Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell Biol Int ; 48(1): 20-30, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37975488

ABSTRACT

Short for pyruvate kinase M2 subtype, PKM2 can be said of all-round player that is notoriously known for its metabolic involvement in glycolysis. Holding a dural role as a metabolic or non-metabolic (kinase) enzyme, PKM2 has drawn extensive attention over its biological roles implicated in tumor cells, including proliferation, migration, invasion, metabolism, and so on. wandering PKM2 can be transboundary both intracellularly and extracellularly. Specifically, PKM2 can be nuclear, cytoplasmic, mitochondrial, exosomal, or even circulate within the body. Importantly, PKM2 can function as an RNA-binding protein (RBP) to self-support its metabolic function. Despite extensive investigations or reviews available surrounding the biological roles of PKM2 from different angles in tumor cells, little has been described regarding some novel role of PKM2 that has been recently found, including, for example, acting as RNA-binding protein, protection of Golgi apparatus, and remodeling of microenvironment, and so forth. Given these findings, in this review, we summarize the recent advancements made in PKM2 research, mainly from non-metabolic respects. By the way, PKM1, another paralog of PKM2, seems to have been overlooked or under-investigated since its discovery. Some recent discoveries made about PKM1 are also preliminarily mentioned and discussed.


Subject(s)
Glycolysis , Neoplasms , Pyruvate Kinase , Cell Line, Tumor , Pyruvate Kinase/metabolism , RNA-Binding Proteins/metabolism , Neoplasms/metabolism , Humans
2.
Dig Dis Sci ; 69(5): 1562-1570, 2024 May.
Article in English | MEDLINE | ID: mdl-38580886

ABSTRACT

Esophageal carcinoma (ESCA) is an aggressive solid tumor. The 5-year survival rate for patients with ESCA is estimated to be less than 20%, mainly due to tumor invasion and metastasis. Therefore, it is urgent to improve early diagnostic tools and effective treatments for ESCA patients. Tumor microenvironment (TME) enhances the ability of tumor cells to proliferate, migrate, and escape from the immune system, thus promoting the occurrence and development of tumor. TME contains chemokines. Chemokines consist of four major families, which are mainly composed of CC and CXC families. The main purpose of this review is to understand the CC and CXC chemokines and their receptors in ESCA, to improve the understanding of tumorigenesis of ESCA and determine new biomarkers for the diagnosis and prognosis of ESCA. We reviewed the literature on CC and CXC chemokines and their receptors in ESCA identified by PubMed database. This article introduces the general structures and functions of CC, CXC chemokines and their receptors in TME, as well as their roles in the progress of ESCA. Chemokines are involved in the development of ESCA, such as cancer cell invasion, metastasis, angiogenesis, and radioresistance, and are key determinants of disease progression, which have a great impact on patient prognosis and treatment response. In addition, a full understanding of their mechanism of action is essential to further verify that these chemokines and their receptors may serve as biomarkers or therapeutic targets of ESCA.


Subject(s)
Chemokines , Esophageal Neoplasms , Tumor Microenvironment , Humans , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/immunology , Chemokines/metabolism , Receptors, Chemokine/metabolism , Biomarkers, Tumor/metabolism , Prognosis
3.
Odontology ; 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528238

ABSTRACT

Chronic apical periodontitis (CAP) is characterized by inflammation and destruction of the apical periodontium that is of pulpal origin, appearing as an apical radiolucent area, and does not produce clinical symptoms. Little is known about whether the PD-1/PD-L1 ratio is associated with the balance between RANKL and OPG in CAP. The relationship between PD-1/PD-L1 and RANKL/OPG in CAP is investigated in this study. A CAP rat model was established using Sprague-Dawley rats. The pulp chambers were exposed to the oral cavity to allow bacterial contamination. The apical tissues of the bilateral mandibular first molars were analyzed for histological morphology using hematoxylin and eosin (H&E) staining. Immunohistochemistry and qRT-PCR were used to determine the expression of PD-1, PD-L1, OPG, and RANKL mRNA and proteins in periapical tissues and mandibular samples, respectively. The radiological images indicated a poorly defined low-density shadow and alveolar bone resorption after periodontitis induction. Histological analysis revealed an infiltration of inflammatory cells and alveolar bone resorption in the periapical tissues. Mandibular mRNA and periapical protein expression of PD-1, PD-L1, and RANKL was upregulated 7-28 days after periodontitis induction, while the expression of OPG was downregulated. No significant relationship was observed between PD-1/PD-L1 and RANKL/OPG at either mRNA or protein levels in CAP. There is an increased expression of PD-1, PD-L1, and RANKL and a decreased expression of OPG, indicating progression of CAP.

4.
Yi Chuan ; 46(5): 373-386, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38763772

ABSTRACT

Cardioembolic stroke, characterized by severe illness, poor prognosis, and high recurrence rate, is one of the important causes of ischemic stroke. In the field of genetic research, numerous genes associated with cardioembolic stroke have been identified, and their potential in predicting disease risk and evaluating risk factors has been progressively explored. Here, we provide an overview of the latest advancements in genetics for cardioembolic stroke, including genome-wide association studies, copy number variation studies, whole-genome sequencing studies. Furthermore, we also summarize the application of genetic datasets in polygenic risk score and Mendelian randomization. The aim of this overview is to provide insights and references from multiple perspectives for future investigations on the genetic information for cardioembolic stroke.


Subject(s)
DNA Copy Number Variations , Embolic Stroke , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Embolic Stroke/genetics , Embolic Stroke/etiology , Risk Factors
5.
J Cell Physiol ; 236(7): 5373-5386, 2021 07.
Article in English | MEDLINE | ID: mdl-33368292

ABSTRACT

CXCL6, contraction of C-X-C motif chemokine ligand 6, whose biological roles have been rarely described in esophageal squamous cell carcinoma (ESCC). To understand the clinicopathological and biological roles played by CXCL6 in the growth and metastasis of ESCC, immunohistochemistry was used to detect the expression of CXCL6 in ESCC tissues, totaling 105 cases; and the correlation was statistically analyzed between CXCL6 expression and clinicopathological parameters. The role mediated in migration and invasion was evaluated using wound-healing and Transwell assays. MTT and flow cytometry were used to assay the proliferative variation. In vivo, tail vein injection model was established in nude mice xenografted with human ESCC cell lines whose CXCL6 were artificially manipulated. It was found that relative to normal control, CXCL6 was profoundly higher in ESCC; upregulated CXCL6 only significantly correlated with differentiation degree. In vitro, CXCL6 was found to promote the proliferation, migration, and invasion of ESCC cells; which was fully corroborated by nude mice experiment that CXCL6 can promote the growth and metastases of ESCC cells in vivo. Mechanistically, CXCL6 was discovered to be capable of promoting epithelial-mesenchymal transition and upregulating PD-L1 expression through activation of the STAT3 pathway. Collectively, all the data we showed here demonstrate that CXCL6 can enhance the growth and metastases of ESCC cells both in vivo and in vitro.


Subject(s)
B7-H1 Antigen/metabolism , Chemokine CXCL6/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , STAT3 Transcription Factor/metabolism , Animals , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Invasiveness/pathology , Signal Transduction/physiology , Up-Regulation
6.
Lab Invest ; 101(7): 908-920, 2021 07.
Article in English | MEDLINE | ID: mdl-33958701

ABSTRACT

Long non-coding RNAs (lncRNAs) play important roles in a range of different human cancers. However, the role of lncRNA solute carrier organic anion transporter family member 4A1-AS1 (SLCO4A1-AS1) in colon cancer remains enigmatic. Hence, we aimed to explore the specific role of SLCO4A1-AS1 in colon cancer stem cells. Colon cancer-related differentially expressed lncRNA and mRNA were screened using microarray-based analysis, and the expression of SLCO4A1-AS1 and SLCO4A1 in colon cancer tissues was determined using reverse transcription quantitative polymerase chain reaction and western blot analysis. The interaction among SLCO4A1-AS1, microRNA-150-3p (miR-150-3p) and SLCO4A1 was verified using dual-luciferase reporter assay, RNA immunoprecipitation and RNA pull-down. Moreover, SLCO4A1-AS1, miR-150-3p and/or SLCO4A1 were overexpressed or depleted in colon cancer cells to detect their effects on migration, invasion, sphere formation, apoptosis and tumorigenesis abilities of colon cancer stem CD133+CD44+ cells using both in vitro and in vivo assays. SLCO4A1-AS1 and SLCO4A1 were screened as the differentially expressed lncRNA and mRNA in colon cancer tissues. SLCO4A1-AS1 was confirmed to competitively bind to miR-150-3p to elevate SLCO4A1 expression. Moreover, knockdown of SLCO4A1-AS1 decreased SLCO4A1 expression, thus inhibiting cell migration, invasion, sphere formation, and tumorigenesis abilities and enhancing the apoptosis of CD133+CD44+ cells. Collectively, these findings provide evidence demonstrating that depleting SLCO4A1-AS1 competitively binds to miR-150-3p, which downregulates SLCO4A1 expression, thus hindering colon cancer progression.


Subject(s)
Colonic Neoplasms , MicroRNAs , Neoplastic Stem Cells/metabolism , Organic Anion Transporters/genetics , RNA, Long Noncoding , Animals , Cell Movement , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Gene Knockdown Techniques , HCT116 Cells , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Xenograft Model Antitumor Assays
7.
Cancer Cell Int ; 21(1): 121, 2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33602237

ABSTRACT

BACKGROUND: Calmodulin1 (CALM1) has been identified as one of the overexpression genes in a variety of cancers and EGFR inhibitor have been widely used in clinical treatment but it is unknown whether CALM1 and epidermal growth factor receptor (EGFR) have a synergistic effect in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to explore the synergistic effects of knock-out CALM1 combined with EGFR inhibitor (Afatinib) and to elucidate the role of CALM1 in sensitizing the resistance to Afatinib in ESCC. METHOD: Immunohistochemistry (IHC) and qRT-PCR were used to examine the expression of CALM1 and EGFR in ESCC tissues. Kaplan-Meier survival analysis was used to analyze the clinical and prognostic significance of CALM1 and EGFR expression in ESCC. Furthermore, to evaluate the biological function of CALM1 in ESCC, the latest gene editing technique CRISPR/Cas9(Clustered regularly interspaced short palindromic repeats)was applied to knockout CALM1 in ESCC cell lines KYSE150, Eca109 and TE-1. MTT, flow cytometry, Transwell migration, scratch wound-healing and colony formation assays were performed to assay the combined effect of knock-out CALM1 and EGFR inhibitor on ESCC cell proliferation and migration. In addition, nude mice xenograft model was used to observe the synergistic inhibition of knock-out CALM1 and Afatinib. RESULTS: Both CALM1 and EGFR were found to be significantly over-expressed in ESCC compared with paired normal control. Over-expressed CALM1 and EGFR were significantly associated with clinical stage, T classification and poor overall prognosis, respectively. In vitro, the combined effect of knock-out CALM1 mediated by the lentivirus and EGFR inhibitor was shown to be capable of inhibiting the proliferation, inducing cell cycle arrest at G1/S stage and increasing apoptosis of KYSE-150 and Eca109 cells; invasion and migration were also suppressed. In vivo, the results of tumor weight and total fluorescence were markedly reduced compared with the sgCtrl-infected group and sgCAML1 group. CONCLUSION: Our data demonstrated that knock-out of CALM1 could sensitize ESCC cells to EGFR inhibitor, and it may exert oncogenic role via promotion of EMT. Taken together, CALM1 may be a tempting target to overcome Afatinib resistance.

8.
Chaos ; 31(5): 053105, 2021 May.
Article in English | MEDLINE | ID: mdl-34240922

ABSTRACT

The essence of logical stochastic resonance is the dynamic manipulation of potential wells. The effect of time delay on the depth of potential wells and the width of a bistable region can be inferred by logic operations in the bistable system with time delay. In a time-delayed synthetic gene network, time delay in the synthesis process can increase the depth of the potential wells, while that in the degradation process, it can reduce the depth of the potential wells, which will result in a decrease in the width of the bistable region (the reason for time delay to induce logic operations without external driving force) and the instability of the system (oscillation). These two opposite effects imply stretching and folding, leading to complex dynamical behaviors of the system, including period, chaos, bubble, chaotic bubble, forward and reverse period doubling bifurcation, intermittency, and coexisting attractors.


Subject(s)
Gene Regulatory Networks , Genes, Synthetic
9.
Biochem Biophys Res Commun ; 526(1): 29-34, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32192776

ABSTRACT

NME4, also named Nm23-H4, is a contraction of NME/NM23 Nucleoside Diphosphate Kinase 4, whose major role is the synthesis of nucleoside triphosphates. However, its association with programmed death ligand 1 (PD-L1) remains far from understood. Herein, it was discovered that silencing NME4 can lead to the marked downregulation of PD-L1, with phosphorylated STAT3 at the 705th serine being inactivated in vitro in esophageal squamous cell carcinoma (ESCC) cell lines. To further validate the association between NME4 and PD-L1 that was observed in cell lines, Pearson correlation analysis was performed on the data regarding the transcriptomic RNA sequencing of NME4 and PD-L1 in cervical squamous cell carcinoma (CSCC), which pathologically highly resembles ESCC in terms of tumor origin, obtained from the GEPIA database. It was demonstrated that their correlation was significant but negative between NME4 and PD-L1 in CSCC. To the best of our knowledge, this is the first report describing a modulation exerted by NME4 over PD-L1 in the background of squamous cell carcinoma, strongly suggestive of the underlying role of NME4 working to exclude CD8 T cells from infiltrating into the squamous cell carcinoma microenvironment.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Nucleoside Diphosphate Kinase D/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , B7-H1 Antigen/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Down-Regulation , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptome/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
10.
Mol Cell Biochem ; 471(1-2): 81-89, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32504364

ABSTRACT

NME4, also designated nm23-H4 or NDPK-D, has been known for years for its well-established roles in the synthesis of nucleoside triphosphates, though; little has been known regarding the differential metabolites involved as well as the biological roles NME4 plays in proliferation and invasion of esophageal squamous cell carcinoma (ESCC) cells. To understand the biological roles of NME4 in ESCC cells, lentiviral-based short hairpin RNA interference (shRNA) vectors were constructed and used to stably knock down NME4. Then, the proliferative and invasive variations were assessed using MTT, Colony formation and Transwell assays. To understand the metabolites involved after silencing of NME4 in ESCC cells, widely targeted metabolomic screening was taken. It was discovered that silencing of NME4 can profoundly suppress the proliferation and invasion in ESCC cells in vitro. Metabolically, a total of 11 differential metabolites were screened. KEGG analyses revealed that Tryptophan, Riboflavin, Purine, Nicotinate, lysine degradation, and Linoleic acid metabolism were also involved in addition to the well-established nucleotides metabolism. Some of these differential metabolites, say, 2-Picolinic Acid, Nicotinic Acid and Pipecolinic Acid were suggested to be associated with tumor immunomodulation. The data we described here support the idea that metabolisms occurred in mitochondrial was closely related to tumor immunity.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic , Metabolome , Mitochondria/metabolism , Nucleoside Diphosphate Kinase D/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Humans , Mitochondria/pathology , Neoplasm Invasiveness , Nucleoside Diphosphate Kinase D/antagonists & inhibitors , Nucleoside Diphosphate Kinase D/genetics
11.
J Cell Biochem ; 120(7): 11539-11550, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30756445

ABSTRACT

Recent studies have demonstrated pleiotropic roles of pyruvate kinase isoenzyme type M2 (PKM2) in tumor progression. However, the precise mechanisms underlying the effects of PKM2 on esophageal squamous cell carcinoma (ESCC) metastasis and transforming growth factor ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) remain to be established. In this study, we observed upregulation of PKM2 in ESCC tissues that was markedly associated with lymph node metastasis and poor prognosis. High PKM2 expression in tumor tissues frequently coincided with the high pSTAT3Tyr705 expression and low E-cadherin expression. Furthermore, altered PKM2 expression was significantly associated with proliferation, migration, and invasion of ESCC cells, in addition to expression patterns of EMT markers (Snail, E-cadherin, and vimentin) and pSTAT3Tyr705 /STAT3 ratio. Overexpression of STAT3 significantly attenuated the effects of PKM2 knockdown on cell proliferation and motility as well as expression of pSTAT3 Tyr705 and EMT markers. Consistently, stable short hairpin RNA (shRNA)-mediated silencing of PKM2 reversed the effects of TGF-ß1 treatment, specifically, upregulation of PKM2, phosphorylation of STAT3 at Tyr705, and increased EMT, migration, and invasion. We propose that PKM2 regulates cell proliferation, migration, and invasion via phosphorylation of STAT3 through TGF-ß1-induced EMT. Our findings collectively provide mechanistic insights into the tumor-promoting role of PKM2, supporting its prognostic value and the therapeutic utility of PKM2 inhibitors as potential antitumor agents in ESCC.

12.
Cell Biol Int ; 43(6): 634-641, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30958603

ABSTRACT

KIAA1377 has been found to be linked with lymph node metastasis in esophageal squamous cell carcinoma (SCC) in our previous study; however, the regulation of KIAA1377 remains far from understood. Herein, to understand the regulation of KIAA1377 from the angle of microRNA (miRNA)-messenger RNA (mRNA) modulation in the setting of SCC cells, the basal level of KIAA1377 was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot analysis in KYSE-150 and HeLa cells; biological roles of KIAA1377 contributing in the proliferation, migration, and invasion were evaluated using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), wound-healing and Transwell assays, respectively, after KIAA1377 was knocked out mediated by the CRISPR-Cas9 system. Bioinformatic prediction revealed that let-7b-5p was a putative miRNA regulating KIAA1377, which was ensuingly validated by the luciferase reporter assay; after which, variation of KIAA1377 expression was further verified by qRT-PCR and western blot analysis. Moreover, the biological roles of let-7b-5p in proliferation, migration, and invasion of KYSE-150 and HeLa cells were also evaluated. It was exhibited that KIAA1377 was able to promote the proliferation and motility of both KYSE-150 and HeLa cells, which can be reverted by re-expression of let-7b-5p. The luciferase reporter assay verified that let-7b-5p can diametrically target KIAA1377. Collectively, our data demonstrated that let-7b-5p can directly but negatively regulate KIAA1377 in SCC cell lines, Ecal109, and HeLa cells.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphatic Metastasis , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction
13.
J Cell Biochem ; 119(8): 7040-7052, 2018 08.
Article in English | MEDLINE | ID: mdl-29737564

ABSTRACT

Despite the phenotype has been established that M2 macrophages promotes the metastasis of ESCC, question still remains as to how the M2 macrophages facilitated metastasis of ESCC cells. To begin with, immunohistochemistry was performed to detect the expression of CD163, one typical surface marker of M2 macrophages in 90 paired ESCC and its normal controls after meta-analyzing the relevant studies regarding M2 macrophages in ESCC, confirming that infiltration of M2 macrophages was significantly linked with lymph node metastasis, T classification, and inferior overall survival of ESCC. To explore the mechanism behind, protein factors secreted by M2 macrophages were identified using antibody microarray. Six different significantly differential protein factors were screened out, including IL-1ß, TIMP1, IL-1α, MDC, TGF-ß1, and TGF-ß2. Among which, IL-1ß was picked up as cytokine as interest based on previous reports and its absolute fold. Functional analysis of IL-1ß showed that IL-1ß was able to promote migration and invasion of ESCC cells, enhancing epithelial-mesenchymal transition, and activating NF-κB pathway. Our study supports the promoting role of M2 macrophages in metastasis of ESCC cells, enriching the profile of protein factors released from M2 macrophages.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , NF-kappa B/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Humans , Interleukin-1beta/genetics , Macrophages/pathology , Male , NF-kappa B/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , THP-1 Cells
14.
FASEB J ; 31(5): 2114-2122, 2017 05.
Article in English | MEDLINE | ID: mdl-28174206

ABSTRACT

Numerous studies have reported that the role played by miR-26a in cancer is controversial, but whether miR-26a regulates metadherin (MTDH) expression in esophageal squamous cell carcinoma (ESCC) is unclear. We performed this study to investigate the clinical relevance of miR-26a expression in ESCC. miR-26a was detected by using the in situ hybridization method. To functionally analyze the role of miR-26a in ESCC cell lines in vitro, KYSE-450 and Eca109 cells were employed, whose endogenous miR-26a was artificially down- or up-regulated, respectively, by using lentiviral-based transfection. There was significant association between miR-26a expression and clinical stage (P = 0.049), lymph node metastasis (P = 0.023), tumor volume (P = 0.003), and poor overall prognosis (P = 0.026). miR-26a was able to suppress proliferation and migration of ESCC cells in vitro Moreover, we have confirmed that miR-26a can negatively regulate MTDH in ESCC cells by using luciferase reporter assay. In addition, to investigate the role miR-26a plays in cell proliferation, we nude mice were xenografted with ESCC cells whose miR-26a was stably down- and up-regulated. Together, our results show that miR-26a is capable of suppressing the proliferation and migration of ESCC cells via negative regulation of MTDH. Moreover, miR-26a expression was clinically relevant in cancer progression and poor prognosis, which supports the idea that miR-26a acts as a tumor suppressor in ESCC.-Yang, C., Zheng, S., Liu, T., Liu, Q., Dai, F., Zhou, J., Chen, Y., Sheyhidin, I., Lu, X. Down-regulated miR-26a promotes proliferation, migration, and invasion via negative regulation of MTDH in esophageal squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell/genetics , Cell Adhesion Molecules/metabolism , Cell Movement , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , RNA-Binding Proteins
15.
Cell Biol Int ; 42(3): 365-372, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29148603

ABSTRACT

MCP2, aliased CCL8, has been suggested to be implicated in the metastasis of cancer cells; however, no direct evidence has been established in esophageal squamous cell carcinoma (ESCC). In our present study, to investigate the role MCP2 played in the metastasis of ESCC cells; in vitro cell co-culture system was established. Wound-healing and Transwell assays were used to evaluate the migratory and invasive variation of ESCC cells before and after treatment with recombinant human MCP2. It was shown that MCP2 was able to activate the NF-κB signaling pathway inducing the epithelial-mesenchymal transition (EMT) and promoting the migration and invasion of ESCC cells in vitro. Our study provides an alternative working mechanism for M2 macrophage mediated the metastasis in tumor microenvironment in ESCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Chemokine CCL8/metabolism , Esophageal Neoplasms/metabolism , NF-kappa B/metabolism , Carcinoma, Squamous Cell/pathology , Cell Count , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Coculture Techniques , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Neoplasm Invasiveness , Signal Transduction
16.
Tumour Biol ; 37(6): 7255-66, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26666822

ABSTRACT

P38ß, p38γ, and p38δ have been sporadically and scarcely reported to be involved in the carcinogenesis of cancers, compared with p38α isoform. However, little has been known regarding their clinicopathological significance and biological roles in esophageal squamous cell carcinoma (ESCC). Expression status of p38ß, p38γ, and p38δ was assayed using immunohistochemistry with ESCC tissue microarray; ensuing clinicopathological significance was statistically analyzed. To define its biological roles on proliferation, migration and invasion of ESCC cell line Eca109 in vitro, MTT, wound healing, and Transwell assays were employed, respectively. As confirmation, athymic nude mice were taken to verify the effect over proliferation in vivo. It was found that both p38ß and p38δ expression, other than p38γ, were significantly higher in ESCC tissues compared with paired normal controls. In terms of prognosis, only p38ß expression was observed to be significantly associated with overall prognosis. Clinicopathologically, there was significant association between p38γ expression and clinical stage, lymph nodes metastases, and tumor volume. No significant association was found for p38ß and p38δ between its expression and other clinicopathological parameters other than significant difference of expression between ESCC versus normal control. In Eca109, it was observed that p38ß, p38γ, and p38δ can promote the cell growth and motility. As verification, over-expression of p38δ can promote, whereas knockdown of p38γ can prevent, the tumorigenesis in nude mice model xenografted with Eca109 cells whose basal level of p38δ was stably over-expressed and p38γ was stably knocked down. Together, our results demonstrate that p38ß, p38γ, and p38δ played oncogenic roles in ESCC.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Esophageal Neoplasms/enzymology , Mitogen-Activated Protein Kinase 11/physiology , Mitogen-Activated Protein Kinase 12/physiology , Mitogen-Activated Protein Kinase 13/physiology , Neoplasm Proteins/physiology , Animals , Apoptosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle , Cell Line, Tumor , Cell Movement , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Female , Heterografts , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 12/genetics , Mitogen-Activated Protein Kinase 13/genetics , Neoplasm Proteins/genetics , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Recombinant Fusion Proteins/metabolism , Tumor Burden
17.
Tumour Biol ; 37(9): 12767-12777, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27448819

ABSTRACT

Despite PIK3CD has been extensively reported in cancers, however, little evidence has been available regarding its role in the setting of diffuse large B cell lymphoma (DLBCL). In the present study, to investigate the role of PIK3CD in DLBCL, relevant experiments were carried out on both in vivo clinical tissue level and in vitro cell line level. Prognostic and clinicopathological significance were analyzed after immunohistochemical assay of PIK3CD expression on DLBCL tissue microarray. MTT assay and flow cytometry were employed to evaluate the proliferative variation, cell cycle, and apoptosis. Athymic nude mice xenografted with DLBCL cell line were employed to confirm the role of PIK3CD. It was found that there was a significant difference between expression of PIK3CD and international prognosis index (IPI), performance state (PS), and inferior overall prognosis. Furthermore, PIK3CD can promote proliferation and prevent apoptosis in DLBCL cells in vitro through upregulation of c-myc and p-AKT and in contrast downregulation of p21 and p27. In nude mice model, knock-down of PIK3CD was shown to be able to suppress the proliferation of DLBCL but not significantly compared with control group. Taken together, our study showed that PIK3CD can promote proliferation of DLBCL cells both in vitro and in vivo, suggesting that PIK3CD could be druggable in the therapy of DLBCL.


Subject(s)
Cell Proliferation , Class I Phosphatidylinositol 3-Kinases/biosynthesis , Lymphoma, Large B-Cell, Diffuse/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Up-Regulation , Animals , Apoptosis/genetics , Blotting, Western , Cell Cycle/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Transplantation, Heterologous
18.
Tumour Biol ; 37(11): 14595-14604, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27619676

ABSTRACT

Accumulated evidence suggests that miR-106b played a key role in the promotion of the metastases of cancer; however, little is known about miR-106b in esophageal squamous cell carcinoma (ESCC). To investigate expression level of miR-106b in ESCC tissues, quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect miR-106b expression in 35 Kazakh's ESCC and paired normal adjacent tissues (NATs). To evaluate the role mediated by miR-106b in the proliferation, migration, and invasion, MTT, wound healing, and transwell assays were employed, respectively. Luciferase reporter assay was used to identify the downstream target through miR-106b. To understand the regulation between miR-106b and Smad 7, qRT-PCR and western blot were performed. The present study showed that miR-106b was pronouncedly upregulated in ESCC relative to paired NAT and that upregulated miR-106b was significantly associated with lymph node metastases. MiR-106b was found to be able to promote proliferation, migration, and invasion of ESCC cells in vitro. Smad 7 was confirmed as a downstream target of miR-106b in our experimental setting. Smad 7 was remarkably downregulated in ESCC compared with paired NAT. In addition, upregulation of miR-106b can promote epithelial mesenchymal transition (EMT) in ESCC cell in vitro. Our results indicated that miR-106b can promote migration and invasion of ESCC cells through enhancing EMT process via downregulation of Smad 7, suggesting that miR-106b can be a potential molecular phenotype in ESCC metastases.


Subject(s)
Carcinoma, Squamous Cell/secondary , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Smad7 Protein/metabolism , Apoptosis , Blotting, Western , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Smad7 Protein/genetics , Tumor Cells, Cultured
19.
Tumour Biol ; 37(2): 2665-72, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26404132

ABSTRACT

The objectives of the present study are to explore role of pyruvate kinase isoenzyme type M2 (PKM2) in progression of Kazakh's esophageal squamous cell carcinoma (ESCC) in Xinjiang, China, and to clarify mechanism of PKM2 in malignant phenotype. PKM2 expression was examined using immunohistochemistry (IHC) in 101 matched pairs of ESCC and normal adjacent tissues (NATs) and using enzyme-linked immunosorbent assay (ELISA) in 35 serum samples of Kazakh's ESCC and 8 serum samples of healthy subjects. To investigate mechanism, small interfering RNA (siRNA)-PKM2 was transfected into ESCC cells. Cell migration and invasion were evaluated by wound healing and Transwell assays. Apoptosis and cell cycle were analyzed by flow cytometry (FCM). PKM2 expression was significantly higher in ESCC tissues (77.2 %, 78/101) compared with matched NAT (P = 0.003) and also higher in serum samples of Kazakh's ESCC patients (78.84 ng/mL) compared with healthy subjects (13.55 ng/mL) (P = 0.001). Patients with overexpression of PKM2 had a poor prognosis (P = 0.032). After knockdown of PKM2, cell proliferation, migration, and invasion were significantly reduced (P = 0.001), apoptosis increased (P = 0.001), and cell cycle was arrested at G1 phase. PKM2 overexpression was significantly correlated with the worse outcome of Kazakh's ESCC. Furthermore, PKM2 was involved in progression of ESCC by promoting proliferation and suppressing apoptosis, accelerating invasion, and influencing cell cycle. PKM2 could be a potential biomarker for molecular classification of ESCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carrier Proteins/genetics , Cell Movement/genetics , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Membrane Proteins/genetics , Thyroid Hormones/genetics , Up-Regulation/genetics , Apoptosis/genetics , Carcinoma, Squamous Cell/pathology , Cell Cycle/genetics , Cell Line, Tumor , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , RNA, Small Interfering/genetics , Thyroid Hormone-Binding Proteins
20.
Mol Carcinog ; 54(9): 779-88, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24668792

ABSTRACT

The aim of the study was to identify candidate biomarkers for esophageal squamous cell carcinoma (ESCC) in Kazakh ethnic in Xinjiang as well as to reveal the potential role of Annexin A2 in ESCC carcinogenesis and progression. Five paired of Kazakh's ESCC tissues (T) and matched adjacent morphologically normal tissues (N) were separated by two-dimensional electrophoresis (2-DE) and differential proteins were identified by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). Annexin A2 was identified as a down-regulated protein in Kazakh's ESCC and further validated by immunohistochemistry (IHC) in 77 Kazakh's ESCC formalin-fixed paraffin-embeded (FFPE) samples. The expression level of Annexin A2 protein significantly correlated with the degree of ESCC differentiation and depth of invasion. For clarification of the role of Annexin A2 in regulating cell phenotype, in vitro eukaryotic expression vectors harboring full length Annexin A2 (pCMV-XL5-Annexin A2) was tranfected into Eca109 cells, and transfection effects were evaluated by RT-PCR and Western blotting analysis, respectively. Functionally, there was a significant decrease in cell proliferation, migration, and invasion capability in Eca109 with transfected pCMV-XL5-Annexin A2 compared to the controls. Furthermore, up-regulating Annexin A2 can significantly cause cell cycle arrest at the G2 phase, but no apoptosis was induced. Together, our findings suggested that Annexin A2 was involved in malignant phenotype and was a potential biomark for molecular classification in ESCC.


Subject(s)
Annexin A2/analysis , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Esophagus/pathology , Annexin A2/genetics , Carcinoma, Squamous Cell/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Electrophoresis, Gel, Two-Dimensional , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma , Esophagus/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
SELECTION OF CITATIONS
SEARCH DETAIL