Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters











Publication year range
1.
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
2.
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
3.
Journal of Medical Research ; (12): 107-112, 2024.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1023607

ABSTRACT

Objective To analyze the expression level of kinesin family member 26B(KIF26B)in bladder cancer based on the pub-lic database,and to investigate the effect of silencing KIF26B on the proliferation,invasion and migration of bladder cancer cells.Methods The expression of KIF26B in bladder cancer and its relationship with survival and prognosis were analyzed based on GEO and UaLcan databases.Real-time quantitative polymerase chain reaction(RT-qPCR)and Western blot were used to detect the expression of KIF26B in bladder cancer cell lines(T24,J82)and normal bladder epithelial cells SV-HUV-1.si-KIF26B and si-NC fragments were transfected into T24 cells,and the effects of silencing KIF26B on the proliferation,invasion and migration of T24 cells were detected by methyl thiazolyl tetrazolium(MTT)assay,Transwell assay and scratch assay.Western blot was used to detect the expression levels of p-MEK,MEK,ERK and p-ERK proteins after the silencing of KIF26B.Results KIF26B was highly expressed in bladder cancer tis-sues,and the prognosis of patients with high expression of KIF26B was worse(P<0.05).The expression level of KIF26B in bladder cancer cell lines T24 and J82 was significantly higher than that in normal bladder epithelial cells SV-HUV-1(P<0.05).After the si-lencing of KIF26B gene,MTT,Transwell and scratch assay Results showed that the proliferation,invasion and migration ability of T24 cells were significantly decreased(P<0.05);After silencing KIF26B,the expression of p-MEK and p-ERK proteins in T24 cells was down-regulated(P<0.05),while MEK and ERK proteins had no significant changes(P>0.05).Conclusion KIF26B is highly ex-pressed in bladder cancer tissues and cells,which is associated with poor prognosis of patients.Silencing KIF26B can inhibit the prolifera-tion,invasion and migration of bladder cancer cells,and the mechanism may play a role through the MEK/ERK pathway.

4.
J Biochem ; 174(6): 561-570, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37793168

ABSTRACT

Mutations of proline-rich transmembrane protein 2 (PRRT2) lead to dyskinetic disorders such as paroxysmal kinesigenic dyskinesia (PKD), which is characterized by attacks of involuntary movements precipitated by suddenly initiated motion, and some convulsive disorders. Although previous studies have shown that PKD might be caused by cerebellar dysfunction, PRRT2 has not been sufficiently analyzed in some motor-related regions, including the basal ganglia, where dopaminergic neurons are most abundant in the brain. Here, we generated several types of Prrt2 knock-in (KI) mice harboring mutations, such as c.672dupG, that mimics the human pathological mutation c.649dupC and investigated the contribution of Prrt2 to dopaminergic regulation. Regardless of differences in the frameshift sites, all truncating mutations abolished Prrt2 expression within the striatum and cerebral cortex, consistent with previous reports of similar Prrt2 mutant rodents, confirming the loss-of-function nature of these mutations. Importantly, administration of l-dopa, a precursor of dopamine, exacerbated rotarod performance, especially in Prrt2-KI mice. These findings suggest that dopaminergic dysfunction in the brain by the PRRT2 mutation might be implicated in a part of motor symptoms of PKD and related disorders.


Subject(s)
Dopamine , Dystonia , Animals , Humans , Mice , Dystonia/genetics , Membrane Proteins/genetics , Mutation
5.
Cell Cycle ; 22(14-16): 1726-1742, 2023.
Article in English | MEDLINE | ID: mdl-37436127

ABSTRACT

BACKGROUND: The protein kinesin family member 26B (KIF26B) is aberrantly expressed in various cancers. However, its particular role and association with tumor immune infiltration in colon adenocarcinoma (COAD) remain unclear. METHODS: All original data were downloaded directly from The Cancer Genome Atlas (TCGA), UCSC Xena, and Gene Expression Omnibus (GEO) databases and processed with R 3.6.3. KIF26B expression was analyzed using Oncomine, TIMER, TCGA, GEO databases, and our clinical specimens. KIF26B expression at the protein level was explored with Human Protein Atlas (HPA) database. The upstream miRNAs and lncRNAs were predicted by StarBase and validated using RT-qPCR. Correlation of KIF26B expression with the expression of immune-related or immune checkpoint genes and GSEA analysis of KIF26B-related genes were investigated via R software. Relationship of KIF26B expression with immune biomarkers or tumor immune infiltration levels was studied through GEPIA2 and TIMER databases. RESULTS: KIF26B was upregulated, and its overexpression was closely related to overall survival (OS), disease-specific survival (DSS), progression-free interval (PFI), T stage, N stage, and CEA levels in COAD. MIR4435-2HG/hsa-miR-500a-3p/KIF26B axis was identified as the promising regulatory pathway of KIF26B. KIF26B expression was positively correlated with immune-related genes, tumor immune infiltration, and biomarker genes of immune cells in COAD, and KIF26B-related genes were significantly enriched in macrophage activation-related pathways. Expression of immune checkpoint genes, including PDCD1, CD274, and CTLA4, was also closely related to KIF26B expression. CONCLUSIONS: Our results clarified that ncRNA-based increased KIF26B expression was associated with a worse prognosis and high tumor immune infiltration in COAD.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Colonic Neoplasms/genetics , RNA, Untranslated , RNA, Long Noncoding/genetics , MicroRNAs/genetics , Kinesins/genetics
6.
FASEB J ; 36(11): e22606, 2022 11.
Article in English | MEDLINE | ID: mdl-36250931

ABSTRACT

Kinesin family member 26b (Kif26b) is essential for kidney development, and its deletion in mice leads to kidney agenesis. However, the roles of this gene in adult settings remain elusive. Thus, this study aims to investigate the role of Kif26b in the progression of renal fibrosis. A renal fibrosis model with adenine administration using Kif26b heterozygous mice and wild-type mice was established. Renal fibrosis and the underlying mechanism were investigated. The underlying pathways and functions of Kif26b were evaluated in an in vitro model using primary renal fibroblasts. Kif26b heterozygous mice were protected from renal fibrosis with adenine administration. Renal expressions of connective tissue growth factor (CTGF) and myofibroblast accumulation were reduced in Kif26b heterozygous mice. The expression of nonmuscle myosin heavy chain II (NMHCII), which binds to the C-terminus of Kif26b protein, was also suppressed in Kif26b heterozygous mice. The in vitro study revealed reduced expressions of CTGF, α-smooth muscle actin, and myosin heavy chain 9 (Myh9) via transfection with siRNAs targeting Kif26b in renal fibroblasts (RFB). RFBs, which were transfected by the expression vector of Kif26b, demonstrated higher expressions of these genes than non-transfected cells. Finally, Kif26b suppression and NMHCII blockage led to reduced abilities of migration and collagen gel contraction in renal fibroblasts. Taken together, Kif26b contributes to the progression of interstitial fibrosis via migration and myofibroblast differentiation through Myh9 in the renal fibrosis model. Blockage of this pathway at appropriate timing might be a therapeutic approach for renal fibrosis.


Subject(s)
Kidney , Kinesins , Myofibroblasts , Animals , Mice , Actins/genetics , Actins/metabolism , Adenine/metabolism , Collagen/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Fibroblasts/metabolism , Fibrosis , Kidney/metabolism , Kinesins/genetics , Myofibroblasts/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Cell Differentiation , Cell Movement
7.
J Microbiol Biotechnol ; 32(10): 1344-1354, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36224753

