Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.988
Filter
1.
Oncol Res ; 32(7): 1221-1229, 2024.
Article in English | MEDLINE | ID: mdl-38948025

ABSTRACT

At present, the role of many long non-coding RNAs (lncRNAs) as tumor suppressors in the formation and development of cervical cancer (CC) has been studied. However, lncRNA prostate cancer gene expression marker 1 (PCGEM1), whose high expression not only aggravates ovarian cancer but also can induce tumorigenesis and endometrial cancer progression, has not been studied in CC. The objective of this study was to investigate the expression and the underlying role of PCGEM1 in CC. The relative expression of PCGEM1 in CC cells was detected by real-time PCR. After the suppression of PCGEM1 expression by shRNA, the changes in the proliferation, migration, and invasion capacities were detected via CCK-8 assay, EdU assay, and colony formation assay wound healing assay. Transwell assay and the changes in expressions of epithelial-to-mesenchymal transition (EMT) markers were determined by western blot and immunofluorescence. The interplay among PCGEM1, miR-642a-5p, and kinesin family member 5B (KIF5B) was confirmed by bioinformatics analyses and luciferase reporter assay. Results showed that PCGEM1 expressions were up-regulated within CC cells. Cell viabilities, migration, and invasion were remarkably reduced after the suppression of PCGEM1 expression by shRNA in Hela and SiHa cells. N-cadherin was silenced, but E-cadherin expression was elevated by sh-PCGEM1. Moreover, by sponging miR-642a-5p in CC, PCGEM1 was verified as a competitive endogenous RNA (ceRNA) that modulates KIF5B levels. MiR-642a-5p down-regulation partially rescued sh-PCGEM1's inhibitory effects on cell proliferation, migration, invasion, and EMT process. In conclusion, the PCGEM1/miR-642a-5p/KIF5B signaling axis might be a novel therapeutic target in CC. This study provides a research basis and new direction for targeted therapy of CC.


Subject(s)
Cell Movement , Cell Proliferation , Disease Progression , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Kinesins , MicroRNAs , RNA, Long Noncoding , Uterine Cervical Neoplasms , Humans , RNA, Long Noncoding/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , MicroRNAs/genetics , Female , Kinesins/genetics , Kinesins/metabolism , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Cell Movement/genetics , Cell Line, Tumor , HeLa Cells , Neoplasm Invasiveness
2.
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
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.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-39000337

ABSTRACT

Few efficacious treatment options are available for patients with small cell lung carcinoma (SCLC), indicating the need to develop novel therapeutic approaches. In this study, we explored kinesin family member 11 (KIF11), a potential therapeutic target in SCLC. An analysis of publicly available data suggested that KIF11 mRNA expression levels are significantly higher in SCLC tissues than in normal lung tissues. When KIF11 was targeted by RNA interference or a small-molecule inhibitor (SB743921) in two SCLC cell lines, Lu-135 and NCI-H69, cell cycle progression was arrested at the G2/M phase with complete growth suppression. Further work suggested that the two cell lines were more significantly affected when both KIF11 and BCL2L1, an anti-apoptotic BCL2 family member, were inhibited. This dual inhibition resulted in markedly decreased cell viability. These findings collectively indicate that SCLC cells are critically dependent on KIF11 activity for survival and/or proliferation, as well as that KIF11 inhibition could be a new strategy for SCLC treatment.


Subject(s)
Cell Survival , Kinesins , Lung Neoplasms , Small Cell Lung Carcinoma , Humans , Kinesins/metabolism , Kinesins/genetics , Kinesins/antagonists & inhibitors , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Cell Line, Tumor , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/drug therapy , Cell Survival/drug effects , Cell Survival/genetics , Cell Proliferation , bcl-X Protein/metabolism , bcl-X Protein/genetics , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Benzamides , Quinazolines
5.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38829962

