Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.285
Filtrar
1.
J Phys Chem Lett ; 15(14): 3893-3899, 2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38563569

RESUMO

Kinesin is a typical motor protein that can use the chemical energy of ATP hydrolysis to step processively on microtubules, alternating between one-head-bound and two-head-bound states. Some published experimental results showed that the duration of the one-head-bound state increases greatly with a decrease in ATP concentration, whereas the duration of the two-head-bound state is independent of ATP concentration, indicating that ATP binding occurs in the one-head-bound state. On the contrary, other experimental results showed that the duration of the two-head-bound state increases greatly with a decrease in ATP concentration, whereas the duration of the one-head-bound state increases slightly with a decrease in ATP concentration, indicating that ATP binding occurs mainly in the two-head-bound state. Here, we explain consistently and quantitatively these contradictory experimental results, resolving the controversy that is critical to the chemomechanical coupling mechanism of the kinesin motor.


Assuntos
Trifosfato de Adenosina , Cinesinas , Cinesinas/metabolismo , Trifosfato de Adenosina/metabolismo , Microtúbulos/metabolismo , Cinética
2.
Int J Oncol ; 64(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38426627

RESUMO

Despite advances in treatment and early detection, breast cancer remains one of the most common types of cancer and is the second leading cause of cancer death after lung cancer in women. Therefore, there is an urgent need to develop new biomarkers and therapeutic targets for the treatment of breast cancer. Based on gene expression profiles and subsequent screening performed in a preliminary study, kinesin family member 20B (KIF20B) was selected as a candidate target molecule, because it was highly and frequently expressed in all subtypes of breast cancer and barely detected in normal tissues. Reverse transcription­quantitative PCR and western blotting revealed that KIF20B mRNA and protein expression levels were upregulated in most breast cancer cell lines but were scarcely expressed in normal mammary epithelial cells. Immunohistochemical staining of a tissue microarray showed that KIF20B was detected in 145 out of 251 (57.8%) breast cancer tissues. Strong KIF20B expression was significantly related to advanced pathological N stage. Moreover, patients with breast cancer and strong KIF20B expression exhibited a significantly worse prognosis than those with weak or negative KIF20B expression (P<0.0001, log­rank test). In multivariate analysis, strong expression was an independent prognostic factor for patients with breast cancer. Furthermore, knockdown of KIF20B expression by small interfering RNA inhibited breast cancer cell proliferation and induced apoptosis. In addition, Matrigel cell invasion assays revealed that the invasiveness of breast cancer cells was significantly decreased by KIF20B silencing. Since KIF20B is an oncoprotein that is strongly expressed in highly malignant clinical breast cancer and serves a pivotal role in breast cancer cell proliferation, survival and invasion, KIF20B could be considered a candidate biomarker for prognostic prediction and a potential molecular target for developing new therapeutics, such as small molecule inhibitors, for a wide variety of breast cancers.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/genética , Prognóstico , RNA Interferente Pequeno , Células MCF-7 , Proliferação de Células/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Movimento Celular/genética , Cinesinas/metabolismo
3.
J Cell Sci ; 137(7)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38477340

