kinesin superfamily
Recently Published Documents


TOTAL DOCUMENTS

163
(FIVE YEARS 24)

H-INDEX

40
(FIVE YEARS 2)

2021 ◽  
Vol 11 ◽  
Author(s):  
Yang Yang ◽  
Lanyang Gao ◽  
Ning-Na Weng ◽  
Jun-Jun Li ◽  
Jin Lu Liu ◽  
...  

BackgroundKinesin superfamily of proteins (KIFs) has been broadly reported to play an indispensable role in the biological process. Recently, emerging evidence reveals its oncogenic role in various cancers. However, the prognostic, oncological, and immunological values of KIFs have not been comprehensively explored in pancreatic ductal adenocarcinoma (PDAC) patients. We aimed to illustrate the relationship between KIFs and pancreatic ductal adenocarcinoma by using bioinformatical analysis.MethodsWe use GEPIA, Oncomine datasets, cBioPortal, LOGpc, TIMER, and STRING bioinformatics tools and web servers to investigate the aberrant expression, prognostic values, and oncogenic role of KIFs. The two-gene prognostic model and the correlation between KIFs and KRAS and TP53 mutation were performed using an R-based computational framework.ResultsOur results demonstrated that KIFC1/2C/4A/11/14/15/18A/18B/20B/23 (we name it prognosis-related KIFs) were upregulated and associated with unfavorable clinical outcome in pancreatic cancer patients. KIF21B overexpression is associated with better clinical outcome. The KIFC1/2C/4A/11/14/15/18A/18B/20B/23 profiles were significantly increased compared to grade 1 and grade 2/3. Besides, KIFC1/2C/4A/11/14/15/18A/18B/20B/23 was significantly associated with the mutation status of KRAS and TP53.Notably, most prognosis-related KIFs have strong correlations with tumor growth and myeloid-derived suppressor cells infiltration (MDSCs). A prognostic signature based on KIF20B and KIF21B showed a reliable predictive performance. Receiver operating characteristic (ROC) curve was employed to assess the predictive power of two-gene signature. Consequently, the gene set enrichment analysis (GSEA) showed that KIF20B and KIF21B’s overexpression was associated with the immunological and oncogenic pathway activation in pancreatic cancer. Finally, real-time quantitative PCR (RT-qPCR) was utilized to investigate the expression pattern of KIF20B and KIF21B in pancreatic cancer cell lines and normal pancreatic cell.ConclusionsKnowledge of the expression level of the KIFs may provide novel therapeutic molecular targets and potential prognostic biomarkers to pancreatic cancer patients.


2021 ◽  
Author(s):  
Huong T Vu ◽  
Zhechun Zhang ◽  
Riina Tehver ◽  
Dave Thirumalai

Many members in the kinesin superfamily walk predominantly towards the plus end of the microtubule (MT) in a hand-over-hand manner. Despite great progress in elucidating the mechanism of stepping kinetics, the origin of stepping directionality is not fully understood. To provide quantitative insights into this important issue, we represent the structures of conventional kinesin (Kin1), MT, and the Kin1-MT complex using the elastic network model, and calculate the residue-dependent responses to a local perturbation in these constructs. Fluctuations in the residues in the β domain of the α/β-tubulin are distinct from the α domain. Surprisingly, the Kin1-induced asymmetry, which is more pronounced in α/β-tubulin in the plus end of MT than in the minus end, propagates spatially across multiple α/β-tubulin dimers. Kin1 binding expands the MT lattice by mechanical stresses, resulting in a transition in the cleft of α/β tubulin dimer between a closed (CC for closed cleft) state (not poised for Kin1 to bind) to an open (OC for open cleft) binding competent state. The long-range asymmetric responses in the MT, leading to the creation of OC states with high probability in several α/β dimers on the plus end of the bound Kin1, is needed for the motor to take multiple steps towards the plus end of the MT. Reciprocally, kinesin binding to the MT stiffens the residues in the MT binding region, induces correlations between switches I and II in the motor, and enhances fluctuations in ADP and the residues in the binding pocket. Our findings explain both the directionality of stepping and MT effects on a key step in the catalytic cycle of Kin1.


2021 ◽  
Author(s):  
Lin Li ◽  
Jing Hao ◽  
Jin-Xiu Liu ◽  
Peng-Fei Wang ◽  
Chao-Fei Zhao ◽  
...  

Abstract Background KIF15, a member of kinesin superfamily proteins, has been found that play a of vital role in the carcinogenesis of various malignant tumor. But whether KIF15 can facilitate the evolution of prostate cancer (PCa) is still unknown. This study aims to explore its biological function in PCa cells and its relevance to prognosis and clinical features in PCa patients. Material and Methods KIF15 expression at mRNA and protein level in tumor and normal tissues was detected by quantitative real-time PCR (RT-PCR) and immunohistochemistry. Then the correlations between KIF15 expression and PCa patients’ clinical characteristics was analyzed. After inhibiting the expression of KIF15 by shRNA, the role of KIF15 on proliferative capacity of PCa was evaluated by using MTT assay. The function of KIF15 on metastatic potential of PCa was determined by using transwell assay. The prognostic value of KIF15 was determined by using bioinformatics analysis. Results Compared with normal tissues, KIF15 was overexpressed in PCa tissues. After knocking down KIF15 in C4-2 and Lncap cell lines, the proliferation (P < 0.001) and invasion (P < 0.001) capabilities of tumor cells are significantly reduced compared to the shCON group. The proliferation marker Ki67 and the metastasis-related marker MMP9 were also significantly reduced in two cell lines after silencing KIF15. Except that, increased KIF15 in tumor tissue is associated with clinical stage (P = 0.004), seminal vesicle invasion (P = 0.02), lymph node metastasis (P = 0.03), and poor disease-free survival (P < 0.05) in PCa patients. Conclusions The results proved that KIF15 might served as a prognostic factor and therapeutic target in prostate cancer, and play as a vital regulatory factor in tumorigenesis and cancer development of prostate cancer.


2021 ◽  
Author(s):  
Zehao Chen ◽  
Jie Wang ◽  
Jian Xu ◽  
Wenjie Zhu ◽  
Jianxin Jiang

Abstract BackgroundKinesin superfamily (KIFs) has a long-reported significant influence on the initiation, development, and progress of pancreatic adenocarcinoma (PAAD). However, the expression level of different KIFs in PAAD and its relationship with the prognosis and immune infiltration in patients with PAAD have not been fully elucidated.MethodsComprehensive bioinformatics analyses were done using data from UCSC XENA data hubs, TCGA database,Oncomine databases, GEO datasets, GEPIA, GTEx, The human protein atlas, Kaplan Meier plotter, cBioPortal, STRING and KEGG database. Then, the relationship between KIFs expression and tumor immune infiltrates was studied by using the TIMER database. ResultsA total of 24 differentially expressed KIFs at transcriptional levels were identified between tumor tissues and normal tissues with 1 (KIF1A) downregulated and 23 (KIF2A, KIF2C, KIF3A, KIF3B, KIF3C, KIF4A, KIF5B, KIF7, KIF9, KIF10, KIF11, KIF13A, KIF13B, KIF15, KIF16B, KIF18B, KIF20A, KIF20B, KIF21B, KIF22, KIF23, KIF26B, KIFC1) overexpressed by GEPIA. The protein expression level of KIF3A, KIF3B, KIF4A, KIF5B, KIF7, KIF9, CENPE, KIF11, KIF13A, KIF13B, KIF15, KIF16B, KIF18B, KIF20A, KIF20B, KIF21B, KIF22, KIF23, KIF26B, KIFC1 are higher in PAAD tissues than in the adjacent tissues, which is almost the same as the results of KIFs at transcriptional levels in PAAD. In addition, the expression levels of KIF1A, KIF1C, KIF3A, KIF5A, KIF5C, KIF6, KIF9, KIF13B, KIF19A, KIF21A, KIF24, KIF26B, KIFC1 are significantly correlated with the tumor stage of PAAD patients. As for prognosis value, the abnormal expression of KIF2C, KIF4A, KIF11, KIF15, KIF18B, KIF20A, KIF20B, KIF21B and KIF23 in PAAD patients is significantly related to the worse overall survival (OS) and relapse-free survival (RFS), and based on this, a 5-KIFs-based (KIF15, KIF20A, KIF20B, KIF21B, KIF23) risk score was generated by LASSO regression with a nomogram validated an accurate predictive efficacy. GO and KEGG enrichments revealed functions and pathways affected in PAAD. We also found that the expression of the 5-KIFs was significantly correlated with immune infiltrates, including B cells, CD8+ T cells, CD4+T cells, neutrophils, macrophages, and dendritic cells.ConclusionOur study may provide new insights into the choice of prognostic biomarkers and immunotherapy targets in PAAD patients.


2021 ◽  
Author(s):  
Valentina Gifford ◽  
Anna M Woskowicz ◽  
Noriko Ito ◽  
Stefan Balint ◽  
Michael Dustin ◽  
...  