ABSTRACT

Two sets of motor proteins underpin motile cilia/flagella function. The axoneme-associated inner and outer dynein arms drive sliding of adjacent axoneme microtubule doublets to periodically bend the flagellum for beating, while intraflagellar transport (IFT) kinesins and dyneins carry IFT trains bidirectionally along the axoneme. Despite assembling motile cilia and flagella, IFT train speeds have only previously been quantified in immobilized flagella-mechanical immobilization or genetic paralysis. This has limited investigation of the interaction between IFT and flagellar beating. Here, in uniflagellate Leishmania parasites, we use high-frequency, dual-color fluorescence microscopy to visualize IFT train movement in beating flagella. We discovered that adhesion of flagella to a microscope slide is detrimental, reducing IFT train speed and increasing train stalling. In flagella free to move, IFT train speed is not strongly dependent on flagella beat type; however, permanent disruption of flagella beating by deletion of genes necessary for formation or regulation of beating showed an inverse correlation of beat frequency and IFT train speed.


Subject(s)
Flagella , Leishmania , Microtubules , Axoneme/metabolism , Axoneme/genetics , Biological Transport , Cilia/metabolism , Cilia/genetics , Dyneins/metabolism , Dyneins/genetics , Flagella/metabolism , Flagella/genetics , Kinesins/metabolism , Kinesins/genetics , Leishmania/cytology , Leishmania/genetics , Leishmania/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Microtubules/metabolism
6.
Genes (Basel) ; 15(6)2024 May 24.
Article in English | MEDLINE | ID: mdl-38927616

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disorder characterized by progressive damage to both upper and lower motor neurons. Genetic factors are known to play a crucial role in ALS, as genetic studies not only advance our comprehension of disease mechanisms but also help unravel the complex phenotypes exhibited by patients. To gain further insights into the genetic landscape of ALS in the Chinese population and explore genotype-phenotype correlations among individuals, we conducted whole-genome sequencing to screen genes in 34 Chinese familial ALS (FALS) probands lacking the most common ALS-associated genes. Within this cohort, we identified a rare heterozygous missense mutation in the N-terminal domain of KIF5A (c.86A>G) in one of the probands. This finding is significant as mutations in the KIF5A gene have been implicated in ALS in European cohorts since 2018, predominantly characterized by C-terminal mutations. Analysis of the clinical phenotype within this familial lineage revealed a delayed onset of symptoms, an extended survival duration, and initial manifestations in both upper limbs. These observations underscore the clinical heterogeneity observed in ALS patients harboring KIF5A mutations. In conclusion, our study contributes to the growing body of evidence linking KIF5A to ALS and enhances our understanding of the intricate genetic landscape of this disease.


Subject(s)
Amyotrophic Lateral Sclerosis , Kinesins , Mutation, Missense , Whole Genome Sequencing , Adult , Aged , Female , Humans , Male , Middle Aged , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , China , East Asian People/genetics , Kinesins/genetics , Mutation , Pedigree , Phenotype
7.
Sci Adv ; 10(25): eadl6153, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38896608

ABSTRACT

Platelet-producing megakaryocytes (MKs) primarily reside in the bone marrow, where they duplicate their DNA content with each cell cycle resulting in polyploid cells with an intricate demarcation membrane system. While key elements of the cytoskeletal reorganizations during proplatelet formation have been identified, what initiates the release of platelets into vessel sinusoids remains largely elusive. Using a cell cycle indicator, we observed a unique phenomenon, during which amplified centrosomes in MKs underwent clustering following mitosis, closely followed by proplatelet formation, which exclusively occurred in G1 of interphase. Forced cell cycle arrest in G1 increased proplatelet formation not only in vitro but also in vivo following short-term starvation of mice. We identified that inhibition of the centrosomal protein kinesin family member C1 (KIFC1) impaired clustering and subsequent proplatelet formation, while KIFC1-deficient mice exhibited reduced platelet counts. In summary, we identified KIFC1- and cell cycle-mediated centrosome clustering as an important initiator of proplatelet formation from MKs.