ABSTRACT

Laryngeal cancer is one of the highest incidence, most prevalently diagnosed head and neck cancers, making it critically necessary to probe effective targets for laryngeal cancer treatment. Here, real-time quantitative reverse transcription PCR (qRT-PCR) and western blot analysis were used to detect gene expression levels in laryngeal cancer cell lines. Fluorescence in situ hybridization (FISH) and subcellular fractionation assays were used to detect the subcellular location. Functional assays encompassing Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), transwell and wound healing assays were performed to examine the effects of target genes on cell proliferation and migration in laryngeal cancer. The in vivo effects were proved by animal experiments. RNA-binding protein immunoprecipitation (RIP), RNA pulldown and luciferase reporter assays were used to investigate the underlying regulatory mechanisms. The results showed that KIF26B antisense RNA 1 (KIF26B-AS1) propels cell proliferation and migration in laryngeal cancer and regulates the toll-like receptor 4 (TLR4) signaling pathway. KIF26B-AS1 also recruits FUS to stabilize TLR4 mRNA, consequently activating the TLR4 signaling pathway. Furthermore, KIF26B-AS1 plays an oncogenic role in laryngeal cancer via upregulating TLR4 expression as well as the FUS/TLR4 pathway axis, findings which offer novel insight for targeted therapies in the treatment of laryngeal cancer patients.


Subject(s)
Laryngeal Neoplasms , MicroRNAs , RNA, Long Noncoding , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Toll-Like Receptor 4/genetics , MicroRNAs/genetics , In Situ Hybridization, Fluorescence , Cell Line, Tumor , Cell Proliferation/genetics , Signal Transduction , Cell Movement/genetics
8.
Int J Mol Sci ; 23(16)2022 Aug 16.
Article in English | MEDLINE | ID: mdl-36012474

ABSTRACT

The formation of pathological bone deposits within soft tissues, termed heterotopic ossification (HO), is common after trauma. However, the severity of HO formation varies substantially between individuals, from relatively isolated small bone islands through to extensive soft tissue replacement by bone giving rise to debilitating symptoms. The aim of this study was to identify novel candidate therapeutic molecular targets for severe HO. We conducted a genome-wide scan in men and women with HO of varying severity following hip replacement for osteoarthritis. HO severity was dichotomized as mild or severe, and association analysis was performed with adjustment for age and sex. We next confirmed expression of the gene encoded by the lead signal in human bone and in primary human mesenchymal stem cells. We then examined the effect of gene knockout in a murine model of osseous trans-differentiation, and finally we explored transcription factor phosphorylation in key pathways perturbed by the gene. Ten independent signals were suggestively associated with HO severity, with KIF26B as the lead. We subsequently confirmed KIF26B expression in human bone and upregulation upon BMP2-induced osteogenic differentiation in primary human mesenchymal stem cells, and also in a rat tendo-Achilles model of post-traumatic HO. CRISPR-Cas9 mediated knockout of Kif26b inhibited BMP2-induced Runx2, Sp7/Osterix, Col1A1, Alp, and Bglap/Osteocalcin expression and mineralized nodule formation in a murine myocyte model of osteogenic trans-differentiation. Finally, KIF26B deficiency inhibited ERK MAP kinase activation during osteogenesis, whilst augmenting p38 and SMAD 1/5/8 phosphorylation. Taken together, these data suggest a role for KIF26B in modulating the severity of post-traumatic HO and provide a potential novel avenue for therapeutic translation.


Subject(s)
Kinesins , Ossification, Heterotopic , Osteogenesis , Animals , Cell Differentiation/genetics , Female , Humans , Kinesins/genetics , Male , Mice , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism , Osteocalcin/metabolism , Osteogenesis/genetics , Rats
9.
Tissue Cell ; 76: 101750, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35182988

ABSTRACT

Kinesin family member 26B (KIF26B) is reported differently expressed in multiple neoplasms and exerts a pivotal role in carcinogenesis. To date, the relationship between KIF26B and non-small cell lung cancer (NSCLC) is unaddressed. This study explored the possible roles and mechanisms of KIF26B in NSCLC. We observed high levels of KIF26B in NSCLC, and demonstrated that high KIF26B levels predicted an overall shorter duration of survival. Functional experiments demonstrated that restraint of KIF26B by gene knockdown exhibited remarkable tumor-suppressive effects in NSCLC in vitro, including repression of cell proliferation, induction of G0/G1 cell cycle arrest, suppression of cell invasion and epithelial-mesenchymal transition, and enhancement of chemotherapeutic sensitivity. The study further revealed that inhibition of KIF26B was able to affect the activation of ß-catenin via regulation of the AKT/GSK-3ß axis. Moreover, forced expression of ß-catenin could reverse KIF26B-silencing-evoked tumor-suppressive effects. Importantly, NSCLC cells with KIF26B silencing exhibited decreased growth potential in nude mice in vivo. Collectively, our data indicate that restraint of KIF26B has a tumor-suppressive role in NSCLC by affecting the AKT/GSK-3ß/ß-catenin pathway. This work unveils a pivotal role of KIF26B in NSCLC and suggests it as a viable target for anti-NSCLC therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Lung Neoplasms/genetics , Mice , Mice, Nude , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism
10.
Biochem Biophys Res Commun ; 592: 125-133, 2022 02 12.
Article in English | MEDLINE | ID: mdl-35066304

ABSTRACT

Emerging evidence delineates that obesity, a complex metabolic disorder, impairs the structure and function of stromal cells residing in various tissues. The exuberant adipose tissue mass observed in obesity is, in part, associated with hyperplasia of adipocytes resulting from recruitment of multipotent stromal cells within the stromal vascular fraction of adipose tissues. However, a clear understanding of the causal role of stromal cells and biological factors in obesity is lacking. In our quest to understanding the role of kinesin family member 26B (KIF26B), we found that KIF26B regulates osteogenic and chondrogenic differentiation of stromal/progenitor cells. In this study, we sought to examine the effects of Kif26b loss-of-function on adipogenic differentiation of murine C3H10T1/2 multipotent stromal cells. In vitro loss-of-function studies demonstrated that Kif26b knockdown by lentivirus mediated shRNA markedly dampened the differentiation potential of C3H10T1/2 cells to adipocytes and suppressed the expression of adipogenesis-related genes e.g., Pparg, C/ebpα, Fabp4 and Adipoq. Analysis of cell cycle revealed that Kif26b knockdown resulted in elevated expression of cyclins (Ccnd1, Ccnb1, Ccna2) along with rapid cell cycle progression from G0/G1 to S and G2 phases. Mechanistically, reduced adipogenic differentiation of Kif26b-deficient cells was partly dependent on PPARγ, a key transcription factor implicated in adipogenesis. This observation was experimentally supported as loss of adipogenesis was partially rescued by the addition of PPARγ agonist, rosiglitazone in Kif26b-deficient cells. We further found that silencing of Kif26b lessened the protein levels of phospho-AKT(Ser473), phospho-S6(Ser235/236), and phospho-mTOR(Ser2448), the major component of AKT/mTOR complex 1 (mTORC1) signaling at the basal level. Together, these data define a novel role of Kif26b in regulating the commitment of C3H10T1/2 multipotent stromal cells to the adipocyte lineage and provide a practical framework for further experiments to establish its therapeutic potential for the treatment of problems associated with adipogenesis such as obesity at the cellular and molecular level.


Subject(s)
Adipogenesis , Kinesins/metabolism , Multipotent Stem Cells/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Cycle , Cell Line , Gene Knockdown Techniques , Mice , PPAR gamma/metabolism , Signal Transduction , Stromal Cells/metabolism
11.
J Bone Miner Res ; 37(2): 349-368, 2022 02.
Article in English | MEDLINE | ID: mdl-34787331

ABSTRACT

Ectopic calcification is an osteogenic process that leads to the formation of inappropriate bone within intra-articular soft tissues, often in response to injury or surgery. The molecular mechanisms governing this phenotype have yet to be determined. Using a population genetics approach, we identified an association of the kinesin superfamily member 26b (Kif26b) with injury-induced ectopic calcification through quantitative trait locus analysis of recombinant inbred mouse strains, consistent with a genomewide association study that identified KIF26B as a severity locus for ectopic calcification in patients with hip osteoarthritis. Despite these associations of KIF26B with ectopic calcification, its mechanistic role and functional implications have not yet been fully elucidated. Here, we aim to decipher the functional role of KIF26B in osseous and chondrogenic transdifferentiation of human and murine progenitor/stem cells and in a murine model of non-invasive injury-induced intra-articular ectopic calcification. We found that KIF26B ablation via lentivirus-mediated shRNA significantly arrested osteogenesis of progenitor/stem cells and suppressed the expression of typical osteogenic marker genes. Conversely, KIF26B loss-of-function increased chondrogenesis as demonstrated by enhanced Safranin-O staining and by the elevated expression of chondrogenic marker genes. Furthermore, cell function analysis revealed that KIF26B knockdown significantly decreased cell viability and proliferation and induced cellular apoptosis. Mechanistically, loss of osteogenesis was reverted by the addition of a Wnt agonist, SKL2001, demonstrating a role of KIF26B in canonical Wnt/ß-catenin signaling. Finally, intra-articular delivery of Kif26b shRNA in B6-129SF2/J mice significantly hampered the development of intra-articular ectopic calcification at 8 weeks after injury compared with mice treated with non-target scrambled shRNA. In summary, these observations highlight that KIF26B plays a crucial role in ectopic bone formation by repressing osteogenesis, but not chondrogenesis, potentially via modulating Wnt/ß-catenin signaling. These findings establish KIF26B as a critical determinant of the osteogenic process in pathologic endochondral bone formation and an actionable target for pharmacotherapy to mitigate ectopic calcification (and heterotopic ossification). © 2021 American Society for Bone and Mineral Research (ASBMR).


Subject(s)
Calcinosis/genetics , Cell Transdifferentiation , Kinesins , Osteogenesis , Stem Cells/cytology , Animals , Disease Models, Animal , Gene Silencing , Kinesins/genetics , Mice , Wnt Signaling Pathway
12.
J Ginseng Res ; 45(6): 734-743, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34764728

ABSTRACT

BACKGROUND: The underlying mechanisms of the potential tumor-suppressive effects of ginsenoside Rh2 are complex. N6-methyladenosine (m6A) RNA methylation is usually dysregulated in cancer. This study explored the regulatory effect of ginsenoside Rh2 on m6A RNA methylation in cancer.Methods: m6A RNA quantification and gene-specific m6A RIP-qPCR assays were applied to assess total and gene-specific m6A RNA levels. Co-immunoprecipitation, fractionation western blotting, and immunofluorescence staining were performed to detect protein interactions and distribution. QRT-PCR, dual-luciferase, and ChIP-qPCR assays were conducted to check the transcriptional regulation. RESULTS: Ginsenoside Rh2 reduces m6A RNA methylation and KIF26B expression in a dose-dependent manner in some cancers. KIF26B interacts with ZC3H13 and CBLL1 in the cytoplasm of cancer cells and enhances their nuclear distribution. KIF26B inhibition reduces m6A RNA methylation level in cancer cells. SRF bound to the KIF26B promoter and activated its transcription. SRF mRNA m6A abundance significantly decreased upon KIF26B silencing. SRF knockdown suppressed cancer cell proliferation and growth both in vitro and in vivo, the effect of which was partly rescued by KIF26B overexpression.Conclusion: ginsenoside Rh2 reduces m6A RNA methylation via downregulating KIF26B expression in some cancer cells. KIF26B elevates m6A RNA methylation via enhancing ZC3H13/CBLL1 nuclear localization. KIF26B-SRF forms a positive feedback loop facilitating tumor growth.

13.
Med Oncol ; 39(1): 15, 2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34817735

ABSTRACT

KIF26B is a member of the kinesin superfamily that is up-regulated in various tumors, including breast cancer (BC), which can promote tumor progression. This study aimed to investigate the potential function of KIF26B in BC, and the underlying mechanisms, focusing mainly on cell proliferation. KIF26B expression was examined in BC tissue samples obtained from 99 patients. Then, we performed MTS, EdU and flow cytometry assays to detect cell proliferation, and western blotting to measure the expression of cell cycle-related proteins in MDA-MB-231 and MDA-MB-468 cells following KIF26B knockdown. Promoter analysis was used to study the upstream regulatory mechanism of KIF26B. KIF26B was upregulated in BC tissues. High expression of KIF26B was associated with clinicopathological parameters, such as positive lymph node metastasis, higher tumor grade, and higher proliferative index in BC. Furthermore, knockdown of KIF26B expression inhibited MDA-MB-231 and MDA-MB-468 cell proliferation, arresting cells in the G1 phase of the cell cycle in vitro. Similarly, KIF26B silencing decreased the expression levels of Wnt, ß-catenin, and cell cycle-related proteins such as c-Myc, cyclin D1, and cyclin-dependent kinase 4, while increasing the expression of p27. Moreover, ELK1 could bind to the core promoter region of KIF26B and activate its transcription. KIF26B acts as an oncogene in BC by regulating multiple proteins involved in the cell cycle. ELK1 activates KIF26B transcription.


Subject(s)
Breast Neoplasms/genetics , Cell Cycle/genetics , Kinesins/genetics , ets-Domain Protein Elk-1/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Kinesins/metabolism , Middle Aged , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Transcriptional Activation , ets-Domain Protein Elk-1/genetics
14.
Iran J Public Health ; 50(2): 397-406, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33748005

ABSTRACT

BACKGROUND: KIF26B gene is found to play essential roles in regulating different aspects of cell proliferation and development of the nervous system. We aimed to determine if rs12407427 T/C polymorphism could affect susceptibility to schizophrenia (SZN) and breast cancer (BC), the two genetically correlated diseases. METHODS: The current case-control study was performed from Aug 2018 to Dec 2018. Briefly, 159 female pathologically confirmed BC cases referring to Alzahra Hospital, Isfahan, Iran, and 102 psychologically confirmed SZN patients (60 males and 42 females) admitted to Baharan Hospital, Zahedan, Iran, were enrolled. Using the salting-out method, genomic DNA was extracted, and variants were genotyped using allele-specific amplification refractory mutation system polymerase chain reaction (ARMS-PCR) method. RESULTS: The results revealed a significant association between the KIF26B rs12407427 codominant CT (P=0.001), CC (P=0.0001), dominant CT+CC, and recessive CC (P=0.001) genotypes with the risk of developing SZN. Significant correlations were also found regarding rs12407427 and BC susceptibility in different inheritance models, including over-dominant CT (P=0.026), dominant CT+CC (P=0.001), recessive CC (P=0.009), and codominant CT and CC (P=0.001) genotypes. The over-presence of the C allele was also correlated with an increased risk for SZN (P=0.0001) and BC (P=0.0001). Finally, computational analysis predicted that T/C variation in this polymorphism could change the binding sites in proteins involved in splicing. CONCLUSION: rs12407427 T/C as a de novo KIF26B variant might be a novel genetic biomarker for SZN and/or BC susceptibility in a sample of the Iranian population.

15.
Cancer Cell Int ; 19: 205, 2019.
Article in English | MEDLINE | ID: mdl-31388332

ABSTRACT

BACKGROUND: Kinesin family member 26B (KIF26B) is unveiled acted as important role in many solid tumors, however, the function of KIF26B in hepatocellular carcinoma (HCC) is unclear. METHODS: The expression of KIF26B in HCC tissues and cell lines were measured with immunochemistry, real-time PCR and western blotting. The correlation between KIF26B expression and clinicopathological characteristics were analyzed by SPSS19.0. Functional experiments of KIF26B was conducted by CCK-8, transwell, EDU, colony formation in vitro and tumorigenesis in vivo. The gene set enrichment analysis was used to search the downstream pathway, luciferase reporter experiment was used to find the upstream regulatory factor of KIF26B. RESULTS: In this study, we found that KIF26B was overexpressed both in HCC tissues and cell lines. High expression of KIF26B was associated with poor overall survival (OS), late TNM stage and poor differentiation. Loss of function experiments showed that suppression of KIF26B could inhibit cell viability, proliferation rate and invasion ability of HCC cells. KEGG and GO analysis showed that expression of KIF26B was highly relevant with PI3K/AKT signal pathway, and suppression of KIF26B could decrease the expression of m-TOR, p-PI3K and p-AKT. Further study demonstrated that expression of KIF26B was negative correlated with miR-450b-5p level in HCC tissues, and miR-450b-5p could inhibit cell viability, proliferation rate and invasion ability of HCC cells via targeted inhibiting KIF26B. CONCLUSION: Our study demonstrated that miR-450-5p/KIF26B/AKT axis is critical for progression of HCC, and might provide novel prognostic biomarker and therapeutic target for HCC.

16.
Int Ophthalmol ; 39(11): 2621-2628, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31077021

ABSTRACT

PURPOSE: Keratoconus (KTCN) is a congenital corneal eye disorder which correlates with abnormal distribution of the collagen fiber and causes loss of visual acuity. COLA4A gene has a substantive role in collagen synthesis, whereas KIF26B as a new candidate gene belonging to kinesin superfamily (KIFs) has been suggested to be associated with this disease. So, in this preliminary study, we simultaneously evaluated the effects of two single nucleotide polymorphisms, 222855rs7C/T and rs12407427C/T, on KTCN susceptibility in a sample of Iranian population. METHODS: The present case-control study consists of 144 patients confirmed with KTCN and 153 healthy controls. The variants are genotyped by using amplification refractory mutation system-polymerase chain reaction method. RESULTS: The findings disclosed that rs2228557C/T and rs12407427C/T polymorphisms significantly increased the risk of KTCN in measured (codominant1; p = 0.0001, codominant2; p = 0.0001, codominant3; p = 0.0006, dominant; p = 0.0001, over-dominant; p = 0.0005) and (codominant1; p = 0.0001, codominant3; p = 0.0005, recessive; p = 0.0001) inheritance patterns, respectively. CONCLUSION: Our results did prove a statistical association of both rs2228557 and rs12407427 genotypes (TT and CT + CC) and allele (T) with KTCN susceptibility in Iranian population. Further studies in other ethnicities are required to verify our results.


Subject(s)
Collagen Type IV/genetics , DNA/genetics , Genetic Predisposition to Disease , Keratoconus/genetics , Kinesins/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Child , Collagen Type IV/metabolism , Female , Gene Frequency , Genotype , Humans , Incidence , Iran/epidemiology , Keratoconus/epidemiology , Keratoconus/metabolism , Kinesins/metabolism , Male , Middle Aged , Risk Factors , Young Adult
17.
Biomed Pharmacother ; 108: 766-773, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30248545

ABSTRACT

BACKGROUND: Many studies have suggested that high KIF26B expression is directly linked to poor prognostic outcomes in breast cancer. However, the exact role of KIF26B in breast cancer progression is not fully understood. In this study, we aimed to explore the function and mechanism of KIF26B in breast cancer progression. METHODS: Quantitative real-time PCR and immunohistochemistry analysis were used to detect KIF26B expression in breast cancer cell lines and patient samples. Cell proliferation was assessed by CCK-8 assay, and cell migration and invasion were evaluated by wound healing assay and transwell assay. Western blot analysis was carried out to assess the underlying molecular mechanisms. Tumor formation and metastasis were determined by in vivo mouse experiments. RESULTS: KIF26B levels were significantly increased in breast cancer cells and patient samples. KIF26B level correlated with tumor size, TNM grade, and differentiation in patients with breast cancer. Overexpressing KIF26B in vitro promoted breast cancer cell proliferation and migration by activating FGF2/ERK signaling, while silencing KIF26B had the opposite effects. Similarly, KIF26B knockdown repressed tumor formation and metastasis in nude mice. CONCLUSION: KIF26B promoted the development and progression of breast cancer and might act as a potential therapeutic target for treating breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Fibroblast Growth Factor 2/metabolism , Kinesins/metabolism , MAP Kinase Signaling System/physiology , Signal Transduction/physiology , Animals , Breast Neoplasms/pathology , Cell Differentiation/physiology , Cell Line , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/physiology , Humans , MCF-7 Cells , Mice , Mice, Nude
18.
Am J Med Genet A ; 176(12): 2623-2629, 2018 12.
Article in English | MEDLINE | ID: mdl-30151950

ABSTRACT

KIF26B is a member of the kinesin superfamily with evolutionarily conserved functions in controlling aspects of embryogenesis, including the development of the nervous system, though its function is incompletely understood. We describe an infant with progressive microcephaly, pontocerebellar hypoplasia, and arthrogryposis secondary to the involvement of anterior horn cells and ventral (motor) nerves. We performed whole exome sequencing on the trio and identified a de novo KIF26B missense variant, p.Gly546Ser, in the proband. This variant alters a highly conserved amino acid residue that is part of the phosphate-binding loop motif and motor-like domain and is deemed pathogenic by several in silico methods. Functional analysis of the variant protein in cultured cells revealed a reduction in the KIF26B protein's ability to promote cell adhesion, a defect that potentially contributes to its pathogenicity. Overall, KIF26B may play a critical role in the brain development and, when mutated, cause pontocerebellar hypoplasia with arthrogryposis.


Subject(s)
Kinesins/genetics , Olivopontocerebellar Atrophies/genetics , Spinal Muscular Atrophies of Childhood/diagnosis , Spinal Muscular Atrophies of Childhood/genetics , Amino Acid Sequence , Animals , Brain/abnormalities , Brain/diagnostic imaging , Cell Adhesion , Disease Models, Animal , Gene Expression , Humans , Kinesins/chemistry , Magnetic Resonance Imaging/methods , Mice , Models, Molecular , Protein Conformation , Exome Sequencing
19.
Onco Targets Ther ; 11: 3195-3203, 2018.
Article in English | MEDLINE | ID: mdl-29881293

ABSTRACT

BACKGROUND: Kinesin family member 26B (KIF26B) plays a key role in the development and progression of many human cancers. However, the role and underlying mechanisms of KIF26B in breast cancer cells remain unknown. MATERIALS AND METHODS: In this study, we inhibited the expression of KIF26B in MDA-MB-231 and MCF-7 cells using lentivirus-delivered shRNA. RESULTS: Lentivirus-mediated KIF26B knockdown significantly suppressed cell proliferation, colony formation, migration, and invasion. Furthermore, cell cycle analyses revealed that the percentage of cells in the G0/G1 phase was significantly increased in KIF26B knockdown cells. Moreover, the knockdown of KIF26B significantly promoted cell apoptosis via the upregulation of cleaved caspase-3 and Bax. CONCLUSION: Our data indicate that KIF26B plays a pivotal role in tumor growth and metastasis in breast cancer cells and may be a potential therapeutic target for treating breast cancer.

20.
Bio Protoc ; 8(6)2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29770348

ABSTRACT

Noncanonical Wnt signaling functions independently of the ß-catenin pathway to control diverse developmental processes, and dysfunction of the pathway contributes to a number of human pathological conditions, including birth defects and metastatic cancer. Progress in the field, however, has been hampered by the scarcity of functional assays for measuring noncanonical Wnt signaling activity. We recently described the Wnt5a-Ror-Kif26b (WRK) reporter assay, which directly monitors a post-transcriptional regulatory event in noncanonical Wnt signaling. In this protocol, we describe the generation of the stable GFP-Kif26b reporter cell line and a quantitative reporter assay for detecting and measuring Wnt5a signaling activities in live cells via flow cytometry.

SELECTION OF CITATIONS
SEARCH DETAIL