RESUMO

Axonal transport in neurons is essential for cargo movement between the cell body and synapses. Caenorhabditis elegans UNC-104 and its homolog KIF1A are kinesin-3 motors that anterogradely transport precursors of synaptic vesicles (pre-SVs) and are degraded at synapses. However, in C. elegans, touch neuron-specific knockdown of the E1 ubiquitin-activating enzyme, uba-1, leads to UNC-104 accumulation at neuronal ends and synapses. Here, we performed an RNAi screen and identified that depletion of fbxb-65, which encodes an F-box protein, leads to UNC-104 accumulation at neuronal distal ends, and alters UNC-104 net anterograde movement and levels of UNC-104 on cargo without changing synaptic UNC-104 levels. Split fluorescence reconstitution showed that UNC-104 and FBXB-65 interact throughout the neuron. Our theoretical model suggests that UNC-104 might exhibit cooperative cargo binding that is regulated by FBXB-65. FBXB-65 regulates an unidentified post-translational modification (PTM) of UNC-104 in a region beside the cargo-binding PH domain. Both fbxb-65 and UNC-104, independently of FBXB-65, regulate axonal pre-SV distribution, transport of pre-SVs at branch points and organismal lifespan. FBXB-65 regulates a PTM of UNC-104 and the number of motors on the cargo surface, which can fine-tune cargo transport to the synapse.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas F-Box , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Cinesinas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas F-Box/metabolismo , Domínios de Homologia à Plecstrina , Proteínas do Tecido Nervoso/metabolismo , Processamento de Proteína Pós-Traducional , Transporte Axonal/fisiologia
4.
Cell Commun Signal ; 22(1): 199, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553728

RESUMO

KIFC3 is a member of Kinesin-14 family motor proteins, which play a variety of roles such as centrosome cohesion, cytokinesis, vesicles transportation and cell proliferation in mitosis. Here, we investigated the functional roles of KIFC3 in meiosis. Our findings demonstrated that KIFC3 exhibited expression and localization at centromeres during metaphase I, followed by translocation to the midbody at telophase I throughout mouse oocyte meiosis. Disruption of KIFC3 activity resulted in defective polar body extrusion. We observed aberrant meiotic spindles and misaligned chromosomes, accompanied by the loss of kinetochore-microtubule attachment, which might be due to the failed recruitment of BubR1/Bub3. Coimmunoprecipitation data revealed that KIFC3 plays a crucial role in maintaining the acetylated tubulin level mediated by Sirt2, thereby influencing microtubule stability. Additionally, our findings demonstrated an interaction between KIFC3 and PRC1 in regulating midbody formation during telophase I, which is involved in cytokinesis regulation. Collectively, these results underscore the essential contribution of KIFC3 to spindle assembly and cytokinesis during mouse oocyte meiosis.


Assuntos
Citocinese , Cinesinas , Animais , Camundongos , Cinesinas/genética , Cinesinas/metabolismo , Meiose , Microtúbulos/metabolismo , Oócitos/metabolismo
5.
J Comput Aided Mol Des ; 38(1): 16, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38556596

RESUMO

The kinesin spindle protein (Eg5) is a mitotic protein that plays an essential role in the formation of the bipolar spindles during the mitotic phase. Eg5 protein controls the segregation of the chromosomes in mitosis which renders it a vital target for cancer treatment. In this study our approach to identifying novel scaffold for Eg5 inhibitors is based on targeting the novel allosteric pocket (α4/α6/L11). Extensive computational techniques were applied using ligand-based virtual screening and molecular docking by two approaches, MOE and AutoDock, to screen a library of commercial compounds. We identified compound 8-(3-(1H-imidazol-1-ylpropylamino)-3-methyl-7-((naphthalen-3-yl)methyl)-1H-purine-2, 6 (3H,7H)-dione (compound 5) as a novel scaffold for Eg5 inhibitors. This compound inhibited cancer cell Eg5 ATPase at 2.37 ± 0.15 µM. The molecular dynamics simulations revealed that the identified compound formed stable interactions in the allosteric pocket (α4/α6/L11) of the receptor, indicating its potential as a novel Eg5 inhibitor.


