Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Cell Commun Signal ; 22(1): 211, 2024 Apr 02.
Article En | MEDLINE | ID: mdl-38566191

The EP300-ZNF384 fusion gene is an oncogenic driver in B-cell acute lymphoblastic leukemia (B-ALL). In the present study, we demonstrated that EP300-ZNF384 substantially induces the transcription of IL3RA and the expression of IL3Rα (CD123) on B-ALL cell membranes. Interleukin 3 (IL-3) supplementation promotes the proliferation of EP300-ZNF348-positive B-ALL cells by activating STAT5. Conditional knockdown of IL3RA in EP300-ZF384-positive cells inhibited the proliferation in vitro, and induced a significant increase in overall survival of mice, which is attributed to impaired propagation ability of leukemia cells. Mechanistically, the EP300-ZNF384 fusion protein transactivates the promoter activity of IL3RA by binding to an A-rich sequence localized at -222/-234 of IL3RA. Furthermore, forced EP300-ZNF384 expression induces the expression of IL3Rα on cell membranes and the secretion of IL-3 in CD19-positive B precursor cells derived from healthy individuals. Doxorubicin displayed a selective killing of EP300-ZNF384-positive B-ALL cells in vitro and in vivo. Collectively, we identify IL3RA as a direct downstream target of EP300-ZNF384, suggesting CD123 is a potent biomarker for EP300-ZNF384-driven B-ALL. Targeting CD123 may be a novel therapeutic approach to EP300-ZNF384-positive patients, alternative or, more likely, complementary to standard chemotherapy regimen in clinical setting.


Precursor Cell Lymphoblastic Leukemia-Lymphoma , Trans-Activators , Animals , Humans , Mice , Doxorubicin , E1A-Associated p300 Protein , Interleukin-3 , Interleukin-3 Receptor alpha Subunit , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Trans-Activators/metabolism
2.
Exp Hematol Oncol ; 12(1): 19, 2023 Feb 17.
Article En | MEDLINE | ID: mdl-36797781

Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a refractory and recurrent subtype of B-cell ALL enriched with kinase-activating rearrangements. Incomplete understanding of the heterogeneity within the tumor cells presents a major challenge for the diagnosis and therapy of Ph-like ALL. Here, we exhibited a comprehensive cell atlas of one Ph-like ALL patient with a novel TPR-PDGFRB fusion gene at diagnosis and relapse by using single-cell RNA sequencing (scRNA-seq). Twelve heterogeneous B-cell clusters, four with strong MKI67 expression indicating highly proliferating B cells, were identified. A relapse-enriched B-cell subset associated with poor prognosis was discovered, implicating the transcriptomic evolution during disease progression. Integrative single-cell analysis was performed on Ph-like ALL and Ph+ ALL patients, and revealed Ph-like specific B-cell subpopulations and shared malignant B cells characterized by the ectopic expression of the inhibitory receptor CLEC2D. Collectively, scRNA-seq of Ph-like ALL with a novel TPR-PDGFRB fusion gene provides valuable insights into the underlying heterogeneity associated with disease progression and offers useful information for the development of immunotherapeutic techniques in the future.

3.
Front Oncol ; 12: 887035, 2022.
Article En | MEDLINE | ID: mdl-36568166

Breast cancer stem cells are responsible for cancer initiation, progression, and drug resistance. However, effective targeting strategies against the cell subpopulation are still limited. Here, we unveil two splice variants of very-low-density lipoprotein receptor, VLDLR-I and -II, which are highly expressed in breast cancer stem cells. In breast cancer cells, VLDLR silencing suppresses sphere formation abilities in vitro and tumor growth in vivo. We find that VLDLR knockdown induces transition from self-renewal to quiescence. Surprisingly, ligand-binding activity is not involved in the cancer-promoting functions of VLDLR-I and -II. Proteomic analysis reveals that citrate cycle and ribosome biogenesis-related proteins are upregulated in VLDLR-I and -II overexpressed cells, suggesting that VLDLR dysregulation is associated with metabolic and anabolic regulation. Moreover, high expression of VLDLR in breast cancer tissues correlates with poor prognosis of patients. Collectively, these findings indicate that VLDLR may be an important therapeutic target for breast cancer treatment.

4.
Biomaterials ; 290: 121848, 2022 11.
Article En | MEDLINE | ID: mdl-36306684

Mesenchymal stem cells (MSCs) play a critical role in tumor metastasis. However, the dynamic process of MSCs-mediated cancer cell invasion remains inconclusive. In breast cancer mouse models, we observed that MSCs promoted lung metastasis. We constructed a microfluidic-based 3D co-culture device to monitor MSCs-mediated cancer cell invasion in a nutrient-deficient hypoxic microenvironment. On biomimetic microfluidic devices, MSCs guided cancer cell migration in a "cluster-sprout-infiltrating" mode. Importantly, hypoxic conditions significantly promoted MSCs migration at the infiltration stage, leading to accelerated breast cancer cell invasion. Moreover, hypoxia related LncRNA analysis showed that H19 was dramatically upregulated in response to hypoxic conditions. Conversely, H19 depletion impaired MSCs-directed breast cancer cell invasion. Mechanistically, H19 functions as a competitive endogenous RNA (ceRNA) which sequesters miRNA let-7 to release its target matrix metalloproteinase-1 (MMP1). Intriguingly, aspirin dramatically suppressed H19 and MMP1 expression and blocked MSCs infiltration under hypoxic conditions, resulting in alleviated breast cancer cell invasion. These findings point to the metastatic promoting role of MSCs in tumor stroma and suggest that MSCs might be a therapeutic target for metastatic breast cancer.


Mesenchymal Stem Cells , RNA, Long Noncoding , Mice , Animals , Matrix Metalloproteinase 1/metabolism , Microfluidics , Cell Line, Tumor , Cell Movement/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mesenchymal Stem Cells/metabolism , Hypoxia/metabolism , Neoplasm Invasiveness , Cell Proliferation , Tumor Microenvironment
5.
Adv Sci (Weinh) ; 9(22): e2104823, 2022 08.
Article En | MEDLINE | ID: mdl-35652200

AURKA is a potential kinase target in various malignancies. The kinase-independent oncogenic functions partially disclose the inadequate efficacy of the kinase inhibitor in a Phase III clinical trial. Simultaneously targeting the catalytic and noncatalytic functions of AURKA may be a feasible approach. Here, a set of AURKA proteolysis targeting chimeras (PROTACs) are developed. The CRBN-based dAurA383 preferentially degrades the highly abundant mitotic AURKA, while cIAP-based dAurA450 degrades the lowly abundant interphase AURKA in acute myeloid leukemia (AML) cells. The proteomic and transcriptomic analyses indicate that dAurA383 triggers the "mitotic cell cycle" and "stem cell" processes, while dAurA450 inhibits the "MYC/E2F targets" and "stem cell" processes. dAurA383 and dAurA450 are combined as a PROTAC cocktail. The cocktail effectively degrades AURKA, relieves the hook effect, and synergistically inhibits AML stem cells. Furthermore, the PROTAC cocktail induces AML regression in a xenograft mouse model and primary patient blasts. These findings establish the PROTAC cocktail as a promising spatial-temporal drug administration strategy to sequentially eliminate the multifaceted functions of oncoproteins, relieve the hook effect, and prevent cancer stem cell-mediated drug resistance.


Aurora Kinase A , Leukemia, Myeloid, Acute , Animals , Humans , Mice , Aurora Kinase A/metabolism , Cell Line, Tumor , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proteomics
7.
Signal Transduct Target Ther ; 7(1): 97, 2022 04 01.
Article En | MEDLINE | ID: mdl-35361747

Aberrant RNA splicing produces alternative isoforms of genes to facilitate tumor progression, yet how this process is regulated by oncogenic signal remains largely unknown. Here, we unveil that non-canonical activation of nuclear AURKA promotes an oncogenic RNA splicing of tumor suppressor RBM4 directed by m6A reader YTHDC1 in lung cancer. Nuclear translocation of AURKA is a prerequisite for RNA aberrant splicing, specifically triggering RBM4 splicing from the full isoform (RBM4-FL) to the short isoform (RBM4-S) in a kinase-independent manner. RBM4-S functions as a tumor promoter by abolishing RBM4-FL-mediated inhibition of the activity of the SRSF1-mTORC1 signaling pathway. Mechanistically, AURKA disrupts the binding of SRSF3 to YTHDC1, resulting in the inhibition of RBM4-FL production induced by the m6A-YTHDC1-SRSF3 complex. In turn, AURKA recruits hnRNP K to YTHDC1, leading to an m6A-YTHDC1-hnRNP K-dependent exon skipping to produce RBM4-S. Importantly, the small molecules that block AURKA nuclear translocation, reverse the oncogenic splicing of RBM4 and significantly suppress lung tumor progression. Together, our study unveils a previously unappreciated role of nuclear AURKA in m6A reader YTHDC1-dependent oncogenic RNA splicing switch, providing a novel therapeutic route to target nuclear oncogenic events.


Alternative Splicing , Aurora Kinase A , Nerve Tissue Proteins , RNA Splicing Factors , RNA-Binding Proteins , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Nucleus/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA Splicing , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
8.
Cancer Cell Int ; 22(1): 40, 2022 Jan 26.
Article En | MEDLINE | ID: mdl-35081975

BACKGROUND: Rearrangements involving the fibroblast growth factor receptor 1 (FGFR1) gene result in 8p11 myeloproliferative syndrome (EMS), which is a rare and aggressive hematological malignancy that is often initially diagnosed as myelodysplastic syndrome (MDS). Clinical outcomes are typically poor due to relative resistance to tyrosine kinase inhibitors (TKIs) and rapid transformation to acute leukemia. Deciphering the transcriptomic signature of FGFR1 fusions may open new treatment strategies for FGFR1 rearrangement patients. METHODS: DNA sequencing (DNA-seq) was performed for 20 MDS patients and whole exome sequencing (WES) was performed for one HOOK3-FGFR1 fusion positive patient. RNA sequencing (RNA-seq) was performed for 20 MDS patients and 8 healthy donors. Fusion genes were detected using the STAR-Fusion tool. Fluorescence in situ hybridization (FISH), quantitative real-time PCR (qRT-PCR), and Sanger sequencing were used to confirm the HOOK3-FGFR1 fusion gene. The phosphorylation antibody array was performed to validate the activation of nuclear factor-kappaB (NF-kappaB) signaling. RESULTS: We identified frequently recurrent mutations of ASXL1 and U2AF1 in the MDS cohort, which is consistent with previous reports. We also identified a novel in-frame HOOK3-FGFR1 fusion gene in one MDS case with abnormal monoclonal B-cell lymphocytosis and ring chromosome 8. FISH analysis detected the FGFR1 break-apart signal in myeloid blasts only. qRT-PCR and Sanger sequencing confirmed the HOOK3-FGFR1 fusion transcript with breakpoints located at the 11th exon of HOOK3 and 10th exon of FGFR1, and Western blot detected the chimeric HOOK3-FGFR1 fusion protein that is presumed to retain the entire tyrosine kinase domain of FGFR1. The transcriptional feature of HOOK3-FGFR1 fusion was characterized by the significant enrichment of the NF-kappaB pathway by comparing the expression profiling of FGFR1 fusion positive MDS with 8 healthy donors and FGFR1 fusion negative MDS patients. Further validation by phosphorylation antibody array also showed NF-kappaB activation, as evidenced by increased phosphorylation of p65 (Ser 536) and of IKBalpha (Ser 32). CONCLUSIONS: The HOOK3-FGFR1 fusion gene may contribute to the pathogenesis of MDS and activate the NF-kappaB pathway. These findings highlight a potential novel approach for combination therapy for FGFR1 rearrangement patients.

9.
Neurosci Lett ; 770: 136305, 2022 01 23.
Article En | MEDLINE | ID: mdl-34699942

Serotonin (5-hydroxytraptamine, 5-HT) is a neurotransmitter plays important roles in emotion and motivation. The action of 5-HT varies across nucleus and the receptor sub-types. Lateral habenula (LHb) in a brain area reciprocally connects with raphe nucleus and plays important roles in emotion and depression. In this study, we aimed to study the role of 5-HT1 receptor in LHb on fear learning. 15 min before or immediate after the fear conditioning, 5-Carboxyamidotrypamine maleate salt (5-CT), an agonist of 5-HT1 receptor, was bilaterally delivered into LHb (1 µg/µl, 1 µl/side) in rats. We found that activation of 5-HT1 receptor in LHb impaired the acquisition but not consolidation of fear memory in rats, which was accompanied by impaired long-term potentiation (LTP) and decreased phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR) subunit 1 (GluA1) at the Ser845 site in hippocampus. In addition, 5-CT decreased the time spent in center area of the open field and time spent in open arm in elevated plus maze. These results suggest that activation of 5-HT1 receptor in LHb impaired acquisition of hippocampal dependent fear memory and increased anxiety- like behavior in rats.


Habenula/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Memory , Receptors, Serotonin, 5-HT1/metabolism , Animals , Fear , Habenula/drug effects , Habenula/physiology , Hippocampus/drug effects , Hippocampus/physiology , Male , Maze Learning , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Serotonin 5-HT1 Receptor Agonists/pharmacology
10.
Signal Transduct Target Ther ; 6(1): 333, 2021 09 04.
Article En | MEDLINE | ID: mdl-34482361

Application of differentiation therapy targeting cellular plasticity for the treatment of solid malignancies has been lagging. Nasopharyngeal carcinoma (NPC) is a distinctive cancer with poor differentiation and high prevalence of Epstein-Barr virus (EBV) infection. Here, we show that the expression of EBV latent protein LMP1 induces dedifferentiated and stem-like status with high plasticity through the transcriptional inhibition of CEBPA. Mechanistically, LMP1 upregulates STAT5A and recruits HDAC1/2 to the CEBPA locus to reduce its histone acetylation. HDAC inhibition restored CEBPA expression, reversing cellular dedifferentiation and stem-like status in mouse xenograft models. These findings provide a novel mechanistic epigenetic-based insight into virus-induced cellular plasticity and propose a promising concept of differentiation therapy in solid tumor by using HDAC inhibitors to target cellular plasticity.


CCAAT-Enhancer-Binding Proteins/genetics , Histone Deacetylase Inhibitors/pharmacology , Nasopharyngeal Carcinoma/drug therapy , STAT5 Transcription Factor/genetics , Viral Matrix Proteins/genetics , Animals , Cell Dedifferentiation/drug effects , Cell Line, Tumor , Cell Plasticity/drug effects , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/pathology , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/pathogenicity , Heterografts , Humans , Mice , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/virology
11.
Ying Yong Sheng Tai Xue Bao ; 32(5): 1783-1790, 2021 May.
Article Zh | MEDLINE | ID: mdl-34042374

Root exudates are important carriers for material exchange and information transfer between plant and soil, and important regulators of crop-soil-microorganism interaction in intercropping systems. We examined the interaction between crops in intercropping system by setting three treatments, monoculture Chinese milk vetch, monoculture rape and Chinese milk vetch intercropped with rape. The responses of root exudates were emphatically analyzed. The results showed that 391 root exudates were detected, with 93 of which being identified and divided into nine types of metabo-lites. Among them, organooxygen compounds were the most abundant, mainly in the form of ribitol. Under different planting patterns, root exudates of Chinese milk vetch and rape were significantly different. The characteristics of root exudates in intercropping were similar to monoculture rape, but significantly different from monoculture Chinese milk vetch. Among the root exudates in different planting modes, only 9-fluorenone 1 was negatively correlated with others. The differential root exudates were mainly benzenoids, lipids and lipid-like molecules, organic acids and derivatives, and organooxygen compounds. The benzenoids, lipids and lipid-like molecules were important types that characterized the changes of root exudates of Chinese milk vetch and rape. Chinese milk vetch intercropping with rape changed the characteristics of root exudates, which were closely related to benzenoids, lipids, and lipid-like molecules.


Astragalus Plant , Rape , Agriculture , China , Exudates and Transudates , Soil
12.
Cell Death Dis ; 9(11): 1120, 2018 11 02.
Article En | MEDLINE | ID: mdl-30389909

Estrogen receptor ß (ERß) plays critical roles in thyroid cancer progression. However, its role in thyroid cancer stem cell maintenance remains elusive. Here, we report that ERß is overexpressed in papillary thyroid cancer stem cells (PTCSCs), whereas ablation of ERß decreases stemness-related factors expression, diminishes ALDH+ cell populations, and suppresses sphere formation ability and tumor growth. Screening estrogen-responsive lncRNAs in PTC spheroid cells, we find that lncRNA-H19 is highly expressed in PTCSCs and PTC tissue specimens, which is correlated with poor overall survival. Mechanistically, estradiol (E2) significantly promotes H19 transcription via ERß and elevates H19 expression. Silencing of H19 inhibits E2-induced sphere formation ability. Furthermore, H19 acting as a competitive endogenous RNA sequesters miRNA-3126-5p to reciprocally release ERß expression. ERß depletion reverses H19-induced stem-like properties upon E2 treatment. Appropriately, ERß is upregulated in PTC tissue specimens. Notably, aspirin attenuates E2-induced cancer stem-like traits through decreasing both H19 and ERß expression. Collectively, our findings reveal that ERß-H19 positive feedback loop has a compelling role in PTCSC maintenance under E2 treatment and provides a potential therapeutic targeting strategy for PTC.


Estrogen Receptor beta/genetics , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Antineoplastic Agents/pharmacology , Aspirin/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Feedback, Physiological , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA, Long Noncoding/metabolism , Signal Transduction , Survival Analysis , Thyroid Cancer, Papillary/drug therapy , Thyroid Cancer, Papillary/mortality , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology
13.
Eur J Med Chem ; 145: 805-812, 2018 Feb 10.
Article En | MEDLINE | ID: mdl-29358147

The inhibition of the members of aurora kinase family using ATP-competitive small molecules is an effective method for anticancer therapeutics. Based on our previous work, we synthesized 12 new N-trisubstituted pyrimidine derivatives and evaluated their biological activities and stabilities. Among them, compound 11j showed the best inhibition against aurora A kinase (IC50 = 7.1 nM), human leukemia cell line U937 (IC50 = 12.2 nM) and the growth of U937 xenograft tumors in vivo. By the flow cytometry and immunofluorescence analysis of U937, we found that compound 11j can induced polyploidy formation including (4N, 8N and 16N) and induce defects in both chromosome alignment and spindle formation. Furthermore, compound 11j exhibited good chemical, physical, and thermal stabilities. All these results suggested that 11j is a promising lead compound for further development of anticancer drugs.


Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , U937 Cells
14.
Cell Physiol Biochem ; 44(3): 1051-1063, 2017.
Article En | MEDLINE | ID: mdl-29179207

BACKGROUND/AIMS: Cancer stem cells (CSCs) are considered to be responsible for tumor relapse and metastasis, which serve as a potential therapeutic target for cancer. Aspirin has been shown to reduce cancer risk and mortality, particularly in colorectal cancer. However, the CSCs-suppressing effect of aspirin and its relevant mechanisms in colorectal cancer remain unclear. METHODS: CCK8 assay was employed to detect the cell viability. Sphere formation assay, colony formation assay, and ALDH1 assay were performed to identify the effects of aspirin on CSC properties. Western blotting was performed to detect the expression of the stemness factors. Xenograft model was employed to identify the anti-cancer effects of aspirin in vivo. Unpaired Student t test, ANOVA test and Kruskal-Wallis test were used for the statistical comparisons. RESULTS: Aspirin attenuated colonosphere formation and decreased the ALDH1 positive cell population of colorectal cancer cells. Aspirin inhibited xenograft tumor growth and reduced tumor cells stemness in nude mice. Consistently, aspirin decreased the protein expression of stemness-related transcription factors, including c-Myc, OCT4 and NANOG. Suppression of NANOG blocked the effect of aspirin on sphere formation. Conversely, ectopic expression of NANOG rescued the aspirin-repressed sphere formation, suggesting that NANOG is a key downstream target. Moreover, we found that aspirin repressed NANOG expression in protein level by decreasing its stability. CONCLUSION: We have provided new evidence that aspirin attenuates CSC properties through down-regulation of NANOG, suggesting aspirin as a promising therapeutic agent for colorectal cancer treatment.


Aspirin/toxicity , Cell Proliferation/drug effects , Nanog Homeobox Protein/metabolism , Animals , Aspirin/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nanog Homeobox Protein/antagonists & inhibitors , Nanog Homeobox Protein/genetics , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Protein Stability/drug effects , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , RNA, Small Interfering/metabolism , SOXB1 Transcription Factors/metabolism , Transplantation, Heterologous
15.
Biochim Biophys Acta Gen Subj ; 1861(7): 1788-1800, 2017 Jul.
Article En | MEDLINE | ID: mdl-28341484

BACKGROUND: The mechanisms of breast cancer collective invasion are poorly understood limiting the metastasis therapy. The ATPase RUVBL1 is frequently overexpressed in various cancers and plays a crucial role in oncogenic process. We further investigated the role of RUVBL1 in promoting collective invasion and uncovered that targeting RUVBL1 could inhibit metastatic progression. METHODS: The expression levels of RUVBL1 and ITFG1 were examined by Western blot and qRT-PCR. Co-localization and interaction of RUVBL1 and ITFG1 were determined by immunofluorescence and co-immunoprecipitation. The invasive ability was examined by transwell assay and microfluidic assay. The metastatic and tumorigenic abilities of breast cancer cells were revealed in BALB/c nude mice by xenograft and tail vein injection. RESULTS: ATPase RUVBL1 is highly expressed in breast cancer and predicts the poor prognosis. Elevated expression of RUVBL1 is found in high metastatic breast cancer cells. Silencing RUVBL1 suppresses cancer cell expansion and invasion in vitro and in vivo. RUVBL1 interacts with a conserved transmembrane protein ITFG1 in cytoplasm and plasma membrane to promote the collective invasion. Using a microfluidic model, we demonstrated that silencing RUVBL1 or ITFG1 individually compromises collective invasion of breast cancer cells. CONCLUSION: RUVBL1 is a vital regulator for collective invasion. The interaction between RUVBL1 and ITFG1 is required for breast cancer cell collective invasion and progression. GENERAL SIGNIFICANCE: Targeting collective invasion promoted by RUVBL1-ITFG1 complex provides a novel therapeutic strategy to improve the prognosis of invasive breast cancer.


Breast Neoplasms/pathology , Carrier Proteins/physiology , DNA Helicases/physiology , Membrane Proteins/physiology , ATPases Associated with Diverse Cellular Activities , Animals , Carrier Proteins/analysis , Cell Line, Tumor , DNA Helicases/analysis , Epithelial-Mesenchymal Transition , Female , Humans , Membrane Proteins/analysis , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Binding
16.
Oncotarget ; 6(8): 6326-40, 2015 Mar 20.
Article En | MEDLINE | ID: mdl-25811972

Cancer stem-like cell (CS-like cell) is considered to be responsible for recurrence and drug resistance events in breast cancer, which makes it a potential target for novel cancer therapeutic strategy. The FDA approved flubendazole, has been widely used in the treatment of intestinal parasites. Here, we demonstrated a novel effect of flubendazole on breast CS-like cells. Flubendazole inhibited breast cancer cells proliferation in dose- and time-dependent manner and delayed tumor growth in xenograft models by intraperitoneal injection. Importantly, flubendazole reduced CD44high/CD24low subpopulation and suppressed the formation of mammosphere and the expression of self-renewal related genes including c-myc, oct4, sox2, nanog and cyclinD1. Moreover, we found that flubendazole induced cell differentiation and inhibited cell migration. Consistently, flubendazole reduced mesenchymal markers (ß-catenin, N-cadherin and Vimentin) expression and induced epithelial and differentiation marker (Keratin 18) expression in breast cancer cells. Mechanism study revealed that flubendazole arrested cell cycle at G2/M phase and induced monopolar spindle formation through inhibiting tubulin polymerization. Furthermore, flubendazole enhanced cytotoxic activity of conventional therapeutic drugs fluorouracil and doxorubicin against breast cancer cells. In conclusion, our findings uncovered a remarkable effect of flubendazole on suppressing breast CS-like cells, indicating a novel utilization of flubendazole in breast cancer therapy.


Breast Neoplasms/drug therapy , Mebendazole/analogs & derivatives , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Antinematodal Agents/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , MCF-7 Cells , Mebendazole/administration & dosage , Mebendazole/pharmacology , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Random Allocation , Xenograft Model Antitumor Assays
17.
Cancer Biol Ther ; 16(3): 466-76, 2015.
Article En | MEDLINE | ID: mdl-25778879

Glucocorticoid (GC) resistance remains a major obstacle to successful treatment of lymphoid malignancies. Till now, the precise mechanism of GC resistance remains unclear. In the present study, dexamethasone (Dex) inhibited cell proliferation, arrested cell cycle in G0/G1-phase, and induced apoptosis in Dex-sensitive acute lymphoblastic leukemia cells. However, Dex failed to cause cell death in Dex-resistant lymphoid malignant cells. Intriguingly, we found that autophagy was induced by Dex in resistant cells, as indicated by autophagosomes formation, LC3-I to LC3-II conversion, p62 degradation, and formation of acidic autophagic vacuoles. Moreover, the results showed that Dex reduced the activity of mTOR pathway, as determined by decreased phosphorylation levels of mTOR, Akt, P70S6K and 4E-BP1 in resistant cells. Inhibition of autophagy by either chloroquine (CQ) or 3-methyladenine (3-MA) overcame Dex-resistance in lymphoid malignant cells by increasing apoptotic cell death in vitro. Consistently, inhibition of autophagy by stably knockdown of Beclin1 sensitized Dex-resistant lymphoid malignant cells to induction of apoptosis in vivo. Thus, inhibition of autophagy has the potential to improve lymphoid malignancy treatment by overcoming GC resistance.


Autophagy/drug effects , Dexamethasone/administration & dosage , Glucocorticoids/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Apoptosis/drug effects , Autophagy/genetics , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics
18.
Mol Cancer Ther ; 13(8): 1991-2003, 2014 Aug.
Article En | MEDLINE | ID: mdl-24899685

Chemoresistance is a major cause of cancer treatment failure. Tumor-initiating cells (TIC) have attracted a considerable amount of attention due to their role in chemoresistance and tumor recurrence. Here, we evaluated the small molecule Aurora kinase inhibitor AKI603 as a novel agent against TICs in breast cancer. AKI603 significantly inhibited Aurora-A (AurA) kinase and induced cell-cycle arrest. In addition, the intragastric administration of AKI603 reduced xenograft tumor growth. Interestingly, we found that breast cancer cells that were resistant to epirubicin expressed a high level of activated AurA and also have a high CD24(Low)/CD44(High) TIC population. The inhibition of AurA kinase by AKI603 abolished the epirubicin-induced enrichment of TICs. Moreover, AKI603 suppressed the capacity of cells to form mammosphere and also suppressed the expression of self-renewal genes (ß-catenin, c-Myc, Sox2, and Oct4). Thus, our work suggests the potential clinical use of the small molecule Aurora kinase inhibitor AKI603 to overcome drug resistance induced by conventional chemotherapeutics in breast cancer.


Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm , Neoplastic Stem Cells/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Aurora Kinase A/antagonists & inhibitors , Breast Neoplasms/pathology , Cell Cycle Checkpoints , Cell Proliferation , Drug Synergism , Epirubicin/pharmacology , Female , Humans , MCF-7 Cells , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Spheroids, Cellular/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
19.
Clin Transl Oncol ; 15(2): 106-10, 2013 Feb.
Article En | MEDLINE | ID: mdl-23011854

Annexin family proteins are a well-known multigene family of Ca(2+)-regulated phospholipid- and membrane-binding proteins. As one of the annexin family genes/proteins, accumulated researches have begun to reveal that annexin A3 (Anxa3) exhibits important roles in tumor development, metastasis and drug resistance. The summarized research evidences in recent years indicate Anxa3 might specifically functionalize either as a tumor suppressor or as a tumor promoter candidate for different cancers depending on the types of tumor cells and tissues. The up-regulation of Anxa3 was found to be correlated with enhanced drug resistance of ovarian cancer, to promote the developments of colorectal adenocarcinoma and pancreatic carcinoma, and to facilitate the metastases of lung adenocarcinoma and hepatocarcinoma; meanwhile, the decreased Anxa3 expressions was negatively correlated with the developments of prostatic carcinoma and renal carcinoma. It is conceivable that Anxa3 could be regarded as a target for therapeutic intervention and a biological indicator for tumor development, invasion and metastasis as well as for the prognosis of tumor patients.


Annexin A3/metabolism , Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Humans
...