Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Biomed Res ; : 1-14, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38828848

ABSTRACT

Although the p21-activated kinase 2 (PAK2) is an essential serine/threonine protein kinase, its role in lung squamous cell carcinoma (LUSC) progression has yet to be fully understood. We analyzed PAK2 mRNA levels and DNA copy numbers as well as protein levels by quantitative real-time PCR and immunohistochemical staining, respectively, in human LUSC tissues and adjacent normal tissues. Then, we used colony formation assays, cell counting kit-8 assays, matrigel invasion assays, wound healing assays and xenograft models in nude mice to investigate the functions of PAK2 in LUSC progression. We demonstrated that the mRNA levels, DNA copy numbers, and protein levels of PAK2 were up-regulated in human LUSC tissues than in adjacent normal tissues. In addition, a higher PAK2 expression was correlated with a poorer prognosis in LUSC patients. In the in vitro study, we found that PAK2 promoted cell growth, migration, invasion, EMT process, and cell morphology regulation in LUSC cells. Furthermore, PAK2 enhanced tumor cell proliferation, migration, and invasion by regulating actin dynamics through the LIMK1/cofilin signaling. Our findings implicated that the PAK2/LIMK1/cofilin signaling pathway is likely a potential clinical marker and therapeutic target for LUSC.

2.
BMC Genomics ; 24(1): 217, 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37098483

ABSTRACT

BACKGROUND: Small cell lung cancer (SCLC) is a highly lethal malignant tumor. It accounts for approximately 15% of newly diagnosed lung cancers. Long non-coding RNAs (lncRNAs) can regulate gene expression and contribute to tumorigenesis through interactions with microRNAs (miRNAs). However, there are only a few studies reporting the expression profiles of lncRNAs, miRNAs, and mRNAs in SCLC. Also, the role of differentially expressed lncRNAs, miRNAs, and mRNAs in relation to competitive endogenous RNAs (ceRNA) network in SCLC remain unclear. RESULTS: In the present study, we first performed next generation sequencing (NGS) with six pairs of SCLC tumors and adjacent non-cancerous tissues obtained from SCLC patients. Overall, 29 lncRNAs, 48 miRNAs, and 510 mRNAs were found to be differentially expressed in SCLC samples (|log2[fold change] |> 1; P < 0.05). Bioinformatics analysis was performed to predict and construct a lncRNA-miRNA-mRNA ceRNA network, which included 9 lncRNAs, 11 miRNAs, and 392 mRNAs. Four up-regulated lncRNAs and related mRNAs in the ceRNA regulatory pathways were selected and validated by quantitative PCR. In addition, we examined the role of the most upregulated lncRNA, TCONS_00020615, in SCLC cells. We found that TCONS_00020615 may regulate SCLC tumorigenesis through the TCONS_00020615-hsa-miR-26b-5p-TPD52 pathway. CONCLUSIONS: Our study provided the comprehensive analysis of the expression profiles of lncRNAs, miRNAs, and mRNAs of SCLC tumors and adjacent non-cancerous tissues. We constructed the ceRNA networks which may provide new evidence for the underlying regulatory mechanism of SCLC. We also found that the lncRNA TCONS_00020615 may regulate the carcinogenesis of SCLC.


Subject(s)
Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Small Cell Lung Carcinoma , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Small Cell Lung Carcinoma/genetics , Gene Regulatory Networks , RNA, Messenger/genetics , RNA, Messenger/metabolism , Lung Neoplasms/genetics , Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics
3.
Exp Biol Med (Maywood) ; 247(18): 1619-1629, 2022 09.
Article in English | MEDLINE | ID: mdl-35861209

