Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 17.619
1.
Cells ; 13(9)2024 Apr 24.
Article En | MEDLINE | ID: mdl-38727272

Microtubules are an indispensable component of all eukaryotic cells due to their role in mitotic spindle formation, yet their organization and number can vary greatly in the interphase. The last common ancestor of all eukaryotes already had microtubules and microtubule motor proteins moving along them. Sponges are traditionally regarded as the oldest animal phylum. Their body does not have a clear differentiation into tissues, but it contains several distinguishable cell types. The choanocytes stand out among them and are responsible for creating a flow of water with their flagella and increasing the filtering and feeding efficiency of the sponge. Choanocyte flagella contain microtubules, but thus far, observing a developed system of cytoplasmic microtubules in non-flagellated interphase sponge cells has been mostly unsuccessful. In this work, we combine transcriptomic analysis, immunofluorescence, and electron microscopy with time-lapse recording to demonstrate that microtubules appear in the cytoplasm of sponge cells only when transdifferentiation processes are activated. We conclude that dynamic cytoplasmic microtubules in the cells of sponges are not a persistent but rather a transient structure, associated with cellular plasticity.


Cell Differentiation , Interphase , Microtubules , Porifera , Microtubules/metabolism , Animals , Porifera/cytology
2.
FASEB J ; 38(10): e23661, 2024 May 31.
Article En | MEDLINE | ID: mdl-38733310

Itching is an aversive somatosensation that triggers the desire to scratch. Transient receptor potential (TRP) channel proteins are key players in acute and chronic itch. However, whether the modulatory effect of fibroblast growth factor 13 (FGF13) on acute and chronic itch is associated with TRP channel proteins is unclear. Here, we demonstrated that conditional knockout of Fgf13 in dorsal root ganglion neurons induced significant impairment in scratching behaviors in response to acute histamine-dependent and chronic dry skin itch models. Furthermore, FGF13 selectively regulated the function of the TRPV1, but not the TRPA1 channel on Ca2+ imaging and electrophysiological recordings, as demonstrated by a significant reduction in neuronal excitability and current density induced by TRPV1 channel activation, whereas TRPA1 channel activation had no effect. Changes in channel currents were also verified in HEK cell lines. Subsequently, we observed that selective modulation of TRPV1 by FGF13 required its microtubule-stabilizing effect. Furthermore, in FGF13 knockout mice, only the overexpression of FGF13 with a tubulin-binding domain could rescue TRP channel function and the impaired itch behavior. Our findings reveal a novel mechanism by which FGF13 is involved in TRPV1-dependent itch transduction and provide valuable clues for alleviating pathological itch syndrome.


Fibroblast Growth Factors , Mice, Knockout , Microtubules , Pruritus , TRPV Cation Channels , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Pruritus/metabolism , Pruritus/genetics , Animals , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Mice , Humans , HEK293 Cells , Microtubules/metabolism , Ganglia, Spinal/metabolism , Male , Mice, Inbred C57BL , TRPA1 Cation Channel/metabolism , TRPA1 Cation Channel/genetics
3.
Nat Commun ; 15(1): 3793, 2024 May 07.
Article En | MEDLINE | ID: mdl-38714822

Across the cell cycle, mitochondrial dynamics are regulated by a cycling wave of actin polymerization/depolymerization. In metaphase, this wave induces actin comet tails on mitochondria that propel these organelles to drive spatial mixing, resulting in their equitable inheritance by daughter cells. In contrast, during interphase the cycling actin wave promotes localized mitochondrial fission. Here, we identify the F-actin nucleator/elongator FMNL1 as a positive regulator of the wave. FMNL1-depleted cells exhibit decreased mitochondrial polarization, decreased mitochondrial oxygen consumption, and increased production of reactive oxygen species. Accompanying these changes is a loss of hetero-fusion of wave-fragmented mitochondria. Thus, we propose that the interphase actin wave maintains mitochondrial homeostasis by promoting mitochondrial content mixing. Finally, we investigate the mechanistic basis for the observation that the wave drives mitochondrial motility in metaphase but mitochondrial fission in interphase. Our data indicate that when the force of actin polymerization is resisted by mitochondrial tethering to microtubules, as in interphase, fission results.


