Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 81(1): 189, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38643448

ABSTRACT

Peritoneal metastasis, the third most common metastasis in colorectal cancer (CRC), has a poor prognosis for the rapid progression and limited therapeutic strategy. However, the molecular characteristics and pathogenesis of CRC peritoneal metastasis are poorly understood. Here, we aimed to elucidate the action and mechanism of adipose-derived stem cells (ADSCs), a prominent component of the peritoneal microenvironment, in CRC peritoneal metastasis formation. Database analysis indicated that ADSCs infiltration was increased in CRC peritoneal metastases, and high expression levels of ADSCs marker genes predicted a poor prognosis. Then we investigated the effect of ADSCs on CRC cells in vitro and in vivo. The results revealed that CRC cells co-cultured with ADSCs exhibited stronger metastatic property and anoikis resistance, and ADSCs boosted the intraperitoneal seeding of CRC cells. Furthermore, RNA sequencing was carried out to identify the key target gene, angiopoietin like 4 (ANGPTL4), which was upregulated in CRC specimens, especially in peritoneal metastases. Mechanistically, TGF-ß1 secreted by ADSCs activated SMAD3 in CRC cells, and chromatin immunoprecipitation assay showed that SMAD3 facilitated ANGPTL4 transcription by directly binding to ANGPTL4 promoter. The ANGPTL4 upregulation was essential for ADSCs to promote glycolysis and anoikis resistance in CRC. Importantly, simultaneously targeting TGF-ß signaling and ANGPTL4 efficiently reduced intraperitoneal seeding in vivo. In conclusion, this study indicates that tumor-infiltrating ADSCs promote glycolysis and anoikis resistance in CRC cells and ultimately facilitate peritoneal metastasis via the TGF-ß1/SMAD3/ANGPTL4 axis. The dual-targeting of TGF-ß signaling and ANGPTL4 may be a feasible therapeutic strategy for CRC peritoneal metastasis.


Subject(s)
Colorectal Neoplasms , Peritoneal Neoplasms , Humans , Peritoneal Neoplasms/genetics , Transforming Growth Factor beta1 , Glycolysis , Colorectal Neoplasms/genetics , Stem Cells , Tumor Microenvironment , Smad3 Protein/genetics , Angiopoietin-Like Protein 4/genetics
2.
Cancer Lett ; 576: 216420, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37778684

ABSTRACT

LIM kinase 1 (LIMK1) is a member of the LIMK family that has been considered to be involved in chemoresistance in various tumors, and N6-methyladenosine (m6A) is the most abundant nucleotide modification on mRNA. However, whether elevated expression of LIMK1 leads to chemoresistance due to m6A modification remains to be further studied. The findings of our study indicate that high LIMK1 expression in colorectal cancer (CRC) cells promotes cell proliferation and increases resistance to 5-fluorouracil (5-FU). Moreover, downregulation of YTH domain-containing 2 (YTHDC2), an m6A "reader", in CRC cells resulted in decreased recognition and binding to the m6A site "GGACA" in LIMK1 mRNA, thereby increasing LIMK1 mRNA stability and expression. Furthermore, the overexpression of LIMK1 facilitated eIF2α phosphorylation, which induced endoplasmic reticulum (ER) stress and promoted stress granule (SG) formation, ultimately leading to 5-FU resistance. This study evaluated the specificity of the YTHDC2/LIMK1/eIF2α signalling axis and the efficacy of related drugs in modulating 5-FU sensitivity in CRC.