Assuntos
Cinesinas , Simulação de Dinâmica Molecular , Simulação de Acoplamento Molecular , Cinesinas/metabolismo , Ligantes , Mitose
6.
Mol Biol Cell ; 35(5): ar61, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38446634

RESUMO

Neurons are polarized cells that require accurate membrane trafficking to maintain distinct protein complements at dendritic and axonal membranes. The Kinesin-3 family members KIF13A and KIF13B are thought to mediate dendrite-selective transport, but the mechanism by which they are recruited to polarized vesicles and the differences in the specific trafficking role of each KIF13 have not been defined. We performed live-cell imaging in cultured hippocampal neurons and found that KIF13A is a dedicated dendrite-selective kinesin. KIF13B confers two different transport modes, dendrite- and axon-selective transport. Both KIF13s are maintained at the trans-Golgi network by interactions with the heterotetrameric adaptor protein complex AP-1. Interference with KIF13 binding to AP-1 resulted in disruptions to both dendrite- and axon-selective trafficking. We propose that AP-1 is the molecular link between the sorting of polarized cargoes into vesicles and the recruitment of kinesins that confer polarized transport.


Assuntos
Complexo 1 de Proteínas Adaptadoras , Complexo de Golgi , Cinesinas , Rede trans-Golgi , Células Cultivadas , Complexo de Golgi/metabolismo , Cinesinas/metabolismo , Neurônios/metabolismo , Transporte Proteico/genética , Transporte Proteico/fisiologia , Complexo 1 de Proteínas Adaptadoras/metabolismo , Rede trans-Golgi/metabolismo
7.
Nat Methods ; 21(4): 569-573, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38480903

RESUMO

Here we show that MINSTED localization, a method whereby the position of a fluorophore is identified with precisely controlled beams of a STED microscope, tracks fluorophores and hence labeled biomolecules with nanometer/millisecond spatiotemporal precision. By updating the position for each detected photon, MINSTED recognizes fluorophore steps of 16 nm within <250 µs using about 13 photons. The power of MINSTED tracking is demonstrated by resolving the stepping of the motor protein kinesin-1 walking on microtubules and switching protofilaments.


Assuntos
Cinesinas , Microtúbulos , Microtúbulos/metabolismo , Cinesinas/metabolismo , Microscopia
8.
Mil Med Res ; 11(1): 17, 2024 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-38475827

RESUMO

BACKGROUND: Tactile and mechanical pain are crucial to our interaction with the environment, yet the underpinning molecular mechanism is still elusive. Endophilin A2 (EndoA2) is an evolutionarily conserved protein that is documented in the endocytosis pathway. However, the role of EndoA2 in the regulation of mechanical sensitivity and its underlying mechanisms are currently unclear. METHODS: Male and female C57BL/6 mice (8-12 weeks) and male cynomolgus monkeys (7-10 years old) were used in our experiments. Nerve injury-, inflammatory-, and chemotherapy-induced pathological pain models were established for this study. Behavioral tests of touch, mechanical pain, heat pain, and cold pain were performed in mice and nonhuman primates. Western blotting, immunostaining, co-immunoprecipitation, proximity ligation and patch-clamp recordings were performed to gain insight into the mechanisms. RESULTS: The results showed that EndoA2 was primarily distributed in neurofilament-200-positive (NF200+) medium-to-large diameter dorsal root ganglion (DRG) neurons of mice and humans. Loss of EndoA2 in mouse NF200+ DRG neurons selectively impaired the tactile and mechanical allodynia. Furthermore, EndoA2 interacted with the mechanically sensitive ion channel Piezo2 and promoted the membrane trafficking of Piezo2 in DRG neurons. Moreover, as an adaptor protein, EndoA2 also bound to kinesin family member 5B (KIF5B), which was involved in the EndoA2-mediated membrane trafficking process of Piezo2. Loss of EndoA2 in mouse DRG neurons damaged Piezo2-mediated rapidly adapting mechanically activated currents, and re-expression of EndoA2 rescued the MA currents. In addition, interference with EndoA2 also suppressed touch sensitivity and mechanical hypersensitivity in nonhuman primates. CONCLUSIONS: Our data reveal that the KIF5B/EndoA2/Piezo2 complex is essential for Piezo2 trafficking and for sustaining transmission of touch and mechanical hypersensitivity signals. EndoA2 regulates touch and mechanical allodynia via kinesin-mediated Piezo2 trafficking in sensory neurons. Our findings identify a potential new target for the treatment of mechanical pain.


