Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (186)

Search Parameters:
Keywords = amyloid enhancing factor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
42 pages, 989 KB  
Review
The Neuroprotective Role of Curcumin: From Molecular Pathways to Clinical Translation—A Narrative Review
by Andrea Lehoczki, Mónika Fekete, Tamás Jarecsny, Virág Zábó, Ágnes Szappanos, Tamás Csípő, Ágnes Lipécz, Dávid Major, Vince Fazekas-Pongor, Péter Varga and János Tamás Varga
Nutrients 2025, 17(17), 2884; https://doi.org/10.3390/nu17172884 - 6 Sep 2025
Viewed by 1258
Abstract
Neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and post-stroke cognitive impairment (PSCI), represent an escalating global health and economic challenge. In the quest for disease-modifying interventions, natural polyphenols—most notably curcumin, the principal bioactive compound of Curcuma longa—have attracted considerable interest due [...] Read more.
Neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and post-stroke cognitive impairment (PSCI), represent an escalating global health and economic challenge. In the quest for disease-modifying interventions, natural polyphenols—most notably curcumin, the principal bioactive compound of Curcuma longa—have attracted considerable interest due to their pleiotropic neuroprotective effects. This narrative review critically synthesizes findings from a selection of peer-reviewed articles published between 2000 and 2025, chosen for their relevance to curcumin’s molecular mechanisms and translational potential. Curcumin’s complex chemical structure confers antioxidant, anti-inflammatory, and epigenetic modulatory properties; however, its clinical application is limited by poor oral bioavailability. Mechanistically, curcumin attenuates oxidative stress and suppresses key inflammatory mediators, including nuclear factor kappa B (NF-κB), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS). Additionally, it modulates apoptosis, inhibits amyloid-beta aggregation, and enhances cellular quality control processes such as autophagy and mitophagy, while upregulating neurotrophic factors such as brain-derived neurotrophic factor (BDNF). Preclinical studies employing rodent models of AD, PD, and ischemic stroke have demonstrated curcumin’s dose-dependent neuroprotective efficacy, with improved outcomes observed using nanoparticle-based delivery systems. Early-phase clinical trials further support curcumin’s favorable safety profile and potential cognitive benefits, although challenges remain regarding pharmacokinetics, formulation standardization, and therapeutic reproducibility. Future directions include the development of advanced drug delivery platforms, combinatory therapeutic regimens, and personalized medicine approaches integrating curcumin within multifaceted neurotherapeutic strategies. Collectively, this narrative review highlights curcumin as a promising multi-targeted candidate for combating neurodegenerative diseases, while emphasizing the need for further translational and clinical validation. Full article
(This article belongs to the Special Issue Therapeutic Potential of Phytochemicals in Neurodegenerative Diseases)
Show Figures

Figure 1

12 pages, 4072 KB  
Article
A Comparative Analysis of Cardiac Amyloidosis and Cardiac Sarcoidosis: A Single-Center Experience
by Luka Katic, Sanjay Sivalokanathan, James Choi, Darren Kong, Vincent A. Torelli, Alexander Silverman, Alexander Nagourney, Usman Saeedullah, Komail Jafri, Syed Zaidi, Serdar Farhan and Ashish Correa
J. Clin. Med. 2025, 14(17), 6056; https://doi.org/10.3390/jcm14176056 - 27 Aug 2025
Viewed by 641
Abstract
Background/Objectives: Cardiac amyloidosis (CA) and cardiac sarcoidosis (CS) are two distinct infiltrative cardiomyopathies that can present with overlapping clinical features, including heart failure and arrhythmias. However, they arise from fundamentally different pathophysiological mechanisms: amyloid protein deposition in CA versus granulomatous inflammation in [...] Read more.
Background/Objectives: Cardiac amyloidosis (CA) and cardiac sarcoidosis (CS) are two distinct infiltrative cardiomyopathies that can present with overlapping clinical features, including heart failure and arrhythmias. However, they arise from fundamentally different pathophysiological mechanisms: amyloid protein deposition in CA versus granulomatous inflammation in CS. These differing pathophysiologies result in divergent imaging patterns, clinical trajectories, and treatment strategies. This study aims to compare the clinical presentations, imaging characteristics, and outcomes of patients with CA and CS to identify key differentiating factors that can improve diagnostic precision and guide therapy. Methods: This single-center, retrospective, cross-sectional study analyzed electronic medical records of patients diagnosed with CA (limited to transthyretin CA) or CS at Mount Sinai Morningside system from January 2017 until October 2023. Patients were identified using diagnostic codes and confirmed by histology or disease-specific imaging criteria. Clinical data, transthoracic echocardiography (TTE), cardiac magnetic resonance (CMR) imaging, pyrophosphate scintigraphy (PYP), and fluorodeoxyglucose positron emission tomography (FDG-PET) findings were collected. Statistical comparisons between groups were performed using chi-square tests and independent t-tests, with p < 0.05 considered statistically significant. Results: A total of 16,834 patients were screened and 216 patients were included in the analysis (125 CA, 92 CS). CA patients were older (78.2 vs. 62.0 years, p = 0.01), had greater interventricular septal thickness (1.57 vs. 1.10 cm, p = 0.01), and exhibited diffuse late gadolinium enhancement (LGE) and elevated extracellular volume (ECV) on CMR. CS patients had higher rates of ventricular tachycardia (53.3% vs. 10.7%, p = 0.01), increased myocardial fluorodeoxyglucose (FDG) uptake on positron emission tomography (PET) (90%), and more frequent implantable cardioverter-defibrillator (ICD) placement (66.3% vs. 13.0%, p = 0.01). Conclusions: CA and CS demonstrate distinct imaging profiles, arrhythmic risks, and treatment patterns. Early differentiation using advanced imaging is crucial for implementing disease-modifying therapies in CA and for immunosuppression and ICD implantation in CS, thereby improving patient outcomes. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