Actins , Homeostasis , Interphase , Mitochondria , Mitochondrial Dynamics , Actins/metabolism , Mitochondria/metabolism , Humans , Formins/metabolism , Reactive Oxygen Species/metabolism , HeLa Cells , Microtubules/metabolism , Mitochondrial Proteins/metabolism , Mitochondrial Proteins/genetics , Animals
4.
J Cell Biol ; 223(8)2024 Aug 05.
Article En | MEDLINE | ID: mdl-38743010

Basal bodies (BBs) are conserved eukaryotic structures that organize cilia. They are comprised of nine, cylindrically arranged, triplet microtubules (TMTs) connected to each other by inter-TMT linkages which stabilize the structure. Poc1 is a conserved protein important for BB structural integrity in the face of ciliary forces transmitted to BBs. To understand how Poc1 confers BB stability, we identified the precise position of Poc1 in the Tetrahymena BB and the effect of Poc1 loss on BB structure. Poc1 binds at the TMT inner junctions, stabilizing TMTs directly. From this location, Poc1 also stabilizes inter-TMT linkages throughout the BB, including the cartwheel pinhead and the inner scaffold. The full localization of the inner scaffold protein Fam161A requires Poc1. As ciliary forces are increased, Fam161A is reduced, indicative of a force-dependent molecular remodeling of the inner scaffold. Thus, while not essential for BB assembly, Poc1 promotes BB interconnections that establish an architecture competent to resist ciliary forces.


Basal Bodies , Cilia , Microtubules , Protozoan Proteins , Microtubules/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Cilia/metabolism , Basal Bodies/metabolism , Tetrahymena thermophila/metabolism , Tetrahymena thermophila/genetics , Tetrahymena/metabolism , Tetrahymena/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Protein Binding
5.
J Cell Biol ; 223(7)2024 Jul 01.
Article En | MEDLINE | ID: mdl-38722279

In addition to its well-established role in actin assembly, profilin 1 (PFN1) has been shown to bind to tubulin and alter microtubule growth. However, whether PFN1's predominant control over microtubules in cells occurs through direct regulation of tubulin or indirectly through the polymerization of actin has yet to be determined. Here, we manipulated PFN1 expression, actin filament assembly, and actomyosin contractility and showed that reducing any of these parameters for extended periods of time caused an adaptive response in the microtubule cytoskeleton, with the effect being significantly more pronounced in neuronal processes. All the observed changes to microtubules were reversible if actomyosin was restored, arguing that PFN1's regulation of microtubules occurs principally through actin. Moreover, the cytoskeletal modifications resulting from PFN1 depletion in neuronal processes affected microtubule-based transport and mimicked phenotypes that are linked to neurodegenerative disease. This demonstrates how defects in actin can cause compensatory responses in other cytoskeleton components, which in turn significantly alter cellular function.


Actins , Microtubules , Profilins , Animals , Humans , Mice , Actin Cytoskeleton/metabolism , Actins/metabolism , Actins/genetics , Actomyosin/metabolism , Microtubules/metabolism , Neurons/metabolism , Profilins/metabolism , Profilins/genetics , Tubulin/metabolism , Tubulin/genetics
6.
Proc Natl Acad Sci U S A ; 121(20): e2316266121, 2024 May 14.
Article En | MEDLINE | ID: mdl-38709923

Neurons regulate the microtubule-based transport of certain vesicles selectively into axons or dendrites to ensure proper polarization of function. The mechanism of this polarized vesicle transport is still not fully elucidated, though it is known to involve kinesins, which drive anterograde transport on microtubules. Here, we explore how the kinesin-3 family member KIF13A is regulated such that vesicles containing transferrin receptor (TfR) travel only to dendrites. In experiments involving live-cell imaging, knockout of KIF13A, BioID assay, we found that the kinase MARK2 phosphorylates KIF13A at a 14-3-3 binding motif, strengthening interaction of KIF13A with 14-3-3 such that it dissociates from TfR-containing vesicles, which therefore cannot enter axons. Overexpression of KIF13A or knockout of MARK2 leads to axonal transport of TfR-containing vesicles. These results suggest a unique kinesin-based mechanism for polarized transport of vesicles to dendrites.