Subject(s)
Blood Platelets , Cell Cycle , Centrosome , Kinesins , Megakaryocytes , Centrosome/metabolism , Animals , Megakaryocytes/metabolism , Megakaryocytes/cytology , Mice , Blood Platelets/metabolism , Kinesins/metabolism , Kinesins/genetics , Mice, Knockout , Humans , Mitosis
8.
BMC Gastroenterol ; 24(1): 205, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890649

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. Hepatitis B virus (HBV) is one of the major causes of liver cirrhosis (LC) and HCC. Therefore, the discovery of common markers for hepatitis B or LC and HCC is crucial for the prevention of HCC. METHODS: Expressed genes for to chronic active hepaititis B (CAH-B), LC and HCC were obtained from the GEO and TCGA databases, and co-expressed genes were screened using Protein-protein interaction (PPI) networks, least absolute shrinkage and selection operator (LASSO), random forest (RF) and support vector machine - recursive feature elimination (SVM-RFE). The prognostic value of genes was assessed using Kaplan-Meier (KM) survival curves. Columnar line plots, calibration curves and receiver operating characteristic (ROC) curves of individual genes were used for evaluation. Validation was performed using GEO datasets. The association of these key genes with HCC clinical features was explored using the UALCAN database ( https://ualcan.path.uab.edu/index.html ). RESULTS: Based on WGCNA analysis and TCGA database, the co-expressed genes (565) were screened. Moreover, the five algorithms of MCODE (ClusteringCoefficient, MCC, Degree, MNC, and DMNC) was used to select one of the most important and most closely linked clusters (the top 50 genes ranked). Using, LASSO regression model, RF model and SVM-RFE model, four key genes (UBE2T, KIF4A, CDCA3, and CDCA5) were identified for subsequent research analysis. These 4 genes were highly expressed and associated with poor prognosis and clinical features in HCC patients. CONCLUSION: These four key genes (UBE2T, KIF4A, CDCA3, and CDCA5) may be common biomarkers for CAH-B and HCC or LC and HCC, promising to advance our understanding of the molecular basis of CAH-B/LC/HCC progression.


Subject(s)
Carcinoma, Hepatocellular , Cell Cycle Proteins , Computational Biology , Kinesins , Liver Cirrhosis , Liver Neoplasms , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Humans , Kinesins/genetics , Liver Cirrhosis/genetics , Computational Biology/methods , Cell Cycle Proteins/genetics , Prognosis , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/complications , Biomarkers, Tumor/genetics , Protein Interaction Maps/genetics , Male , Kaplan-Meier Estimate , Support Vector Machine
9.
J Cell Biol ; 223(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38869473

ABSTRACT

At each cell division, nanometer-scale motors and microtubules give rise to the micron-scale spindle. Many mitotic motors step helically around microtubules in vitro, and most are predicted to twist the spindle in a left-handed direction. However, the human spindle exhibits only slight global twist, raising the question of how these molecular torques are balanced. Here, we find that anaphase spindles in the epithelial cell line MCF10A have a high baseline twist, and we identify factors that both increase and decrease this twist. The midzone motors KIF4A and MKLP1 are together required for left-handed twist at anaphase, and we show that KIF4A generates left-handed torque in vitro. The actin cytoskeleton also contributes to left-handed twist, but dynein and its cortical recruitment factor LGN counteract it. Together, our work demonstrates that force generators regulate twist in opposite directions from both within and outside the spindle, preventing strong spindle twist during chromosome segregation.


