Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters











Publication year range
1.
Int Immunopharmacol ; 131: 111613, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38489970

ABSTRACT

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


Subject(s)
MicroRNAs , RNA, Circular , Urinary Bladder Neoplasms , Animals , Humans , Mice , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Kinesins/genetics , Kinesins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , RNA, Circular/metabolism
2.
Sci Prog ; 106(1): 368504221148340, 2023.
Article in English | MEDLINE | ID: mdl-36594221

ABSTRACT

Kinesin family member 3A is an important motor protein that participates in various physiological and pathological processes, including normal tissue development, homeostasis maintenance, tumor infiltration, and migration. The wingless-related integration site/ß-catenin signaling pathway is essential for critical molecular mechanisms such as embryonic development, organogenesis, tissue regeneration, and carcinogenesis. Recent studies have examined the molecular mechanisms of kinesin family member 3A, among which the wingless-related integration site/ß-catenin signaling pathway has gained attention. The interaction between kinesin family member 3A and the wingless-related integration site/ß-catenin signaling pathway is compact and complex but is fascinating and worthy of further study. The upregulation and downregulation of kinesin family member 3A influence many diseases and patient survival through the wingless-related integration site/ß-catenin signaling pathway. Therefore, this review mainly focuses on describing the kinesin family member 3A and wingless-related integration site/ß-catenin signaling pathways and their associated diseases.


Subject(s)
Kinesins , beta Catenin , Humans , beta Catenin/genetics , beta Catenin/metabolism , Kinesins/genetics , Kinesins/metabolism , Wnt Signaling Pathway , Family
3.
J Mol Cell Biol ; 14(7)2022 09 27.
Article in English | MEDLINE | ID: mdl-35981808

ABSTRACT

Meiosis is essential for evolution and genetic diversity in almost all sexual eukaryotic organisms. The mechanisms of meiotic recombination, such as synapsis, have been extensively investigated. However, it is still unclear whether signals from the cytoplasm or even from outside of the cell can regulate the meiosis process. Cilia are microtubule-based structures that protrude from the cell surface and function as signaling hubs to sense extracellular signals. Here, we reported an unexpected and critical role of cilia during meiotic recombination. During gametogenesis of zebrafish, cilia were specifically present in the prophase stages of both primary spermatocytes and primary oocytes. By developing a germ cell-specific CRISPR/Cas9 system, we demonstrated that germ cell-specific depletion of ciliary genes resulted in compromised double-strand break repair, reduced crossover formation, and increased germ cell apoptosis. Our study reveals a previously undiscovered role for cilia during meiosis and suggests that extracellular signals may regulate meiotic recombination via this particular organelle.


Subject(s)
Cilia , Zebrafish , Animals , Male , Meiosis , Chromosome Pairing , DNA Repair
4.
Cell Mol Gastroenterol Hepatol ; 14(5): 1025-1051, 2022.
Article in English | MEDLINE | ID: mdl-35835391

ABSTRACT

BACKGROUND & AIMS: Efforts to characterize the signaling mechanisms that underlie gastroenteropancreatic neoplasms (GEP-NENs) are precluded by a lack of comprehensive models that recapitulate pathogenesis. Investigation into a potential cell-of-origin for gastrin-secreting NENs revealed a non-cell autonomous role for loss of menin in neuroendocrine cell specification, resulting in an induction of gastrin in enteric glia. Here, we investigated the hypothesis that cell autonomous Men1 inactivation in glial fibrillary acidic protein (GFAP)-expressing cells induced neuroendocrine differentiation and tumorigenesis. METHODS: Transgenic GFAPΔMen1 mice were generated by conditional GFAP-directed Men1 deletion in GFAP-expressing cells. Cre specificity was confirmed using a tdTomato reporter. GFAPΔMen1 mice were evaluated for GEP-NEN development and neuroendocrine cell hyperplasia. Small interfering RNA-mediated Men1 silencing in a rat enteric glial cell line was performed in parallel. RESULTS: GFAPΔMen1 mice developed pancreatic NENs, in addition to pituitary prolactinomas that phenocopied the human MEN1 syndrome. GFAPΔMen1 mice exhibited gastric neuroendocrine hyperplasia that coincided with a significant loss of GFAP expression. Men1 deletion induced loss of glial-restricted progenitor lineage markers and an increase in neuroendocrine genes, suggesting a reprogramming of GFAP+ cells. Deleting Kif3a, a mediator of Hedgehog signaling, in GFAP-expressing cells attenuated neuroendocrine hyperplasia by restricting the neuroendocrine cell fate. Similar results in the pancreas were observed when Sox10 was used to delete Men1. CONCLUSIONS: GFAP-directed Men1 inactivation exploits glial cell plasticity in favor of neuroendocrine differentiation.