14-3-3 Proteins , Dendrites , Kinesins , Protein Serine-Threonine Kinases , Receptors, Transferrin , Kinesins/metabolism , Kinesins/genetics , 14-3-3 Proteins/metabolism , Dendrites/metabolism , Phosphorylation , Receptors, Transferrin/metabolism , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Humans , Binding Sites , Microtubules/metabolism , Rats , Mice , Protein Binding
7.
Sci Rep ; 14(1): 10276, 2024 05 04.
Article En | MEDLINE | ID: mdl-38704483

Fuchs endothelial corneal dystrophy (FECD) is a complex corneal disease characterized by the progressive decline and morphological changes of corneal endothelial cells (CECs) that leads to corneal edema and vision loss. The most common mutation in FECD is an intronic CTG repeat expansion in transcription factor 4 (TCF4) that leads to its altered expression. Corneal endothelial wound healing occurs primarily through cell enlargement and migration, and FECD CECs have been shown to display increased migration speeds. In this study, we aim to determine whether TCF4 can promote cellular migration in FECD CECs. We generated stable CEC lines derived from FECD patients that overexpressed different TCF4 isoforms and investigated epithelial-to-mesenchymal (EMT) expression, morphological analysis and cellular migration speeds. We found that full length TCF4-B isoform overexpression promotes cellular migration in FECD CECs in an EMT-independent manner. RNA-sequencing identified several pathways including the negative regulation of microtubules, with TUBB4A (tubulin beta 4A class IVa) as the top upregulated gene. TUBB4A expression was increased in FECD ex vivo specimens, and there was altered expression of cytoskeleton proteins, tubulin and actin, compared to normal healthy donor ex vivo specimens. Additionally, there was increased acetylation and detyrosination of microtubules in FECD supporting that microtubule stability is altered in FECD and could promote cellular migration. Future studies could be aimed at investigating if targeting the cytoskeleton and microtubules would have therapeutic potential for FECD by promoting cellular migration and regeneration.


Cell Movement , Endothelium, Corneal , Fuchs' Endothelial Dystrophy , Microtubules , Transcription Factor 4 , Humans , Fuchs' Endothelial Dystrophy/genetics , Fuchs' Endothelial Dystrophy/metabolism , Fuchs' Endothelial Dystrophy/pathology , Cell Movement/genetics , Microtubules/metabolism , Transcription Factor 4/metabolism , Transcription Factor 4/genetics , Endothelium, Corneal/metabolism , Endothelium, Corneal/pathology , Male , Female , Epithelial-Mesenchymal Transition/genetics , Aged , Endothelial Cells/metabolism , Endothelial Cells/pathology , Tubulin/metabolism , Tubulin/genetics , Middle Aged , Protein Isoforms/metabolism , Protein Isoforms/genetics
8.
Nat Commun ; 15(1): 3779, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710684