Subject(s)
Colorectal Neoplasms , Lim Kinases , Humans , Lim Kinases/genetics , Lim Kinases/metabolism , Methylation , Drug Resistance, Neoplasm/genetics , Stress Granules , RNA, Messenger/metabolism , Fluorouracil/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Endoplasmic Reticulum Stress , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , RNA Helicases/genetics , RNA Helicases/metabolism
3.
Front Plant Sci ; 14: 1257672, 2023.
Article in English | MEDLINE | ID: mdl-37780520

ABSTRACT

Introduction: Adjuvants can effectively enhance the utilization rate of pesticides, but the application of adjuvants in plant growth regulators is rarely studied. Methods: This work explored the effects of adjuvants dioctyl sulfosuccinate sodium salt (AOT) and methyl oleate (MO) on lime sulfur (LS), especially the drop behavior on flower and paraffin surface. Results: The results showed that the addition of AOT and AOT+MO can significantly reduce the static and dynamic surface tension of LS from 72mN/m to 28mN/m and 32mN/m respectively, and increase the spreading factor from 0.18 to 1.83 and 3.10 respectively, reduce the bounce factor from 2.72 to 0.37 and 0.27 respectively. The fluorescence tracer test showed that the addition of adjuvants could promote the spreading and permeation of droplets. The field test results revealed that the flower thinning rate of adjuvant and non-adjuvant were 80.55% and 54.4% respectively, and the flower thinning effect of adding adjuvant was the same as that of artificial which the flower thinning rate was 84.77%. The quality of apples treated with adjuvants was similar to that treated with artificial, and the weight of single fruit increased by 24.08% compared with CK (spray water). Discussion: The application of tank-mixture adjuvant could reduce the dosage of LS for thinning agent application, improve apple's quality, and decrease labor cost and improve the economic benefits of fruit planting and the environmental benefits of plant growth regulators.

4.
Cell Death Dis ; 14(2): 148, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36810285

ABSTRACT

miR-17-5p has been found to be involved in the proliferation and metastasis of colorectal cancer (CRC), and N6-methyladenosine (m6A) modification is the most common RNA modification in eukaryotes. However, whether miR-17-5p contributes to chemotherapy sensitivity in CRC via m6A modification is unclear. In this study, we found that overexpression of miR-17-5p led to less apoptosis and lower drug sensitivity in vitro and in vivo under the 5-fluorouracil (5-FU) treatment, which indicated miR-17-5p led to 5-FU chemotherapy resistance. Bioinformatic analysis suggested that miR-17-5p-mediated chemoresistance was associated with mitochondrial homeostasis. miR-17-5p directly bound to the 3' untranslated region of Mitofusin 2 (MFN2), leading to decreased mitochondrial fusion and enhanced mitochondrial fission and mitophagy. Meanwhile, methyltransferase-like protein 14 (METTL14) was downregulated in CRC, resulting in lower m6A level. Moreover, the low level of METTL14 promoted the expression of pri-miR-17 and miR-17-5p. Further experiments suggested that m6A mRNA methylation initiated by METTL14 inhibits pri-miR-17 mRNA decay via reducing the recognition of YTHDC2 to the "GGACC" binding site. The METTL14/miR-17-5p/MFN2 signaling axis may play a critical role in 5-FU chemoresistance in CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Colorectal Neoplasms/pathology , MicroRNAs/genetics , Fluorouracil/pharmacology , Methyltransferases/metabolism , Homeostasis , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics
5.
Cancer Cell Int ; 22(1): 230, 2022 Jul 17.
Article in English | MEDLINE | ID: mdl-35843949

ABSTRACT

BACKGROUND: The critical role of thioredoxin-interacting protein (TXNIP) in cellular sulfhydryl redox homeostasis and inflammasome activation is already widely known, however, no pan-cancer analysis is currently available. METHODS: We thus first explored the potential roles of TXNIP across thirty-three tumors mainly based on The Cancer Genome Atlas and Gene Expression Omnibus datasets. RESULTS: TXNIP is lowly expressed in most cancers, and distinct associations exist between TXNIP expression and the prognosis of tumor patients. TXNIP expression was associated with tumor mutational burden, microsatellite instability, mismatch repair genes, tumor infiltrating immune cell abundance as well as cancer-associated fibroblasts. Moreover, ubiquitin mediated proteolysis, protein post-translational modification and other related pathways were involved in the functional mechanisms of TXNIP. CONCLUSIONS: Our first pan-cancer study comprehensively revealed the carcinostatic role of TXNIP across different tumors. And this molecule may be considered as a potential immunological and prognostic biomarker.