ABSTRACT

As a major co-factor of F-actin depolymerization, WD-repeat domain 1 (WDR1) affects the cellular microenvironment by cytoskeleton remodeling, thereby influencing cell molecular behavior. Our previous study showed that WDR1 activates YAP (Yes-associated protein) signaling in non-small-cell lung cancer (NSCLC) cells, but the mechanism remains unclear. We discovered that knockdown WDR1 in NSCLC cells decreased the expression of YAP and the nucleus-to-cytoplasm ratio. Disruption of cortical stress by drugs significantly inhibited YAP nuclear trafficking and enhanced YAP phosphorylation. In WDR1-knockdown NSCLC cells, inhibition of Hippo pathway reduced the nuclear exclusion of YAP and phosphorylated YAP. Our data suggest that WDR1-mediated cortical stress might be involved in regulating YAP signaling, thereby affecting the proliferation and migration of NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Actins/metabolism , Lung Neoplasms/metabolism , YAP-Signaling Proteins , Cell Line, Tumor , Cell Proliferation/physiology , Tumor Microenvironment , Microfilament Proteins/physiology
4.
Mol Cells ; 43(8): 749-762, 2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32868491

ABSTRACT

The migration, dedifferentiation, and proliferation of vascular smooth muscle cells (VSMCs) are responsible for intimal hyperplasia, but the mechanism of this process has not been elucidated. WD repeat domain 1 (WDR1) promotes actin-depolymerizing factor (ADF)/cofilin-mediated depolymerization of actin filaments (F-actin). The role of WDR1 in neointima formation and progression is still unknown. A model of intimal thickening was constructed by ligating the left common carotid artery in Wdr1 deletion mice, and H&E staining showed that Wdr1 deficiency significantly inhibits neointima formation. We also report that STAT3 promotes the proliferation and migration of VSMCs by directly promoting WDR1 transcription. Mechanistically, we clarified that WDR1 promotes the proliferation and migration of VSMCs and neointima formation is regulated by the activation of the JAK2/STAT3/WDR1 axis.


Subject(s)
Microfilament Proteins/deficiency , Animals , Carotid Arteries/cytology , Carotid Arteries/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Janus Kinase 2/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Neointima/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , WD40 Repeats
5.
Epigenomics ; 12(8): 685-700, 2020 04.
Article in English | MEDLINE | ID: mdl-32079426

ABSTRACT

Aim: We aimed to explore the circular RNA (circRNA) profile of small-cell lung cancer (SCLC). Materials & methods: Total RNA was extracted from six paired SCLC tumors and adjacent noncancerous tissues. Next-generation sequencing was performed to identify the circRNA expression profile of SCLC. Results: We found that five circRNAs were significantly upregulated and 30 circRNAs were significantly downregulated in the SCLC tissues. We confirmed the five upregulated and four randomly selected downregulated circRNAs using real-time quantitative PCR. Notably, circ-STXBP5L was selectively upregulated in SCLC samples, but undetectable in the normal control tissues. Bioinformatics analysis demonstrated that circ-STXBP5L may participate in SCLC carcinogenesis by regulating numerous cancer-related pathways. Conclusion: This study may provide new insights into the early diagnosis and development of targeted therapies for SCLC.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , RNA, Circular/genetics , Small Cell Lung Carcinoma/genetics , Cell Transformation, Neoplastic/genetics , Computational Biology/methods , Early Detection of Cancer , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/therapy , RNA-Seq , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/therapy
6.
Gene ; 697: 40-47, 2019 May 20.
Article in English | MEDLINE | ID: mdl-30794912

ABSTRACT

WDR1 is a major cofactor of the actin depolymerizing factor (ADF)/cofilin, accelerating ADF/cofilin-mediated actin disassembly. We had previously showed that WDR1-mediated actin dynamics is required for postnatal myocardial growth and adult myocardial maintenance in mice, in which the detailed phenotypes of adult cardiomyocyte-specific Wdr1 deletion mice had not been analyzed. In this study, we systematically analyzed the role of Wdr1 in adult mouse heart. Adult cardiomyocyte-specific Wdr1 deletion mice (cKO) exhibited cardiac hypertrophy and myocardial fibrosis. Echocardiographic study and electrocardiography revealed impaired contractile function, prolonged QT interval and Tpeak-Tend interval, and abnormal T-wave amplitude in cKO mice. Increased levels of sarcomeric proteins, adherens junction proteins and cofilin, and severe actin filament (F-actin) accumulations were observed in cKO mice heart. Taken together, this finding demonstrates that WDR1 is a critical factor for normal structure and function of adult mouse heart.


Subject(s)
Cardiomegaly/genetics , Microfilament Proteins/genetics , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Actins/metabolism , Animals , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Destrin/metabolism , Female , Heart/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/metabolism , Myocytes, Cardiac/physiology , Organogenesis
7.
Int J Biol Sci ; 14(9): 1067-1080, 2018.
Article in English | MEDLINE | ID: mdl-29989053

ABSTRACT

The characteristic of carcinoma is cell migration and invasion, which involve in strong actin dynamics. Regulations of actin dynamics have been implicated in cancer cell migration and tumor progression. WDR1 (WD-repeat domain 1) is a major cofactor of the actin depolymerizing factor (ADF)/cofilin, strongly accelerating ADF/cofilin-mediated actin disassembly. The role of WDR1 in non-small cell lung cancer (NSCLC) progression has been unknown. Here, we show that the expression levels of WDR1 are increased in human NSCLC tissues compared with adjacent non-tumor tissues, and high WDR1 level correlates with poor prognosis in NSCLC patients. Knockdown of WDR1 in NSCLC cells significantly inhibits cell migration, invasion, EMT process and tumor cell growth in vitro and in vivo. Otherwise, overexpression of WDR1 promotes NSCLC cell proliferation and migration. Mechanically, our data suggested WDR1 regulated tumor cells proliferation and migration might through actin cytoskeleton-mediated regulation of YAP, and we demonstrated that WDR1 contributes to NSCLC progression through ADF/cofilin-mediated actin disassembly. Our findings implicate that the ADF/cofilin-WDR1-actin axis as an activator of malignant phenotype that will be a promising therapeutic target in lung cancer.


Subject(s)
Actins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Destrin/metabolism , Lung Neoplasms/metabolism , Microfilament Proteins/metabolism , A549 Cells , Actins/genetics , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Destrin/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Male , Mice , Mice, Nude , Microfilament Proteins/genetics , Microfilament Proteins/physiology , Xenograft Model Antitumor Assays
8.
Dev Biol ; 438(2): 124-137, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29654745

ABSTRACT

Outflow tract (OFT) anomalies account for about 30% of human congenital heart defects detected at birth. The second heart field (SHF) progenitors contribute to OFT and right ventricle (RV) development, but the process largely remains unknown. WDR1 (WD-repeat domain 1) is a major co-factor of actin depolymerizing factor (ADF)/cofilin that actively disassembles ADF/cofilin-bound actin filaments. Its function in embryonic heart development has been unknown. Using Wdr1 floxed mice and Nkx2.5-Cre, we deleted Wdr1 in embryonic heart (Wdr1F/F;Nkx2.5-Cre) and found that these mice exhibited embryonic lethality, and hypoplasia of OFT and RV. To investigate the role of WDR1 in OFT and RV development, we generated SHF progenitors-specific Wdr1 deletion mice (shfKO). shfKO mice began to die at embryonic day 11.5 (E11.5), and displayed decreased size of the proximal OFT and RV at E10.5. In shfKO embryos, neither the number of SHF cells deployment to OFT nor cell proliferation and the cell number were changed, whereas the cellular organization and myofibrillar assembly of cardiomyocytes were severely disrupted. In the proximal OFT and RV of both shfKO and Wdr1F/F;Nkx2.5-Cre embryos, cardiomyocytes were dissociated from the outer compact myocardial layer and loosely and disorderly arranged into multilayered myocardium. Our results demonstrate that WDR1 is indispensable for normal OFT and RV development, and suggest that WDR1-mediated actin dynamics functions in controlling the size of OFT and RV, which might through regulating the spatial arrangement of cardiomyocytes.


Subject(s)
Heart Ventricles/embryology , Microfilament Proteins/physiology , Actins/genetics , Actins/metabolism , Animals , Embryo, Mammalian/embryology , Embryonic Development/genetics , Embryonic Development/physiology , Gene Expression Regulation, Developmental/genetics , Heart/embryology , Heart Defects, Congenital/genetics , Mice , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Myocardium , Myocytes, Cardiac , Organogenesis , Signal Transduction , Ventricular Outflow Obstruction
9.
Exp Cell Res ; 350(1): 73-82, 2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27871850

ABSTRACT

Uterine fibroids, also known as uterine leiomyomas, are a benign tumor of the human uterus and the commonest estrogen-dependent benign tumor found in women. Myocardin is an important transcriptional regulator in smooth and cardiac muscle development. The role of myocardin and its relationship with ERα in uterine fibroids have barely been addressed. We noticed that the expression of myocardin was markedly reduced in human uterine fibroid tissue compared with corresponding normal or adjacent myometrium tissue. Here we reported that myocardin induced the transcription and expression of differentiation markers SM22α and alpha smooth muscle actin (α-SMA) in rat primary uterine smooth muscle cells (USMCs) and this effect was inhibited by ERα. Notably, we showed that, ERα induced expression of proliferation markers PCNA and ki-67 in rat primary USMCs. We also found ERα interacted with myocardin and formed complex to bind to CArG box and inhibit the SM22α promoter activity. Furthermore, ERα inhibited the transcription and expression of myocardin, and reduced the levels of transcription and expression of downstream target SM22α, a SMC differentiation marker. Our data thus provided important and novel insights into how ERα and myocardin interact to control the cell differentiation and proliferation of USMCs. Thus, it may provide potential therapeutic target for uterine fibroids.


Subject(s)
Cell Differentiation/drug effects , Estrogen Receptor alpha/metabolism , Leiomyoma/metabolism , Nuclear Proteins/pharmacology , Trans-Activators/pharmacology , Animals , Cell Differentiation/physiology , Gene Expression Regulation/genetics , Humans , Leiomyoma/chemically induced , Leiomyoma/drug therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Rats , Serum Response Factor/metabolism , Trans-Activators/metabolism
10.
Cell Rep ; 13(4): 723-732, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26489465

ABSTRACT

RNA G-quadruplexes (G4s) play important roles in RNA biology. However, the function and regulation of mRNA G-quadruplexes in embryonic development remain elusive. Previously, we identified RHAU (DHX36, G4R1) as an RNA helicase that resolves mRNA G-quadruplexes. Here, we find that cardiac deletion of Rhau leads to heart defects and embryonic lethality in mice. Gene expression profiling identified Nkx2-5 mRNA as a target of RHAU that associates with its 5' and 3' UTRs and modulates its stability and translation. The 5' UTR of Nkx2-5 mRNA contains a G-quadruplex that requires RHAU for protein translation, while the 3' UTR of Nkx2-5 mRNA possesses an AU-rich element (ARE) that facilitates RHAU-mediated mRNA decay. Thus, we uncovered the mechanisms underlying Nkx2-5 post-transcriptional regulation during heart development. Meanwhile, this study demonstrates the function of mRNA 5' UTR G-quadruplex-mediated protein translation in organogenesis.


Subject(s)
DEAD-box RNA Helicases/metabolism , G-Quadruplexes , Homeodomain Proteins/genetics , Transcription Factors/genetics , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Blotting, Northern , COS Cells , Cell Line , Chlorocebus aethiops , DEAD-box RNA Helicases/genetics , Heart/embryology , Homeobox Protein Nkx-2.5 , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism
11.
Am J Pathol ; 184(7): 1967-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24840128

ABSTRACT

Actin dynamics are critical for muscle development and function, and mutations leading to deregulation of actin dynamics cause various forms of heritable muscle diseases. AIP1 is a major cofactor of the actin depolymerizing factor/cofilin in eukaryotes, promoting actin depolymerizing factor/cofilin-mediated actin disassembly. Its function in vertebrate muscle has been unknown. To investigate functional roles of AIP1 in myocardium, we generated conditional knockout (cKO) mice with cardiomyocyte-specific deletion of Wdr1, the mammalian homolog of yeast AIP1. Wdr1 cKO mice began to die at postnatal day 13 (P13), and none survived past P24. At P12, cKO mice exhibited cardiac hypertrophy and impaired contraction of the left ventricle. Electrocardiography revealed reduced heart rate, abnormal P wave, and abnormal T wave at P10 and prolonged QT interval at P12. Actin filament (F-actin) accumulations began at P10 and became prominent at P12 in the myocardium of cKO mice. Within regions of F-actin accumulation in myofibrils, the sarcomeric components α-actinin and tropomodulin-1 exhibited disrupted patterns, indicating that F-actin accumulations caused by Wdr1 deletion result in disruption of sarcomeric structure. Ectopic cofilin colocalized with F-actin aggregates. In adult mice, Wdr1 deletion resulted in similar but much milder phenotypes of heart hypertrophy, F-actin accumulations within myofibrils, and lethality. Taken together, these results demonstrate that AIP1-regulated actin dynamics play essential roles in heart function in mice.


Subject(s)
Actins/physiology , Heart/growth & development , Microfilament Proteins/physiology , Muscle Development/physiology , Myocytes, Cardiac/physiology , Actin Cytoskeleton/physiology , Actins/genetics , Animals , Cofilin 2/physiology , Heart/physiopathology , Hypertrophy , Mice, Knockout , Microfilament Proteins/genetics
12.
Int J Mol Sci ; 14(12): 24380-98, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24351823

ABSTRACT

Ras homologue enriched in brain 1 (Rheb1) plays an important role in a variety of cellular processes. In this study, we investigate the role of Rheb1 in the post-natal heart. We found that deletion of the gene responsible for production of Rheb1 from cardiomyocytes of post-natal mice resulted in malignant arrhythmias, heart failure, and premature death of these mice. In addition, heart growth impairment, aberrant metabolism relative gene expression, and increased cardiomyocyte apoptosis were observed in Rheb1-knockout mice prior to the development of heart failure and arrhythmias. Also, protein kinase B (PKB/Akt) signaling was enhanced in Rheb1-knockout mice, and removal of phosphatase and tensin homolog (Pten) significantly prolonged the survival of Rheb1-knockouts. Furthermore, signaling via the mammalian target of rapamycin complex 1 (mTORC1) was abolished and C/EBP homologous protein (CHOP) and phosphorylation levels of c-Jun N-terminal kinase (JNK) were increased in Rheb1 mutant mice. In conclusion, this study demonstrates that Rheb1 is important for maintaining cardiac function in post-natal mice via regulation of mTORC1 activity and stress on the endoplasmic reticulum. Moreover, activation of Akt signaling helps to improve the survival of mice with advanced heart failure. Thus, this study provides direct evidence that Rheb1 performs multiple important functions in the heart of the post-natal mouse. Enhancing Akt activity improves the survival of infant mice with advanced heart failure.


Subject(s)
Apoptosis , Endoplasmic Reticulum/metabolism , Heart Failure/etiology , Monomeric GTP-Binding Proteins/metabolism , Neuropeptides/metabolism , Animals , Animals, Newborn , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Cells, Cultured , Heart/growth & development , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuropeptides/deficiency , Neuropeptides/genetics , PTEN Phosphohydrolase/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Ras Homolog Enriched in Brain Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...