The α-Aurora kinase is a crucial regulator of spindle microtubule organization during mitosis in plants. Here, we report a post-mitotic role for α-Aurora in reorganizing the phragmoplast microtubule array. In Arabidopsis thaliana, α-Aurora relocated from spindle poles to the phragmoplast midzone, where it interacted with the microtubule cross-linker MAP65-3. In a hypomorphic α-Aurora mutant, MAP65-3 was detected on spindle microtubules, followed by a diffuse association pattern across the phragmoplast midzone. Simultaneously, phragmoplast microtubules remained belatedly in a solid disk array before transitioning to a ring shape. Microtubules at the leading edge of the matured phragmoplast were often disengaged, accompanied by conspicuous retentions of MAP65-3 at the phragmoplast interior edge. Specifically, α-Aurora phosphorylated two residues towards the C-terminus of MAP65-3. Mutation of these residues to alanines resulted in an increased association of MAP65-3 with microtubules within the phragmoplast. Consequently, the expansion of the phragmoplast was notably slower compared to wild-type cells or cells expressing a phospho-mimetic variant of MAP65-3. Moreover, mimicking phosphorylation reinstated disrupted MAP65-3 behaviors in plants with compromised α-Aurora function. Overall, our findings reveal a mechanism in which α-Aurora facilitates cytokinesis progression through phosphorylation-dependent restriction of MAP65-3 associating with microtubules at the phragmoplast midzone.


Arabidopsis Proteins , Arabidopsis , Cytokinesis , Microtubule-Associated Proteins , Microtubules , Arabidopsis/metabolism , Arabidopsis/genetics , Microtubules/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Phosphorylation , Mutation , Spindle Apparatus/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Plants, Genetically Modified , Mitosis
9.
Nat Commun ; 15(1): 3992, 2024 May 11.
Article En | MEDLINE | ID: mdl-38734767

Visual proteomics attempts to build atlases of the molecular content of cells but the automated annotation of cryo electron tomograms remains challenging. Template matching (TM) and methods based on machine learning detect structural signatures of macromolecules. However, their applicability remains limited in terms of both the abundance and size of the molecular targets. Here we show that the performance of TM is greatly improved by using template-specific search parameter optimization and by including higher-resolution information. We establish a TM pipeline with systematically tuned parameters for the automated, objective and comprehensive identification of structures with confidence 10 to 100-fold above the noise level. We demonstrate high-fidelity and high-confidence localizations of nuclear pore complexes, vaults, ribosomes, proteasomes, fatty acid synthases, lipid membranes and microtubules, and individual subunits inside crowded eukaryotic cells. We provide software tools for the generic implementation of our method that is broadly applicable towards realizing visual proteomics.


Cryoelectron Microscopy , Electron Microscope Tomography , Proteasome Endopeptidase Complex , Proteomics , Ribosomes , Software , Electron Microscope Tomography/methods , Cryoelectron Microscopy/methods , Ribosomes/ultrastructure , Ribosomes/metabolism , Proteasome Endopeptidase Complex/ultrastructure , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/chemistry , Humans , Proteomics/methods , Nuclear Pore/ultrastructure , Nuclear Pore/metabolism , Microtubules/ultrastructure , Microtubules/metabolism , Fatty Acid Synthases/metabolism , Machine Learning , Imaging, Three-Dimensional/methods , Algorithms , Image Processing, Computer-Assisted/methods
10.
J Transl Med ; 22(1): 441, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730481

Microtubule targeting agents (MTAs) are commonly prescribed to treat cancers and predominantly kill cancer cells in mitosis. Significantly, some MTA-treated cancer cells escape death in mitosis, exit mitosis and become malignant polyploid giant cancer cells (PGCC). Considering the low number of cancer cells undergoing mitosis in tumor tissues, killing them in interphase may represent a favored antitumor approach. We discovered that ST-401, a mild inhibitor of microtubule (MT) assembly, preferentially kills cancer cells in interphase as opposed to mitosis, a cell death mechanism that avoids the development of PGCC. Single cell RNA sequencing identified mRNA transcripts regulated by ST-401, including mRNAs involved in ribosome and mitochondrial functions. Accordingly, ST-401 induces a transient integrated stress response, reduces energy metabolism, and promotes mitochondria fission. This cell response may underly death in interphase and avoid the development of PGCC. Considering that ST-401 is a brain-penetrant MTA, we validated these results in glioblastoma cell lines and found that ST-401 also reduces energy metabolism and promotes mitochondria fission in GBM sensitive lines. Thus, brain-penetrant mild inhibitors of MT assembly, such as ST-401, that induce death in interphase through a previously unanticipated antitumor mechanism represent a potentially transformative new class of therapeutics for the treatment of GBM.


Cell Death , Giant Cells , Interphase , Microtubules , Polyploidy , Humans , Interphase/drug effects , Microtubules/metabolism , Microtubules/drug effects , Cell Line, Tumor , Cell Death/drug effects , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/pathology , Mitochondrial Dynamics/drug effects , Energy Metabolism/drug effects , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/genetics , Neoplasms/pathology , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Gene Expression Regulation, Neoplastic/drug effects
11.
J Cell Sci ; 137(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38587100

During development, neurons achieve a stereotyped neuron type-specific morphology, which relies on dynamic support by microtubules (MTs). An important player is the augmin complex (hereafter augmin), which binds to existing MT filaments and recruits the γ-tubulin ring complex (γ-TuRC), to form branched MTs. In cultured neurons, augmin is important for neurite formation. However, little is known about the role of augmin during neurite formation in vivo. Here, we have revisited the role of mammalian augmin in culture and then turned towards the class four Drosophila dendritic arborization (c4da) neurons. We show that MT density is maintained through augmin in cooperation with the γ-TuRC in vivo. Mutant c4da neurons show a reduction of newly emerging higher-order dendritic branches and in turn also a reduced number of their characteristic space-filling higher-order branchlets. Taken together, our data reveal a cooperative function for augmin with the γ-TuRC in forming enough MTs needed for the appropriate differentiation of morphologically complex dendrites in vivo.


Dendrites , Drosophila Proteins , Microtubule-Associated Proteins , Microtubules , Animals , Microtubules/metabolism , Dendrites/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Drosophila melanogaster/metabolism , Tubulin/metabolism , Drosophila/metabolism , Humans , Neurons/metabolism , Neurons/cytology
12.
J Cell Sci ; 137(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38606636

Microtubules are nucleated by γ-tubulin ring complexes (γ-TuRCs) and are essential for neuronal development. Nevertheless, γ-TuRC depletion has been reported to perturb only higher-order branching in elaborated Drosophila larval class IV dendritic arborization (da) neurons. This relatively mild phenotype has been attributed to defects in microtubule nucleation from Golgi outposts, yet most Golgi outposts lack associated γ-TuRCs. By analyzing dendritic arbor regrowth in pupae, we show that γ-TuRCs are also required for the growth and branching of primary and secondary dendrites, as well as for higher-order branching. Moreover, we identify the augmin complex (hereafter augmin), which recruits γ-TuRCs to the sides of pre-existing microtubules, as being required predominantly for higher-order branching. Augmin strongly promotes the anterograde growth of microtubules in terminal dendrites and thus terminal dendrite stability. Consistent with a specific role in higher-order branching, we find that augmin is expressed less strongly and is largely dispensable in larval class I da neurons, which exhibit few higher-order dendrites. Thus, γ-TuRCs are essential for various aspects of complex dendritic arbor development, and they appear to function in higher-order branching via the augmin pathway, which promotes the elaboration of dendritic arbors to help define neuronal morphology.


Dendrites , Drosophila Proteins , Microtubules , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Dendrites/metabolism , Microtubules/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/genetics , Tubulin/metabolism , Larva/metabolism , Larva/growth & development , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Drosophila/metabolism
13.
Cell Mol Life Sci ; 81(1): 193, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38652325

The acetylation of α-tubulin on lysine 40 is a well-studied post-translational modification which has been associated with the presence of long-lived stable microtubules that are more resistant to mechanical breakdown. The discovery of α-tubulin acetyltransferase 1 (ATAT1), the enzyme responsible for lysine 40 acetylation on α-tubulin in a wide range of species, including protists, nematodes, and mammals, dates to about a decade ago. However, the role of ATAT1 in different cellular activities and molecular pathways has been only recently disclosed. This review comprehensively summarizes the most recent knowledge on ATAT1 structure and substrate binding and analyses the involvement of ATAT1 in a variety of cellular processes such as cell motility, mitosis, cytoskeletal organization, and intracellular trafficking. Finally, the review highlights ATAT1 emerging roles in human diseases and discusses ATAT1 potential enzymatic and non-enzymatic roles and the current efforts in developing ATAT1 inhibitors.


Acetyltransferases , Microtubule Proteins , Tubulin , Humans , Acetyltransferases/metabolism , Acetyltransferases/chemistry , Tubulin/metabolism , Tubulin/chemistry , Animals , Protein Processing, Post-Translational , Acetylation , Microtubules/metabolism , Mitosis , Cell Movement , Neoplasms/pathology , Neoplasms/enzymology , Neoplasms/metabolism , Cytoskeleton/metabolism
14.
Mol Biol Cell ; 35(5): ar72, 2024 May 01.
Article En | MEDLINE | ID: mdl-38568782

Cilia generate three-dimensional waveforms required for cell motility and transport of fluid, mucus, and particles over the cell surface. This movement is driven by multiple dynein motors attached to nine outer doublet microtubules that form the axoneme. The outer and inner arm dyneins are organized into 96-nm repeats tandemly arrayed along the length of the doublets. Motility is regulated in part by projections from the two central pair microtubules that contact radial spokes located near the base of the inner dynein arms in each repeat. Although much is known about the structures and protein complexes within the axoneme, many questions remain about the regulatory mechanisms that allow the cilia to modify their waveforms in response to internal or external stimuli. Here, we used Chlamydomonas mbo (move backwards only) mutants with altered waveforms to identify at least two conserved proteins, MBO2/CCDC146 and FAP58/CCDC147, that form part of a L-shaped structure that varies between doublet microtubules. Comparative proteomics identified additional missing proteins that are altered in other motility mutants, revealing overlapping protein defects. Cryo-electron tomography and epitope tagging revealed that the L-shaped, MBO2/FAP58 structure interconnects inner dynein arms with multiple regulatory complexes, consistent with its function in modifying the ciliary waveform.


Axoneme , Dyneins , Axoneme/metabolism , Dyneins/metabolism , Microtubules/metabolism , Cilia/metabolism , Proteins/metabolism , Flagella/metabolism
15.
Nanoscale ; 16(16): 7976-7987, 2024 Apr 25.
Article En | MEDLINE | ID: mdl-38567463

Collective functionalization of the phytochemicals of medicinal herbs on nanoparticles is emerging as a potential cancer therapeutic strategy. This study presents the facile synthesis of surface-functionalized gold nanoparticles using Bacopa monnieri (Brahmi; Bm) phytochemicals and their therapeutically relevant mechanism of action in the colorectal cancer cell line, HT29. The nanoparticles were characterized using UV-visible spectroscopy, TEM-EDAX, zeta potential analysis, TGA, FTIR and 1H NMR spectroscopy, and HR-LC-MS. The particles (Bm-GNPs) were of polygonal shape and were stable against aggregation. They entered the target cells and inhibited the viability and clonogenicity of the cells with eight times more antiproliferative efficacy (25 ± 1.5 µg mL-1) than Bm extract (Bm-EX). In vitro studies revealed that Bm-GNPs bind tubulin (a protein crucial in cell division and a target of anticancer drugs) and disrupt its helical structure without grossly altering its tertiary conformation. Like other antitubulin agents, Bm-GNPs induced G2/M arrest and ultimately killed the cells, as confirmed using flow cytometry analyses. ZVAD-FMK-mediated global pan-caspase inhibition and the apparent absence of cleaved caspase-3 in treated cells indicated that the death did not involve the classic apoptosis pathway. Cellular ultrastructure analyses, western immunoblots, and in situ immunofluorescence visualization of cellular microtubules revealed microtubule-acetylation-independent induction of autophagy as the facilitator of cell death. Together, the data indicate strong antiproliferative efficacy and a possible mechanism of action for these designer nanoparticles. Bm-GNPs, therefore, merit further investigations, including preclinical evaluations, for their therapeutic potential as inducers of non-apoptotic cell death.


Autophagy , Colorectal Neoplasms , Gold , Metal Nanoparticles , Humans , Gold/chemistry , Gold/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Metal Nanoparticles/chemistry , Autophagy/drug effects , Acetylation , Microtubules/metabolism , Microtubules/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/drug therapy , HT29 Cells , Caspases/metabolism , Phytochemicals/pharmacology , Phytochemicals/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Tubulin/metabolism , Tubulin/chemistry
16.
Cells ; 13(7)2024 Apr 07.
Article En | MEDLINE | ID: mdl-38607086

Miro GTPases are key components in the machinery responsible for transporting mitochondria and peroxisomes along microtubules, and also play important roles in regulating calcium homeostasis and organizing contact sites between mitochondria and the endoplasmic reticulum. Moreover, Miro GTPases have been shown to interact with proteins that actively regulate cytoskeletal organization and dynamics, suggesting that these GTPases participate in organizing cytoskeletal functions and organelle transport. Derailed mitochondrial transport is associated with neuropathological conditions such as Parkinson's and Alzheimer's diseases. This review explores our recent understanding of the diverse roles of Miro GTPases under cytoskeletal control, both under normal conditions and during the course of human diseases such as neuropathological disorders.


GTP Phosphohydrolases , Mitochondria , Humans , GTP Phosphohydrolases/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Biological Transport , Microtubules/metabolism
17.
Reprod Biol Endocrinol ; 22(1): 36, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38570783

Microtubule-associated protein 1a (Map1a) is a microtubule (MT) regulatory protein that binds to the MT protofilaments in mammalian cells to promote MT stabilization. Maps work with MT cleavage proteins and other MT catastrophe-inducing proteins to confer MT dynamics to support changes in the Sertoli cell shape to sustain spermatogenesis. However, no functional studies are found in the literature to probe its role in spermatogenesis. Using an RNAi approach, coupled with the use of toxicant-induced testis (in vivo)- and Sertoli cell (in vitro)-injury models, RNA-Seq analysis, transcriptome profiling, and relevant bioinformatics analysis, immunofluorescence analysis, and pertinent biochemical assays for cytoskeletal organization, we have delineated the functional role of Map1a in Sertoli cells and testes. Map1a was shown to support MT structural organization, and its knockdown (KD) also perturbed the structural organization of actin, vimentin, and septin cytoskeletons as these cytoskeletons are intimately related, working in concert to support spermatogenesis. More importantly, cadmium-induced Sertoli cell injury that perturbed the MT structural organization across the cell cytoplasm was associated with disruptive changes in the distribution of Map1a and a surge in p-p38-MAPK (phosphorylated p38-mitogen-activated protein kinase) expression but not total p38-MAPK. These findings thus support the notion that p-p38-MAPK activation is involved in cadmium-induced Sertoli cell injury. This conclusion was supported by studies using doramapimod, a specific p38-MAPK phosphorylation (activation) inhibitor, which was capable of restoring the cadmium-induced disruptive structural organization of MTs across the Sertoli cell cytoplasm. In summary: this study provides mechanistic insights regarding restoration of toxicant-induced Sertoli cell and testis injury and male infertility.


Actins , Sertoli Cells , Rats , Animals , Male , Actins/metabolism , Sertoli Cells/metabolism , Cadmium , Rats, Sprague-Dawley , Blood-Testis Barrier/metabolism , Microtubules/metabolism , Testis/metabolism , Spermatogenesis/physiology , Mammals
18.
Chem Biol Drug Des ; 103(4): e14513, 2024 Apr.
Article En | MEDLINE | ID: mdl-38570322