Assuntos
Aciltransferases , Hiperalgesia , Canais Iônicos , Tato , Animais , Feminino , Masculino , Camundongos , Hiperalgesia/patologia , Canais Iônicos/metabolismo , Cinesinas/metabolismo , Mecanotransdução Celular/fisiologia , Camundongos Endogâmicos C57BL , Dor , Primatas , Tato/fisiologia , Aciltransferases/metabolismo
9.
PLoS One ; 19(3): e0295652, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38478520

RESUMO

In intracellular active transport, molecular motors are responsible for moving biological cargo along networks of microtubules that serve as scaffolds. Cargo dynamics can be modified by different features of microtubule networks such as geometry, density, orientation modifications. Also, the dynamical behaviour of the molecular motors is determined by the microtubule network and by the individual and/or collective action of the motors. For example, unlike single kinesins, the mechanistic behavior of multiple kinesins varies from one experiment to another. However, the reasons for this experimental variability are unknown. Here we show theoretically how non-radial and quasi-radial microtubule architectures modify the collective behavior of two kinesins attached on a cargo. We found out under which structural conditions transport is most efficient and the most likely way in which kinesins are organized in active transport. In addition, with motor activity, mean intermotor distance and motor organization, we determined the character of the collective interaction of the kinesins during transport. Our results demonstrate that two-dimensional microtubule structures promote branching due to crossovers that alter directionality in cargo movement and may provide insight into the collective organization of the motors. Our article offers a perspective to analyze how the two-dimensional network can modify the cargo-motor dynamics for the case in which multiple motors move in different directions as in the case of kinesin and dynein.


Assuntos
Dineínas , Cinesinas , Cinesinas/metabolismo , Transporte Biológico , Transporte Biológico Ativo , Dineínas/metabolismo , Microtúbulos/metabolismo
10.
Cell Death Dis ; 15(3): 222, 2024 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-38493150

RESUMO

Unlike normal cells, cancer cells frequently exhibit supernumerary centrosomes, leading to formation of multipolar spindles that can trigger cell death. Nevertheless, cancer cells with supernumerary centrosomes escape the deadly consequences of unequal segregation of genomic material by coalescing their centrosomes into two poles. This unique trait of cancer cells presents a promising target for cancer therapy, focusing on selectively attacking cells with supernumerary centrosomes. Nek2A is a kinase involved in mitotic regulation, including the centrosome cycle, where it phosphorylates linker proteins to separate centrosomes. In this study, we investigated if Nek2A also prevents clustering of supernumerary centrosomes, akin to its separation function. Reduction of Nek2A activity, achieved through knockout, silencing, or inhibition, promotes centrosome clustering, whereas its overexpression results in inhibition of clustering. Significantly, prevention of centrosome clustering induces cell death, but only in cancer cells with supernumerary centrosomes, both in vitro and in vivo. Notably, none of the known centrosomal (e.g., CNAP1, Rootletin, Gas2L1) or non-centrosomal (e.g., TRF1, HEC1) Nek2A targets were implicated in this machinery. Additionally, Nek2A operated via a pathway distinct from other proteins involved in centrosome clustering mechanisms, like HSET and NuMA. Through TurboID proximity labeling analysis, we identified novel proteins associated with the centrosome or microtubules, expanding the known interaction partners of Nek2A. KIF2C, in particular, emerged as a novel interactor, confirmed through coimmunoprecipitation and localization analysis. The silencing of KIF2C diminished the impact of Nek2A on centrosome clustering and rescued cell viability. Additionally, elevated Nek2A levels were indicative of better patient outcomes, specifically in those predicted to have excess centrosomes. Therefore, while Nek2A is a proposed target, its use must be specifically adapted to the broader cellular context, especially considering centrosome amplification. Discovering partners such as KIF2C offers fresh insights into cancer biology and new possibilities for targeted treatment.


Assuntos
Centrossomo , Neoplasias , Humanos , Ciclo Celular , Morte Celular , Centrossomo/metabolismo , Análise por Conglomerados , Cinesinas/genética , Cinesinas/metabolismo , Microtúbulos/metabolismo , Mitose , Neoplasias/genética , Neoplasias/metabolismo , Fuso Acromático/metabolismo
11.
J Cell Biol ; 223(5)2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38470363

RESUMO

Mitochondria transport is crucial for axonal mitochondria distribution and is mediated by kinesin-1-based anterograde and dynein-based retrograde motor complexes. While Miro and Milton/TRAK were identified as key adaptors between mitochondria and kinesin-1, recent studies suggest the presence of additional mechanisms. In C. elegans, ric-7 is the only single gene described so far, other than kinesin-1, that is absolutely required for axonal mitochondria localization. Using CRISPR engineering in C. elegans, we find that Miro is important but is not essential for anterograde traffic, whereas it is required for retrograde traffic. Both the endogenous RIC-7 and kinesin-1 act at the leading end to transport mitochondria anterogradely. RIC-7 binding to mitochondria requires its N-terminal domain and partially relies on MIRO-1, whereas RIC-7 accumulation at the leading end depends on its disordered region, kinesin-1, and metaxin2. We conclude that transport complexes containing kinesin-1 and RIC-7 polarize at the leading edge of mitochondria and are required for anterograde axonal transport in C. elegans.


Assuntos
Transporte Axonal , Cinesinas , Animais , Axônios , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Cinesinas/metabolismo , Mitocôndrias/metabolismo
12.
Nat Commun ; 15(1): 1948, 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38431715

RESUMO

Microtubules (MTs) are key components of the eukaryotic cytoskeleton and are essential for intracellular organization, organelle trafficking and mitosis. MT tasks depend on binding and interactions with MT-associated proteins (MAPs). MT-associated protein 7 (MAP7) has the unusual ability of both MT binding and activating kinesin-1-mediated cargo transport along MTs. Additionally, the protein is reported to stabilize MTs with its 112 amino-acid long MT-binding domain (MTBD). Here we investigate the structural basis of the interaction of MAP7 MTBD with the MT lattice. Using a combination of solid and solution-state nuclear magnetic resonance (NMR) spectroscopy with electron microscopy, fluorescence anisotropy and isothermal titration calorimetry, we shed light on the binding mode of MAP7 to MTs at an atomic level. Our results show that a combination of interactions between MAP7 and MT lattice extending beyond a single tubulin dimer and including tubulin C-terminal tails contribute to formation of the MAP7-MT complex.


Assuntos
Proteínas Associadas aos Microtúbulos , Tubulina (Proteína) , Cinesinas/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Organelas/metabolismo , Tubulina (Proteína)/metabolismo , Humanos
13.
Int Immunopharmacol ; 131: 111613, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38489970

RESUMO

BACKGROUND: Bladder cancer (BCa) is a common malignant disease with high recurrence and poor prognosis. Several circular RNAs (circRNAs) have been found to be associated with the malignant progression of bladder cancer (BCa). Here, the aim of this study was to investigate the expression, role and mechanism of circRAPGEF5 in BCa progression. METHODS: Quantitative real-time PCR (qRT-PCR) and immunoblotting were used to detect gene and protein expression levels. In vitro functional studies were performed using CCK-8, colony formation, wound healing and Transwell assays, respectively, and a mouse xenograft tumor model was established to perform in vivo experiments. Bioinformatic predictions as well as luciferase reporter assays and RNA pull-down assays were used to probe circRAPGEF5-mediated competitive endogenous RNA (ceRNA) network. RESULTS: CircRAPGEF5 was significantly overexpressed in BCa patients (p < 0.05), indicating a potential unsatisfactory prognosis. Functionally, knockdown of circRAPGEF5 inhibited the growth, migration and invasion of BCa cells in vitro (p < 0.05), as well as BCa growth in vivo (p < 0.05). Mechanistically, circRAPGEF5 acted as a sponge for miR-582-3p and targeted kinesin family member 3A (KIF3A). In addition, rescue experiments showed that inhibition of miR-582-3p or overexpression of KIF3A reversed the anticancer effects of circRAPGEF5 knockdown on BCa cells (p < 0.05). CONCLUSION: Silencing circRAPGEF5 inhibits BCa proliferation, migration and invasion via the miR-582-3p/KIF3A axis, demonstrating a promising target for BCa-targeted therapy.


Assuntos
MicroRNAs , RNA Circular , Neoplasias da Bexiga Urinária , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Cinesinas/genética , Cinesinas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , RNA Circular/metabolismo
14.
Dig Dis Sci ; 69(4): 1274-1286, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38446308

RESUMO

BACKGROUND & AIMS: Kinesin family member 18A (KIF18A) is notable for its aberrant expression across various cancer types and its pivotal role is driving cancer progression. In this study, we aim to investigate the intricate molecular mechanisms underlying the impact of KIF18A on the progression of HCC. METHODS: Western blotting assays, a quantitative real-time PCR and immunohistochemical analyses were performed to quantitatively assess KIF18A expression in HCC tissues. We then performed genetic manipulations within HCC cells by silencing endogenous KIF18A using short hairpin RNA (shRNA) and introducing exogenous plasmids to overexpress KIF18A. We monitored cell progression, analyzed cell cycle and cell apoptosis and assessed cell migration and invasion both in vitro and in vivo. Moreover, we conducted RNA-sequencing to explore KIF18A-related signaling pathways utilizing Reactome and KEGG enrichment methods and validated these critical mediators in these pathways. RESULTS: Analysis of the TCGA-LIHC database revealed pronounced overexpression of KIF18A in HCC tissues, the finding was subsequently confirmed through the analysis of clinical samples obtained from HCC patients. Notably, silencing KIF18A in cells led to an obvious inhibition of cell proliferation, migration and invasion in vitro. Furthermore, in subcutaneous and orthotopic xenograft models, suppression of KIF18A sgnificantly redudce tumor weight and the number of lung metastatic nodules. Mechanistically, KIF18A appears to facilitate cell proliferation by upregulating MAD2 and CDK1/CyclinB1 expression levels, with the activation of SMAD2/3 signaling contributing to KIF18A-driven metastasis. CONCLUSION: Our study elucidates the molecular mechanism by which KIF18A mediates proliferation and metastasis in HCC cells, offering new insights into potential therapeutic targets.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Prognóstico , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias Hepáticas/patologia , Biomarcadores Tumorais/genética , Proliferação de Células , RNA Interferente Pequeno , Família , Regulação Neoplásica da Expressão Gênica , Movimento Celular , Linhagem Celular Tumoral
15.
Cancer Lett ; 588: 216815, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38490329

RESUMO

Epigenetic modifiers are upregulated during the process of prostate cancer, acquiring resistance to castration therapy and becoming lethal metastatic castration-resistant prostate cancer (CRPC). However, the relationship between regulation of histone modifications and chromatin structure in CRPC has yet not fully been validated. Here, we reanalyzed publicly available clinical transcriptome and clinical outcome data and identified NSD2, a histone methyltransferase that catalyzes H3K36me2, as an epigenetic modifier that was upregulated in CRPC and whose increased expression in prostate cancer correlated with higher recurrence rate. We performed ChIP-seq, RNA-seq, and Hi-C to conduct comprehensive epigenomic and transcriptomic analyses to identify epigenetic reprogramming in CRPC. In regions where H3K36me2 was increased, H3K27me3 was decreased, and the compartment was shifted from inactive to active. In these regions, 68 aberrantly activated genes were identified as candidate downstream genes of NSD2 in CRPC. Among these genes, we identified KIF18A as critical for CRPC growth. Under NSD2 upregulation in CRPC, epigenetic alteration with H3K36me2-gain and H3K27me3-loss occurs accompanying with an inactive-to-active compartment shift, suggesting that histone modification and chromatin structure cooperatively change prostate carcinogenesis.


Assuntos
Cromatina , Neoplasias de Próstata Resistentes à Castração , Masculino , Humanos , Cromatina/genética , Histonas/genética , Histonas/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Receptores Androgênicos/metabolismo , Cinesinas/metabolismo
16.
Crit Rev Eukaryot Gene Expr ; 34(3): 37-48, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38305287

RESUMO

Gastric cancer (GC) is a main cause of cancer death in the world, and improving the chemotherapy sensitivity can enhance the chemotherapy efficacy of GC. The study objective is to explore the differential KIF18B expression in GC and its effect on GC chemotherapy sensitivity. The KIF18B expression in GC tissues and adjacent normal tissues was analyzed by real-time quantitative polymerase chain reaction. The relationship between differential KIF18B expression and different clinicopathological features was detected. It was found that KIF18B was highly expressed in GC tissues, and KIF18B expression was differential in patients with different clinicopathological features. The upregulation of KIF18B has a positive correlation with the poor therapeutic effect and high KIF18 was associated with lower 3-year overall survival and disease-free survival. The KIF18B-downregulated NCI-N87 cells were constructed and tested by cell counting kit-8 assay and colony formation. Cell migration and invasion were detected by Transwell assay. The xenograft tumor model was established to observe the effect of KIF18B on the efficacy of chemotherapy. The upregulation of KIF18B reduced the chemotherapy sensitivity of GC cells and enhanced their proliferation, migration, and invasion. Silencing KIF18B inhibited tumor growth and promoted chemotherapy efficacy in vivo. In summary, KIF18B inhibitor may have a potential function for improving the efficacy of chemotherapy in GC.


Assuntos
Cinesinas , Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Cinesinas/genética , Cinesinas/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Regulação para Cima , Animais
17.
PLoS Pathog ; 20(2): e1012000, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38300973

RESUMO

The early branching eukaryote Trypanosoma brucei divides uni-directionally along the longitudinal cell axis from the cell anterior toward the cell posterior, and the cleavage furrow ingresses along the cell division plane between the new and the old flagella of a dividing bi-flagellated cell. Regulation of cytokinesis in T. brucei involves actomyosin-independent machineries and trypanosome-specific signaling pathways, but the molecular mechanisms underlying cell division plane positioning remain poorly understood. Here we report a kinesin-13 family protein, KIN13-5, that functions downstream of FPRC in the cytokinesis regulatory pathway and determines cell division plane placement. KIN13-5 localizes to multiple cytoskeletal structures, interacts with FPRC, and depends on FPRC for localization to the site of cytokinesis initiation. Knockdown of KIN13-5 causes loss of microtubule bundling at both ends of the cell division plane, leading to mis-placement of the cleavage furrow and unequal cytokinesis, and at the posterior cell tip, causing the formation of a blunt posterior. In vitro biochemical assays demonstrate that KIN13-5 bundles microtubules, providing mechanistic insights into the role of KIN13-5 in cytokinesis and posterior morphogenesis. Altogether, KIN13-5 promotes microtubule bundle formation to ensure cleavage furrow placement and to maintain posterior cytoskeleton morphology in T. brucei.