Subject(s)
Cell Plasticity , Neuroglia , Animals , Mice , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Plasticity/genetics , Cell Plasticity/physiology , Gastrins , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Hedgehog Proteins , Hyperplasia/pathology , Multiple Endocrine Neoplasia Type 1/genetics , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Neuroendocrine Cells/physiology , Neuroglia/metabolism , Proto-Oncogene Proteins , RNA, Small Interfering
5.
Int Immunopharmacol ; 108: 108686, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35378445

ABSTRACT

Colorectal cancer (CRC) is a common digestive system malignancy characterized by the high morbidity and mortality rates. Due to the limited therapeutic options, the majority of CRC patients do not receive timely treatment, which has thus increased the risk of tumor recurrence. Long noncoding RNAs (lncRNAs), as the essential regulators, are involved in numerous pivotal physiological and pathological processes in CRC. By qRT-PCR, it could be discovered in this study that MIR600HG expression obviously increased in CRC tissues and cell lines compared with the normal group. Moreover, the high expression of MIR600HG was correlated with tumor size, tumor volume and TNM stage. Overexpression of MIR600HG affected the expression of downstream molecules, such as miR-144-3p and KIF3A, providing the possible mechanisms of MIR600HG in the development and progression of CRC.


Subject(s)
Colorectal Neoplasms , Kinesins , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , Kinesins/genetics , MicroRNAs/genetics , Neoplasm Recurrence, Local/genetics , RNA, Long Noncoding/genetics
6.
Matrix Biol ; 107: 1-23, 2022 03.
Article in English | MEDLINE | ID: mdl-35122963

ABSTRACT

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 motor 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 KIF3A/KIF13A dependent MT1-MMP vesicles transport takes over upon KIF9 knockdown. Live-cell imaging analyses have indicated that KIF3A and KIF13A coordinate to transport the same MT1-MMP-containing vesicles from the trans-Golgi to the endosomes, and KIF13A alone transports the vesicle from the endosome to the plasma membrane. 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.


Subject(s)
Kinesins , Matrix Metalloproteinase 14 , Cell Line, Tumor , Cell Movement , Endosomes/metabolism , Extracellular Matrix/metabolism , Humans , Kinesins/genetics , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Neoplasm Invasiveness
7.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166335, 2022 04 01.
Article in English | MEDLINE | ID: mdl-34973373

ABSTRACT

BACKGROUND & AIMS: Loss of primary cilia in epithelial cells is known to cause cystic diseases of the liver and kidney. We have previously shown that during experimental and human cirrhosis that primary cilia were predominantly expressed on biliary cells in the ductular reaction. However, the role of primary cilia in the pathogenesis of the ductular reaction is not fully understood. METHODS: Primary cilia were specifically removed in biliary epithelial cells (BECs) by the administration of tamoxifen to Kif3af/f;CK19CreERT mice at week 2 of a 20-week course of TAA treatment. Biliary progenitor cells were isolated and grown as organoids from gallbladders. Cells and tissue were analysed using histology, immunohistochemistry and Western blot assays. RESULTS: At the end of 20 weeks TAA administration, primary cilia loss in liver BECs resulted in multiple microscopic cystic lesions within an unaltered ductular reaction. These were not seen in control mice who did not receive TAA. There was no effect of biliary primary cilia loss on the development of cirrhosis. Increased cellular proliferation was seen within the cystic structures associated with a decrease in hepatocyte lobular proliferation. Loss of primary cilia within biliary organoids was initially associated with reduced cell passage survival but this inhibitory effect was diminished in later passages. ERK but not WNT signalling was enhanced in primary cilia loss-induced cystic lesions in vivo and its inhibition reduced the expansion of primary cilia deficient biliary progenitor cells in vitro. CONCLUSIONS: TAA-treated kif3a BEC-specific knockout mice had an unaltered progression to cirrhosis, but developed cystic lesions that showed increased proliferation.


Subject(s)
Cilia/pathology , Cysts/pathology , Kinesins/genetics , Liver Diseases/pathology , Animals , Biliary Tract/cytology , Cell Proliferation , Cilia/metabolism , Cysts/chemically induced , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Keratin-19/genetics , Keratin-19/metabolism , Kinesins/deficiency , Liver/metabolism , Liver/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Thioacetamide/toxicity
8.
Biochem Biophys Res Commun ; 584: 19-25, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34753064

ABSTRACT

The primary cilium is a sensory organelle at the cell surface with integral functions in cell signaling. It contains a microtubular axoneme that is rooted in the basal body (BB) and serves as a scaffold for the movement of intraflagellar transport (IFT) particles by Kinesin-2 along the cilium. Ift88, a member of the anterograde moving IFT-B1 complex, as well as the Kinesin-2 subunit Kif3a are required for cilia formation. To facilitate signaling, the cilium restricts the access of molecules to its membrane ("ciliary gate"). This is thought to be mediated by cytoskeletal barriers ("subciliary domains") originating from the BB subdistal/distal appendages, the periciliary membrane compartment (PCMC) as well as the transition fibers and zone (TF/TZ). The PCMC is a poorly characterized membrane domain surrounding the ciliary base with exclusion of certain apical membrane proteins. Here we describe that Ift88, but not Kinesin-2, is required for the establishment of the PCMC in MDCK cells. Likewise, in C. elegans mutants of the Ift88 ortholog osm-5 fail to establish the PCMC, while Kinesin-2 deficient osm-3 mutants form PCMCs normally. Furthermore, disruption of IFT-B1 into two subcomplexes, while disrupting ciliogenesis, does not interfere with PCMC formation. Our findings suggest that cilia are not a prerequisite for the formation of the PCMC, and that separate machineries with partially overlapping functions are required for the establishment of each.


Subject(s)
Cell Membrane/metabolism , Cilia/metabolism , Epithelial Cells/metabolism , Kinesins/metabolism , Membrane Transport Proteins/metabolism , Animals , Basal Bodies/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cytoskeleton/metabolism , Dogs , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Signal Transduction
9.
Reprod Toxicol ; 105: 136-147, 2021 10.
Article in English | MEDLINE | ID: mdl-34492310

ABSTRACT

Neurulation-stage alcohol exposure (NAE; embryonic day [E] 8-10) is associated with midline craniofacial and CNS defects that likely arise from disruption of morphogen pathways, such as Sonic hedgehog (Shh). Notably, midline anomalies are also a hallmark of genetic ciliopathies such as Joubert syndrome. We tested whether NAE alters Shh pathway signaling and the number and function of primary cilia, organelles critical for Shh pathway transduction. Female C57BL/6 J mice were administered two doses of alcohol (2.9 g/kg/dose) or vehicle on E9. Embryos were collected 6, 12, or 24 h later, and changes to Shh, cell cycle genes, and primary cilia were measured in the rostroventral neural tube (RVNT). Within the first 24 h post-NAE, reductions in Shh pathway and cell cycle gene expression and the ratio of Gli3 forms in the full-length activator state were observed. RVNT volume and cell layer width were reduced at 12 h. In addition, altered expression of multiple cilia-related genes was observed at 6 h post-NAE. As a further test of cilia gene-ethanol interaction, mice heterozygous for Kif3a exhibited perturbed behavior during adolescence following NAE compared to vehicle-treated mice, and Kif3a heterozygosity exacerbated the hyperactive effects of NAE on exploratory activity. These data demonstrate that NAE downregulates the Shh pathway in a region of the neural tube that gives rise to alcohol-sensitive brain structures and identifies disruption of primary cilia function, or a "transient ciliopathy", as a possible cellular mechanism of prenatal alcohol pathogenesis.


Subject(s)
Cilia/genetics , Ethanol/adverse effects , Hedgehog Proteins/genetics , Neural Tube/metabolism , Prenatal Exposure Delayed Effects/genetics , Animals , Behavior, Animal , Cell Cycle/genetics , Female , Gene Expression Regulation, Developmental , Kinesins/genetics , Male , Maternal-Fetal Exchange , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy
10.
Gene ; 798: 145795, 2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34175396

ABSTRACT

Spermiogenesis is the final phase of spermatogenesis, wherein the spermatids differentiate into mature spermatozoa via complex morphological transformation. In this process, kinesin plays an important role. Here, we observed the morphological transformation of spermatids and analyzed the characterization, dynamic transcription, and potential function of kinesin KIF3A/KIF3B during spermiogenesis in Chinese hook snout carp (Opsariichthys bidens). We found that the full-length cDNAs of O. bidens kif3a and kif3b were 2544 and 2806 bp in length comprising 119 bp and 259 bp 5' untranslated region (UTR), 313 bp and 222 bp 3' UTR, and 2112 bp and 2325 bp open reading frame encoding 703 and 774 amino acids, respectively. Ob-KIF3A/KIF3B proteins have three domains, namely N-terminal head, coiled-coil stalk, and C-terminal tail, and exhibit high similarity with homologous proteins in vertebrates and invertebrates. Ob-kif3a/kif3b mRNAs were ubiquitously expressed in all tissues examined, with the highest expression in the brain and stage-IV testis. Immunofluorescence results showed that Ob-KIF3A was co-localized with tubulin and the mitochondria. Particularly, in early spermatids, Ob-KIF3A, tubulin, and the mitochondrial signals were evenly distributed in the cytoplasm, whereas in middle spermatids, they were distributed around the nucleus. In the late stage, the signals were concentrated on one side of the nucleus, where the tail is formed, whereas in mature sperms, they were detected in the midpiece and flagellum. These results indicate that Ob-KIF3A/KIF3B may participate in nuclear reshaping, flagellum formation, and mitochondrial aggregation in the midpiece during spermiogenesis.


Subject(s)
Cyprinidae/physiology , Kinesins/physiology , Spermatogenesis/physiology , Animals , Cyprinidae/genetics , Kinesins/chemistry , Kinesins/genetics , Male , Microtubules/metabolism , Mitochondria/metabolism , Phylogeny , Protein Conformation , RNA, Messenger/metabolism , Sequence Alignment , Sequence Analysis, DNA , Sperm Tail/physiology , Spermatids/physiology , Spermatids/ultrastructure , Spermatogenesis/genetics , Testis/metabolism , Transcription, Genetic
11.
Neoplasia ; 23(1): 169-180, 2021 01.
Article in English | MEDLINE | ID: mdl-33341566

ABSTRACT

Primary cilium is a microtubule-based organelle that projects from the surfaces of most mammalian cell types and protrudes into the extracellular milieu as an antenna-like sensor to senses extracellular physical and biochemical signals, and then transmits signals into cytoplasm or nucleus to regulate numerous physical and developmental processes. Therefore, loss of primary cilia is associated to multiple cancer progression, including skin, breast, pancreas, ovarian, prostate, and kidney cancers. Our previous studies demonstrate that high prevalent loss of DAB2 Interacting Protein (DAB2IP) is associated with renal cell carcinoma, and we found a kinesin-like protein, kinesin family member 3A (KIF3a), was significantly increased in DAB2IP-interacting protein fraction. KIF3 is one of the most abundant kinesin-2 family proteins expressed in cells, and it is necessary for ciliogenesis. In this study, we observed that loss of DAB2IP in normal kidney epithelial cell significantly impair primary cilia formation. We unveiled a new mechanism of primary cilia stability via DAB2IP and KIF3a physical interaction at DAB2IP-PH domain. Furthermore, we found that KIF3a also act as a tumor suppressor in renal cell carcinoma, affect tumor development and patient survival.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cilia/metabolism , Kidney Neoplasms/etiology , Kidney Neoplasms/metabolism , ras GTPase-Activating Proteins/genetics , Animals , Cell Line , Cell Transformation, Neoplastic/metabolism , Disease Susceptibility , Humans , Kidney Neoplasms/pathology , Kinesins/genetics , Kinesins/metabolism , Mice , Protein Binding , Protein Processing, Post-Translational , ras GTPase-Activating Proteins/metabolism
12.
Cell Mol Gastroenterol Hepatol ; 11(5): 1251-1266, 2021.
Article in English | MEDLINE | ID: mdl-33347972

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori infection in humans typically begins with colonization of the gastric antrum. The initial Th1 response occasionally coincides with an increase in gastrin secretion. Subsequently, the gastritis segues to chronic atrophic gastritis, metaplasia, dysplasia and distal gastric cancer. Despite these well characterized clinical events, the link between inflammatory cytokines and non-cardia gastric cancer remains difficult to study in mouse models. Prior studies have demonstrated that overexpression of the Hedgehog (HH) effector GLI2 induces loss of gastrin (atrophy) and antral hyperplasia. To determine the link between specific cytokines, HH signaling and pre-neoplastic changes in the gastric antrum. METHODS: Mouse lines were created to conditionally direct IL1ß or IFN-γ to the antrum using the Gastrin-CreERT2 and Tet activator. Primary cilia, which transduces HH signaling, on G cells were disrupted by deleting the ciliary motor protein KIF3a. Phenotypic changes were assessed by histology and western blots. A subclone of GLUTag enteroendocrine cells selected for gastrin expression and the presence of primary cilia was treated with recombinant SHH, IL1ß or IFN-γ with or without kif3a siRNA. RESULTS: IFN-γ increased gastrin and induced antral hyperplasia. However, antral expression of IL1ß suppressed tissue and serum gastrin, while also inducing antral hyperplasia. IFN-γ treatment of GLUTAg cells suppressed GLI2 and induced gastrin, without affecting cilia length. By contrast, IL1ß treatment doubled primary cilia length, induced GLI2 and suppressed gastrin gene expression. Knocking down kif3a in GLUTAg cells mitigated SHH or IL1ß suppression of gastrin. CONCLUSIONS: Overexpression of IL1ß in the antrum was sufficient to induce antral hyperplasia coincident with suppression of gastrin via primary cilia. ORCID: #0000-0002-6559-8184.


Subject(s)
Cilia/pathology , Gastrins/metabolism , Helicobacter Infections/complications , Hyperplasia/pathology , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Pyloric Antrum/pathology , Animals , Antiviral Agents/pharmacology , Cilia/metabolism , Gastrins/genetics , Helicobacter Infections/microbiology , Helicobacter pylori/isolation & purification , Hyperplasia/etiology , Hyperplasia/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyloric Antrum/drug effects , Pyloric Antrum/metabolism , Pyloric Antrum/microbiology
13.
Glia ; 69(5): 1184-1203, 2021 05.
Article in English | MEDLINE | ID: mdl-33368703

ABSTRACT

Primary cilia are small microtubule-based organelles capable of transducing signals from growth factor receptors embedded in the cilia membrane. Developmentally, oligodendrocyte progenitor cells (OPCs) express genes associated with primary cilia assembly, disassembly, and signaling, however, the importance of primary cilia for adult myelination has not been explored. We show that OPCs are ciliated in vitro and in vivo, and that they disassemble their primary cilia as they progress through the cell cycle. OPC primary cilia are also disassembled as OPCs differentiate into oligodendrocytes. When kinesin family member 3a (Kif3a), a gene critical for primary cilium assembly, was conditionally deleted from adult OPCs in vivo (Pdgfrα-CreER™:: Kif3a fl/fl transgenic mice), OPCs failed to assemble primary cilia. Kif3a-deletion was also associated with reduced OPC proliferation and oligodendrogenesis in the corpus callosum and motor cortex and a progressive impairment of fine motor coordination.


Subject(s)
Adult Stem Cells , Oligodendrocyte Precursor Cells , Animals , Cell Differentiation , Cilia , Kinesins/genetics , Mice , Mice, Transgenic , Oligodendroglia
14.
FEBS Open Bio ; 11(5): 1487-1496, 2021 05.
Article in English | MEDLINE | ID: mdl-31774623

ABSTRACT

Bladder cancer is one of the most common malignant tumors of the urinary system, with high morbidity and mortality. At present, the survival rates and prognosis of patients with bladder cancer are still relatively low; thus, there remains a need to improve prognosis by identifying novel targets. Kinesins (kinesin superfamily proteins) are a series of microtubule-based motor proteins that mediate various types of cellular processes. Kinesin family member 3A (KIF3A) is critical for cytoplasm separation in mitosis, and it has been reported to be misexpressed in multiple types of cancer. However, its effects on the progression and development of bladder cancer remain unclear. Herein, we report that KIF3A is highly expressed in human bladder cancer. We identified a significant correlation between KIF3A and clinical features, including clinical stage (P = 0.047), pathological tumor status (P = 0.045), lymph node status (P = 0.041) and metastasis (P = 0.035). KIF3A expression was also correlated with poor prognosis of patients with bladder cancer. Our results further indicated that KIF3A ablation resulted in cell cycle arrest; blocked the proliferation, migration and invasion of bladder cancer cells in vitro; and restrained tumor growth in mice in a microtubule-dependent manner. In summary, our findings suggest that KIF3A is a potential therapeutic target for bladder cancer.


Subject(s)
Kinesins/metabolism , Urinary Bladder Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , China , Female , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Kinesins/genetics , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , Urinary Bladder Neoplasms/genetics , Xenograft Model Antitumor Assays
15.
Oncol Lett ; 20(6): 348, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33123259

ABSTRACT

Kinesin family member 3A (KIF3A) plays a crucial role in the carcinogenesis of different types of human cancer. The present study aimed to identify the role of KIF3A in the carcinogenesis of non-small cell lung cancer (NSCLC). KIF3A protein expression was determined in 163 patients with NSCLC using immunohistochemistry staining. The prognosis of patients with NSCLC was determined using Kaplan-Meier survival and Cox regression analyses. The function of KIF3A on the carcinogenesis and metastasis of NSCLC was determined in vitro. Furthermore, a protein-protein interaction (PPI) network of KIF3A was constructed and the potential interacting molecules were identified using bioinformatic analysis. The protein expression levels of KIF3A were significantly lower in the NSCLC tissues compared with that in the adjacent tissues, and low KIF3A expression level was associated with unfavorable survival outcomes in patients with NSCLC. Furthermore, KIF3A knockdown increased proliferation, invasion and metastasis, and inhibited apoptosis of NSCLC cells. KIF3A was demonstrated to interact with intraflagellar transport 57 (IFT57) in the PPI network. In addition, validation analyses indicated that KIF3A mRNA expression levels were positively correlated with IFT57 mRNA expression levels in clinical NSCLC samples and NSCLC cell lines. Taken together, the results of the present study suggested that KIF3A is a key tumor suppressor gene for carcinogenesis and metastasis of NSCLC, it may also function as a biomarker and interacts with IFT57 in the progression of NSCLC.

16.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(2): 219-224, 2020 Feb 29.
Article in Chinese | MEDLINE | ID: mdl-32376524

ABSTRACT

OBJECTIVE: To investigate the expression of KIF3A in mice with unilateral ureteral obstruction (UUO) and TGF-ß1-induced NRK-52E cells and the role of KIF3A in renal tubular epithelial cell transdifferentiation. METHODS: Thirty-six C57BL/6J mice were randomly divided into the sham group (n=18) and UUO group (n=18). Six mice in each group were sacrificed at 7, 14 and 21days after the operation. The degree of renal tubulointerstitial fibrosis of the mice was observed by HE staining, Masson trichrome staining and Sirius red staining. The expression and distribution of KIF3A in the kidney of the mice was detected using RT-PCR, Western blotting and immunohistochemistry. Western blotting was used to detect the expression of KIF3A, E-cadherin and α-SMA proteins in the renal tissue of the mice. The expressions of KIF3A, E-cadherin, α-SMA, Wnt4 and ß-catenin proteins in NRK-52E cells with TGF-ß1-induced transdifferentiation were detected by Western blotting. RESULTS: Compared with the sham-operated mice, the mice with UUO showed worsened renal interstitial fibrosis with the increase of obstruction time, indicating successful modeling. The expressions of KIF3A mRNA and protein increased progressively and reached the peaked level at 21 days after UUO. The expression of α-SMA protein was significantly increased while E-cadherin protein expression was significantly reduced after UUO. The transdifferentiated NRK-52E cells showed significantly increased expressions of KIF3A (P < 0.001), Wnt4 (P < 0.05) and ß-catenin proteins (P < 0.0001). CONCLUSIONS: KIF3A may participate in the development of renal fibrosis through epithelial-mesenchymal transition mediated by wnt/ß-catenin signaling pathway.


Subject(s)
Ureteral Obstruction , Animals , Fibrosis , Kidney , Kidney Diseases , Kinesins , Mice , Mice, Inbred C57BL , Transforming Growth Factor beta1
17.
J Mol Histol ; 51(3): 241-250, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32399704

ABSTRACT

Using a large-scale quantitative mesenchymal stem cells (MSCs) membrane proteomics analysis, we identified a large group of ciliary proteins in the MSCs membrane fraction, which prompted us to examine the cilia, intricate organelles that were originally discovered approximately 100 years ago. Here we characterize their primary structure and function in MSCs. We first characterized the primary cilia on undifferentiated human MSCs by immunostaining and verified these observation with scanning and 3D electronic microscopy. To investigate the function of the primary cilia of the MSCs we induced loss of function by means of siRNA knockdown (targeted to two known ciliary proteins: IFT172 and KIF3A). After either of these two proteins was knocked down by the respective siRNA, the MSCs showed fewer and shortened primary cilia. The MSCs proliferation assays showed increased cell proliferative activity under confluent conditions after the siRNA knockdown of IFT172 or KIF3A; among these MSCs, the proportion in S phase was increased in the IFT172 siRNA knockdown group. The expression of stem cell markers on the MSCs, namely, Oct4, Nanog and Sox2, were downregulated after the siRNA-induced knockdown of IFT172 or KIF3A, and the gene expression upregulation of ectoderm lineage markers was notable. Furthermore, manipulation of the primary cilia-dependent Shh pathway, using the Shh activator SAG (smoothened agonist), upregulated the gene expression of pluripotency markers, including Nanog and Oct4, and transcriptional target genes in the Shh pathway. This study confirms that MSCs have primary cilia and provides evidence that primary cilia-dependent signaling pathways play functional roles in MSCs proliferation and stemness maintenance.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Cilia/ultrastructure , Cytoskeletal Proteins/genetics , Kinesins/genetics , Mesenchymal Stem Cells/ultrastructure , Pluripotent Stem Cells/ultrastructure , Adaptor Proteins, Signal Transducing/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cells, Cultured , Cilia/genetics , Cilia/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression , Gene Knockdown Techniques , Humans , Kinesins/metabolism , Mesenchymal Stem Cells/metabolism , Microscopy, Electron, Transmission , Pluripotent Stem Cells/metabolism , Proteomics , RNA, Small Interfering/genetics
18.
Cell Tissue Res ; 381(3): 527-541, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32458081

ABSTRACT

The Wnt/ß-catenin pathway participates in many important physiological events such as cell proliferation and differentiation in the male reproductive system. We found that Kinesin-2 motor KIF3A is highly expressed during spermatogenesis in Eriocheir sinensis; it may potentially promote the intracellular transport of cargoes in this process. However, only a few studies have focused on the relationship between KIF3A and the Wnt/ß-catenin pathway in the male reproductive system of decapod crustaceans. In this study, we cloned and characterized the CDS of ß-catenin in E. sinensis for the first time. Fluorescence in situ hybridization and immunofluorescence results showed the colocalization of Es-KIF3A and Es-ß-catenin at the mRNA and the protein level respectively. To further explore the regulatory function of Es-KIF3A to the Wnt/ß-catenin pathway, the es-kif3a was knocked down by double-stranded RNA (dsRNA) in vivo and in primary cultured cells in testes of E. sinensis. Results showed that the expression of es-ß-catenin and es-dvl were decreased in the es-kif3a knockdown group. The protein expression level of Es-ß-catenin was also reduced and the location of Es-ß-catenin was changed from nucleus to cytoplasm in the late stage of spermatogenesis when es-kif3a was knocked down. Besides, the co-IP result demonstrated that Es-KIF3A could bind with Es-ß-catenin. In summary, this study indicates that Es-KIF3A can positively regulate the Wnt/ß-catenin pathway during spermatogenesis and Es-KIF3A can bind with Es-ß-catenin to facilitate the nuclear translocation of Es-ß-catenin.


Subject(s)
Kinesins/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Anomura , Female , Humans , Male , Mice , Spermatogenesis/physiology , Transfection
19.
Cancer Sci ; 111(4): 1422-1434, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32011034

ABSTRACT

Triple negative breast cancer (TNBC) displays higher heterogeneity, stronger invasiveness, higher risk of metastasis and poorer prognosis compared with major breast cancer subtypes. KIF3A, a member of the kinesin family of motor proteins, serves as a microtubule-directed motor subunit and has been found to regulate early development, ciliogenesis and tumorigenesis. To explore the expression, regulation and mechanism of KIF3A in TNBC, 3 TNBC cell lines, 98 cases of primary TNBC and paired adjacent tissues were examined. Immunohistochemistry, real-time PCR, western blot, flow cytometry, short hairpin RNA (shRNA) interference, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation techniques, transwell assays, scratch tests, and xenograft mice models were used. We found that KIF3A was overexpressed in TNBC and such high KIF3A expression was also associated with tumor recurrence and lymph node metastasis. Silencing of KIF3A suppressed TNBC cell proliferation by repressing the Rb-E2F signaling pathway and inhibited migration and invasion by repressing epithelial-mesenchymal transition. The tumor size was smaller and the number of lung metastatic nodules was lower in KIF3A depletion MDA-MB-231 cell xenograft mice than in the negative control group. In addition, KIF3A overexpression correlated with chemoresistance. These results suggested that high expression of KIF3A in TNBC was associated with the tumor progression and metastasis.


Subject(s)
E2F Transcription Factors/genetics , Kinesins/genetics , Retinoblastoma Binding Proteins/genetics , Triple Negative Breast Neoplasms/drug therapy , Ubiquitin-Protein Ligases/genetics , Aged , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinesins/antagonists & inhibitors , Mice , Middle Aged , Neoplasm Metastasis , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
20.
Biotechnol Lett ; 42(6): 905-916, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32048127

ABSTRACT

OBJECTIVE: Asthma is a chronic immune disease that has become a serious public health problem. The currently available medications are not ideal because of their limitations and side effects; hence, new target proteins and signaling cascades for precise and safe therapy treatment are needed. This work established an ovalbumin-induced asthma rat model and treated it with total flavonoid extract from the Xinjiang chamomile. The proteins that were differentially expressed in the chamomile extract-treated asthmatic rats and the asthma and healthy rat groups were identified using isobaric tagging followed by LC-MS/MS. Kyoto encyclopedia of genes and genomes pathway analysis of the differentially expressed proteins was performed. RESULTS: Pathways involved in purine metabolism, herpes simplex infection, and JNK phosphorylation and activation mediated by activated human TAK1 were enriched, indicating the intrinsic links between the mechanism of asthma development and treatment effects. Furthermore, we constructed a protein-protein interaction network and identified KIF3A as a potential target protein of chamomile extract that affected the Hedgehog signaling pathway. CONCLUSIONS: This study may provide new insights into the pathogenesis of asthma and reveal several proteins and pathways that could be exploited to develop novel treatment approaches.


Subject(s)
Asthma/metabolism , Chamomile/chemistry , Flavonoids/pharmacology , Proteome/drug effects , Animals , Hedgehog Proteins/metabolism , Kinesins/metabolism , Lung/chemistry , Lung/drug effects , Lung/metabolism , Plant Extracts/pharmacology , Protein Interaction Maps/drug effects , Proteomics , Rats , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL