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

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (190)

Search Parameters:
Keywords = microtubule disruption

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 1576 KB  
Article
A Supra-Physiological Dose of 2-Hydroxyestradiol Impairs Meiotic Progression and Developmental Competence of Mouse Antral Oocytes
by Valeria Merico, Paola Rebuzzini, Mario Zanoni, Maurizio Zuccotti and Silvia Garagna
J. Dev. Biol. 2025, 13(4), 37; https://doi.org/10.3390/jdb13040037 - 15 Oct 2025
Abstract
Estrogen metabolites (EMs) play a local regulatory role in mammalian ovarian function. Among them, 2-hydroxyestradiol (2-OHE2) exerts dose-dependent effects on reproductive physiology, supporting either normal ovarian processes or contributing to pathological conditions. Specifically, 2-OHE2 modulates ovarian vasculature and progesterone biosynthesis, and at 1–10 [...] Read more.
Estrogen metabolites (EMs) play a local regulatory role in mammalian ovarian function. Among them, 2-hydroxyestradiol (2-OHE2) exerts dose-dependent effects on reproductive physiology, supporting either normal ovarian processes or contributing to pathological conditions. Specifically, 2-OHE2 modulates ovarian vasculature and progesterone biosynthesis, and at 1–10 nM concentrations, it enhances in vitro developmental competence and blastocyst quality in mouse oocytes. Conversely, doses below 1 nM show no appreciable effects, suggesting the existence of a biological activity threshold. However, the impact of supra-physiological concentrations remains largely unexplored. In this study, we investigated the effects of increasing 2-OHE2 doses (0.05, 0.50, and 5.00 µM) on oocyte meiotic progression and quality. Exposure to 0.50 and 5.00 µM significantly impaired oocyte maturation, while only the highest dose notably reduced the percentage of embryos developing to the blastocyst stage. Morphometric analysis during the GV-to-MII transition revealed altered first polar body morphology, defective asymmetric division, and disruptions in cytoskeletal organization, including enlarged meiotic spindles, increased F-actin cap angles, and aberrant microtubule-organizing centers distribution. These structural alterations were paralleled by distinct changes in cytoplasmic movement velocity patterns observed through time-lapse imaging during meiotic resumption. Together, these findings demonstrate that supra-physiological exposure to 2-OHE2 compromises oocyte maturation and developmental competence by perturbing key cytoskeletal dynamics and cellular architecture necessary for successful meiosis and early embryogenesis. Full article
Show Figures

Figure 1

23 pages, 5324 KB  
Article
Vincristine Beyond Mitosis: Uncovering a First Link to G-Quadruplex DNA in Cancer Cells
by Anna Di Porzio, Carolina Persico, Francesca Romano, Alessandra Barra, Immacolata Aiello, Ludovica D’Auria, Sara Abate, Federica D’Aria, Concetta Giancola, Elpidio Cinquegrana, Francesco Saverio Di Leva, Jussara Amato, Simona Marzano, Nunzia Iaccarino and Antonio Randazzo
Int. J. Mol. Sci. 2025, 26(19), 9606; https://doi.org/10.3390/ijms26199606 - 1 Oct 2025
Viewed by 357
Abstract
Vincristine is a classical chemotherapeutic agent widely used for its ability to disrupt microtubule polymerization, yet additional molecular effects may contribute to its anticancer activity. G-quadruplexes (G4s), non-canonical nucleic acid structures enriched in regulatory regions of the genome and in mitochondrial DNA, have [...] Read more.
Vincristine is a classical chemotherapeutic agent widely used for its ability to disrupt microtubule polymerization, yet additional molecular effects may contribute to its anticancer activity. G-quadruplexes (G4s), non-canonical nucleic acid structures enriched in regulatory regions of the genome and in mitochondrial DNA, have emerged as relevant modulators of cellular homeostasis. In this study, we investigated whether vincristine can influence G4 biology. Cancer cells treated with vincristine were analyzed by immunofluorescence, revealing a consistent increase in nuclear and mitochondrial G4 foci. In particular, mitochondrial G4s were significantly elevated by approximately 1.5–2.5 fold compared to untreated cells, an effect accompanied by a detectable reduction in membrane potential, indicative of impaired organelle function. In addition, biophysical analyses on representative G4-forming sequences were carried out. Proton nuclear magnetic resonance titrations showed localized chemical shift perturbations upon vincristine addition, circular dichroism confirmed preservation of G4 topology, and isothermal titration calorimetry indicated weak but enthalpically favorable interactions. Taken together, these results suggest that vincristine perturbs both the cellular G4 landscape and mitochondrial homeostasis, while also engaging G4 DNA in vitro. Although additional studies are required to establish the mechanistic details, this work provides proof-of-concept for a previously unrecognized dimension of vincristine’s anticancer action. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

28 pages, 3546 KB  
Article
SCAMP3-Driven Regulation of ERK1/2 and Autophagy Phosphoproteomics Signatures in Triple-Negative Breast Cancer
by Beatriz M. Morales-Cabán, Yadira M. Cantres-Rosario, Eduardo L. Tosado-Rodríguez, Abiel Roche-Lima, Loyda M. Meléndez, Nawal M. Boukli and Ivette J. Suarez-Arroyo
Int. J. Mol. Sci. 2025, 26(19), 9577; https://doi.org/10.3390/ijms26199577 - 1 Oct 2025
Viewed by 355
Abstract
Extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitors show therapeutic potential in triple-negative breast cancer (TNBC), but resistance through compensatory signaling limits their efficacy. We previously identified the secretory carrier membrane protein 3 (SCAMP3) as a regulator of TNBC progression and ERK1/2 activation. Here, we [...] Read more.
Extracellular signal-regulated kinase 1/2 (ERK1/2) inhibitors show therapeutic potential in triple-negative breast cancer (TNBC), but resistance through compensatory signaling limits their efficacy. We previously identified the secretory carrier membrane protein 3 (SCAMP3) as a regulator of TNBC progression and ERK1/2 activation. Here, we investigated the role of SCAMP3 in ERK1/2 signaling and therapeutic response using TMT-based LC-MS/MS phosphoproteomics of wild-type (WT) and SCAMP3 knockout (SC3KO) SUM-149 cells under basal conditions, after epidermal growth factor (EGF) stimulation, and during ERK1/2 inhibition with MK-8353. A total of 4408 phosphosites were quantified, with 1093 significantly changed. SC3KO abolished residual ERK activity under MK-8353 and affected the compensatory activation of oncogenic pathways observed in WT cells. SC3KO reduced the phosphorylation of ERK feedback regulators RAF proto-oncogene serine/threonine-protein kinase Raf-1 (S43) and the dual-specificity mitogen-activated protein kinase kinase 2 (MEK2) (T394), affected other ERK targets, including nucleoporins, transcription factors, and metabolic enzymes triosephosphate isomerase (TPI1) (S21) and ATP-citrate lyase (ACLY) (S455). SCAMP3 loss also impaired the mammalian target of rapamycin complex I (mTORC1) signaling and disrupted autophagic flux, evidenced by elevated sequestosome-1 (SQSTM1/p62) and microtubule-associated protein light chain 3 (LC3B-II) with reduced levels of the autophagosome lysosome maturation marker, Rab7A. Beyond ERK substrates, SC3KO affected phosphorylation events mediated by other kinases. These findings position SCAMP3 as a central coordinator of ERK signaling and autophagy. Our results support SCAMP3 as a potential therapeutic target to enhance ERK1/2 inhibitor clinical efficacy and overcome adaptive resistance mechanisms in TNBC. Full article
Show Figures

Figure 1

37 pages, 801 KB  
Review
Tau-Targeted Therapeutic Strategies: Mechanistic Targets, Clinical Pipelines, and Analysis of Failures
by Xinai Shen, Huan Li, Beiyu Zhang, Yunan Li and Zheying Zhu
Cells 2025, 14(19), 1506; https://doi.org/10.3390/cells14191506 - 26 Sep 2025
Viewed by 984
Abstract
Tau protein, a neuron-enriched microtubule-associated protein encoded by the MAPT gene, plays pivotal roles in microtubule stabilisation, axonal transport, and synaptic plasticity. Aberrant post-translational modifications (PTMs), hyperphosphorylation, acetylation, ubiquitination, oxidative stress and neuroinflammation disrupt tau’s normal functions, drive its mislocalization, and promote aggregation [...] Read more.
Tau protein, a neuron-enriched microtubule-associated protein encoded by the MAPT gene, plays pivotal roles in microtubule stabilisation, axonal transport, and synaptic plasticity. Aberrant post-translational modifications (PTMs), hyperphosphorylation, acetylation, ubiquitination, oxidative stress and neuroinflammation disrupt tau’s normal functions, drive its mislocalization, and promote aggregation into neurofibrillary tangles, a hallmark of Alzheimer’s disease (AD) and related tauopathies. Over the past two decades, tau-targeted therapies have advanced into clinical development, yet most have failed to demonstrate efficacy in human trials. This review synthesises mechanistic insights into tau biology and pathology, highlighting phosphorylation and acetylation pathways, aggregation-prone motifs, and immune-mediated propagation. We analyse the current therapeutic landscape, including kinase and phosphatase modulators, O-GlcNAcase inhibitors, aggregation blockers, immunotherapies, and microtubule-stabilising agents, while examining representative clinical programs and the reasons underlying their limited success. By combining mechanistic understanding with clinical experience, this review outlines emerging opportunities for rational treatment development, aiming to inform future tau-targeted strategies for AD and other tauopathies. Full article
(This article belongs to the Special Issue Recent Advances in the Study of Tau Protein)
Show Figures

Figure 1

39 pages, 13134 KB  
Article
Mitotic Disruption and Cytoskeletal Alterations Induced by Acorus calamus Essential Oil: Implications for Bioherbicidal Potential
by Mateusz Wróblewski, Natalia Gocek, Aneta Żabka and Justyna T. Polit
Int. J. Mol. Sci. 2025, 26(18), 8933; https://doi.org/10.3390/ijms26188933 - 13 Sep 2025
Viewed by 411
Abstract
Essential oils are increasingly recognized as promising agents for sustainable weed control due to their selectivity and complex modes of action. This study evaluated the effects of Acorus calamus essential oil (SEO) on mitosis in two Fabaceae species (Vicia faba, Lupinus [...] Read more.
Essential oils are increasingly recognized as promising agents for sustainable weed control due to their selectivity and complex modes of action. This study evaluated the effects of Acorus calamus essential oil (SEO) on mitosis in two Fabaceae species (Vicia faba, Lupinus luteus) and two Brassicaceae species (Brassica napus, Arabidopsis thaliana) treated with species-specific IC50 concentrations (0.03%, 0.025%, 0.01%, and 0.005%, respectively). Previous research showed that SEO induces oxidative stress and S-phase delay via genome instability. Here, SEO consistently disrupted mitosis across all species, reducing mitotic index by 50–60%, decreasing Cdc2 (CDKA homolog) levels, and causing chromosomal aberrations, including uneven chromatin condensation, sticky chromosomes, bridges, and micronuclei. Cells accumulated in metaphase and exhibited abnormal karyokinetic and cytokinetic spindles. Immunolabeling revealed thick, tightly packed microtubules and actin filaments, indicating excessive stabilization and impaired reorganization. Epigenetic regulation was also affected: H3T3 phosphorylation was abnormally strong, widely distributed, and persistent into anaphase/telophase, while H3S10Ph intensity was weakened. These results suggest that SEO targets multiple components of mitotic machinery and epigenetic control, regardless of species. The observed selectivity depends on dosage, not mechanism. This multi-targeted action may limit the development of plant resistance, supporting the potential of SEO as a bioherbicide in sustainable agriculture. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Figure 1

13 pages, 2086 KB  
Article
Bioactivity-Guided Fractionation and Mechanistic Insights into Aristolochia ringens Root Extract-Induced G1 Phase Arrest and Mitochondria-Mediated Apoptosis in Human Colon Adenocarcinoma Cells
by Saheed O. Anifowose, Abdalrhaman M. Salih, Musa K. Oladejo, Ahmad Rady, Mobarak S. Al Mosallam, Hasan A. Aljohi, Mansour I. Almansour, Saad Hussin Alkahtani, Ibrahim O. Alanazi and Badr A. Al-Dahmash
Pharmaceuticals 2025, 18(9), 1250; https://doi.org/10.3390/ph18091250 - 23 Aug 2025
Viewed by 723
Abstract
Background/Objectives: Aristolochia ringens, a medicinal plant widely used in traditional medicine, has shown potential therapeutic applications. This study aimed to investigate the anticancer mechanism of action of its crude extract against human colorectal adenocarcinoma cells (Caco-2 and HT-29). Methods: Cell [...] Read more.
Background/Objectives: Aristolochia ringens, a medicinal plant widely used in traditional medicine, has shown potential therapeutic applications. This study aimed to investigate the anticancer mechanism of action of its crude extract against human colorectal adenocarcinoma cells (Caco-2 and HT-29). Methods: Cell viability was assessed using the MTT assay to determine IC50 values. Immunofluorescence microscopy was used to examine nuclear morphology and microtubule integrity. Flow cytometry with PI staining was used for cell cycle analysis and Annexin V-FITC/PI staining for apoptosis detection. Mitochondrial membrane potential was evaluated using JC-1 dye. Bioactivity-guided fractionation was performed via HPLC, and GC–MS was used to profile active constituents. Results: The extract exhibited dose-dependent cytotoxicity with IC50 values below 30 µg/mL in colon adenocarcinoma cell lines. Treated Caco-2 cells showed nuclear shrinkage and disrupted microtubules. PI-based flow cytometry revealed G1 phase arrest, and Annexin V-FITC/PI staining indicated enhanced late apoptosis. JC-1 staining demonstrated mitochondrial depolarization. HPLC fractionation identified fractions 2 and 3 as active, and preliminary GC–MS analysis tentatively annotated the presence of alkaloids, sesquiterpenes/diterpenes, and steroidal compounds. Conclusions: A. ringens exerts anticancer effects through a mitochondria-mediated apoptotic pathway, involving G1 checkpoint arrest and cytoskeletal disruption. These findings provide the first integrated cellular and mechanistic evidence of its anticancer potential in colorectal cancer, supporting its promise as a source of novel therapeutic lead compounds. Full article
Show Figures

Graphical abstract

11 pages, 4923 KB  
Article
Expanding the Phenotypic Spectrum of SPG4: Autism Spectrum Disorder in Early-Onset and Complex SPAST-HSP and Case Study
by Carlo Alberto Quaranta, Alice Gardani, Giulia Andorno, Anna Pichiecchio, Simone Gana, Renato Borgatti and Simona Orcesi
Genes 2025, 16(8), 970; https://doi.org/10.3390/genes16080970 - 18 Aug 2025
Viewed by 846
Abstract
Background/Objectives: Hereditary spastic paraplegias (HSPs) comprise a heterogenous spectrum of rare neurogenetic disorders predominantly characterized by progressive spasticity and weakness of the lower extremities. Among autosomal-dominant forms of HSP, molecular defects in the SPAST gene—particularly those associated with the SPG4 subtype—represent the most [...] Read more.
Background/Objectives: Hereditary spastic paraplegias (HSPs) comprise a heterogenous spectrum of rare neurogenetic disorders predominantly characterized by progressive spasticity and weakness of the lower extremities. Among autosomal-dominant forms of HSP, molecular defects in the SPAST gene—particularly those associated with the SPG4 subtype—represent the most frequent genetic cause. SPAST encodes spastin, a microtubule-severing ATPase, crucial for cytoskeletal remodeling, neuronal connectivity, and intracellular trafficking. Disruption of spastin function can impair neurite outgrowth and synaptic formation, processes increasingly implicated in neurodevelopmental disorders (NDDs). Methods: We conducted a comprehensive clinical, neurological, and dysmorphological evaluation of a 4-year-old male. Standardized neuropsychological assessments were administered. Whole-exome sequencing (WES) was performed to identify underlying genetic causes. EEG and 3T-brain MRI were also acquired. Results: The proband harbored two novel de novo heterozygous missense variants in cis of the SPAST gene, displaying the typical features of early-onset and complex HSP, in addition to global developmental delay and severe autism spectrum disorder (ASD), an underexplored manifestation in this rare genetic disorder. Conclusions: These findings broaden the clinical and mutational spectrum of SPG4, underscoring the importance of considering SPAST gene analysis in patients with ASD in the early years of life and early motor delay, even in the presence of only subtle pyramidal signs. We advocate for comprehensive neuropsychiatric assessment in the diagnostic pathway of early-onset complex HSP presentations and support further investigation into the role of spastin in neuronal connectivity. Full article
(This article belongs to the Section Neurogenomics)
Show Figures

Figure 1

24 pages, 4513 KB  
Article
Anticancer Activity of Paclitaxel-Loaded Mesoporous Silica Nanoparticles in B16F10 Melanoma-Bearing Mice
by Jihoon Lee, Jung Mo Kim, Yeon-Ju Baek, Hyojeung Kang, Min-Koo Choi and Im-Sook Song
Pharmaceutics 2025, 17(8), 1042; https://doi.org/10.3390/pharmaceutics17081042 - 11 Aug 2025
Viewed by 837
Abstract
Background/Objectives: Paclitaxel (PTX) faces clinical limitations in melanoma treatment due to poor solubility, P-glycoprotein (P-gp)-mediated efflux, and systemic toxicity. This study aimed to develop PTX-loaded mesoporous silica nanoparticles (PS), which would be co-administered with curcumin (CUR) and D-α-tocopherol polyethylene glycol 1000 succinate [...] Read more.
Background/Objectives: Paclitaxel (PTX) faces clinical limitations in melanoma treatment due to poor solubility, P-glycoprotein (P-gp)-mediated efflux, and systemic toxicity. This study aimed to develop PTX-loaded mesoporous silica nanoparticles (PS), which would be co-administered with curcumin (CUR) and D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) to enhance intracellular accumulation and improve anti-tumor activity. CUR and TPGS were integrated with PS to inhibit P-gp-mediated PTX-efflux, to enhance the intracellular accumulation of PTX, and to improve anti-tumor activity in B16F10 cells. Methods: The physicochemical properties of PS were analyzed using standard characterization methods. The antitumor activity of PS co-administered with CUR and TPGS was evaluated using two-dimensional (2D) culture and three-dimensional (3D) spheroid assays, and also assessed in B16F10 tumor-bearing mice. The therapeutic mechanism of the PS combination was compared using apoptosis and microtubule disruption through flow cytometry and confocal microscopy. The pharmacokinetics and biodistribution of the PS combination were compared in B16F10 tumor-bearing mice. Results: PS formulations exhibited amorphous transformation with an approximate particle size of 200 nm. PS co-administered with CUR and TPGS reduced the IC50 to 178.7 nM compared with 283.3 nM for free PTX in B16F10 melanoma cells and achieved significant tumor growth inhibition in B16F10 melanoma spheroid culture. The intracellular accumulation of PTX correlated with its therapeutic efficacy. Flow cytometry revealed a significant induction of both early and late apoptosis in cells treated with the PS + CUR + TPGS combination, while confocal imaging confirmed enhanced microtubule disruption. In B16F10 tumor-bearing mice, PS co-administered with CUR and TPGS demonstrated higher and selective distribution of PTX into tumor tissue without affecting systemic exposure of PTX in B16F10-xenografted mice. Conclusions: PS + CUR + TPGS combination enhanced PTX delivery by improving solubility and enhancing distribution to tumor tissue through P-gp inhibition, thereby increasing its therapeutic potential. The combination of CUR and TPGS offers synergistic apoptosis induction and microtubule disruption. Thus, the PS + CUR + TPGS combination represents a promising approach for treating drug-resistant melanomas. Full article
(This article belongs to the Special Issue Targeted Drug Delivery to Improve Cancer Therapy, 2nd Edition)
Show Figures

Graphical abstract

38 pages, 3745 KB  
Review
Recent Advances in Microtubule Targeting Agents for Cancer Therapy
by Henrique C. Assunção, Patrícia M. A. Silva, Hassan Bousbaa and Honorina Cidade
Molecules 2025, 30(16), 3314; https://doi.org/10.3390/molecules30163314 - 8 Aug 2025
Cited by 1 | Viewed by 1835
Abstract
Cancer mortality and the development of cancer resistance present significant challenges that must be addressed to ensure global health. Among anticancer agents, microtubule-targeting agents (MTAs) represent a well-recognized therapeutic approach that disrupts microtubule dynamics, thereby inhibiting cell division, and has been widely used [...] Read more.
Cancer mortality and the development of cancer resistance present significant challenges that must be addressed to ensure global health. Among anticancer agents, microtubule-targeting agents (MTAs) represent a well-recognized therapeutic approach that disrupts microtubule dynamics, thereby inhibiting cell division, and has been widely used to treat several types of cancers. However, even though MTAs are widely regarded as effective, their potential is limited primarily due to cancer resistance and toxicity. Consequently, in the last years, the exploration of new MTAs with the aim of identifying agents with improved cytotoxicity, selectivity, and adequate pharmacokinetic profile, as well as the ability to evade drug resistance mechanisms, has remained a major concern in the development of anticancer treatment. This review highlights the discovery of new MTAs since 2020, with the goal of understanding the advancements made in this field and its future directions. Special attention is given to structure–activity relationship (SAR) studies that could be important for the discovery of more effective MTAs in the future. Full article
(This article belongs to the Special Issue Nature-Inspired Antitumor Agents, 2nd Edition)
Show Figures

Figure 1

27 pages, 1680 KB  
Review
Microtubule-Targeting Agents: Advances in Tubulin Binding and Small Molecule Therapy for Gliomas and Neurodegenerative Diseases
by Maya Ezzo and Sandrine Etienne-Manneville
Int. J. Mol. Sci. 2025, 26(15), 7652; https://doi.org/10.3390/ijms26157652 - 7 Aug 2025
Viewed by 2281
Abstract
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central [...] Read more.
Microtubules play a key role in cell division and cell migration. Thus, microtubule-targeting agents (MTAs) are pivotal in cancer therapy due to their ability to disrupt cell division microtubule dynamics. Traditionally divided into stabilizers and destabilizers, MTAs are increasingly being repurposed for central nervous system (CNS) applications, including brain malignancies such as gliomas and neurodegenerative diseases like Alzheimer’s and Parkinson’s. Microtubule-stabilizing agents, such as taxanes and epothilones, promote microtubule assembly and have shown efficacy in both tumour suppression and neuronal repair, though their CNS use is hindered by blood–brain barrier (BBB) permeability and neurotoxicity. Destabilizing agents, including colchicine-site and vinca domain binders, offer potent anticancer effects but pose greater risks for neuronal toxicity. This review highlights the mapping of nine distinct tubulin binding pockets—including classical (taxane, vinca, colchicine) and emerging (tumabulin, pironetin) sites—that offer new pharmacological entry points. We summarize the recent advances in structural biology and drug design, enabling MTAs to move beyond anti-mitotic roles, unlocking applications in both cancer and neurodegeneration for next-generation MTAs with enhanced specificity and BBB penetration. We further discuss the therapeutic potential of combination strategies, including MTAs with radiation, histone deacetylase (HDAC) inhibitors, or antibody–drug conjugates, that show synergistic effects in glioblastoma models. Furthermore, innovative delivery systems like nanoparticles and liposomes are enhancing CNS drug delivery. Overall, MTAs continue to evolve as multifunctional tools with expanding applications across oncology and neurology, with future therapies focusing on optimizing efficacy, reducing toxicity, and overcoming therapeutic resistance in brain-related diseases. Full article
(This article belongs to the Special Issue New Drugs Regulating Cytoskeletons in Human Health and Diseases)
Show Figures

Figure 1

26 pages, 2011 KB  
Review
Substance Abuse and Cognitive Decline: The Critical Role of Tau Protein as a Potential Biomarker
by Liliana Rebolledo-Pérez, Jorge Hernández-Bello, Alicia Martínez-Ramos, Rolando Castañeda-Arellano, David Fernández-Quezada, Flavio Sandoval-García and Irene Guadalupe Aguilar-García
Int. J. Mol. Sci. 2025, 26(15), 7638; https://doi.org/10.3390/ijms26157638 - 7 Aug 2025
Cited by 1 | Viewed by 3565
Abstract
Tau protein is essential for the structural stability of neurons, particularly through its role in microtubule assembly and axonal transport. However, when abnormally hyperphosphorylated or cleaved, Tau can aggregate into insoluble forms that disrupt neuronal function, contributing to the pathogenesis of neurodegenerative diseases [...] Read more.
Tau protein is essential for the structural stability of neurons, particularly through its role in microtubule assembly and axonal transport. However, when abnormally hyperphosphorylated or cleaved, Tau can aggregate into insoluble forms that disrupt neuronal function, contributing to the pathogenesis of neurodegenerative diseases such as Alzheimer’s disease (AD). Emerging evidence suggests that similar Tau-related alterations may occur in individuals with chronic exposure to psychoactive substances. This review compiles experimental, clinical, and postmortem findings that collectively indicate a substance-specific influence on Tau dynamics. Alcohol and opioids, for instance, promote Tau hyperphosphorylation and fragmentation through the activation of kinases such as GSK-3β and CDK5, as well as proteases like caspase-3, leading to neuroinflammation and microglial activation. Stimulants and dissociatives disrupt insulin signaling, increase oxidative stress, and impair endosomal trafficking, all of which can exacerbate Tau pathology. In contrast, cannabinoids and psychedelics may exert protective effects by modulating kinase activity, reducing inflammation, or enhancing neuroplasticity. Psychedelic compounds such as psilocybin and harmine have been demonstrated to decrease Tau phosphorylation and facilitate cognitive restoration in animal models. Although the molecular mechanisms differ across substances, Tau consistently emerges as a convergent target altered in substance-related cognitive disorders. Understanding these pathways may provide not only mechanistic insights into drug-induced neurotoxicity but also identify Tau as a valuable biomarker and potential therapeutic target for the prevention or treatment of cognitive decline associated with substance use. Full article
(This article belongs to the Special Issue Neurobiological Mechanisms of Addictive Disorders)
Show Figures

Figure 1

15 pages, 1466 KB  
Article
Effect of Tau Fragment and Membrane Interactions on Membrane Permeabilization and Peptide Aggregation
by Majedul Islam, Md Raza Ul Karim, Emily Argueta, Mohammed N. Selim, Ewa P. Wojcikiewicz and Deguo Du
Membranes 2025, 15(7), 208; https://doi.org/10.3390/membranes15070208 - 13 Jul 2025
Viewed by 1690
Abstract
Aggregation of tau protein is a hallmark feature of tauopathies such as Alzheimer’s disease. The microtubule-binding domain of tau plays a crucial role in the tau aggregation process. In this study, we investigated the dual effects of membrane interactions of tau298–317, [...] Read more.
Aggregation of tau protein is a hallmark feature of tauopathies such as Alzheimer’s disease. The microtubule-binding domain of tau plays a crucial role in the tau aggregation process. In this study, we investigated the dual effects of membrane interactions of tau298–317, a fragment peptide from the microtubule-binding domain, on peptide-induced membrane disruption and membrane-mediated peptide self-assembly. Our results show that neither wild-type tau298–317 nor its P301L or Ser305-phosphorylated mutants aggregate in the presence of zwitterionic POPC vesicles or cause lipid vesicle leakage, indicating weak peptide–membrane interactions. In contrast, tau298–317 strongly interacts with negatively charged POPG liposomes, leading to a rapid transition of the peptide conformation from random coils to α-helical intermediate conformation upon membrane adsorption, which may further promote peptide self-association to form oligomers and β-sheet-rich fibrillar structures. Tau298–317-induced rapid POPG membrane leakage indicates a synergistic process of the peptide self-assembly at the membrane interface and the aggregation-induced membrane disruption. Notably, phosphorylation at Ser305 disrupts favorable electrostatic interactions between the peptide and POPG membrane surface, thus preventing peptide aggregation and membrane leakage. In contrast, the P301L mutation significantly enhances membrane-mediated peptide aggregation and peptide-induced membrane disruption, likely due to alleviation of local conformational constraints and enhancement of local hydrophobicity, which facilitates fast conformational conversion to β-sheet structures. These findings provide mechanistic insights into the molecular mechanisms underlying membrane-mediated aggregation of crucial regions of tau and peptide-induced membrane damage, indicating potential strategies to prevent tau aggregation and membrane rupture by targeting critical electrostatic interactions between membranes and key local regions of tau. Full article
(This article belongs to the Section Biological Membranes)
Show Figures

Figure 1

31 pages, 705 KB  
Review
Molecular Guardians of Oocyte Maturation: A Systematic Review on TUBB8, KIF11, and CKAP5 in IVF Outcomes
by Charalampos Voros, Ioakeim Sapantzoglou, Diamantis Athanasiou, Antonia Varthaliti, Despoina Mavrogianni, Kyriakos Bananis, Antonia Athanasiou, Aikaterini Athanasiou, Georgios Papadimas, Athanasios Gkirgkinoudis, Ioannis Papapanagiotou, Kyriaki Migklis, Dimitrios Vaitsis, Aristotelis-Marios Koulakmanidis, Dimitris Mazis Kourakos, Sofia Ivanidou, Maria Anastasia Daskalaki, Marianna Theodora, Panagiotis Antsaklis, Dimitrios Loutradis and Georgios Daskalakisadd Show full author list remove Hide full author list
Int. J. Mol. Sci. 2025, 26(13), 6390; https://doi.org/10.3390/ijms26136390 - 2 Jul 2025
Viewed by 1323
Abstract
The efficacy of in vitro fertilization (IVF) is significantly hindered by early embryonic developmental failure and oocyte maturation arrest. Recent findings in reproductive genetics have identified several oocyte-specific genes—TUBB8, KIF11, and CKAP5—as essential regulators of meiotic spindle formation and [...] Read more.
The efficacy of in vitro fertilization (IVF) is significantly hindered by early embryonic developmental failure and oocyte maturation arrest. Recent findings in reproductive genetics have identified several oocyte-specific genes—TUBB8, KIF11, and CKAP5—as essential regulators of meiotic spindle formation and cytoskeletal dynamics. Mutations in these genes can lead to significant meiotic defects, fertilization failure, and embryo arrest. The links between genotype and phenotype, along with the underlying biological mechanisms, remain inadequately characterized despite the increasing number of identified variations. This systematic review was conducted in accordance with PRISMA 2020 guidelines. Relevant papers were retrieved from the PubMed and Embase databases using combinations of the keywords “TUBB8,” “KIF11,” “CKAP5,” “oocyte maturation arrest,” “embryonic arrest,” and “IVF failure.” Studies were included if they contained clinical, genomic, and functional data on TUBB8, KIF11, or CKAP5 mutations in women undergoing IVF. Molecular data, including gene variant classifications, inheritance models, in vitro tests (such as microtubule network analysis in HeLa cells), and assisted reproductive technology (ART) outcomes, were obtained. Eighteen trials including 35 women with primary infertility were included. Over fifty different variants were identified, the majority of which can be attributed to TUBB8 mutations. TUBB8 disrupted α/β-tubulin heterodimer assembly due to homozygous missense mutations, hence hindering meiotic spindle formation and leading to early embryo fragmentation or the creation of many pronuclei and cleavage failure. KIF11 mutations resulted in spindle disorganization and chromosomal misalignment via disrupting tubulin acetylation and microtubule transport. Mutations in CKAP5 impaired bipolar spindle assembly and microtubule stabilization. In vitro validation studies showed cytoskeletal disturbances, protein instability, and dominant negative effects in transfected animals. Donor egg IVF was the sole effective treatment; however, no viable pregnancies were documented in patients with pathogenic mutations of TUBB8 or KIF11. TUBB8, KIF11, and CKAP5 are essential for safeguarding oocyte meiotic competence and early embryonic development at the molecular level. Genetic differences in these genes disrupt microtubule dynamics and spindle assembly, resulting in various aspects of oocyte maturation and fertilization. Functional validation underscores the necessity of routine genetic screening for women experiencing unresolved IVF failure, as it substantiates their causal role in infertility. Future therapeutic avenues in ART may be enhanced by tailored counseling and innovative rescue methodologies like as gene therapy. Full article
(This article belongs to the Special Issue Molecular Advances in Obstetrical and Gynaecological Disorders)
Show Figures

Figure 1

20 pages, 3202 KB  
Article
Multi-Omic Analysis Identifies Key Genes Driving Testicular Fusion in Spodoptera litura
by Yaqun Dong, Haoyun Luo, Lihua Huang and Lin Liu
Int. J. Mol. Sci. 2025, 26(12), 5564; https://doi.org/10.3390/ijms26125564 - 10 Jun 2025
Viewed by 551
Abstract
The Spodoptera litura, a Lepidopteran pest known for its high fecundity, undergoes a complete metamorphosis, including a distinctive process during which the male testes fuse from two separate organs into a single entity, significantly enhancing its fertility. To elucidate the molecular mechanisms [...] Read more.
The Spodoptera litura, a Lepidopteran pest known for its high fecundity, undergoes a complete metamorphosis, including a distinctive process during which the male testes fuse from two separate organs into a single entity, significantly enhancing its fertility. To elucidate the molecular mechanisms underlying this testicular fusion, this study employed an integrated multi-omics approach to investigate concurrent changes at the transcriptomic and proteomic levels. We identified a series of synchronized alterations on the peritestic larval membrane, including heme binding, peptidase activity, hydrolase activity, metal ion transport, redox reactions, and chitin metabolism, all of which are substantially enriched at specific temporal points during testicular fusion. Nine genes/proteins co-expressed at the mRNA and protein levels were selected for targeted quantitative proteomics (PRM) and quantitative PCR (qPCR) validation, leading to the identification of five genes potentially involved in the testicular fusion process: Sl3030, ARCP, PSLRE, Obstructor-E, and Osris9B. Notably, the gene Sl3030, once knocked out, not only disrupted the normal fusion process but also resulted in reduced testis size, thickened peritestic membranes, and abnormal sperm development. Transcriptomic sequencing of the Sl3030 knockout mutant revealed its primary influence on the fusion process by affecting the assembly of the microtubule system and cytoskeleton. This research, for the first time, provides a multi-omics perspective on the response of key signaling pathways and molecular changes during the testicular fusion of S. litura and validates the role of the previously uncharacterized gene Sl3030 in this process, offering valuable insights into the complex mechanisms of testicular fusion in this species. Full article
(This article belongs to the Special Issue Progress of Molecular Biology and Physiology in Lepidopteran Insects)
Show Figures

Figure 1

28 pages, 831 KB  
Review
Cyanobacterial Peptides in Anticancer Therapy: A Comprehensive Review of Mechanisms, Clinical Advances, and Biotechnological Innovation
by Heayyean Lee, Khuld Nihan and Yale Ryan Kwon
Mar. Drugs 2025, 23(6), 233; https://doi.org/10.3390/md23060233 - 29 May 2025
Cited by 1 | Viewed by 1623
Abstract
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic [...] Read more.
Cyanobacteria-derived peptides represent a promising class of anticancer agents due to their structural diversity and potent bioactivity. They exert cytotoxic effects through mechanisms including microtubule disruption, histone deacetylase inhibition, and apoptosis induction. Several peptides—most notably the dolastatin-derived auristatins—have achieved clinical success as cytotoxic payloads in antibody–drug conjugates (ADCs). However, challenges such as limited tumor selectivity, systemic toxicity, and production scalability remain barriers to broader application. Recent advances in targeted delivery technologies, combination therapy strategies, synthetic biology, and genome mining offer promising solutions. Emerging data from preclinical and clinical studies highlight their therapeutic potential, particularly in treatment-resistant cancers. In this review, we (i) summarize key cyanobacterial peptides and their molecular mechanisms of action, (ii) examine progress toward clinical translation, and (iii) explore biotechnological approaches enabling sustainable production and structural diversification. We also discuss future directions for enhancing specificity and the therapeutic index to fully exploit the potential of these marine-derived peptides in oncology. Full article
(This article belongs to the Special Issue Marine Natural Products as Anticancer Agents, 4th Edition)
Show Figures

Figure 1

Back to TopTop