Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 15207, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956294

ABSTRACT

The creep characteristics and potential deformation patterns of gangue backfill material are crucial in backfill mining operations. This study utilizes crushed gangue from the Gangue Yard in Fuxin City as the research material. An in-house designed, large-scale, triaxial gangue compaction test system was used. Triaxial compaction creep tests were conducted on gangue materials with varying particle size distributions. Analysis was performed based on different particle sizes, stresses, and confinement pressures. The study investigates the creep characteristics of the gangue under different conditions and explores the underlying causes. It reveals the relationship between the creep deformation of gangue materials and the passage of time. Mathematical methods are applied to develop a triaxial compaction creep power law model for gangue backfill materials. Finally, the creep results are fitted using an empirical formula approach.

2.
Front Neurosci ; 17: 1279259, 2023.
Article in English | MEDLINE | ID: mdl-37817804

ABSTRACT

Accumulating evidence suggests a gain of elusive toxicity in pathogenically mutated PFN1. The prominence of PFN1 aggregates as a pivotal pathological hallmark in PFN1 transgenic rats underscores the crucial involvement of protein aggregation in the initiation and progression of neurodegeneration. Detergent-insoluble materials were extracted from the spinal cords of paralyzed rats afflicted with ALS and were intramuscularly administered to asymptomatic recipient rats expressing mutant PFN1, resulting in an accelerated development of PFN1 inclusions and ALS-like phenotypes. This effect diminished when the extracts derived from wildtype PFN1 transgenic rats were employed, as detergent-insoluble PFN1 was detected exclusively in mutant PFN1 transgenic rats. Consequently, the factor influencing the progression of ALS pathology in recipient rats is likely associated with the presence of detergent-insoluble PFN1 within the extracted materials. Noteworthy is the absence of disease course modification upon administering detergent-insoluble extracts to rats that already displayed PFN1 inclusions, suggesting a seeding rather than augmenting role of such extracts in initiating neuropathological changes. Remarkably, pathogenic PFN1 exhibited an enhanced affinity for the molecular chaperone DNAJB6, leading to the sequestration of DNAJB6 within protein inclusions, thereby depleting its availability for cellular functions. These findings shed light on a novel mechanism that underscores the prion-like characteristics of pathogenic PFN1 in driving neurodegeneration in the context of PFN1-related ALS.

3.
Sci Rep ; 13(1): 12550, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37532768

ABSTRACT

The method of filling mining can solve the problem of surface subsidence caused by coal mining. Among them, it is crucial to study the mechanism of filler strength improvement timeliness and filler mining to control rock movement for filler mining. In this paper, by combining theoretical analysis and similar simulation experiments, compressive strength is used as the research parameter to conduct proportioning test research on paste filling similar materials such as coal gangue, fly ash, and cement. The results prove that the strengths of the test ratios can meet the strength design criteria and lay the foundation for the requirements of similar simulation experiments. In order to study the characteristics of overburden failure, stress and displacement in the process of filling mining, the key technical parameters of overburden movement are determined. Similar simulation experiments were conducted to study the movement and deformation of overburden rock and the displacement and stress distribution law of overburden rock in the coal mine under different filling rates and filling steps conditions. The results show that the filling rate and filling step are the keys to preventing the overlying rock from rupture and collapse, and the larger the filling rate is, the smaller the stress and displacement of the overburden; the larger the filling step is, the larger the displacement and stress change of the overburden, and vice versa. In addition, the displacement curve along the strike is basically an "arch" type distribution, and the stress variation trend is "large-small-large" with a "Z" type distribution. The research results are of great significance to guide the practice of filling mining and can provide the theoretical basis for its further promotion.

4.
Theranostics ; 12(16): 7051-7066, 2022.
Article in English | MEDLINE | ID: mdl-36276640

ABSTRACT

Rationale: The mitogen-activated protein kinase pathway (MAPK) is one of the major cancer-driving pathways found in non-small cell lung cancer (NSCLC) patients. ERK inhibitors (ERKi) have been shown to be effective in NSCLC patients with MAPK pathway mutations. However, like other MAPK inhibitors, ERKi rarely confers complete and durable responses. The mechanism of tumor relapse after ERKi treatment is yet defined. Methods: To best study the mechanism of tumor relapse after ERK inhibitor treatment in NSCLC patients, we treated various NSCLC cell lines and patient-derived xenograft (PDX) with ERK inhibitors and evaluated the enrichment of cancer stem cell (CSC) population. We then performed a Next-generation sequencing (NGS) to identify potential pathways that are responsible for the CSC enrichment. Further, the involvement of specific pathways was examined using molecular and cellular methods. Finally, we investigated the therapeutic benefits of ERKi treatment combined with JAK/STAT pathway inhibitor using cellular and xenograft NSCLC models. Results: We found that ERKi treatment expands the CSC population in NSCLC cells through enhanced epithelial-to-mesenchymal transition (EMT)-mediated cancer cell dedifferentiation. Mechanistically, ERK inactivation induces EMT via pSTAT3-mediated upregulation of Slug, in which, upregulation of miR-204 and downregulation of SPDEF, a transcription repressor of Slug, are involved. Finally, the JAK/STAT pathway inhibitor Ruxolitinib blocks the ERK inactivation-induced EMT and CSC expansion, as well as the tumor progression in xenograft models after ERKi treatment. Conclusions: This study revealed a potential tumor relapse mechanism of NSCLC after ERK inhibition through the unintended activation of the EMT program, ascertained the pSTAT-miR-204-SPDEF-Slug axis, and provided a promising combination inhibitor approach to prevent tumor relapse in patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Janus Kinases/metabolism , Cell Line, Tumor , Cell Movement , Signal Transduction , STAT Transcription Factors/metabolism , Neoplasm Recurrence, Local/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Transcription Factors/metabolism , Mitogen-Activated Protein Kinases/metabolism , MicroRNAs/pharmacology , Gene Expression Regulation, Neoplastic
5.
Am J Transl Res ; 14(2): 1220-1233, 2022.
Article in English | MEDLINE | ID: mdl-35273724

ABSTRACT

Post-therapeutic relapse remains the biggest challenge to breast cancer management. The re-initiation of proliferation of dormant tumor cells in either metastatic or primary tumor location marks the final rate-limiting step of malignancy and mortality. The underlying molecular mechanisms remain poorly understood. We have recently demonstrated that KLF8 promotes breast cancer metastasis via CXCR4 upregulation. Here we report a role and mechanisms for KLF8 in driving the recurrence-like tumor outgrowth in both secondary and primary sites in a CXCR4-dependent manner. Treatment of an MDA-MB-231 breast cancer cell variant with the CXCR4 ligand, CXCL12, induces formation of filopodia in monolayer culture and filopodium-like protrusions (FLPs) in 3D culture. The FLP+ cells proliferate significantly faster than FLP- cells in the 3D culture supplemented with CXCL12. Both the FLP formation and enhanced proliferation in the 3D culture can be prevented by silencing KLF8 expression in the cells. From this prevention, the cells can be rescued by overexpressing wild-type CXCR4 but not its inactive mutant form in the cells. Overexpression of KLF8 or CXCR4 in the cells dramatically enhances their invasive outgrowth and metastasis after being implanted into immunocompromised mice. Mechanistically, we found that the activated FAK was recruited to the nascent FLPs and that proliferation of the cells was completely prevented with a FAK-specific inhibitor. Taken together, these results shed new light on the role of KLF8 in promoting breast cancer recurrence, the fatal episode of the disease, by inducing CXCR4-dependent FLP formation.

6.
Am J Cancer Res ; 11(10): 4884-4899, 2021.
Article in English | MEDLINE | ID: mdl-34765298

ABSTRACT

Herceptin-resistant tumor relapse remains a major clinical issue responsible for the poor prognosis of HER2+ breast cancer. Understanding the underlying mechanisms and finding a therapeutic solution are of paramount urgency to improve the patient management. Here we report that anticancer redox active cerium oxide nanoparticles (CONPs) can potently sensitize the cancer cells to the cytotoxicity of Herceptin. By comparing between Herceptin-sensitive and Herceptin-resistant human breast cancer cell lines under normoxic as well as hypoxic culture conditions, we found that in the presence of CONPs, Herceptin can kill the Herceptin-resistant cells equally effectively as it kills the Herceptin-sensitive cells under the hypoxic, but not normoxic, culture conditions by inhibiting the cell viability, survival and proliferation. Signaling analysis reveals that under the normoxic conditions, the levels of hypoxia induced factor 1α as well as vascular endothelial growth factor are higher in the Herceptin-resistant cells than that in the Herceptin-sensitive cells and are strongly induced once the culture is switched to the hypoxic conditions, which can be potently suppressed by CONPs. Treatment with CONPs plus Herceptin significantly slows down the primary tumor growth and lung metastasis of the Herceptin-resistant cells in a xenograft mouse model of orthotopic breast cancer through inhibiting the cell proliferation and survival as well as tumor angiogenesis. These results shed new lights on the mechanisms underlying the Herceptin resistance of the HER2+ breast cancer and provide insights into introducing CONPs-like agents to Herceptin-based therapy to improve treatment outcomes.

7.
Am J Cancer Res ; 11(5): 2188-2201, 2021.
Article in English | MEDLINE | ID: mdl-34094677

ABSTRACT

Metastasis and drug resistance are intertwined processes that are responsible for the vast majority of patient deaths from breast cancer. The underlying mechanisms remain incompletely understood. We previously demonstrated that KLF8 activates CXCR4 transcription in metastatic breast cancer. Here, we report a novel role of KLF8-CXCR4 signaling for converting single organ metastasis into multiple organ metastasis associated with chemotherapeutic resistance. We show that KLF8 expression in metastatic breast cancer cells can be over-induced by chemotherapeutic drugs. Analysis of data from large-cohorts of patients indicates that post-chemotherapy there is a close correlation between the aberrant high levels of KLF8 and CXCR4 and that this correlation is well associated with drug resistance, metastasis, and poor prognosis. To mimic their aberrant high levels, we overexpressed KLF8 or CXCR4 in a human breast cancer cell line known to metastasize only to the lungs after intravenous injection in nude mice. As expected, these cells become more resistant to chemotherapeutic drugs. Surprisingly, these KLF8 or CXCR4 overexpressing cells, even implanted orthotopically, metastasized extensively to multiple organs particularly the CXCL12-rich organs. Tube formation assay, Ki67 staining and Western blotting revealed that KLF8 or CXCR4 overexpression enhanced angiogenesis involving increased expression and secretion of VEGF protein. We also found that KLF8 or CXCR4 overexpression strongly enhanced formation of filopodium-like protrusions and proliferation via CXCR4 stimulation in a 3D culture model mimicking the colonization step of metastasis. Taken together, these results suggest that the chemo-induction of KLF8 upregulation be critical for drug resistance and systemic metastasis through enhanced tumor angiogenesis and colonization via CXCR4 over-activation and that KLF8-CXCR4 signaling axis may be a new therapeutic target for drug-resistant breast cancer metastasis.

8.
J Neurochem ; 157(4): 1244-1252, 2021 05.
Article in English | MEDLINE | ID: mdl-32754913

ABSTRACT

Mutation of profilin 1 (PFN1) can cause amyotrophic lateral sclerosis (ALS). To assess how PFN1 mutation causes the disease, we created transgenic rats with human genomic DNA that harbors both the coding and the regulatory sequences of the human PFN1 gene. Selected transgenic lines expressed human PFN1 with or without the pathogenic mutation C71G at a moderate and a comparable level and in the similar pattern of spatial and temporal expression to rat endogenous PFN1. The artificial effects of arbitrary transgene expression commonly observed in cDNA transgenic animals were minimized in PFN1 transgenic rats. Expression of the mutant, but not the wild type, human PFN1 in rats recapitulated the cardinal features of ALS including the progressive loss of motor neurons and the subsequent denervation atrophy of skeletal muscles. Detergent-insoluble PFN1 inclusions were detected as the first pathology in otherwise asymptomatic transgenic rats expressing mutant human PFN1. The findings suggest that protein aggregation is involved in the neurodegeneration of ALS associated with PFN1 mutation. The resulting rat model is useful to mechanistic study on the ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Inclusion Bodies/pathology , Motor Neurons/pathology , Profilins/genetics , Animals , Mice , Muscle, Skeletal/pathology , Rats, Sprague-Dawley , Rats, Transgenic
9.
Am J Transl Res ; 11(3): 1521-1530, 2019.
Article in English | MEDLINE | ID: mdl-30972179

ABSTRACT

Krüppel-like factor 8 (KLF8) plays many important roles in various diseases, especially cancer. Previous studies have shown that KLF8 is regulated by ubiquitylation. The molecular mechanism underlying this posttranslational modification of KLF8, however, has not been investigated. Reported here is our identification of the neural precursor cell expressed, developmentally down-regulated 4 (NEDD4) as the E3 ubiquitin ligase for this modification. By co-immunoprecipitation and ubiquitylation assays, we determined that KLF8 interacts with NEDD4 and is ubiquitylated by NEDD4. By site-directed mutagenesis and pharmacological inhibition of MEK, we found that the ubiquitylation of KLF8 by NEDD4 depends upon the phosphorylation of KLF8 at serine 48 by ERK. Cycloheximide chase analysis, target gene promoter reporter assay and fluorescent staining indicated that NEDD4 plays a critical role in promoting the stability and transcriptional activity of KLF8 in the nucleus. Taken together, this work identified NEDD4 as a novel E3 ubiquitin ligase for KLF8 that provides insights into targeting the KLF8-NEDD4 axis to treat various types of cancer associated with overexpression of both proteins.

10.
Cancers (Basel) ; 10(9)2018 Sep 01.
Article in English | MEDLINE | ID: mdl-30200491

ABSTRACT

Side effects of radiation therapy (RT) remain the most challenging issue for pancreatic cancer treatment. Cerium oxide nanoparticles (CONPs) are currently being tested in pre-clinical trials as an adjuvant to sensitize pancreatic cancer cells to RT and protect normal tissues from the harmful side effects. CONPs were not able to significantly affect RT-induced DNA damage in cancer cells, thereby ruling out sensitization through increased mitotic catastrophe. However, activation of c-Jun terminal kinase (JNK), a key driver of RT-induced apoptosis, was significantly enhanced by co-treatment with CONPs and RT in pancreatic cancer cells in vitro and human pancreatic tumors in nude mice in vivo compared to CONPs or RT treatment alone. Further, CONP-driven increase in RT-induced JNK activity was associated with a marked increase in Caspase 3/7 activation, indicative of apoptosis. We have previously shown that CONPs increase reactive oxygen species (ROS) production in cancer cells. ROS has been shown to drive the oxidation of thioredoxin 1 (TRX1) which results in the activation of apoptosis signaling kinase 1 (ASK1). The increase in ASK1 activation following the co-treatment with CONPs followed by RT suggests that the increased JNK activation is the result of increased TRX1 oxidation. The ability of CONPs to sensitize pancreatic cancer cells to RT was mitigated when the TRX1 oxidation was prevented by mutagenesis of a cysteine residue or when the JNK activation was blocked by an inhibitor. Taken together, these data demonstrate an important mechanism for CONPs in specifically killing cancer cells and provide novel insights into the utilization of CONPs as a radiosensitizer and therapeutic agent for pancreatic cancer.

11.
Phys Chem Chem Phys ; 20(16): 11285-11295, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29637202

ABSTRACT

A series of CO2-responsive oil-in-water (O/W) emulsions were prepared by introducing hydrophobic tertiary amines (TAs) with varying alkane carbon numbers (ACNs) into the emulsion stabilized by sodium dodecyl benzene sulfonate (SDBS). TAs are converted to bicarbonate salts upon bubbling of CO2, which can form ion pairs with SDBS via electrostatic interaction, and then disrupt the stability of the emulsion. The reversible switch can be triggered by the removal of CO2. The ACN of TA, the concentration of SDBS/TA, the bubbling time of CO2, and the number of cycles are taken into account in order to study the controllable mechanism of these CO2-responsive emulsions. Because of the improved miscibility with oil, the ion pairs with TAs of larger ACNs can much more easily adhere to the oil phase, and then speed up the rupture rate of the oil droplets. The corresponding demulsification process is tracked by studying the interfacial tension, the zeta potential of the droplets, and microscope snapshots of all the systems. The UV-vis spectrophotometer analysis of the water phase and the 1H-nuclear magnetic resonance (1H NMR) test are further designed to comprehend the significance of ACNs and the solubility product of the formed ion pairs.

12.
Am J Cancer Res ; 6(5): 910-23, 2016.
Article in English | MEDLINE | ID: mdl-27293988

ABSTRACT

Krüppel-like factor 8 (KLF8) plays important roles in cancer and is strictly regulated by various post-translational modifications such as sumoylation, acetylation, ubiquitylation and PARylation. Here we report a novel phosphorylation of KLF8 by ERK2 responsible and critical for the stability of KLF8 protein. The full-length KLF8 protein displays a doublet in SDS-PAGE gel. The upper band of the doublet, however, disappeared when the N-terminal 50 amino acids were deleted. In its full-length the upper band disappeared upon phosphatase treatment or mutation of the serine 48 (S48) to alanine whereas the lower band was lost when the S48 was mutated to aspartic acid that mimics phosphorylated S48. These results suggest that S48 phosphorylation is responsible for the motility up-shift of KLF8 protein. Pharmacological and genetic manipulations of various potential kinases identified ERK2 as the likely one that phosphorylates KLF8 at S48. Functional studies indicated that this phosphorylation is crucial for protecting KLF8 protein from degradation in the nucleus and promoting cell migration. Taken together, this study identifies a novel mechanism of phosphorylation critical for KLF8 protein stabilization and function.

13.
Oncotarget ; 7(17): 23552-68, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-26993780

ABSTRACT

Krüppel-like factor 8 (KLF8) has been strongly implicated in breast cancer metastasis. However, the underlying mechanisms remain largely unknown. Here we report a novel signaling from KLF8 to C-X-C cytokine receptor type 4 (CXCR4) in breast cancer. Overexpression of KLF8 in MCF-10A cells induced CXCR4 expression at both mRNA and protein levels, as determined by quantitative real-time PCR and immunoblotting. This induction was well correlated with increased Boyden chamber migration, matrigel invasion and transendothelial migration (TEM) of the cells towards the ligand CXCL12. On the other hand, knockdown of KLF8 in MDA-MB-231 cells reduced CXCR4 expression associated with decreased cell migration, invasion and TEM towards CXCL12. Histological and database mining analyses of independent cohorts of patient tissue microarrays revealed a correlation of aberrant co-elevation of KLF8 and CXCR4 with metastatic potential. Promoter analysis indicated that KLF8 directly binds and activates the human CXCR4 gene promoter. Interestingly, a CXCR4-dependent activation of focal adhesion kinase (FAK), a known upregulator of KLF8, was highly induced by CXCL12 treatment in KLF8-overexpressing, but not KLF8 deficient cells. This activation of FAK in turn induced a further increase in KLF8 expression. Xenograft studies showed that overexpression of CXCR4, but not a dominant-negative mutant of it, in the MDA-MB-231 cells prevented the invasive growth of primary tumor and lung metastasis from inhibition by knockdown of KLF8. These results collectively suggest a critical role for a previously unidentified feed-forward signaling wheel made of KLF8, CXCR4 and FAK in promoting breast cancer metastasis and shed new light on potentially more effective anti-cancer strategies.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Focal Adhesion Kinase 1/metabolism , Lung Neoplasms/secondary , Receptors, CXCR4/metabolism , Repressor Proteins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Proliferation , Chemokine CXCL12/metabolism , Female , Humans , Kruppel-Like Transcription Factors , Lung Neoplasms/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Pharmacol Ther ; 160: 145-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26899500

ABSTRACT

Chemoresistant metastatic relapse of minimal residual disease plays a significant role for poor prognosis of cancer. Growing evidence supports a critical role of cancer stem cell (CSC) behind the mechanisms for this deadly disease. This review briefly introduces the basics of the conventional chemotherapies, updates the CSC theories, highlights the molecular and cellular mechanisms by which CSC smartly designs and utilizes multiple lines of self-defense to avoid being killed by chemotherapy, and concisely summarizes recent progress in studies on CSC-targeted therapies in the end, with the hope to help guide future research toward developing more effective therapeutic strategies to eradicate tumor cells in the patients.


Subject(s)
Drug Resistance, Neoplasm/physiology , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/drug effects , Humans , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects
15.
Oncotarget ; 6(26): 22452-66, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26084291

ABSTRACT

Recent evidence shows the emerging roles of endogenous microRNAs (miRNAs) in repressing gene transcription. However, the miRNAs inhibiting the transcription of matrix metalloproteinase 14 (MMP-14), a membrane-anchored MMP crucial for the tumorigenesis and aggressiveness, still remain largely unknown. In this study, through mining computational algorithm program and genome-wide Argonaute profiling dataset, we identified one binding site of miRNA-337-3p (miR-337-3p) within the MMP-14 promoter. We demonstrated that miR-337-3p was under-expressed and inversely correlated with MMP-14 expression in clinical specimens and cell lines of neuroblastoma (NB), the most common extracranial solid tumor in childhood. Patients with high miR-337-3p expression had greater survival probability. miR-337-3p suppressed the promoter activity, nascent transcription, and expression of MMP-14, resulting in decreased levels of vascular endothelial growth factor, in cultured NB cell lines. Mechanistically, miR-337-3p recognized its binding site and recruited Argonaute 2 to facilitate the enrichment of repressive epigenetic markers and decrease the binding of RNA polymerase II and specificity protein 1 on the MMP-14 promoter. Gain- and loss-of-function studies demonstrated that miR-337-3p suppressed the growth, invasion, metastasis, and angiogenesis of NB cells in vitro and in vivo. In addition, restoration of MMP-14 expression rescued the NB cells from changes in these biological features. Taken together, these data indicate that miR-337-3p directly binds the MMP-14 promoter to repress its transcription, thus suppressing the progression of NB.


Subject(s)
Matrix Metalloproteinase 14/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Animals , Binding Sites , Cell Line, Tumor , Disease Progression , HeLa Cells , Hep G2 Cells , Heterografts , Humans , Male , Matrix Metalloproteinase 14/biosynthesis , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , Neuroblastoma/enzymology , Neuroblastoma/pathology , Promoter Regions, Genetic , Transfection
16.
Oncotarget ; 6(25): 21428-42, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26025929

ABSTRACT

Krüppel-like factor 8 (KLF8) is a dual transcriptional factor critical for breast cancer progression. Epidermal growth factor receptor (EGFR) is frequently overexpressed in aggressive such as triple-negative breast cancer and associated with poor clinical outcomes. Here we report a novel KLF8-EGFR signaling axis in breast cancer. We identified a highly correlated co-overexpression between KLF8 and EGFR in invasive breast cancer cells and patient tumor samples. Overexpression of KLF8 in the non-tumorigenic MCF-10A cells induced the expression of EGFR, whereas knockdown of KLF8 from the MDA-MB-231 cells decreased it. Promoter activation and binding assays indicated that KLF8 promotes the EGFR expression by directly binding its gene promoter. We also revealed that KLF8 directly represses the promoter of miR141 and miR141 targets the 3'-untranslational region of EGFR transcript to inhibit EGFR translation. Treatment with the EGFR inhibitor AG1478 or overexpression of miR141 blocked the activity of ERK downstream of EGFR and inhibited KLF8-depndent cell invasiveness, proliferation and viability in cell culture and invasive growth and lung metastasis in nude mice. Conversely, overexpression of an inhibitory sponge of miR141 led to the opposite phenotypes. Taken together, these findings demonstrate a novel KLF8 to miR141/EGFR signaling pathway potentially crucial for breast cancer malignancy.


Subject(s)
Breast Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Repressor Proteins/metabolism , 3' Untranslated Regions , Animals , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , HEK293 Cells , Humans , Kruppel-Like Transcription Factors , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Metastasis , Phenotype , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Up-Regulation
17.
Cancer Lett ; 359(2): 187-97, 2015 Apr 10.
Article in English | MEDLINE | ID: mdl-25592038

ABSTRACT

Matrix metalloproteinase 14 (MMP-14) is the only membrane-anchored MMP that plays critical roles in tumorigenesis and aggressiveness. However, the regulatory mechanisms underlying the high MMP-14 expression in neuroblastoma (NB), a highly malignant tumor in childhood, still remain unclear. Herein, we applied an integrative approach to analyze the public datasets, and identified hepatocyte nuclear factor 4 alpha (HNF4α) as a crucial transcription factor facilitating the MMP-14 expression in NB. In clinical NB tissues, HNF4α was up-regulated and positively correlated with MMP-14 expression, and was an independent prognostic factor for unfavorable outcome of patients. Luciferase reporter and chromatin immunoprecipitation assays indicated that HNF4α directly targeted the binding site within the MMP-14 promoter to facilitate its transcription. Knockdown of HNF4α suppressed the invasion, metastasis and angiogenesis of NB cells in vitro and in vivo. Conversely, ectopic expression of HNF4α promoted the invasion, metastasis and angiogenesis of NB cells. Importantly, restoration of MMP-14 expression prevented the tumor cells from HNF4α-mediated changes in these biological features. Taken together, HNF4α exhibits oncogenic activity that affects the aggressiveness and angiogenesis of NB through activating the transcription of MMP-14.


Subject(s)
Hepatocyte Nuclear Factor 4/physiology , Matrix Metalloproteinase 14/metabolism , Neovascularization, Pathologic/enzymology , Neuroblastoma/enzymology , Animals , Cell Line, Tumor , Enzyme Induction , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Male , Matrix Metalloproteinase 14/genetics , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neuroblastoma/blood supply , Neuroblastoma/mortality , Neuroblastoma/pathology , Promoter Regions, Genetic , Proportional Hazards Models , Protein Binding , Transcription, Genetic , Transcriptional Activation , Tumor Burden
18.
Oncoscience ; 1(4): 248-249, 2014.
Article in English | MEDLINE | ID: mdl-25485290
19.
J Biol Chem ; 289(9): 5914-24, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24407292

ABSTRACT

The mammalian heart, the body's largest energy consumer, has evolved robust mechanisms to tightly couple fuel supply with energy demand across a wide range of physiologic and pathophysiologic states, yet, when compared with other organs, relatively little is known about the molecular machinery that directly governs metabolic plasticity in the heart. Although previous studies have defined Kruppel-like factor 15 (KLF15) as a transcriptional repressor of pathologic cardiac hypertrophy, a direct role for the KLF family in cardiac metabolism has not been previously established. We show in human heart samples that KLF15 is induced after birth and reduced in heart failure, a myocardial expression pattern that parallels reliance on lipid oxidation. Isolated working heart studies and unbiased transcriptomic profiling in Klf15-deficient hearts demonstrate that KLF15 is an essential regulator of lipid flux and metabolic homeostasis in the adult myocardium. An important mechanism by which KLF15 regulates its direct transcriptional targets is via interaction with p300 and recruitment of this critical co-activator to promoters. This study establishes KLF15 as a key regulator of myocardial lipid utilization and is the first to implicate the KLF transcription factor family in cardiac metabolism.


Subject(s)
DNA-Binding Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lipid Metabolism , Muscle Proteins/metabolism , Myocardium/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Line , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Muscle Proteins/genetics , Myocardium/pathology , Nuclear Proteins/genetics , Oxidation-Reduction , Transcription Factors/genetics
20.
Am J Cancer Res ; 3(4): 356-73, 2013.
Article in English | MEDLINE | ID: mdl-23977446

ABSTRACT

The properties of stem cells can be induced during the epithelial to mesenchymal transition (EMT). The responsible molecular mechanisms, however, remain largely undefined. Here we report the identification of the microRNA-146a (miR-146a) as a common target of Krüppel-like factor 8 (KLF8) and TGF-ß, both of which are known EMT-inducers. Upon KLF8 overexpression or TGF-ß treatment, a significant portion of the MCF-10A cells gained stem cell traits as demonstrated by an increased expression of CD44(high)/CD24low, activity of aldehyde dehydrogenase (ALDH), mammosphere formation and chemoresistance. Along with this change, the expression of miR-146a was highly upregulated in the cells. Importantly, we found that miR-146a was aberrantly co-overexpressed with KLF8 in a panel of invasive human breast cancer cell lines. Ectopic expression of KLF8 failed to induce the stem cell traits in the MCF-10A cells if the cells were pre-treated with miR-146a inhibitor, whereas overexpression of miR-146a in the MCF-10A cells alone was sufficient to induce the stem cell traits. Co-staining and luciferase reporter analyses indicated that miR-146a targets the 3'-UTR of the Notch signaling inhibitor NUMB for translational inhibition. Overexpression of KLF8 dramatically potentiated the tumorigenecity of MCF-10A cells expressing the H-Ras oncogene, which was accompanied by a loss of NUMB expression in the tumors. Taken together, this study identifies a novel role and mechanism for KLF8 in inducing pro-tumorigenic mammary stem cells via miR-146a potentially by activating Notch signaling. This mechanism could be exploited as a therapeutic target against drug resistance of breast cancer.

SELECTION OF CITATIONS
SEARCH DETAIL
...