Assuntos
Citocinese , Trypanosoma brucei brucei , Citocinese/fisiologia , Trypanosoma brucei brucei/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , Citoesqueleto/metabolismo , Microtúbulos/metabolismo , Morfogênese , Proteínas de Protozoários/metabolismo
18.
EMBO J ; 43(7): 1244-1256, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38424239

RESUMO

During mitosis, motor proteins and microtubule-associated protein organize the spindle apparatus by cross-linking and sliding microtubules. Kinesin-5 plays a vital role in spindle formation and maintenance, potentially inducing twist in the spindle fibers. The off-axis power stroke of kinesin-5 could generate this twist, but its implications in microtubule organization remain unclear. Here, we investigate 3D microtubule-microtubule sliding mediated by the human kinesin-5, KIF11, and found that the motor caused right-handed helical motion of anti-parallel microtubules around each other. The sidestepping ratio increased with reduced ATP concentration, indicating that forward and sideways stepping of the motor are not strictly coupled. Further, the microtubule-microtubule distance (motor extension) during sliding decreased with increasing sliding velocity. Intriguingly, parallel microtubules cross-linked by KIF11 orbited without forward motion, with nearly full motor extension. Altering the length of the neck linker increased the forward velocity and pitch of microtubules in anti-parallel overlaps. Taken together, we suggest that helical motion and orbiting of microtubules, driven by KIF11, contributes to flexible and context-dependent filament organization, as well as torque regulation within the mitotic spindle.


Assuntos
Cinesinas , Microtúbulos , Humanos , Cinesinas/metabolismo , Microtúbulos/metabolismo , Fuso Acromático/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Mitose
19.
Microbiol Res ; 282: 127653, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38422859

RESUMO

In multinuclear and multicellular filamentous fungi little is known about how mRNAs encoding secreted enzymes are transcribed and localized spatiotemporally. To better understand this process we analyzed mRNA encoding GlaA, a glucoamylase secreted in large amounts by the industrial filamentous fungus Aspergillus oryzae, by the MS2 system, in which mRNA can be visualized in living cells. We found that glaA mRNA was significantly transcribed and localized near the hyphal tip and septum, which are the sites of protein secretion, in polarity-dependent expression and localization manners. We also revealed that glaA mRNA exhibits long-range dynamics in the vicinity of the endoplasmic reticulum (ER) in a manner that is dependent on the microtubule motor proteins kinesin-1 and kinesin-3, but independent of early endosomes. Moreover, we elucidated that although glaA mRNA localized to stress granules (SGs) and processing bodies (PBs) under high temperature, glaA mRNA was not seen under ER stress, suggesting that there are different regulatory mechanisms of glaA mRNA by SG and PB under high temperature and ER stress. Collectively, this study uncovers a dynamic regulatory mechanism of mRNA encoding a secretory enzyme in filamentous fungi.


Assuntos
Glucana 1,4-alfa-Glucosidase , Cinesinas , Glucana 1,4-alfa-Glucosidase/genética , Glucana 1,4-alfa-Glucosidase/metabolismo , Cinesinas/metabolismo , Retículo Endoplasmático/metabolismo , Transporte Proteico , Fungos/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo
20.
Soft Matter ; 20(11): 2480-2490, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38385209

RESUMO

In active materials, uncoordinated internal stresses lead to emergent long-range flows. An understanding of how the behavior of active materials depends on mesoscopic (hydrodynamic) parameters is developing, but there remains a gap in knowledge concerning how hydrodynamic parameters depend on the properties of microscopic elements. In this work, we combine experiments and multiscale modeling to relate the structure and dynamics of active nematics composed of biopolymer filaments and molecular motors to their microscopic properties, in particular motor processivity, speed, and valency. We show that crosslinking of filaments by both motors and passive crosslinkers not only augments the contributions to nematic elasticity from excluded volume effects but dominates them. By altering motor kinetics we show that a competition between motor speed and crosslinking results in a nonmonotonic dependence of nematic flow on motor speed. By modulating passive filament crosslinking we show that energy transfer into nematic flow is in large part dictated by crosslinking. Thus motor proteins both generate activity and contribute to nematic elasticity. Our results provide new insights for rationally engineering active materials.


Assuntos
Modelos Biológicos , Proteínas Motores Moleculares , Proteínas Motores Moleculares/química , Citoesqueleto/metabolismo , Cinesinas/metabolismo , Elasticidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...