/chemistry , Amyloid/metabolism , Amyloid/chemistry , Mice, Transgenic , Behavior, Animal/drug effects , Peptides/chemistry , Peptides/pharmacology , Protein Aggregation, Pathological/metabolism , Brain/metabolism , Brain/pathology , Brain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL