Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.482
Filter
1.
Elife ; 132024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949652

ABSTRACT

Tubulin posttranslational modifications (PTMs) modulate the dynamic properties of microtubules and their interactions with other proteins. However, the effects of tubulin PTMs were often revealed indirectly through the deletion of modifying enzymes or the overexpression of tubulin mutants. In this study, we directly edited the endogenous tubulin loci to install PTM-mimicking or -disabling mutations and studied their effects on microtubule stability, neurite outgrowth, axonal regeneration, cargo transport, and sensory functions in the touch receptor neurons of Caenorhabditis elegans. We found that the status of ß-tubulin S172 phosphorylation and K252 acetylation strongly affected microtubule dynamics, neurite growth, and regeneration, whereas α-tubulin K40 acetylation had little influence. Polyglutamylation and detyrosination in the tubulin C-terminal tail had more subtle effects on microtubule stability likely by modulating the interaction with kinesin-13. Overall, our study systematically assessed and compared several tubulin PTMs for their impacts on neuronal differentiation and regeneration and established an in vivo platform to test the function of tubulin PTMs in neurons.


Subject(s)
Caenorhabditis elegans , Microtubules , Protein Processing, Post-Translational , Tubulin , Animals , Tubulin/metabolism , Tubulin/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Microtubules/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Acetylation , Axons/metabolism , Axons/physiology , Phosphorylation , Nerve Regeneration , Kinesins/metabolism , Kinesins/genetics
2.
Proc Natl Acad Sci U S A ; 121(28): e2403034121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38954547

ABSTRACT

Nanomaterials acquire a biomolecular corona upon introduction to biological media, leading to biological transformations such as changes in protein function, unmasking of epitopes, and protein fibrilization. Ex vivo studies to investigate the effect of nanoparticles on protein-protein interactions are typically performed in buffer and are rarely measured quantitatively in live cells. Here, we measure the differential effect of silica nanoparticles on protein association in vitro vs. in mammalian cells. BtubA and BtubB are a pair of bacterial tubulin proteins identified in Prosthecobacter strains that self-assemble like eukaryotic tubulin, first into dimers and then into microtubules in vitro or in vivo. Förster resonance energy transfer labeling of each of the Btub monomers with a donor (mEGFP) and acceptor (mRuby3) fluorescent protein provides a quantitative tool to measure their binding interactions in the presence of unfunctionalized silica nanoparticles in buffer and in cells using fluorescence spectroscopy and microscopy. We show that silica nanoparticles enhance BtubAB dimerization in buffer due to protein corona formation. However, these nanoparticles have little effect on bacterial tubulin self-assembly in the complex mammalian cellular environment. Thus, the effect of nanomaterials on protein-protein interactions may not be readily translated from the test tube to the cell in the absence of particle surface functionalization that can enable targeted protein-nanoparticle interactions to withstand competitive binding in the nanoparticle corona from other biomolecules.


Subject(s)
Bacterial Proteins , Nanoparticles , Silicon Dioxide , Tubulin , Tubulin/metabolism , Tubulin/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Silicon Dioxide/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Fluorescence Resonance Energy Transfer , Humans , Microtubules/metabolism , Protein Multimerization , Protein Binding
3.
Nat Commun ; 15(1): 5530, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956021

ABSTRACT

Mutations in the microtubule-associated motor protein KIF1A lead to severe neurological conditions known as KIF1A-associated neurological disorders (KAND). Despite insights into its molecular mechanism, high-resolution structures of KIF1A-microtubule complexes remain undefined. Here, we present 2.7-3.5 Å resolution structures of dimeric microtubule-bound KIF1A, including the pathogenic P305L mutant, across various nucleotide states. Our structures reveal that KIF1A binds microtubules in one- and two-heads-bound configurations, with both heads exhibiting distinct conformations with tight inter-head connection. Notably, KIF1A's class-specific loop 12 (K-loop) forms electrostatic interactions with the C-terminal tails of both α- and ß-tubulin. The P305L mutation does not disrupt these interactions but alters loop-12's conformation, impairing strong microtubule-binding. Structure-function analysis reveals the K-loop and head-head coordination as major determinants of KIF1A's superprocessive motility. Our findings advance the understanding of KIF1A's molecular mechanism and provide a basis for developing structure-guided therapeutics against KAND.


Subject(s)
Cryoelectron Microscopy , Kinesins , Microtubules , Tubulin , Kinesins/metabolism , Kinesins/genetics , Kinesins/chemistry , Microtubules/metabolism , Humans , Tubulin/metabolism , Tubulin/chemistry , Tubulin/genetics , Protein Binding , Mutation , Models, Molecular , Protein Conformation
4.
Cell Mol Biol Lett ; 29(1): 94, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956497

ABSTRACT

BACKGROUND: We have previously identified an unsuspected role for GJB3 showing that the deficiency of this connexin protein induces aneuploidy in human and murine cells and accelerates cell transformation as well as tumor formation in xenograft models. The molecular mechanisms by which loss of GJB3 leads to aneuploidy and cancer initiation and progression remain unsolved. METHODS: GJB3 expression levels were determined by RT-qPCR and Western blot. The consequences of GJB3 knockdown on genome instability were assessed by metaphase chromosome counting, multinucleation of cells, by micronuclei formation and by the determination of spindle orientation. Interactions of GJB3 with α-tubulin and F-actin was analyzed by immunoprecipitation and immunocytochemistry. Consequences of GJB3 deficiency on microtubule and actin dynamics were measured by live cell imaging and fluorescence recovery after photobleaching experiments, respectively. Immunohistochemistry was used to determine GJB3 levels on human and murine bladder cancer tissue sections. Bladder cancer in mice was chemically induced by BBN-treatment. RESULTS: We find that GJB3 is highly expressed in the ureter and bladder epithelium, but it is downregulated in invasive bladder cancer cell lines and during tumor progression in both human and mouse bladder cancer. Downregulation of GJB3 expression leads to aneuploidy and genomic instability in karyotypically stable urothelial cells and experimental modulation of GJB3 levels alters the migration and invasive capacity of bladder cancer cell lines. Importantly, GJB3 interacts both with α-tubulin and F-actin. The impairment of these interactions alters the dynamics of these cytoskeletal components and leads to defective spindle orientation. CONCLUSION: We conclude that deregulated microtubule and actin dynamics have an impact on proper chromosome separation and tumor cell invasion and migration. Consequently, these observations indicate a possible role for GJB3 in the onset and spreading of bladder cancer and demonstrate a molecular link between enhanced aneuploidy and invasive capacity cancer cells during tumor cell dissemination.


Subject(s)
Actins , Aneuploidy , Neoplasm Invasiveness , Tubulin , Urinary Bladder Neoplasms , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , Humans , Animals , Tubulin/metabolism , Tubulin/genetics , Cell Line, Tumor , Mice , Actins/metabolism , Actins/genetics , Urothelium/pathology , Urothelium/metabolism , Cell Movement/genetics , Microtubules/metabolism , Genomic Instability , Protein Binding
5.
Life Sci Alliance ; 7(9)2024 Sep.
Article in English | MEDLINE | ID: mdl-38960623

ABSTRACT

In many animal species, the oocyte meiotic spindle, which is required for chromosome segregation, forms without centrosomes. In some systems, Ran-GEF on chromatin initiates spindle assembly. We found that in Caenorhabditis elegans oocytes, endogenously-tagged Ran-GEF dissociates from chromatin during spindle assembly but re-associates during meiotic anaphase. Meiotic spindle assembly occurred after auxin-induced degradation of Ran-GEF, but anaphase I was faster than controls and extrusion of the first polar body frequently failed. In search of a possible alternative pathway for spindle assembly, we found that soluble tubulin concentrates in the nuclear volume during germinal vesicle breakdown. We found that the concentration of soluble tubulin in the metaphase spindle region is enclosed by ER sheets which exclude cytoplasmic organelles including mitochondria and yolk granules. Measurement of the volume occupied by yolk granules and mitochondria indicated that volume exclusion would be sufficient to explain the concentration of tubulin in the spindle volume. We suggest that this concentration of soluble tubulin may be a redundant mechanism promoting spindle assembly near chromosomes.


Subject(s)
Anaphase , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Oocytes , Spindle Apparatus , Tubulin , Animals , Caenorhabditis elegans/metabolism , Tubulin/metabolism , Spindle Apparatus/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Oocytes/metabolism , Prometaphase , Meiosis/physiology , ran GTP-Binding Protein/metabolism , Guanosine Triphosphate/metabolism , Chromatin/metabolism , Chromosome Segregation
6.
J Extracell Vesicles ; 13(6): e12459, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38868956

ABSTRACT

Isolation of neuron-derived extracellular vesicles (NDEVs) with L1 Cell Adhesion Molecule (L1CAM)-specific antibodies has been widely used to identify blood biomarkers of CNS disorders. However, full methodological validation requires demonstration of L1CAM in individual NDEVs and lower levels or absence of L1CAM in individual EVs from other cells. Here, we used multiple single-EV techniques to establish the neuronal origin and determine the abundance of L1CAM-positive EVs in human blood. L1CAM epitopes of the ectodomain are shown to be co-expressed on single-EVs with the neuronal proteins ß-III-tubulin, GAP43, and VAMP2, the levels of which increase in parallel with the enrichment of L1CAM-positive EVs. Levels of L1CAM-positive EVs carrying the neuronal proteins VAMP2 and ß-III-tubulin range from 30% to 63%, in contrast to 0.8%-3.9% of L1CAM-negative EVs. Plasma fluid-phase L1CAM does not bind to single-EVs. Our findings support the use of L1CAM as a target for isolating plasma NDEVs and leveraging their cargo to identify biomarkers reflecting neuronal function.


Subject(s)
Biomarkers , Extracellular Vesicles , Neural Cell Adhesion Molecule L1 , Neurons , Vesicle-Associated Membrane Protein 2 , Humans , Neural Cell Adhesion Molecule L1/metabolism , Extracellular Vesicles/metabolism , Biomarkers/metabolism , Biomarkers/blood , Neurons/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Tubulin/metabolism
7.
Proc Natl Acad Sci U S A ; 121(25): e2305260121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38857398

ABSTRACT

Human Cep57 is a coiled-coil scaffold at the pericentriolar matrix (PCM), controlling centriole duplication and centrosome maturation for faithful cell division. Genetic truncation mutations of Cep57 are associated with the mosaic-variegated aneuploidy (MVA) syndrome. During interphase, Cep57 forms a complex with Cep63 and Cep152, serving as regulators for centrosome maturation. However, the molecular interplay of Cep57 with these essential scaffolding proteins remains unclear. Here, we demonstrate that Cep57 undergoes liquid-liquid phase separation (LLPS) driven by three critical domains (NTD, CTD, and polybasic LMN). In vitro Cep57 condensates catalyze microtubule nucleation via the LMN motif-mediated tubulin concentration. In cells, the LMN motif is required for centrosomal microtubule aster formation. Moreover, Cep63 restricts Cep57 assembly, expansion, and microtubule polymerization activity. Overexpression of competitive constructs for multivalent interactions, including an MVA mutation, leads to excessive centrosome duplication. In Cep57-depleted cells, self-assembly mutants failed to rescue centriole disengagement and PCM disorganization. Thus, Cep57's multivalent interactions are pivotal for maintaining the accurate structural and functional integrity of human centrosomes.


Subject(s)
Cell Cycle Proteins , Centrioles , Centrosome , Microtubules , Humans , Centrosome/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Microtubules/metabolism , Centrioles/metabolism , Centrioles/genetics , Tubulin/metabolism , Tubulin/genetics , Mutation , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics , Protein Binding , Nuclear Proteins
8.
Exp Cell Res ; 440(1): 114126, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38857838

ABSTRACT

Microtubules are components of the cytoskeleton that perform essential functions in eukaryotes, such as those related to shape change, motility and cell division. In this context some characteristics of these filaments are essential, such as polarity and dynamic instability. In trypanosomatids, microtubules are integral to ultrastructure organization, intracellular transport and mitotic processes. Some species of trypanosomatids co-evolve with a symbiotic bacterium in a mutualistic association that is marked by extensive metabolic exchanges and a coordinated division of the symbiont with other cellular structures, such as the nucleus and the kinetoplast. It is already established that the bacterium division is microtubule-dependent, so in this work, it was investigated whether the dynamism and remodeling of these filaments is capable of affecting the prokaryote division. To this purpose, Angomonas deanei was treated with Trichostatin A (TSA), a deacetylase inhibitor, and mutant cells for histone deacetylase 6 (HDAC6) were obtained by CRISPR-Cas9. A decrease in proliferation, an enhancement in tubulin acetylation, as well as morphological and ultrastructural changes, were observed in TSA-treated protozoa and mutant cells. In both cases, symbiont filamentation occurred, indicating that prokaryote cell division is dependent on microtubule dynamism.


Subject(s)
Cell Division , Microtubules , Symbiosis , Microtubules/metabolism , Microtubules/ultrastructure , Microtubules/drug effects , Trypanosomatina/genetics , Trypanosomatina/metabolism , Trypanosomatina/ultrastructure , Trypanosomatina/physiology , Hydroxamic Acids/pharmacology , Tubulin/metabolism , Tubulin/genetics , Bacteria/metabolism , Bacteria/genetics , Acetylation , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase 6/metabolism , Histone Deacetylase 6/genetics , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure
9.
J Med Chem ; 67(12): 9950-9975, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38865195

ABSTRACT

To improve their aqueous solubility characteristics, water-solubilizing groups were added to some antiproliferative, rigidin-inspired 7-deazahypoxanthine frameworks after molecular modeling seemed to indicate that structural modifications on the C7 and/or C8 phenyl groups would be beneficial. To this end, two sets of 7-deazahypoxanthines were synthesized by way of a multicomponent reaction approach. It was subsequently determined that their antiproliferative activity against HeLa cells was retained for those derivatives with a glycol ether at the 4'-position of the C8 aryl ring system, while also significantly improving their solubility behavior. The best of these compounds were the equipotent 6-[4-(2-ethoxyethoxy)benzoyl]-2-(pent-4-yn-1-yl)-5-phenyl-1,7-dihydro-4H-pyrrolo[2,3-d]pyrimidin-4-one 33 and 6-[4-(2-ethoxyethoxy)benzoyl]-5-(3-fluorophenyl)-2-(pent-4-yn-1-yl)-1,7-dihydro-4H-pyrrolo[2,3-d]pyrimidin-4-one 59. Similarly to the parent 1, the new derivatives were also potent inhibitors of tubulin assembly. In treated HeLa cells, live cell confocal microscopy demonstrated their impact on microtubulin dynamics and spindle morphology, which is the upstream trigger of mitotic delay and cell death.


Subject(s)
Antineoplastic Agents , Cell Proliferation , Humans , HeLa Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Proliferation/drug effects , Structure-Activity Relationship , Drug Screening Assays, Antitumor , Solubility , Models, Molecular , Tubulin/metabolism
10.
Open Biol ; 14(6): 240041, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38835242

ABSTRACT

Platelets are blood cells derived from megakaryocytes that play a central role in regulating haemostasis and vascular integrity. The microtubule cytoskeleton of megakaryocytes undergoes a critical dynamic reorganization during cycles of endomitosis and platelet biogenesis. Quiescent platelets have a discoid shape maintained by a marginal band composed of microtubule bundles, which undergoes remarkable remodelling during platelet activation, driving shape change and platelet function. Disrupting or enhancing this process can cause platelet dysfunction such as bleeding disorders or thrombosis. However, little is known about the molecular mechanisms underlying the reorganization of the cytoskeleton in the platelet lineage. Recent studies indicate that the emergence of a unique platelet tubulin code and specific pathogenic tubulin mutations cause platelet defects and bleeding disorders. Frequently, these mutations exhibit dominant negative effects, offering valuable insights into both platelet disease mechanisms and the functioning of tubulins. This review will highlight our current understanding of the role of the microtubule cytoskeleton in the life and death of platelets, along with its relevance to platelet disorders.


Subject(s)
Blood Platelets , Cytoskeleton , Megakaryocytes , Microtubules , Humans , Blood Platelets/metabolism , Megakaryocytes/metabolism , Megakaryocytes/cytology , Cytoskeleton/metabolism , Microtubules/metabolism , Tubulin/metabolism , Tubulin/genetics , Animals , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/genetics , Blood Platelet Disorders/pathology , Mutation
11.
Sci Rep ; 14(1): 13797, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877139

ABSTRACT

In filamentous fungi, microtubules are important for polar growth and morphological maintenance and serve as rails for intracellular trafficking. The molecular mechanisms associated with microtubules have been analyzed. However, little is known about when and where tubulin, a component of microtubules, is biosynthesized in multinuclear and multicellular filamentous fungi. In this study, we visualized microtubules based on the enhanced green fluorescence protein (EGFP)-labeled α-tubulin and ß-tubulin mRNA tagged by the EGFP-mediated MS2 system in living yellow Koji mold Aspergillus oryzae cells in order to understand the spatiotemporal production mechanism of tubulin. We found that mRNA of btuA, encoding for ß-tubulin, localized at dot-like structures through the apical, middle and basal regions of the hyphal cells. In addition, some btuA mRNA dots showed microtubule-dependent motor protein-like dynamics in the cells. Furthermore, it was found that btuA mRNA dots were decreased in the cytoplasm just before mitosis but increased immediately after mitosis, followed by a gradual decrease. In summary, the localization and abundance of ß-tubulin mRNA is spatiotemporally regulated in living A. oryzae hyphal cells.


Subject(s)
Aspergillus oryzae , Microtubules , RNA, Messenger , Tubulin , Aspergillus oryzae/genetics , Aspergillus oryzae/metabolism , Tubulin/genetics , Tubulin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Microtubules/metabolism , Hyphae/genetics , Hyphae/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Gene Expression Regulation, Fungal , Fungal Proteins/genetics , Fungal Proteins/metabolism
12.
Int J Mol Sci ; 25(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892135

ABSTRACT

Podophyllotoxin (PPT) is an active pharmaceutical ingredient (API) with established antitumor potential. However, due to its systemic toxicity, its use is restricted to topical treatment of anogenital warts. Less toxic PPT derivatives (e.g., etoposide and teniposide) are used intravenously as anticancer agents. PPT has been exploited as a scaffold of new potential therapeutic agents; however, fewer studies have been conducted on the parent molecule than on its derivatives. We have undertaken a study of ultrastructural changes induced by PPT on HaCaT keratinocytes. We have also tracked the intracellular localization of PPT using its fluorescent derivative (PPT-FL). Moreover, we performed molecular docking of both PPT and PPT-FL to compare their affinity to various binding sites of tubulin. Using the Presto blue viability assay, we established working concentrations of PPT in HaCaT cells. Subsequently, we have used selected concentrations to determine PPT effects at the ultrastructural level. Dynamics of PPT distribution by confocal microscopy was performed using PPT-FL. Molecular docking calculations were conducted using Glide. PPT induces a time-dependent cytotoxic effect on HaCaT cells. Within 24 h, we observed the elongation of cytoplasmic processes, formation of cytoplasmic vacuoles, progressive ER stress, and shortening of the mitochondrial long axis. After 48 h, we noticed disintegration of the cell membrane, progressive vacuolization, apoptotic/necrotic vesicles, and a change in the cell nucleus's appearance. PPT-FL was detected within HaCaT cells after ~10 min of incubation and remained within cells in the following measurements. Molecular docking confirmed the formation of a stable complex between tubulin and both PPT and PPT-FL. However, it was formed at different binding sites. PPT is highly toxic to normal human keratinocytes, even at low concentrations. It promptly enters the cells, probably via endocytosis. At lower concentrations, PPT causes disruptions in both ER and mitochondria, while at higher concentrations, it leads to massive vacuolization with subsequent cell death. The novel derivative of PPT, PPT-FL, forms a stable complex with tubulin, and therefore, it is a useful tracker of intracellular PPT binding and trafficking.


Subject(s)
HaCaT Cells , Keratinocytes , Molecular Docking Simulation , Podophyllotoxin , Tubulin , Humans , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Podophyllotoxin/chemistry , Tubulin/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Survival/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/ultrastructure , Fluorescent Dyes/chemistry , Binding Sites , Endoplasmic Reticulum Stress/drug effects
13.
Arch Dermatol Res ; 316(7): 341, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847867

ABSTRACT

Topical tirbanibulin is a highly effective and well tolerated novel treatment option for actinic keratoses (AKs). This study aimed to characterize the mode of action of tirbanibulin in keratinocytes (NHEK) and cutaneous squamous cell carcinoma (cSCC) cell lines (A431, SCC-12) in vitro. Tirbanibulin significantly reduced proliferation in a dose-dependent manner in all investigated cell lines, inhibited migration, and induced G2/M-cell cycle arrest only in the cSCC cell lines analyzed, and induced apoptosis solely in A431, which showed the highest sensitivity to tirbanibulin. In general, we detected low basal expression of phosphorylated SRC in all cell lines analyzed, therefore, interference with SRC signaling does not appear to be the driving force regarding the observed effects of tirbanibulin. The most prominent tirbanibulin-mediated effect was on ß-tubulin-polymerization, which was especially impaired in A431. Additionally, tirbanibulin induced an increase of the proinflammatory cytokines IL-1α, bFGF and VEGF in A431. In conclusion, tirbanibulin mediated anti-tumor effects predominantly in A431, while healthy keratinocytes and more dedifferentiated SCC-12 were less influenced. These effects of tirbanibulin are most likely mediated via dysregulation of ß-tubulin-polymerization and may be supported by proinflammatory aspects.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Movement , Cell Proliferation , Keratinocytes , Skin Neoplasms , Tubulin , Humans , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Line, Tumor , Tubulin/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Apoptosis/drug effects , Cell Proliferation/drug effects , Cell Movement/drug effects , Antineoplastic Agents/pharmacology , Polymerization/drug effects , Keratosis, Actinic/drug therapy , Keratosis, Actinic/pathology , Keratosis, Actinic/metabolism , Signal Transduction/drug effects , Acetamides , Morpholines , Pyridines
14.
Protein Sci ; 33(7): e5078, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38895991

ABSTRACT

Alzheimer's disease is the fastest-growing neurodegenerative disease that affects over six million Americans. The abnormal aggregation of amyloid ß peptide and Tau protein is the expected molecular cause of the loss of neurons in brains of AD patients. A growing body of evidence indicates that lipids can alter the aggregation rate of amyloid ß peptide and modify the toxicity of amyloid ß aggregates. However, the role of lipids in Tau aggregation remains unclear. In this study, we utilized a set of biophysical methods to determine the extent to which phospatidylserine (PS) altered the aggregation properties of Tau isoforms with one (1N4R) and two (2N4R) N terminal inserts that enhance the binding of Tau to tubulin. We found that the length and saturation of fatty acids (FAs) in PS altered the aggregation rate of 2N4R isoform, while no changes in the aggregation rate of 1N4R were observed. These results indicate that N terminal inserts play an important role in protein-lipid interactions. We also found that PS could change the toxicity of 1N4R and 2N4R Tau fibrils, as well as alter molecular mechanisms by which these aggregates exert cytotoxicity to neurons. Finally, we found that although Tau fibrils formed in the presence and absence of PS endocytosed by cells, only fibril species that were formed in the presence of PS exert strong impairment of the cell mitochondria.


Subject(s)
Phosphatidylserines , Tubulin , tau Proteins , tau Proteins/metabolism , tau Proteins/chemistry , tau Proteins/toxicity , Humans , Phosphatidylserines/metabolism , Phosphatidylserines/chemistry , Tubulin/metabolism , Tubulin/chemistry , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Protein Binding , Neurons/metabolism , Neurons/drug effects , Protein Aggregates , Protein Isoforms/metabolism , Protein Isoforms/chemistry
15.
Curr Protoc ; 4(6): e1070, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865215

ABSTRACT

The microtubule (MT) cytoskeleton performs a variety of functions in cell division, cell architecture, neuronal differentiation, and ciliary beating. These functions are controlled by proteins that directly interact with MTs, commonly referred to as microtubule-associated proteins (MAPs). Out of the many proteins reported interact with MTs, only a some have been biochemically and functionally characterized so far. One of the limitations of classical in vitro assays and single-MT reconstitution approaches is that they are typically performed with purified proteins. As purification of proteins can be difficult and time-consuming, many previous studies have only focused on a few proteins, while systematic analyses of many different proteins by in vitro reconstitution assays were not possible. Here we present a detailed protocol using lysates of mammalian cells instead of purified proteins that overcomes this limitation. Those lysates contain all molecular components required for in vitro MT reconstitution including the endogenous tubulin and the recombinant MAPs, which form MT assemblies upon the injection of the lysates into a microscopy chamber. This allows to directly observe the dynamic behavior of growing MTs, as well as the fluorescently labeled associated proteins by total internal reflection fluorescence (TIRF) microscopy. Strikingly, all proteins tested so far were functional in our approach, thus providing the possibility to test virtually any protein of interest. This also opens the possibility to screen the impact of patient mutations on the MT binding behavior of MAPs in a medium-throughput manner. In addition, the lysate approach can easily be adapted to other applications that have predominantly been performed with purified proteins so far, such as investigating other cytoskeletal systems and cytoskeletal crosstalk, or to study structures of MAPs bound to MTs by cryo-electron microscopy. Our approach is thus a versatile, expandable, and easy-to-use method to characterize the impact of a broad spectrum of proteins on cytoskeletal behavior and function. © 2024 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Preparation of lysates of human cells for TIRF reconstitution assays Basic Protocol 2: Quantification of GFP-tagged MAP concentration in cell lysates Support Protocol 1: Purification of KIF5B(N555/T92A) (dead kinesin) protein for TIRF reconstitution assays Support Protocol 2: Preparation of GMPCPP MT seeds for TIRF reconstitution assays Basic Protocol 3: TIRF-based MT-MAP reconstitution assays using cell lysates.


Subject(s)
Microtubule-Associated Proteins , Microtubules , Humans , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Microtubules/chemistry , Animals , Cell-Free System , Tubulin/metabolism , Tubulin/chemistry , Microscopy, Fluorescence
16.
J Med Chem ; 67(11): 9227-9259, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38833507

ABSTRACT

The marine metabolite diazonamide A exerts low nanomolar cytotoxicity against a range of tumor cell lines; however, its highly complex molecular architecture undermines the therapeutic potential of the natural product. We demonstrate that truncation of heteroaromatic macrocycle in natural diazonamide A, combined with the replacement of the challenging-to-synthesize tetracyclic hemiaminal subunit by oxindole moiety leads to considerably less complex analogues with improved drug-like properties and nanomolar antiproliferative potency. The structurally simplified macrocycles are accessible in 12 steps from readily available indolin-2-one and tert-leucine with excellent diastereoselectivity (99:1 dr) in the key macrocyclization step. The most potent macrocycle acts as a tubulin assembly inhibitor and exerts similar effects on A2058 cell cycle progression and induction of apoptosis as does marketed microtubule-targeting agent vinorelbine.


Subject(s)
Antineoplastic Agents , Apoptosis , Microtubules , Tubulin Modulators , Humans , Tubulin Modulators/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/chemical synthesis , Cell Line, Tumor , Microtubules/drug effects , Microtubules/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Structure-Activity Relationship , Cell Proliferation/drug effects , Cell Cycle/drug effects , Biological Products/pharmacology , Biological Products/chemistry , Biological Products/chemical synthesis , Drug Screening Assays, Antitumor , Stereoisomerism , Tubulin/metabolism , Tubulin/chemistry , Indoles/chemistry , Indoles/pharmacology , Indoles/chemical synthesis , Heterocyclic Compounds, 4 or More Rings , Oxazoles
17.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 41(3): 569-576, 2024 Jun 25.
Article in Chinese | MEDLINE | ID: mdl-38932544

ABSTRACT

Tumor treatment fields (TTFields) can effectively inhibit the proliferation of tumor cells, but its mechanism remains exclusive. The destruction of cellular microtubule structure caused by TTFields through electric field force is considered to be the main reason for inhibiting tumor cell proliferation. However, the validity of this hypothesis still lacks exploration at the mesoscopic level. Therefore, in this study, we built force models for tubulins subjected to TTFields, based on the physical and electrical properties of tubulin molecules. We theoretically analyzed and simulated the dynamic effects of electric field force and torque on tubulin monomer polymerization, as well as the alignment and orientation of α/ß tubulin heterodimer, respectively. Research results indicate that the interference of electric field force induced by TTFields on tubulin monomer is notably weaker than the inherent electrostatic binding force among tubulin monomers. Additionally, the electric field torque generated by the TTFileds on α/ß tubulin dimers is also difficult to affect their random alignment. Therefore, at the mesoscale, our study affirms that TTFields are improbable to destabilize cellular microtubule structures via electric field dynamics effects. These results challenge the traditional view that TTFields destroy the microtubule structure of cells through TTFields electric field force, and proposes a new approach that should pay more attention to the "non-mechanical" effects of TTFields in the study of TTFields mechanism. This study can provide reliable theoretical basis and inspire new research directions for revealing the mesoscopic bioelectrical mechanism of TTFields.


Subject(s)
Microtubules , Neoplasms , Tubulin , Tubulin/metabolism , Microtubules/metabolism , Humans , Neoplasms/therapy , Cell Proliferation , Static Electricity , Polymerization , Electromagnetic Fields
18.
J Biol Chem ; 300(6): 107363, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38735475

ABSTRACT

Cryptophycins are microtubule-targeting agents (MTAs) that belong to the most potent antimitotic compounds known to date; however, their exact molecular mechanism of action remains unclear. Here, we present the 2.2 Å resolution X-ray crystal structure of a potent cryptophycin derivative bound to the αß-tubulin heterodimer. The structure addresses conformational issues present in a previous 3.3 Å resolution cryo-electron microscopy structure of cryptophycin-52 bound to the maytansine site of ß-tubulin. It further provides atomic details on interactions of cryptophycins, which had not been described previously, including ones that are in line with structure-activity relationship studies. Interestingly, we discovered a second cryptophycin-binding site that involves the T5-loop of ß-tubulin, a critical secondary structure element involved in the exchange of the guanosine nucleotide and in the formation of longitudinal tubulin contacts in microtubules. Cryptophycins are the first natural ligands found to bind to this new "ßT5-loop site" that bridges the maytansine and vinca sites. Our results offer unique avenues to rationally design novel MTAs with the capacity to modulate T5-loop dynamics and to simultaneously engage multiple ß-tubulin binding sites.


Subject(s)
Maytansine , Tubulin , Tubulin/chemistry , Tubulin/metabolism , Maytansine/chemistry , Maytansine/analogs & derivatives , Humans , Crystallography, X-Ray , Binding Sites , Microtubules/metabolism , Microtubules/chemistry , Vinca Alkaloids/chemistry , Vinca Alkaloids/metabolism
19.
J Biol Chem ; 300(6): 107373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38762183

ABSTRACT

Motile cilia on the cell surface produce fluid flows in the body and abnormalities in motile cilia cause primary ciliary dyskinesia. Dynein axonemal assembly factor 6 (DNAAF6), a causative gene of primary ciliary dyskinesia, was isolated as an interacting protein with La ribonucleoprotein 6 (LARP6) that regulates ciliogenesis in multiciliated cells (MCCs). In MCCs of Xenopus embryos, LARP6 and DNAAF6 were colocalized in biomolecular condensates termed dynein axonemal particles and synergized to control ciliogenesis. Moreover, tubulin alpha 1c-like mRNA encoding α-tubulin protein, that is a major component of ciliary axoneme, was identified as a target mRNA regulated by binding LARP6. While DNAAF6 was necessary for high α-tubulin protein expression near the apical side of Xenopus MCCs during ciliogenesis, its mutant, which abolishes binding with LARP6, was unable to restore the expression of α-tubulin protein near the apical side of MCCs in Xenopus DNAAF6 morphant. These results indicated that the binding of LARP6 and DNAAF6 in dynein axonemal particles regulates highly expressed α-tubulin protein near the apical side of Xenopus MCCs during ciliogenesis.


Subject(s)
Cilia , Ribonucleoproteins , Tubulin , Xenopus Proteins , Xenopus laevis , Cilia/metabolism , Animals , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Tubulin/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Humans , SS-B Antigen , Autoantigens/metabolism , Autoantigens/genetics , Protein Binding , Axoneme/metabolism , RNA, Messenger/metabolism , RNA, Messenger/genetics
20.
Bioorg Med Chem ; 107: 117751, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38762979

ABSTRACT

In previous studies, we developed anti-trypanosome tubulin inhibitors with promising in vitro selectivity and activity against Human African Trypanosomiasis (HAT). However, for such agents, oral activity is crucial. This study focused on further optimizing these compounds to enhance their ligand efficiency, aiming to reduce bulkiness and hydrophobicity, which should improve solubility and, consequently, oral bioavailability. Using Trypanosoma brucei brucei cells as the parasite model and human normal kidney cells and mouse macrophage cells as the host model, we evaluated 30 new analogs synthesized through combinatorial chemistry. These analogs have fewer aromatic moieties and lower molecular weights than their predecessors. Several new analogs demonstrated IC50s in the low micromolar range, effectively inhibiting trypanosome cell growth without harming mammalian cells at the same concentration. We conducted a detailed structure-activity relationship (SAR) analysis and a docking study to assess the compounds' binding affinity to trypanosome tubulin homolog. The results revealed a correlation between binding energy and anti-Trypanosoma activity. Importantly, compound 7 displayed significant oral activity, effectively inhibiting trypanosome cell proliferation in mice.


Subject(s)
Trypanocidal Agents , Trypanosoma brucei brucei , Animals , Trypanosoma brucei brucei/drug effects , Trypanocidal Agents/pharmacology , Trypanocidal Agents/chemical synthesis , Trypanocidal Agents/chemistry , Structure-Activity Relationship , Mice , Humans , Administration, Oral , Cell Proliferation/drug effects , Molecular Structure , Molecular Docking Simulation , Tubulin/metabolism , Parasitic Sensitivity Tests , Dose-Response Relationship, Drug , Tubulin Modulators/pharmacology , Tubulin Modulators/chemical synthesis , Tubulin Modulators/chemistry , Trypanosomiasis, African/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...