22 pages, 2604 KB  
Review
Targeting Metal Imbalance and Oxidative Stress in Alzheimer’s Disease with Novel Multifunctional Compounds
by Eleftherios Charissopoulos and Eleni Pontiki
Molecules 2025, 30(17), 3512; https://doi.org/10.3390/molecules30173512 - 27 Aug 2025
Viewed by 568
Abstract
Alzheimer’s disease (AD) is considered to be one of the most common types of dementia, threatening the health of elderly individuals. Enhancing the brain’s cholinergic activity is currently the primary therapeutic strategy for treating AD patients. Acetylcholine and butyrylcholine are key targets in [...] Read more.
Alzheimer’s disease (AD) is considered to be one of the most common types of dementia, threatening the health of elderly individuals. Enhancing the brain’s cholinergic activity is currently the primary therapeutic strategy for treating AD patients. Acetylcholine and butyrylcholine are key targets in this approach, as they function as neuromodulators within the cerebrum—particularly in its various cholinergic regions responsible for essential functions like memory, thought, inspiration, and excitement. Oxidative stress and free radicals are considered to play a crucial role in the pathogenesis of AD and may be key factors in its etiology. Additionally, oxidants and oxidative stress-induced products can upregulate amyloid precursor protein (APP) expression, promoting Aβ aggregation. Another major factor in the pathogenesis of AD is the imbalance of metal homeostasis in the brain. Notably, the mammalian brain contains significantly higher concentrations of Cu, Zn, and Fe ions compared to other tissues. The present review focuses on novel bifunctional metal chelators with potential antioxidant activity for the treatment of AD. Full article
(This article belongs to the Special Issue Therapeutic Agents for Neurodegenerative Disorders—2nd Edition)
Show Figures

Graphical abstract

28 pages, 1227 KB  
Review
Clinical Significance of APOE4 Genotyping: Potential for Personalized Therapy and Early Diagnosis of Alzheimer’s Disease
by Jelena Rajič Bumber, Valentino Rački, Silvestar Mežnarić, Gordana Pelčić and Jasenka Mršić-Pelčić
J. Clin. Med. 2025, 14(17), 6047; https://doi.org/10.3390/jcm14176047 - 26 Aug 2025
Viewed by 1024
Abstract
Apolipoprotein E (APOE) remains the most robust and widely replicated genetic risk factor for late-onset Alzheimer’s disease (AD) susceptibility, with the ε4 allele (APOE4) demonstrating profound associations with accelerated symptom manifestation, enhanced disease trajectory, and modified therapeutic responsiveness. This comprehensive review [...] Read more.
Apolipoprotein E (APOE) remains the most robust and widely replicated genetic risk factor for late-onset Alzheimer’s disease (AD) susceptibility, with the ε4 allele (APOE4) demonstrating profound associations with accelerated symptom manifestation, enhanced disease trajectory, and modified therapeutic responsiveness. This comprehensive review synthesizes contemporary evidence regarding the clinical utility of APOE4 genotyping, emphasizing its integration into personalized therapeutic frameworks and early diagnostic paradigms. The APOE4 variant exerts pathogenic influence through impaired amyloid-β clearance, enhanced tau pathology, and compromised neuronal repair mechanisms that alter disease phenotype. We systematically examine available genotyping methodologies, encompassing polymerase chain reaction (PCR) and next-generation sequencing (NGS) platforms, and evaluate their practical implementation within clinical environments. Recent investigations demonstrate that APOE4 status profoundly influences therapeutic efficacy, particularly with anti-amyloid interventions such as lecanemab, where carriers exhibit enhanced treatment response alongside increased adverse event susceptibility. Emerging gene therapeutic approaches show promise in mitigating APOE4-associated risks through targeted molecular interventions. The integration of APOE4 genotyping with fluid biomarkers and neuroimaging techniques enables refined patient stratification and enhanced diagnostic precision, facilitating earlier intervention windows that optimize therapeutic outcomes before irreversible neuronal damage occurs. This review underscores APOE4 testing as a transformative component of precision medicine in AD management, emphasizing its contribution to diagnostic refinement, clinical decision support, and targeted therapeutic interventions. Full article
Show Figures

Figure 1

23 pages, 13740 KB  
Article
Mulberroside A: A Multi-Target Neuroprotective Agent in Alzheimer’s Disease via Cholinergic Restoration and PI3K/AKT Pathway Activation
by Jin Li, Jiawen Wang, Yaodong Li, Jingyi Guo, Ziliang Jin, Shourong Qiao, Yunxia Zhang, Guoyin Li, Huazhen Liu and Changjing Wu
Biology 2025, 14(9), 1114; https://doi.org/10.3390/biology14091114 - 22 Aug 2025
Viewed by 515
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia, with current therapies offering only limited symptomatic relief and lacking disease-modifying efficacy. Addressing this critical therapeutic gap, natural multi-target compounds like mulberroside A (MsA)—a bioactive glycoside from Morus alba [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder and the leading cause of dementia, with current therapies offering only limited symptomatic relief and lacking disease-modifying efficacy. Addressing this critical therapeutic gap, natural multi-target compounds like mulberroside A (MsA)—a bioactive glycoside from Morus alba L.—present promising alternatives. This study investigated MsA’s neuroprotective potential using scopolamine-induced AD-like mice and N2a/APP695swe cells. In vivo, MsA significantly ameliorated cognitive deficits and neuronal loss, concurrently enhancing cholinergic neurotransmission through increased acetylcholine levels and inhibited acetylcholinesterase (AChE)/butyrylcholinesterase (BChE) activities. MsA also upregulated neurotrophic factors (BDNF, CREB) in critical brain regions. In vitro, MsA restored cholinergic function, mitigated oxidative stress, and crucially reduced amyloid-β (Aβ) production by dual regulation of APP processing: promoting the non-amyloidogenic pathway via ADAM10 upregulation and inhibiting the amyloidogenic pathway via suppression of BACE1 and γ-secretase components. Mechanistically, these multi-target benefits were mediated by MsA’s activation of the PI3K/AKT pathway, which triggered downstream inhibitory phosphorylation of GSK3β—directly reduced tau hyperphosphorylation—and activation of CREB/BDNF signaling. Collectively, our findings demonstrate that MsA confers comprehensive neuroprotection against AD pathology by simultaneously targeting cholinergic dysfunction, oxidative stress, Aβ accumulation, tau phosphorylation, and impaired neurotrophic signaling, highlighting its strong therapeutic candidacy. Full article
(This article belongs to the Section Neuroscience)
Show Figures

Figure 1

28 pages, 5633 KB  
Article
Investigation into Efficacy and Mechanisms of Neuroprotection of Ashwagandha Root Extract and Water-Soluble Coenzyme Q10 in a Transgenic Mouse Model of Alzheimer’s Disease
by Caleb Vegh, Gabrielle Walach, Keanna Dube, Bromleigh Dobson, Rohan Talukdar, Darcy Wear, Hasana Jayawardena, Kaitlyn Dufault, Lauren Culmone, Subidsa Srikantha, Iva Okaj, Rachel Huggard, Jerome Cohen and Siyaram Pandey
Nutrients 2025, 17(16), 2701; https://doi.org/10.3390/nu17162701 - 20 Aug 2025
Viewed by 1030
Abstract
Background: Alzheimer’s Disease (AD) is one of the most prevalent neurodegenerative disorders and the most common form of dementia. Although current treatments examine disease progression, many have side effects and primarily target symptomatic relief as opposed to halting further neurodegeneration. Objective: The current [...] Read more.
Background: Alzheimer’s Disease (AD) is one of the most prevalent neurodegenerative disorders and the most common form of dementia. Although current treatments examine disease progression, many have side effects and primarily target symptomatic relief as opposed to halting further neurodegeneration. Objective: The current study aims to determine the neuroprotective effects of water-soluble coenzyme Q10 (Ubisol-Q10) and an ethanolic Ashwagandha extract (E-ASH) on a transgenic mouse model of AD. Methods: A variety of immunofluorescence staining of biomarkers was conducted to assess mechanisms commonly implicated in the disease. Additionally, spatial and non-spatial memory tests evaluated cognitive functions at two timepoints throughout the progression of the disease. Results: A substantial reduction in microglial activation and amyloid-β (Aβ) plaques when treated with a combination of natural health products (NHPs), Ubisol-Q10 and E-ASH. Moreover, activation of autophagy was upregulated in both the Ubisol-Q10 and combination (Ubisol-Q10+E-ASH given as a combined “Tonic” solution) groups. Oxidative stress was decreased across treated groups, while astrocyte activation was elevated in both the E-ASH and Tonic group. The Tonic group expressed an elevation in the fluorescent intensity of neuronal nuclei (NeuN) and brain-derived neurotrophic factor (BDNF) levels. Interestingly, treatment with E-ASH and Ubisol-Q10 enhanced synaptic vesicle formation compared to controls. Pre-mortem memory tests revealed the treatments to be effective at preserving cognitive abilities. Conclusions: Based on these findings, the combination of E-ASH and Ubisol-Q10 may effectively mitigate the various mechanisms implicated in AD and ultimately prevent further disease progression. Full article
Show Figures

Figure 1

20 pages, 2920 KB  
Article
The Chelating Abilities of Tertiary Amines with N-O-Donors Towards Cu(II) Ions and the Catalytic Properties of the Resulting Complexes
by Martina Zonzin, Martina Chianese, Andrea Squarcina, Degnet Melese Dereje, Ambra Campofelice, Alessia Da Fermo, Federica Belluti, Nadia Marino, Filip Dębicki, Aleksandra Kotynia, Aleksandra Marciniak, Justyna Brasuń and Mauro Carraro
Molecules 2025, 30(16), 3419; https://doi.org/10.3390/molecules30163419 - 19 Aug 2025
Viewed by 794
Abstract
Oxidative stress, driven by excess reactive oxygen species (ROS), is a key factor in the progression of neurodegenerative diseases like Alzheimer’s disease (AD). In this context, copper dysregulation can also contribute to this imbalance, being responsible for enhanced ROS production, so that copper [...] Read more.
Oxidative stress, driven by excess reactive oxygen species (ROS), is a key factor in the progression of neurodegenerative diseases like Alzheimer’s disease (AD). In this context, copper dysregulation can also contribute to this imbalance, being responsible for enhanced ROS production, so that copper scavenging has been investigated as a possible therapeutic strategy. This study investigates the behavior of two isostructural ligands, featuring an N3O donor set, that effectively chelate Cu(II) in aqueous solution. Interestingly, their resulting mono- or dinuclear copper complexes feature a coordination environment suitable to foster antioxidant activity. By transforming copper’s oxidant potential into antioxidant action, these systems may reduce copper-induced oxidative damage. The work examines the pH-dependent metal-binding behavior of the ligands, the catalytic properties of the resulting complexes under physiological conditions, and their ability to inhibit β-amyloid peptide aggregation. Full article
Show Figures

Figure 1

23 pages, 4468 KB  
Article
Serrapeptase Eliminates Escherichia coli Biofilms by Targeting Curli Fibers, Lipopolysaccharides, and Phosphate Metabolism
by Georgios Katsipis, Michalis Aivaliotis and Anastasia A. Pantazaki
Microorganisms 2025, 13(8), 1875; https://doi.org/10.3390/microorganisms13081875 - 11 Aug 2025
Viewed by 1404
Abstract
Escherichia coli biofilms are implicated in the development of persistent infections and increased antibiotic resistance, posing a significant challenge in clinical settings. These biofilms enhance bacterial survival by forming protective extracellular matrices, rendering conventional treatments less effective. Serrapeptase (SPT), a proteolytic enzyme, has [...] Read more.
Escherichia coli biofilms are implicated in the development of persistent infections and increased antibiotic resistance, posing a significant challenge in clinical settings. These biofilms enhance bacterial survival by forming protective extracellular matrices, rendering conventional treatments less effective. Serrapeptase (SPT), a proteolytic enzyme, has emerged as a potential anti-biofilm agent due to its ability to degrade biofilm components and disrupt bacterial adhesion. In this study, we report the inhibitory effect of SPT against E. coli biofilm and its effect on key virulence factors. In vitro assays, including crystal violet staining, optical and fluorescence microscopy, and viability measurements, revealed the dose-dependent inhibition of biofilm formation (IC50 = 14.2 ng/mL), reduced biofilm (−92%, 500 ng/mL) and planktonic viability (−45%, 500 ng/mL), and a marked loss of amyloid curli fibers. SPT treatment also lowered the levels of key virulence factors: cellular and secreted lipopolysaccharides (−76%, 8 ng/mL; −94%, 32 ng/mL), flagellin (−63%, 8 ng/mL), and peptidoglycan (−29%, 125 ng/mL). Mechanistically, SPT induced a phosphate-dysregulating response: secreted alkaline phosphatase activity rose (+70%, 125 ng/mL) while cellular DING/PstS proteins declined (−84%, 64 ng/mL), correlating strongly with biofilm inhibition. In silico docking further suggests direct interactions between SPT and the curli subunits CsgA and CsgB, potentially blocking fiber polymerization. Together, these findings position SPT as a powerful non-antibiotic biofilm disruptor against E. coli, offering a promising strategy to undermine bacterial persistence and resistance by targeting both structural matrix components and metabolic regulatory pathways. Full article
Show Figures

Figure 1

21 pages, 1786 KB  
Review
Aortic Stiffness and Alzheimer’s Disease: The Medin Connection
by Filippos Triposkiadis, Andrew Xanthopoulos, Harisios Boudoulas and Dirk L. Brutsaert
Biomolecules 2025, 15(8), 1148; https://doi.org/10.3390/biom15081148 - 8 Aug 2025
Viewed by 801
Abstract
Aging is associated with aortic stiffening (AoSt), a condition characterized by diminished aortic elasticity that predisposes individuals to cognitive decline, including Alzheimer’s disease (AD). Emerging evidence implicates medin, which is derived from milk fat globule-EGF factor 8 protein (MFG-E8), as a key link [...] Read more.
Aging is associated with aortic stiffening (AoSt), a condition characterized by diminished aortic elasticity that predisposes individuals to cognitive decline, including Alzheimer’s disease (AD). Emerging evidence implicates medin, which is derived from milk fat globule-EGF factor 8 protein (MFG-E8), as a key link between AoSt and AD. Medin aggregates into aortic medial amyloid (AMA), which is found in approximately 97% of Caucasian individuals aged 50 and above, contributing to vascular inflammation, calcification, and loss of arterial elasticity. These changes may promote hyperpulsatile cerebral blood flow and impair glymphatic clearance, resulting in increased deposition of neurotoxic proteins, such as amyloid-β (Aβ) and possibly medin, which colocalizes with vascular Aβ in the brain. Medin enhances Aβ aggregation, generating heterologous fibrils, and thereby contributes to cerebrovascular dysfunction and neuroinflammation. This interaction (cross-seeding) may deteriorate amyloid pathology in both the vasculature and the parenchyma in AD. Furthermore, medin per se causes endothelial dysfunction, increases oxidative stress, and activates glial cells, promoting the development of a pro-inflammatory environment that enhances cognitive decline. In this manuscript, we contend that medin might act as a bridge connecting the age-related increase in aortic stiffness to AD, and therefore, medin might present a novel therapeutic target within this context. This hypothesis deserves experimental and clinical validation. Full article
Show Figures

Figure 1

24 pages, 1718 KB  
Article
Exploring the Impact of Bioactive Compounds Found in Extra Virgin Olive Oil on NRF2 Modulation in Alzheimer’s Disease
by Marilena M. Bourdakou, Eleni M. Loizidou and George M. Spyrou
Antioxidants 2025, 14(8), 952; https://doi.org/10.3390/antiox14080952 - 2 Aug 2025
Viewed by 801
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder marked by amyloid-β (Aβ) plaques, neurofibrillary tangles, blood–brain barrier dysfunction, oxidative stress (OS), and neuroinflammation. Current treatments provide symptomatic relief, but do not halt the disease’s progression. OS plays a crucial role in AD pathogenesis [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder marked by amyloid-β (Aβ) plaques, neurofibrillary tangles, blood–brain barrier dysfunction, oxidative stress (OS), and neuroinflammation. Current treatments provide symptomatic relief, but do not halt the disease’s progression. OS plays a crucial role in AD pathogenesis by promoting Aβ accumulation. Nuclear factor erythroid 2-related factor 2 (NRF2) is a key regulator of the antioxidant response, influencing genes involved in OS mitigation, mitochondrial function, and inflammation. Dysregulation of NRF2 is implicated in AD, making it a promising therapeutic target. Emerging evidence suggests that adherence to a Mediterranean diet (MD), which is particularly rich in polyphenols from extra virgin olive oil (EVOO), is associated with improved cognitive function and a reduced risk of mild cognitive impairment. Polyphenols can activate NRF2, enhancing endogenous antioxidant defenses. This study employs a computational approach to explore the potential of bioactive compounds in EVOO to modulate NRF2-related pathways in AD. We analyzed transcriptomic data from AD and EVOO-treated samples to identify NRF2-associated genes, and used chemical structure-based analysis to compare EVOO’s bioactive compounds with known NRF2 activators. Enrichment analysis was performed to identify common biological functions between NRF2-, EVOO-, and AD-related pathways. Our findings highlight important factors and biological functions that provide new insight into the molecular mechanisms through which EVOO consumption might influence cellular pathways associated with AD via modulation of the NRF2 pathway. The presented approach provides a different perspective in the discovery of compounds that may contribute to neuroprotective mechanisms in the context of AD. Full article
Show Figures

Graphical abstract

19 pages, 9689 KB  
Article
Anionic Lipid Catalyzes the Generation of Cytotoxic Insulin Oligomers
by Jhinuk Saha, Audrey Wolszczak, Navneet Kaur, Malitha C. Dickwella Widanage, Samuel D. McCalpin, Riqiang Fu, Jamel Ali and Ayyalusamy Ramamoorthy
Biomolecules 2025, 15(7), 994; https://doi.org/10.3390/biom15070994 - 11 Jul 2025
Viewed by 424
Abstract
The misfolding and aggregation of proteins into amyloidogenic assemblies are key features of several metabolic and neurodegenerative diseases. Human insulin has long been known to form amyloid fibrils under various conditions, which affects its bioavailability and function. Clinically, insulin aggregation at recurrent injection [...] Read more.
The misfolding and aggregation of proteins into amyloidogenic assemblies are key features of several metabolic and neurodegenerative diseases. Human insulin has long been known to form amyloid fibrils under various conditions, which affects its bioavailability and function. Clinically, insulin aggregation at recurrent injection sites poses a challenge for diabetic patients who rely on insulin therapy. Furthermore, decreased responsiveness to insulin in type 2 diabetic (T2D) patients may lead to its overproduction and accumulation as aggregates. Earlier reports have reported that various factors such as pH, temperature, agitation, and the presence of lipids or other proteins influence insulin aggregation. Our present study aims to elucidate the effects of non–micellar anionic DMPG (1,2–dimyristoyl–sn–glycero–3–phosphoglycerol) lipids on insulin aggregation. Distinct pathways of insulin aggregation and intermediate formation were observed in the presence of DMPG using a ThT fluorescence assay. The formation of soluble intermediates alongside large insulin fibrils was observed in insulin incubated with DMPG via TEM, DLS, and NMR as opposed to insulin aggregates generated without lipids. 13C magic angle spinning solid–state NMR and FTIR experiments indicated that lipids do not alter the conformation of insulin fibrils but do alter the time scale of motion of aromatic and aliphatic side chains. Furthermore, the soluble intermediates were found to be more cytotoxic than fibrils generated with or without lipids. Overall, our study elucidates the importance of anionic lipids in dictating the pathways and intermediates associated with insulin aggregation. These findings could be useful in determining various approaches to avoid toxicity and enhance the effectiveness of insulin in therapeutic applications. Full article
(This article belongs to the Collection Feature Papers in 'Biomacromolecules: Proteins')
Show Figures

Figure 1

37 pages, 2135 KB  
Review
Neuroprotective Mechanisms of Red Algae-Derived Bioactive Compounds in Alzheimer’s Disease: An Overview of Novel Insights
by Tianzi Wang, Wenling Shi, Zijun Mao, Wei Xie and Guoqing Wan
Mar. Drugs 2025, 23(7), 274; https://doi.org/10.3390/md23070274 - 30 Jun 2025
Viewed by 1302
Abstract
Alzheimer’s disease (AD) is characterized by β-amyloid plaques, neurofibrillary tangles, neuroinflammation, and oxidative stress—pathological features that pose significant challenges for the development of therapeutic interventions. Given these challenges, this review comprehensively evaluates the neuroprotective mechanisms of bioactive compounds derived from red algae, [...] Read more.
Alzheimer’s disease (AD) is characterized by β-amyloid plaques, neurofibrillary tangles, neuroinflammation, and oxidative stress—pathological features that pose significant challenges for the development of therapeutic interventions. Given these challenges, this review comprehensively evaluates the neuroprotective mechanisms of bioactive compounds derived from red algae, including polysaccharides and phycobiliproteins, which are considered a promising source of natural therapeutics for AD. Red algal constituents exhibit neuroprotective activities through multiple mechanisms. Sulfated polysaccharides (e.g., carrageenan, porphyran) suppress NF-κB-mediated neuroinflammation, modulate mitochondrial function, and enhance brain-derived neurotrophic factor (BDNF) expression. Phycobiliproteins (phycoerythrin, phycocyanin) and peptides derived from their degradation scavenge reactive oxygen species (ROS) and activate antioxidant pathways (e.g., Nrf2/HO-1), thus mitigating oxidative damage. Carotenoids (lutein, zeaxanthin) improve cognitive function through the inhibition of acetylcholinesterase and pro-inflammatory cytokines (TNF-α, IL-1β), while phenolic compounds (bromophenols, diphlorethol) provide protection by targeting multiple pathways involved in dopaminergic system modulation and Nrf2 pathway activation. Emerging extraction technologies—including microwave- and enzyme-assisted methods—have been shown to optimize the yield and maintain the bioactivity of these compounds. However, the precise identification of molecular targets and the standardization of extraction techniques remain critical research priorities. Overall, red algae-derived compounds hold significant potential for multi-mechanism AD interventions, providing novel insights for the development of therapeutic strategies with low toxicity. Full article
(This article belongs to the Special Issue Marine-Derived Bioactive Compounds for Neuroprotection)
Show Figures

Figure 1

18 pages, 9930 KB  
Article
The Neuroprotective Potential of Seed Extract from the Indian Trumpet Tree Against Amyloid Beta-Induced Toxicity in SH-SY5Y Cells
by Nut Palachai, Benjaporn Buranrat, Parinya Noisa and Nootchanat Mairuae
Int. J. Mol. Sci. 2025, 26(13), 6288; https://doi.org/10.3390/ijms26136288 - 29 Jun 2025
Viewed by 741
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with an unclear etiology. Multiple factors, including oxidative stress and the accumulation of amyloid beta (Aβ) protein in the brain, contribute to neuronal damage. This study investigated Aβ-induced oxidative stress and cellular damage in SH-SY5Y [...] Read more.
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with an unclear etiology. Multiple factors, including oxidative stress and the accumulation of amyloid beta (Aβ) protein in the brain, contribute to neuronal damage. This study investigated Aβ-induced oxidative stress and cellular damage in SH-SY5Y cells, as well as the neuroprotective potential of Indian trumpet tree seed extract (ITS). SH-SY5Y cells were co-treated with Aβ(25–35) (20 µM) and ITS extract at concentrations of 25 and 50 µg/mL. Cell viability, reactive oxygen species (ROS), malondialdehyde (MDA) levels, and the enzymatic activities of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were assessed. The expression levels of B-cell lymphoma 2 (Bcl-2) and caspase-3, along with the phosphorylation levels of protein kinase B (Akt), extracellular signal-regulated kinases 1 and 2 (ERK1/2), and cAMP response element-binding protein (CREB), were also evaluated. ITS extract at concentrations of 25 and 50 µg/mL significantly improved SH-SY5Y cell viability following Aβ-induced damage; reduced ROS and MDA levels; and enhanced CAT, SOD, and GSH-Px activities. In addition to upregulating Bcl-2 expression, ITS downregulated caspase-3 expression and increased the phosphorylation of Akt, ERK1/2, and CREB. High-performance liquid chromatography (HPLC) analysis identified baicalin, baicalein, and chrysin as major phenolic compounds in ITS extract. In conclusion, ITS extract attenuated Aβ-induced oxidative stress, enhanced antioxidant defenses and cell viability, suppressed apoptotic signaling, and activated key neuroprotective pathways. These findings provide new insights into the neuroprotective potential of ITS extract; however, further in vivo studies are needed to validate its clinical applicability. Full article
(This article belongs to the Special Issue Natural Products for Neuroprotection and Neurodegeneration)
Show Figures

Figure 1

25 pages, 6990 KB  
Article
Study on the Pharmacological Efficacy and Mechanism of Dual-Target Liposome Complex AD808 Against Alzheimer’s Disease
by Chang Liu, Xiaoqing Wang, Wei Xu, Songli Yu, Yueru Zhang, Qiming Xu and Xiangshi Tan
Pharmaceuticals 2025, 18(7), 977; https://doi.org/10.3390/ph18070977 - 29 Jun 2025
Viewed by 806
Abstract
Background/Objectives: To study the efficacy and pharmacological mechanism of the dual-target liposome complex AD808 in the treatment of Alzheimer’s disease. Methods: Using APP/PS1 mouse models, the therapeutic efficacy and pharmacological mechanism of AD808 on Alzheimer’s disease were studied through water maze [...] Read more.
Background/Objectives: To study the efficacy and pharmacological mechanism of the dual-target liposome complex AD808 in the treatment of Alzheimer’s disease. Methods: Using APP/PS1 mouse models, the therapeutic efficacy and pharmacological mechanism of AD808 on Alzheimer’s disease were studied through water maze tests, brain tissue staining, immunofluorescence, and ELISA for inflammatory and neurotrophic factors. Results: AD808 exhibited significant pharmacodynamic effects in improving behavioral and cognitive abilities (70% reduction in escape latency) and repairing damaged nerve cells (90% reduction in Aβ plaque) in Alzheimer’s disease mice. The efficacy of the liposome complex AD808 was significantly better than that of ST707 or gh625-Zn7MT3 alone. AD808 significantly reduced brain inflammation (57.3% and 61.5% reductions in TNF-α and IL-1β, respectively) in AD (Alzheimer’s disease) mouse models and promoted the upregulation of neurotrophic factors and nerve growth factors (142.8% increase in BDNF, 275.9% in GDNF, and 111.3% in NGF-1) in brain homogenates. By activating the PI3K/AKT signaling pathway in brain microglia, AD808 upregulated TREM2 protein expression and removed Aβ amyloid plaques in the brain. Additionally, it promoted the transition of microglia from the pro-inflammatory M1 phenotype to the anti-inflammatory M2 phenotype, regulated the M1/M2 balance, released anti-inflammatory and neurotrophic factors, reduced chronic inflammation, and enhanced neurological repair. Based on these results, the potential pharmacological mechanism of AD808 against Alzheimer’s disease was proposed. Conclusions: As a dual-target liposome complex, AD808 has shown promising therapeutic potential in the treatment of Alzheimer’s disease, providing a new strategy for innovative drug development. Full article
(This article belongs to the Special Issue Pharmacotherapy for Alzheimer’s Disease)
Show Figures

Figure 1

28 pages, 7907 KB  
Article
Estradiol Prevents Amyloid Beta-Induced Mitochondrial Dysfunction and Neurotoxicity in Alzheimer’s Disease via AMPK-Dependent Suppression of NF-κB Signaling
by Pranav Mishra, Ehsan K. Esfahani, Paul Fernyhough and Benedict C. Albensi
Int. J. Mol. Sci. 2025, 26(13), 6203; https://doi.org/10.3390/ijms26136203 - 27 Jun 2025
Viewed by 1485
Abstract
Alzheimer’s disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder characterized by memory loss and cognitive decline. In addition to its two major pathological hallmarks, extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs), recent evidence highlights the [...] Read more.
Alzheimer’s disease (AD), the most common form of dementia, is a progressive neurodegenerative disorder characterized by memory loss and cognitive decline. In addition to its two major pathological hallmarks, extracellular amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs), recent evidence highlights the critical roles of mitochondrial dysfunction and neuroinflammation in disease progression. Aβ impairs mitochondrial function, which, in part, can subsequently trigger inflammatory cascades, creating a vicious cycle of neuronal damage. Estrogen receptors (ERs) are widely expressed throughout the brain, and the sex hormone 17β-estradiol (E2) exerts neuroprotection through both anti-inflammatory and mitochondrial mechanisms. While E2 exhibits neuroprotective properties, its mechanisms against Aβ toxicity remain incompletely understood. In this study, we investigated the neuroprotective effects of E2 against Aβ-induced mitochondrial dysfunction and neuroinflammation in primary cortical neurons, with a particular focus on the role of AMP-activated protein kinase (AMPK). We found that E2 treatment significantly increased phosphorylated AMPK and upregulated the expression of mitochondrial biogenesis regulator peroxisome proliferator-activated receptor gamma coactivator-1 α (PGC-1α), leading to improved mitochondrial respiration. In contrast, Aβ suppressed AMPK and PGC-1α signaling, impaired mitochondrial function, activated the pro-inflammatory nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB), and reduced neuronal viability. E2 pretreatment also rescued Aβ-induced mitochondrial dysfunction, suppressed NF-κB activation, and, importantly, prevented the decline in neuronal viability. However, the pharmacological inhibition of AMPK using Compound C (CC) abolished these protective effects, resulting in mitochondrial collapse, elevated inflammation, and cell death, highlighting AMPK’s critical role in mediating E2’s actions. Interestingly, while NF-κB inhibition using BAY 11-7082 partially restored mitochondrial respiration, it failed to prevent Aβ-induced cytotoxicity, suggesting that E2’s full neuroprotective effects rely on broader AMPK-dependent mechanisms beyond NF-κB suppression alone. Together, these findings establish AMPK as a key mediator of E2’s protective effects against Aβ-driven mitochondrial dysfunction and neuroinflammation, providing new insights into estrogen-based therapeutic strategies for AD. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Graphical abstract

Back to TopTop