Taxol (paclitaxel) is the first approved microtubule-stabilizing agent (MSA) by binding stoichiometrically to tubulin, which is considered to be one of the most significant advances in first-line chemotherapy against diverse tumors. However, a large number of residue missence mutations harboring in the tubulin have been observed to cause acquired drug resistance, largely limiting the clinical application of Taxol and its analogs in chemotherapy. A systematic investigation of the intermolecular interactions between the Taxol and various tubulin mutants would help to establish a comprehensive picture of drug response to tubulin mutations in clinical treatment of cancer, and to design new MSA agents with high potency and selectivity to overcome drug resistance. In this study, we described an integration of in silico analysis and in vitro assay (iSiV) to profile Taxol against a panel of 149 clinically observed, cancer-associated missence mutations in ß-tubulin at molecular and cellular levels, aiming to a systematic understanding of molecular mechanism and biological implication underlying drug resistance and sensitivity conferring from tubulin mutations. It is revealed that the Taxol-resistant mutations can be classified into three types: (I) nonbonded interaction broken due to mutation, (II) steric hindrance caused by mutation, and (III) conformational change upon mutation. In addition, we identified three new Taxol-resistant mutations (C239Y, T274I, and R320P) that can largely reduce the binding affinity of Taxol to tubulin at molecular level, in which the T274I and R320P were observed to considerably impair the antitumor activity of Taxol at cellular level. Moreover, a novel drug-susceptible mutation (M363T) was also identified, which improves Taxol affinity by 2.6-fold and decreases Taxol antitumor EC50 values from 29.4 to 18.7 µM.


Paclitaxel , Tubulin , Paclitaxel/pharmacology , Tubulin/metabolism , Microtubules/metabolism , Mutation , Drug Resistance
19.
Molecules ; 29(8)2024 Apr 15.
Article En | MEDLINE | ID: mdl-38675612

Kinesin-14s, a subfamily of the large superfamily of kinesin motor proteins, function mainly in spindle assembly and maintenance during mitosis and meiosis. KlpA from Aspergillus nidulans and GiKIN14a from Giardia intestinalis are two types of kinesin-14s. Available experimental results puzzlingly showed that while KlpA moves preferentially toward the minus end in microtubule-gliding setups and inside parallel microtubule overlaps, it moves preferentially toward the plus end on single microtubules. More puzzlingly, the insertion of an extra polypeptide linker in the central region of the neck stalk switches the motility direction of KlpA on single microtubules to the minus end. Prior experimental results showed that GiKIN14a moves preferentially toward the minus end on single microtubules in either tailless or full-length forms. The tail not only greatly enhances the processivity but also accelerates the ATPase rate and velocity of GiKIN14a. The insertion of an extra polypeptide linker in the central region of the neck stalk reduces the ATPase rate of GiKIN14a. However, the underlying mechanism of these puzzling dynamical features for KlpA and GiKIN14a is unclear. Here, to understand this mechanism, the dynamics of KlpA and GiKIN14a were studied theoretically on the basis of the proposed model, incorporating potential changes between the kinesin head and microtubule, as well as the potential between the tail and microtubule. The theoretical results quantitatively explain the available experimental results and provide predicted results. It was found that the elasticity of the neck stalk determines the directionality of KlpA on single microtubules and affects the ATPase rate and velocity of GiKIN14a on single microtubules.


Kinesins , Microtubules , Kinesins/metabolism , Kinesins/chemistry , Microtubules/metabolism , Models, Molecular , Aspergillus nidulans/metabolism
20.
Cell Rep ; 43(4): 114050, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38564336

Seo et al.1 shed light on virus-host interactions as they reveal how poxvirus A51R stabilizes microtubules in infected cells, which impacts vaccinia virus virulence in mice by potentially inhibiting reactive-oxygen-species-dependent antiviral responses in macrophages.


Microtubules , Vaccinia virus , Microtubules/metabolism , Animals , Virulence , Vaccinia virus/pathogenicity , Vaccinia virus/physiology , Humans , Mice , Macrophages/virology , Macrophages/metabolism , Poxviridae/pathogenicity , Poxviridae/genetics , Poxviridae/physiology
...