6.
Front Pharmacol ; 12: 727704, 2021.
Article in English | MEDLINE | ID: mdl-34867333

ABSTRACT

Recent epidemiological and preclinical evidence indicates that vitamin D3 inhibits colorectal cancer (CRC) progression, but the mechanism has not been completely elucidated. This study was designed to determine the protective effects of vitamin D3 and identify crucial targets and regulatory mechanisms in CRC. First, we confirmed that 1,25(OH)2D3, the active form of vitamin D3, suppressed the aggressive phenotype of CRC in vitro and in vivo. Based on a network pharmacological analysis, N-acetyltransferase 2 (NAT2) was identified as a potential target of vitamin D3 against CRC. Clinical data of CRC patients from our hospital and bioinformatics analysis by online databases indicated that NAT2 was downregulated in CRC specimens and that the lower expression of NAT2 was correlated with a higher metastasis risk and lower survival rate of CRC patients. Furthermore, we found that NAT2 suppressed the proliferation and migration capacity of CRC cells, and the JAK1/STAT3 signaling pathway might be the underlying mechanism. Moreover, Western blot and immunofluorescence staining assays demonstrated that 1,25(OH)2D3 promoted NAT2 expression, and the chromatin immunoprecipitation assay indicated that the vitamin D receptor (VDR) transcriptionally regulated NAT2. These findings expand the potential uses of vitamin D3 against CRC and introduce VDR signaling via the enzyme NAT2 as a potential diagnostic and therapeutic target for CRC.

7.
Cell Death Dis ; 12(11): 975, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34671029

ABSTRACT

Tumors can use metabolic reprogramming to survive nutrient stress. Epigenetic regulators play a critical role in metabolic adaptation. Here we screened a sgRNA library to identify epigenetic regulators responsible for the vulnerability of colorectal cancer (CRC) cells to glucose deprivation and found that more EZH2-knockout cells survived glucose deprivation. Then, we showed that EZH2 expression was significantly downregulated in response to glucose deprivation in a glucose-sensitive CRC cell line, and EZH2-knockdown cells were more resistant to glucose deprivation. Mechanistically, EZH2 deficiency upregulated the expression of glutaminase (GLS) and promoted the production of glutamate, which in turn led to increased synthesis of intracellular glutathione (GSH) and eventually attenuated the reactive oxygen species (ROS)-mediated cell death induced by glucose deprivation. Although EZH2 functioned as an oncogene in cancer progression and EZH2 knockout abolished colorectal cancer development in a mouse model, here we revealed a mechanistic link between EZH2 and metabolic reprogramming via the direct regulation of GLS expression and observed a negative correlation between EZH2 and GLS expression in colorectal cancer tissues. These findings further confirmed the importance of heterogeneity, provided an explanation for the clinical tolerance of cancer cells to EZH2 inhibitors from the perspective of metabolism, and proposed the possibility of combining EZH2 inhibitors and glutamine metabolism inhibitors for the treatment of cancer.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Glutaminase/antagonists & inhibitors , Neoplasms/genetics , Humans , Neoplasms/pathology
8.
Nat Commun ; 11(1): 5105, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33037191

ABSTRACT

RB1 mutational inactivation is a cancer driver in various types of cancer including lung cancer, making it an important target for therapeutic exploitation. We performed chemical and genetic vulnerability screens in RB1-isogenic lung cancer pair and herein report that aurora kinase A (AURKA) inhibition is synthetic lethal in RB1-deficient lung cancer. Mechanistically, RB1-/- cells show unbalanced microtubule dynamics through E2F-mediated upregulation of the microtubule destabilizer stathmin and are hypersensitive to agents targeting microtubule stability. Inhibition of AURKA activity activates stathmin function via reduced phosphorylation and facilitates microtubule destabilization in RB1-/- cells, heavily impacting the bipolar spindle formation and inducing mitotic cell death selectively in RB1-/- cells. This study shows that stathmin-mediated disruption of microtubule dynamics is critical to induce synthetic lethality in RB1-deficient cancer and suggests that upstream factors regulating microtubule dynamics, such as AURKA, can be potential therapeutic targets in RB1-deficient cancer.


Subject(s)
Aurora Kinase A/genetics , Lung Neoplasms/genetics , Microtubules/metabolism , Retinoblastoma Binding Proteins/genetics , Stathmin/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Nude , Microtubules/genetics , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Retinoblastoma Binding Proteins/metabolism , Stathmin/genetics , Synthetic Lethal Mutations , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
9.
Aging (Albany NY) ; 12(16): 16270-16293, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32688344

ABSTRACT

CTCF is overexpressed in several cancers and plays crucial roles in regulating aggressiveness, but little is known about whether CTCF drives colorectal cancer progression. Here, we identified a tumor-promoting role for CTCF in colorectal cancer. Our study demonstrated that CTCF was upregulated in colorectal cancer specimens compared with adjacent noncancerous colorectal tissues. The overexpression of CTCF promoted colorectal cancer cell proliferation and tumor growth, while the opposite effects were observed in CTCF knockdown cells. Increased GLI1, Shh, PTCH1, and PTCH2 levels were observed in CTCF-overexpressing cells using western blot analyses. CCK-8 and apoptosis assays revealed that 5-fluorouracil chemosensitivity was negatively associated with CTCF expression. Furthermore, we identified that P53 is a direct transcriptional target gene of CTCF in colorectal cancer. Western blot and nuclear extract assays showed that inhibition of P53 can counteract Hedgehog signaling pathway repression induced by CTCF knockdown. In conclusion, we uncovered a crucial role for CTCF regulation that possibly involves the P53-Hedgehog axis and highlighted the clinical utility of colorectal cancer-specific potential therapeutic target as disease progression or clinical response biomarkers.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , CCCTC-Binding Factor/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Fluorouracil/pharmacology , Hedgehog Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , CCCTC-Binding Factor/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
10.
Cancer Lett ; 491: 22-35, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32730779

ABSTRACT

Cancer-associated fibroblasts (CAFs) are the main stromal cells in the tumour microenvironment (TME). We found that the distribution of CAFs was significantly increased with tumour progression and led to a poor prognosis. In vitro and in vivo assays revealed that CAFs enhanced colorectal cancer (CRC) metastasis. Based on extraction and identification of exosomes of CAFs and normal fibroblasts (NFs), CAFs-exo showed higher expression of miR-17-5p than NFs-exo and could deliver exosomal miR-17-5p from parental CAFs to CRC cells. Further exploration verified that miR-17-5p influenced CRC metastasis capacity and directly targeted 3'-untranslated regions (UTRs) of RUNX family transcription factor 3(RUNX3). Our findings further revealed that RUNX3 interacted with MYC proto-oncogene(MYC) and that both RUNX3 and MYC bound to the promoter of transforming growth factor beta1(TGF-ß1) at base pairs 1005-1296, thereby activating the TGF-ß signalling pathway and contributing to tumour progression. In addition, RUNX3/MYC/TGF-ß1 signalling sustained autocrine TGF-ß1 to activate CAFs, and activated CAFs released more exosomal miR-17-5p to CRC cells, forming a positive feedback loop for CRC progression. Taken together, these data provide a new understanding of the potential diagnostic value of exosomal miR-17-5p in CRC.


Subject(s)
Cancer-Associated Fibroblasts/physiology , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/physiology , Exosomes/physiology , MicroRNAs/physiology , Proto-Oncogene Proteins c-myc/physiology , Transforming Growth Factor beta1/physiology , Adult , Aged , Animals , Cell Line, Tumor , Colorectal Neoplasms/etiology , Feedback, Physiological , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Phenotype , Proto-Oncogene Mas , Signal Transduction , Transforming Growth Factor beta1/genetics , Tumor Microenvironment
11.
Int J Biol Sci ; 16(11): 1774-1784, 2020.
Article in English | MEDLINE | ID: mdl-32398948

ABSTRACT

PTEN, a tumor suppressor, is found loss of function in many cancers, including colorectal cancer. To identify the synthetic lethal compounds working with PTEN deficiency, we performed a synthetic lethality drug screening with PTEN-isogenic colorectal cancer cells. From the screening, we found that PTEN-/- colorectal cancer cells were sensitive to anacardic acid, a p300/CBP histone acetyltransferase (HAT) inhibitor. Anacardic acid significantly reduced the viability of PTEN-/- cells not in PTEN+/+ cells via inducing apoptosis. Inhibition of HAT activity of p300/CBP by anacardic acid reduced the acetylation of histones at the promoter region and inhibited the transcription of Hsp70 family of proteins. The down-regulation of Hsp70 family proteins led to the reduction of AKT-Hsp70 complex formation, AKT destabilization and decreased the level of phosphorylated AKT at Ser473, all of which are vital for the survival of PTEN-/- colorectal cells. The synthetic lethality effect of anacardic acid was further validated in tumor xenograft mice models, where PTEN-/- colorectal tumors showed greater sensitivity to anacardic acid treatment than PTEN+/+ tumors. These data suggest that anacardic acid induced synthetic lethality by inhibiting HAT activity of p300/CBP, thereby reducing Hsp70 transcription and destabilizing AKT in PTEN deficient colorectal cancer cells.


Subject(s)
Anacardic Acids/therapeutic use , Colorectal Neoplasms/drug therapy , PTEN Phosphohydrolase/deficiency , Proto-Oncogene Proteins c-akt , p300-CBP Transcription Factors/antagonists & inhibitors , Anacardic Acids/chemistry , Anacardic Acids/pharmacology , Animals , Colorectal Neoplasms/pathology , Combinatorial Chemistry Techniques , Down-Regulation , Drug Design , Drug Discovery , Gene Deletion , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasms, Experimental , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/drug therapy , Synthetic Lethal Mutations , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p300-CBP Transcription Factors/metabolism
12.
Acta Pharm Sin B ; 10(4): 615-627, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32322466

ABSTRACT

Breast cancer susceptibility gene 1 (BRCA1) is a tumor suppressor gene, which is frequently mutated in breast and ovarian cancers. BRCA1 plays a key role in the homologous recombination directed DNA repair, allowing its deficiency to act as a therapeutic target of DNA damaging agents. In this study, we found that inhibition of the class I histone deacetylases (HDAC) exhibited synthetic lethality with BRCA1 deficiency in breast cancer cells. Transcriptome profiling and validation study showed that HDAC inhibition enhanced the expression of thioredoxin interaction protein (TXNIP), causing reactive oxygen species (ROS)-mediated DNA damage. This effect induced preferential apoptosis in BRCA1 -/- breast cancer cells where DNA repair system is compromised. Two animal experiments and gene expression-associated patients' survival analysis further confirmed in vivo synthetic lethality between BRCA1 and HDAC. Finally, the combination of inhibitors of HDAC and bromodomain and extra-terminal motif (BET), another BRCA1 synthetic lethality target that also works through oxidative stress-mediated DNA damage, showed a strong anticancer effect in BRCA1 -/- breast cancer cells. Together, this study provides a new therapeutic strategy for BRCA1-deficient breast cancer by targeting two epigenetic machineries, HDAC and BET.

13.
Int J Biol Sci ; 14(10): 1175-1185, 2018.
Article in English | MEDLINE | ID: mdl-30123067

ABSTRACT

Cholesterol plays a key role in membrane protein function and signaling in endothelial cells. Thus, disturbing cholesterol trafficking is an effective approach for inhibiting angiogenesis. We recently identified astemizole (AST), an antihistamine drug, as a cholesterol trafficking inhibitor from a phenotypic screen. In this study, we found that AST induced cholesterol accumulation in the lysosome by binding to the sterol-sensing domain of Niemann-Pick disease, type C1 (NPC1), a lysosomal surface protein responsible for cholesterol transport. Inhibition of cholesterol trafficking by AST led to the depletion of membrane cholesterol, causing SREBP1 nuclear localization. The depletion of membrane cholesterol resulted in dissociation of mammalian target of rapamycin (mTOR) from the lysosomal surface and inactivation of mTOR signaling. These effects were effectively rescued by addition of exogenous cholesterol. AST inhibited endothelial cell proliferation, migration and tube formation in a cholesterol-dependent manner. Furthermore, AST inhibited zebrafish angiogenesis in a cholesterol-dependent manner. Together, our data suggest that AST is a new class of NPC1 antagonist that inhibits cholesterol trafficking in endothelial cells and angiogenesis.


Subject(s)
Astemizole/therapeutic use , Cholesterol/metabolism , Neovascularization, Pathologic/drug therapy , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Biological Transport/drug effects , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Fluorescent Antibody Technique , Human Umbilical Vein Endothelial Cells , Humans , Niemann-Pick C1 Protein/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics
14.
Nat Commun ; 9(1): 3212, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097580

ABSTRACT

ARID1A, a component of the SWI/SNF chromatin remodeling complex, is a tumor suppressor with a high frequency of inactivating mutations in many cancers. Therefore, ARID1A deficiency has been exploited therapeutically for treating cancer. Here we show that ARID1A has a synthetic lethal interaction with aurora kinase A (AURKA) in colorectal cancer (CRC) cells. Pharmacological and genetic perturbations of AURKA selectively inhibit the growth of ARID1A-deficient CRC cells. Mechanistically, ARID1A occupies the AURKA gene promoter and negatively regulates its transcription. Cells lacking ARID1A show enhanced AURKA transcription, which leads to the persistent activation of CDC25C, a key protein for G2/M transition and mitotic entry. Inhibiting AURKA activity in ARID1A-deficient cells significantly increases G2/M arrest and induces cellular multinucleation and apoptosis. This study shows a novel synthetic lethality interaction between ARID1A and AURKA and indicates that pharmacologically inhibiting the AURKA-CDC25C axis represents a novel strategy for treating CRC with ARID1A loss-of-function mutations.


Subject(s)
Aurora Kinase A/metabolism , Colorectal Neoplasms/genetics , Nuclear Proteins/deficiency , Signal Transduction , Synthetic Lethal Mutations/genetics , Transcription Factors/deficiency , cdc25 Phosphatases/metabolism , Animals , Apoptosis , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins , Drug Evaluation, Preclinical , Female , G2 Phase , Gene Knockout Techniques , Humans , Mice, Inbred BALB C , Mice, Nude , Mitosis , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic
15.
Cancer Lett ; 436: 28-37, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30118842

ABSTRACT

PTEN is a tumor suppressor found mutated in many cancers. From a synthetic lethality drug screen with PTEN-isogenic colorectal cancer cells, we found that mutant-PTEN cells were resistant to dual inhibitors of FLT3 and aurora kinase-A, including KW2449 and ENMD-2076. KW2449 significantly reduced the viability of wildtype-PTEN cells causing apoptosis, while little effect was observed in mutant-PTEN counterparts. Transcriptome profiling showed that members of PI3K-AKT signaling pathway were strongly changed in cells after KW2449 treatment, indicating a potential role of the pathway in drug resistance. We found that KW2449 caused a dose-dependent, biphasic induction of AKT phosphorylation at Ser473 in mutant-PTEN cells. Co-treatment with the inhibitors of its upstream signaling completely abolished the reactivation of AKT phosphorylation by KW2449 and reversed the drug resistant phenotype. These data suggest that reactivation of AKT phosphorylation at Ser473 is a key factor to confer drug resistant phenotype of mutant-PTEN cells to the dual inhibitors and that proper drug combinations that shut down AKT reactivation is necessary for the effective treatment of mutant-PTEN cancer with the dual inhibitors in clinical settings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Indazoles/pharmacology , PTEN Phosphohydrolase/deficiency , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Female , HCT116 Cells , Humans , Indazoles/administration & dosage , Mice, Nude , Mutation , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Piperazines/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays/methods , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
16.
Oncogene ; 37(49): 6341-6356, 2018 12.
Article in English | MEDLINE | ID: mdl-30042414

ABSTRACT

BRCA1 is a tumor suppressor frequently mutated in breast and ovarian cancer, serving it as a target for therapeutic exploitation. Here, we show that BRCA1 has a synthetic lethality interaction with an epigenetics regulator, bromodomain and extra-terminal domain (BET). BET inhibition led to gene expression changes reversing MYC-dependent transcription repression of a redox regulator, thioredoxin-interacting protein (TXNIP), via switching the promoter occupant from MYC to MondoA:MLX complex. Reversing the MYC-TXNIP axis inhibited thioredoxin activity and elevated cellular oxidative stress, causing DNA damages that are detrimental to BRCA1-deficient breast cancer cells. Tumor xenograft models and breast cancer clinical data analyses further demonstrated an in vivo synthetic lethality interaction and clinical association between BET/TXNIP and BRCA1 deficiency in the survival of breast cancer patients.


Subject(s)
BRCA1 Protein/deficiency , Breast Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , BRCA1 Protein/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Genes, BRCA1 , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Quinazolinones/pharmacology , Xenograft Model Antitumor Assays
17.
Cancer Lett ; 409: 91-103, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28923401

ABSTRACT

Cholesterol is an important modulator of membrane protein function and signaling in endothelial cells, thus making it an emerging target for anti-angiogenic agents. In this study, we employed a phenotypic screen that detects intracellular cholesterol distribution in endothelial cells (HUVEC) and identified 13 existing drugs as cholesterol trafficking inhibitors. Cepharanthine, an approved drug for anti-inflammatory and cancer management use, was amongst the candidates, which was selected for in-depth mechanistic studies to link cholesterol trafficking and angiogenesis. Cepharanthine inhibited the endolysosomal trafficking of free-cholesterol and low-density lipoprotein in HUVEC by binding to Niemann-Pick disease, type C1 (NPC1) protein and increasing the lysosomal pH. The blockade of cholesterol trafficking led to a cholesterol-dependent dissociation of mTOR from the lysosomes and inhibition of its downstream signaling. Cepharanthine inhibited angiogenesis in HUVEC and in zebrafish in a cholesterol-dependent manner. Furthermore, cepharanthine suppressed tumor growth in vivo by inhibiting angiogenesis and it enhanced the antitumor activity of the standard chemotherapy cisplatin in lung and breast cancer xenografts in mice. Altogether, these results strongly support the idea that cholesterol trafficking is a viable drug target for anti-angiogenesis and that the inhibitors identified among existing drugs, such as cepharanthine, could be potential anti-angiogenic and antitumor agents.


Subject(s)
Benzylisoquinolines/pharmacology , Cholesterol/metabolism , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Animals, Genetically Modified , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzylisoquinolines/administration & dosage , Cell Growth Processes/drug effects , Cisplatin/administration & dosage , Cisplatin/pharmacology , Drug Synergism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , Zebrafish
18.
Curr Top Med Chem ; 16(19): 2144-55, 2016.
Article in English | MEDLINE | ID: mdl-26881712

ABSTRACT

Although tremendous effort has been made over the past century to treat cancer effectively, the pace of drug development is far behind the increasing rate of cancer incidence and mortality. There are two major hurdles in anticancer drug development: dose-limiting toxic side effects that reduce either drug effectiveness or the quality of life of patients and complicated drug development processes that are costly and time consuming. Drug repositioning has recently gained increasing attention among cancer researchers as this approach utilizes existing drugs and is significantly cost- and time-effective. Existing drugs, particularly non-cancer drugs, have favorable safety profiles in humans and serve as an ever-increasing source for new anticancer drug discovery. Here we review the recent examples of drug repositioning of existing non-cancer drugs for preclinical and clinical introductions of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Repositioning/methods , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Disulfiram/chemistry , Disulfiram/pharmacology , Doxycycline/chemistry , Doxycycline/pharmacology , High-Throughput Screening Assays , Humans , Mebendazole/chemistry , Mebendazole/pharmacology , Neoplasms/drug therapy , Pyrvinium Compounds/chemistry , Pyrvinium Compounds/pharmacology , Triclosan/chemistry , Triclosan/pharmacology
19.
Nat Commun ; 5: 3149, 2014.
Article in English | MEDLINE | ID: mdl-24476626

ABSTRACT

R-spondins are a family of secreted Wnt agonists. One of the family members, R-spondin 2 (RSPO2), has an important role in embryonic development, bone formation and myogenic differentiation; however, its role in human cancers remains largely unknown. Here we show that RSPO2 expression is downregulated in human colorectal cancers (CRCs) due to promoter hypermethylation, and that the RSPO2 reduction correlates with tumour differentiation, size and metastasis. Overexpression of RSPO2 suppresses CRC cell proliferation and tumorigenicity, whereas the depletion of RSPO2 enhances tumour cell growth. RSPO2 has an inhibitory effect on Wnt/ß-catenin signaling in the CRC cells that show suppressed cell proliferation. In human CRC cells, the RSPO2-induced inhibition of Wnt signaling depends on leucine-rich repeat-containing G-protein-coupled receptor 5 (LGR5); RSPO2 interacts with LGR5 to stabilize the membrane-associated zinc and ring finger 3 (ZNRF3). Our data suggest that RSPO2 functions as a tumour suppressor in human CRCs, and these data reveal a RSPO2-induced, LGR5-dependent Wnt signaling-negative feedback loop that exerts a net growth-suppressive effect on CRC cells.


Subject(s)
Colorectal Neoplasms/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Down-Regulation , Female , Humans , Male , Middle Aged
20.
World J Gastroenterol ; 19(40): 6814-24, 2013 Oct 28.
Article in English | MEDLINE | ID: mdl-24187456

ABSTRACT

AIM: To investigate the effects of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) deficiency on the cytotoxicity of chemotherapeutic agents toward colorectal cancer cells. METHODS: PTEN-deficient colorectal cancer (CRC) cells were generated by human somatic cell gene targeting using the adeno-associated virus system. The cytotoxic effects of compounds including curcumin, 5-fluorouracil (5-FU), dihydroartemisinin (DHA), irinotecan (CPT-11) and oxaliplatin (OXA) on cancer cells were determined using the MTT assay. Enhanced cytotoxicity of curcumin in PTEN-deficient CRC cells was observed, and this was confirmed using clonogenic assays. Apoptosis and cell cycle progression were analyzed by flow cytometry. Levels of apoptosis and cell cycle-related proteins were examined by Western blotting. RESULTS: We developed an isogenic set of CRC cell lines that differed only in their PTEN status. Using this set of cell lines, we found that disruption of the PTEN gene had no effect on the sensitivity of CRC cells to 5-FU, CPT-11, DHA, or OXA, whereas PTEN disruption increased the sensitivity of CRC cells to curcumin. Loss of PTEN did not alter the curcumin-induced apoptosis in CRC cells. However, PTEN deficiency led to an altered pattern of curcumin-mediated cell cycle arrest. In HCT116 PTEN (+/+) cells, curcumin caused a G2/M phase arrest, whereas it caused a G0/G1 phase arrest in HCT116 PTEN (-/-) cells. Levels of cell cycle-related proteins were consistent with these respective patterns of cell cycle arrest. CONCLUSION: Curcumin shows enhanced cytotoxicity toward PTEN-deficient cancer cells, suggesting that it might be a potential chemotherapeutic agent for cancers harboring PTEN mutations.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/enzymology , Curcumin/pharmacology , PTEN Phosphohydrolase/deficiency , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Down-Regulation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , HCT116 Cells , Humans , PTEN Phosphohydrolase/genetics , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...