MT1-MMP plays a crucial role in promoting the cellular invasion of cancer cells by degrading the extracellular matrix to create a path for migration. During this process, its localization at the leading edge of migrating cells is critical, and it is achieved by targeted transport of MT1-MMP-containing vesicles along microtubules by kinesin superfamily proteins (KIFs). Here we identified three KIFs involved in MT1-MMP vesicle transport: KIF3A, KIF13A, and KIF9. Knockdown of KIF3A and KIF13A effectively inhibited MT1-MMP-dependent collagen degradation and invasion, while knockdown of KIF9 increased collagen degradation and invasion. Our data suggest that KIF9 competes with KIF3A/KIF13A to bring MT1-MMP vesicles to different locations in the plasma membrane. Live-cell imaging analyses have indicated that KIF3A and KIF13A coordinate to transport the same MT1-MMP-containing vesicles. Taken together, we have identified a unique interplay between three KIFs to regulate leading edge localization of MT1-MMP and MT1-MMP-dependent cancer cell invasion.


Author(s):  
Kelly Hares ◽  
K. Kemp ◽  
S. Loveless ◽  
C. M. Rice ◽  
N. Scolding ◽  
...  

AbstractThere is increasing interest in the development of multiple sclerosis (MS) biomarkers that reflect central nervous system tissue injury to determine prognosis. We aimed to assess the prognostic value of kinesin superfamily motor protein KIF5A in MS by measuring levels of KIF5A in cerebrospinal fluid (CSF) combined with analysis of single nucleotide polymorphisms (SNPs; rs12368653 and rs703842) located within a MS susceptibility gene locus at chromosome 12q13–14 region. Enzyme-linked immunosorbent assay was used to measure KIF5A in CSF obtained from two independent biobanks comprising non-inflammatory neurological disease controls (NINDC), clinically isolated syndrome (CIS) and MS cases. CSF KIF5A expression was significantly elevated in progressive MS cases compared with NINDCs, CIS and relapsing–remitting MS (RRMS). In addition, levels of KIF5A positively correlated with change in MS disease severity scores (EDSS, MSSS and ARMSSS), in RRMS patients who had documented disease progression at 2-year clinical follow-up. Copies of adenine risk alleles (AG/AA; rs12368653 and rs703842) corresponded with a higher proportion of individuals in relapse at the time of lumbar puncture (LP), higher use of disease-modifying therapies post LP and shorter MS duration. Our study suggests that CSF KIF5A has potential as a predictive biomarker in MS and further studies into the potential prognostic value of analysing MS susceptibility SNPs should be considered.


Author(s):  
Nobutaka Hirokawa ◽  
Yosuke Tanaka ◽  
Reiko Takemura
Keyword(s):  

2020 ◽  
Vol Volume 12 ◽  
pp. 12769-12778
Author(s):  
Fei Zhao ◽  
Yunzhang Feng ◽  
Xueqiang Zhang ◽  
Xiaohui Liu ◽  
Aili Li

2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Yang Gao ◽  
Hui Zheng ◽  
Liangdong Li ◽  
Changshuai Zhou ◽  
Xin Chen ◽  
...  

Kinesin superfamily protein 3C (KIF3C), a motor protein of the kinesin superfamily, is expressed in the central nervous system (CNS). Recently, several studies have suggested that KIF3C may act as a potential therapeutic target in solid tumors. However, the exact function and possible mechanism of the motor protein KIF3C in glioma remain unclear. In this study, a variety of tests including CCK-8, migration, invasion, and flow cytometry assays, and western blot were conducted to explore the role of KIF3C in glioma cell lines (U87 and U251). We found that overexpression of KIF3C in glioma cell lines promoted cell proliferation, migration, and invasion and suppressed apoptosis, while silencing of KIF3C reversed these effects. Ectopic KIF3C also increased the expression of N-cadherin, vimentin, snail, and slug to promote the epithelial-mesenchymal transition (EMT). Mechanistically, overexpression of KIF3C increased the levels of phosphatidylinositol 3-kinase (PI3K) and phosphorylated protein kinase B (p-AKT). These responses were reversed by KIF3C downregulation or AKT inhibition. Our results indicate that KIF3C promotes proliferation, migration, and invasion and inhibits apoptosis in glioma cells, possibly by activating the PI3K/AKT pathway in vitro. KIF3C might act as a potential biomarker or therapeutic target for further basic research or clinical management of glioma.


Sign in / Sign up

Export Citation Format

Share Document