Subject(s)
Anaphase , Kinesins , Microtubules , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Kinesins/metabolism , Kinesins/genetics , Microtubules/metabolism , Dyneins/metabolism , Dyneins/genetics , Torque , Chromosome Segregation , Actin Cytoskeleton/metabolism , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/genetics
10.
Mol Omics ; 20(6): 417-429, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38940931

ABSTRACT

Objective: this study evaluates the prognostic relevance of gene subtypes and the role of kinesin family member 2C (KIF2C) in lung cancer progression. Methods: high-expression genes linked to overall survival (OS) and progression-free interval (PFI) were selected from the TCGA-LUAD dataset. Consensus clustering analysis categorized lung adenocarcinoma (LUAD) patients into two subtypes, C1 and C2, which were compared using clinical, drug sensitivity, and immunotherapy analyses. A random forest algorithm pinpointed KIF2C as a prognostic hub gene, and its functional impact was assessed through various assays and in vivo experiments. Results: The study identified 163 key genes and distinguished two LUAD subtypes with differing OS, PFI, pathological stages, drug sensitivity, and immunotherapy response. KIF2C, highly expressed in the C2 subtype, was associated with poor prognosis, promoting cancer cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), with knockdown reducing tumor growth in mice. Conclusion: The research delineates distinct LUAD subtypes with significant clinical implications and highlights KIF2C as a potential therapeutic target for personalized treatment in LUAD.


Subject(s)
Adenocarcinoma of Lung , Epithelial-Mesenchymal Transition , Kinesins , Lung Neoplasms , Kinesins/genetics , Kinesins/metabolism , Humans , Animals , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Mice , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Epithelial-Mesenchymal Transition/genetics , Prognosis , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Profiling , Xenograft Model Antitumor Assays
11.
Ecotoxicol Environ Saf ; 281: 116602, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944010

ABSTRACT

Di-(2-ethylhexyl) phthalate (DEHP) might led to chronic and long-term effects on human organs due to its widespread use and bioaccumulation. Despite some cohorts reporting an association between DEHP exposure and BPH, its underlying mechanisms have not been investigated. Our findings indicate that exposure to DEHP or MEHP (main metabolites of DEHP in the human body) leads to increased prostate weights, elevated prostate index, and notable epithelial thickening in rats. It has been observed to promote BPH-1 cell proliferation with effects ranging from low to high concentrations. Transcriptome sequencing analysis of rat prostate tissues identified KIF11 as the key hub gene. KIF11 is highly expressed after DEHP/MEHP exposure, and knocking down of KIF11 inhibits the MEHP-induced promotion of cell proliferation. Exposure to MEHP has been observed to increase the expression of p-GSK-3ß and elevate the levels of ß-catenin, thereby activating the Wnt/ß-catenin signaling pathway. Knocking down of KIF11 significantly inhibits these effects. Histone H3 at Lysine 27 acetylation (H3K27ac) is implicated in the upregulation of KIF11 expression, as evidenced by the addition of the acetylation inhibitor C646. In summary, our findings established that DEHP exposure could promote BPH through H3K27ac regulated KIF11/Wnt/ß-catenin signaling pathway.


Subject(s)
Diethylhexyl Phthalate , Kinesins , Prostatic Hyperplasia , Wnt Signaling Pathway , Male , Animals , Diethylhexyl Phthalate/toxicity , Prostatic Hyperplasia/chemically induced , Prostatic Hyperplasia/pathology , Wnt Signaling Pathway/drug effects , Kinesins/genetics , Kinesins/metabolism , Rats , Cell Proliferation/drug effects , Rats, Sprague-Dawley , Humans , beta Catenin/metabolism , beta Catenin/genetics , Prostate/drug effects , Prostate/pathology , Prostate/metabolism
12.
Dev Neurobiol ; 84(3): 203-216, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38830696

ABSTRACT

Formation of the corpus callosum (CC), anterior commissure (AC), and postoptic commissure (POC), connecting the left and right cerebral hemispheres, is crucial for cerebral functioning. Collapsin response mediator protein 2 (CRMP2) has been suggested to be associated with the mechanisms governing this formation, based on knockout studies in mice and knockdown/knockout studies in zebrafish. Previously, we reported two cases of non-synonymous CRMP2 variants with S14R and R565C substitutions. Among the, the R565C substitution (p.R565C) was caused by the novel CRMP2 mutation c.1693C > T, and the patient presented with intellectual disability accompanied by CC hypoplasia. In this study, we demonstrate that crmp2 mRNA could rescue AC and POC formation in crmp2-knockdown zebrafish, whereas the mRNA with the R566C mutation could not. Zebrafish CRMP2 R566C corresponds to human CRMP2 R565C. Further experiments with transfected cultured cells indicated that CRMP2 with the R566C mutation could not bind to kinesin light chain 1 (KLC1). Knockdown of klc1a in zebrafish resulted in defective AC and POC formation, revealing a genetic interaction with crmp2. These findings suggest that the CRMP2 R566C mutant fails to bind to KLC1, preventing axonal elongation and leading to defective AC and POC formation in zebrafish and CC formation defects in humans. Our study highlights the importance of the interaction between CRMP2 and KLC1 in the formation of the forebrain commissures, revealing a novel mechanism associated with CRMP2 mutations underlying human neurodevelopmental abnormalities.


Subject(s)
Intercellular Signaling Peptides and Proteins , Nerve Tissue Proteins , Zebrafish Proteins , Zebrafish , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Prosencephalon/metabolism , Kinesins/metabolism , Kinesins/genetics , Corpus Callosum/metabolism , Humans , Animals, Genetically Modified , Embryo, Nonmammalian
13.
World J Gastroenterol ; 30(20): 2689-2708, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38855156

ABSTRACT

BACKGROUND: The regulatory effects of KIF26B on gastric cancer (GC) have been confirmed, but the specific mechanism still needs further exploration. Pan-cancer analysis shows that the KIF26B expression is highly related to immune infiltration of cancer-associated fibroblasts (CAFs), and CAFs promote macrophage M2 polarization and affect cancers' progression. AIM: To investigate the regulatory functions of KIF26B on immune and metastasis of GC. METHODS: We analyzed genes' mRNA levels by quantitative real-time polymerase chain reaction. Expression levels of target proteins were detected by immunohistochemistry, ELISA, and Western blotting. We injected AGS cells into nude mice for the establishment of a xenograft tumor model and observed the occurrence and metastasis of GC. The degree of inflammatory infiltration in pulmonary nodes was observed through hematoxylin-eosin staining. Transwell and wound healing assays were performed for the evaluation of cell invasion and migration ability. Tube formation assay was used for detecting angiogenesis. M2-polarized macrophages were estimated by immunofluorescence and flow cytometry. RESULTS: KIF26B was significantly overexpressed in cells and tissues of GC, and the higher expression of KIF26B was related to GC metastasis and prognosis. According to in vivo experiments, KIF26B promoted tumor formation and metastasis of GC. KIF26B expression was positively associated with CAFs' degree of infiltration. Moreover, CAFs could regulate M2-type polarization of macrophages, affecting GC cells' migration, angiogenesis, invasion, and epithelial-mesenchymal transition process. CONCLUSION: KIF26B regulated M2 polarization of macrophage through activating CAFs, regulating the occurrence and metastasis of GC.


Subject(s)
Cancer-Associated Fibroblasts , Gene Expression Regulation, Neoplastic , Kinesins , Stomach Neoplasms , Animals , Female , Humans , Male , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Kinesins/metabolism , Kinesins/genetics , Macrophages/metabolism , Macrophages/immunology , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neovascularization, Pathologic , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology
14.
Discov Med ; 36(185): 1280-1288, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38926114

ABSTRACT

BACKGROUND: Kinesin family member 26B (KIF26B) has been closely linked to the occurrence and progression of various tumors. However, there is limited research on its role in oral squamous cell carcinoma (OSCC). This article aims to investigate the expression levels and mechanisms of KIF26B in OSCC. METHODS: Real time quantity polymerase chain reaction (RT-qPCR) and Western blot analyses were conducted to assess the expression levels of KIF26B in 35 OSCC specimens and their corresponding non-cancerous tissues. Overexpression and silencing of KIF26B were achieved in HSC6 and SCC25 cells, respectively, resulting in the establishment of KIF26B-overexpressing and si-KIF26B cell lines, designated as the KIF26B group and si-KIF26B group. Proliferation assays using 5-Ethynyl-2'-deoxyuridine (EdU) labeling and clone formation were performed to evaluate the proliferative capacity of cells in these groups. The invasive and migratory abilities of cells in the KIF26B and si-KIF26B groups were assessed using Transwell assay. Additionally, the influence of KIF26B on the glycogen synthase kinase (GSK)-3ß/ß-catenin pathway was investigated through Western blot analysis. RESULTS: According to the results of RT-qPCR and Western blot analyses, the expression of KIF26B was predominantly higher in OSCC tissues compared to normal tissues (p < 0.01). Overexpression of KIF26B notably accelerated cell migration, invasion, and proliferation (p < 0.01), whereas knockdown of KIF26B significantly inhibited these processes (p < 0.01). Additionally, KIF26B overexpression led to increased levels of active ß-catenin, p-GSK-3, and c-myc (p < 0.01), while KIF26B silencing decreased the levels of these proteins (p < 0.01). CONCLUSION: Our findings suggest that KIF26B may play a role in the pathogenesis and progression of OSCC as an oncogene. This study establishes a foundation for the identification of potential therapeutic targets for OSCC.


Subject(s)
Biomarkers, Tumor , Carcinoma, Squamous Cell , Cell Proliferation , Kinesins , Mouth Neoplasms , Humans , Kinesins/genetics , Kinesins/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Cell Line, Tumor , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Cell Proliferation/genetics , Female , Male , Middle Aged , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Cell Movement/genetics , Aged , Wnt Signaling Pathway/genetics , beta Catenin/metabolism , beta Catenin/genetics
15.
J Cell Sci ; 137(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38853670

ABSTRACT

Intraflagellar transport (IFT) is required for ciliary assembly. The IFT machinery comprises the IFT motors kinesin-2 and IFT dynein plus IFT-A and IFT-B complexes, which assemble into IFT trains in cilia. To gain mechanistic understanding of IFT and ciliary assembly, here, we performed an absolute quantification of IFT machinery in Chlamydomonas reinhardtii cilium. There are ∼756, ∼532, ∼276 and ∼350 molecules of IFT-B, IFT-A, IFT dynein and kinesin-2, respectively, per cilium. The amount of IFT-B is sufficient to sustain rapid ciliary growth in terms of tubulin delivery. The stoichiometric ratio of IFT-B:IFT-A:dynein is ∼3:2:1 whereas the IFT-B:IFT-A ratio in an IFT dynein mutant is 2:1, suggesting that there is a plastic interaction between IFT-A and IFT-B that can be influenced by IFT dynein. Considering diffusion of kinesin-2 during retrograde IFT, it is estimated that one kinesin-2 molecule drives eight molecules of IFT-B during anterograde IFT. These data provide new insights into the assembly of IFT trains and ciliary assembly.


Subject(s)
Chlamydomonas reinhardtii , Cilia , Dyneins , Flagella , Kinesins , Proteomics , Cilia/metabolism , Chlamydomonas reinhardtii/metabolism , Chlamydomonas reinhardtii/genetics , Proteomics/methods , Kinesins/metabolism , Kinesins/genetics , Dyneins/metabolism , Flagella/metabolism , Biological Transport
16.
Pediatr Transplant ; 28(5): e14804, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38853137

ABSTRACT

BACKGROUND: Kinesin family member 12 (KIF12) mutation-related cholestatic disorder represents a rare subtype of progressive familial intrahepatic cholestasis (PFIC), referred to as PFIC Type 8, with only 21 reported cases globally to date. METHODS: Here, we present a unique case of a 6-month-old boy diagnosed with homozygous KIF12 gene mutation, who successfully underwent a living donor liver transplant at our center for end-stage liver disease. RESULTS: This case marks the youngest patient of KIF12-related cholestatic disorder necessitating a liver transplant to date. The child initially presented with neonatal cholestasis and then developed infantile hepatic decompensation. Our report discusses the diagnostic process and management strategies employed. It underscores the importance of prompt diagnosis through clinical suspicion, biochemical parameters, and genetic testing, as well as the adoption of suitable management strategies, including the early contemplation of liver transplant in such exceptional and rare cases of genetic intrahepatic cholestasis. CONCLUSION: KIF12-related genetic disease should be considered in neonatal cholestasis cases with high gamma glutamyl transpeptidase to differentiate from conditions like biliary atresia. Favorable outcomes post liver transplant stress the importance of early genetic testing and referral to liver transplant centers for unresponsive patients, potentially saving lives.


Subject(s)
Cholestasis, Intrahepatic , End Stage Liver Disease , Kinesins , Liver Transplantation , Living Donors , Mutation , Humans , Male , Kinesins/genetics , Infant , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/surgery , End Stage Liver Disease/surgery , End Stage Liver Disease/genetics
17.
FASEB J ; 38(13): e23750, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38888878

ABSTRACT

Kif16A, a member of the kinesin-3 family of motor proteins, has been shown to play crucial roles in inducing mitotic arrest, apoptosis, and mitotic cell death. However, its roles during oocyte meiotic maturation have not been fully defined. In this study, we report that Kif16A exhibits unique accumulation on the spindle apparatus and colocalizes with microtubule fibers during mouse oocyte meiotic maturation. Targeted depletion of Kif16A using gene-targeting siRNA disrupts the progression of the meiotic cell cycle. Furthermore, Kif16A depletion leads to aberrant spindle assembly and chromosome misalignment in oocytes. Our findings also indicate that Kif16A depletion reduces tubulin acetylation levels and compromises microtubule resistance to depolymerizing drugs, suggesting its crucial role in microtubule stability maintenance. Notably, we find that the depletion of Kif16A results in a notably elevated incidence of defective kinetochore-microtubule attachments and the absence of BubR1 localization at kinetochores, suggesting a critical role for Kif16A in the activation of the spindle assembly checkpoint (SAC) activity. Additionally, we observe that Kif16A is indispensable for proper actin filament distribution, thereby impacting spindle migration. In summary, our findings demonstrate that Kif16A plays a pivotal role in regulating microtubule and actin dynamics crucial for ensuring both spindle assembly and migration during mouse oocyte meiotic maturation.


Subject(s)
Kinesins , Meiosis , Microtubules , Oocytes , Spindle Apparatus , Animals , Kinesins/metabolism , Kinesins/genetics , Meiosis/physiology , Oocytes/metabolism , Microtubules/metabolism , Mice , Spindle Apparatus/metabolism , Female , Actins/metabolism , Kinetochores/metabolism
18.
Cell Commun Signal ; 22(1): 312, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38902769

ABSTRACT

African American (AA) women are twice as likely to develop triple-negative breast cancer (TNBC) as women of European descent. Additionally, AA women with TNBC present a much more aggressive disease course than their European American (EA) counterparts. Thus, there is an unmet clinical need to identify race-specific biomarkers and improve survival outcomes in AA patients with TNBC. The minus-end directed microtubule motor protein kinesin family member C1 (KIFC1) promotes centrosome clustering and chromosomal instability and is often overexpressed in TNBC. Previous findings suggest that KIFC1 plays a role in cell proliferation and migration in TNBC cells from AAs and that the levels of nuclear KIFC1 (nKIFC1) are particularly high in AA patients with TNBC. The nuclear localization of KIFC1 in interphase may underlie its previously unrecognized race-specific association. In this study, we found that in TNBC cells derived from AAs, nKIFC1 interacted with the tumor suppressor myosin heavy chain 9 (MYH9) over EA cells. Treatment of AA TNBC cells with commercial inhibitors of KIFC1 and MYH9 disrupted the interaction between KIFC1 and MYH9. To characterize the racial differences in the KIFC1-MYH9-MYC axis in TNBC, we established homozygous KIFC1 knockout (KO) TNBC cell lines. KIFC1 KO significantly inhibited proliferation, migration, and invasion in AA TNBC cells but not in EA TNBC cells. RNA sequencing analysis showed significant downregulation of genes involved in cell migration, invasion, and metastasis upon KIFC1 KO in TNBC cell lines from AAs compared to those from EAs. These data indicate that mechanistically, the role of nKIFC1 in driving TNBC progression and metastasis is stronger in AA patients than in EA patients, and that KIFC1 may be a critical therapeutic target for AA patients with TNBC.


Subject(s)
Kinesins , Myosin Heavy Chains , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/ethnology , Triple Negative Breast Neoplasms/metabolism , Kinesins/genetics , Kinesins/metabolism , Female , Cell Line, Tumor , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Cell Proliferation/genetics , Cell Movement/genetics , Black or African American/genetics , White People/genetics , Protein Binding
19.
Kidney Blood Press Res ; 49(1): 410-429, 2024.
Article in English | MEDLINE | ID: mdl-38901404

ABSTRACT

INTRODUCTION: Vesicular transport (VT) has a complex relationship with tumor progression and immunity. But prognostic significance of VT in clear cell renal cell carcinoma (ccRCC) is unclear. Thus, we aimed to establish a prognostic model according to VT to predict overall survival of ccRCC patients. METHODS: We used patient data from TCGA database and built a prognostic model with 13 VT-related genes (VTRGs) by differential expression analysis, LASSO regression, and univariate/multivariate Cox analysis. The model was validated internally and externally, and survival analysis and ROC curves depicted excellent predictive ability. Furthermore, higher modeled riskscores corresponded to more advanced tumor progression. To further understand the potential reasons for different prognoses in patients, we did enrichment analysis on differentially expressed genes identified by the model in risk groups. The expression levels and roles of SAA1 and KIF18B in ccRCC were verified by qRT-PCR and cell function experiments. RESULTS: Humoral immune response and cAMP signaling pathway may be the biological processes and pathways leading to poor prognosis. Further analysis of immune microenvironment presented that ccRCC patients with poor prognoses had highly immune-infiltrated characteristics. We compared the drug response data of samples from different prognostic patients in the GDSC database and identified drug sensitivity differences associated with prognosis. Finally, we demonstrated that SAA1 and KIF18B could increase the proliferation, migration, and invasion ability of ccRCC cells using cellular experiments. CONCLUSION: In summary, we further revealed the importance of VTRGs in ccRCC prognosis.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/mortality , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Kidney Neoplasms/mortality , Prognosis , Female , Male , Tumor Microenvironment/genetics , Kinesins/genetics , Middle Aged , Clinical Relevance
20.
Nat Cell Biol ; 26(7): 1077-1092, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38926505

ABSTRACT

Cargo translocation across membranes is a crucial aspect of secretion. In conventional secretion signal peptide-equipped proteins enter the endoplasmic reticulum (ER), whereas a subset of cargo lacking signal peptides translocate into the ER-Golgi intermediate compartment (ERGIC) in a process called unconventional protein secretion (UcPS). The regulatory events at the ERGIC in UcPS are unclear. Here we reveal the involvement of ERGIC-localized small GTPases, Rab1 (Rab1A and Rab1B) and Rab2A, in regulating UcPS cargo transport via TMED10 on the ERGIC. Rab1 enhances TMED10 translocator activity, promoting cargo translocation into the ERGIC, whereas Rab2A, in collaboration with KIF5B, regulates ERGIC compartmentalization, establishing a UcPS-specific compartment. This study highlights the pivotal role of ERGIC-localized Rabs in governing cargo translocation and specifying the ERGIC's function in UcPS.


Subject(s)
Endoplasmic Reticulum , Golgi Apparatus , Protein Transport , Endoplasmic Reticulum/metabolism , Humans , Golgi Apparatus/metabolism , HeLa Cells , Kinesins/metabolism , Kinesins/genetics , HEK293 Cells , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Animals , rab1 GTP-Binding Proteins/metabolism , rab1 GTP-Binding Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...