Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters











Publication year range
3.
Oncogene ; 42(12): 869-880, 2023 03.
Article in English | MEDLINE | ID: mdl-36721000

ABSTRACT

Targeting cyclin-dependent kinases (CDKs) has recently emerged as a promising therapeutic approach against cancer. However, the anticancer mechanisms of different CDK inhibitors (CDKIs) are not well understood. Our recent study revealed that selective CDK4/6 inhibitors sensitize colorectal cancer (CRC) cells to therapy-induced apoptosis by inducing Death Receptor 5 (DR5) via the p53 family member p73. In this study, we investigated if this pathway is involved in anticancer effects of different CDKIs. We found that less-selective CDKIs, including flavopiridol, roscovitine, dinaciclib, and SNS-032, induced DR5 via p73-mediated transcriptional activation. The induction of DR5 by these CDKIs was mediated by dephosphorylation of p73 at Threonine 86 and p73 nuclear translocation. Knockdown of a common target of these CDKIs, including CDK1, 2, or 9, recapitulated p73-mediated DR5 induction. CDKIs strongly synergized with 5-fluorouracil (5-FU), the most commonly used CRC chemotherapy agent, in vitro and in vivo to promote growth suppression and apoptosis, which required DR5 and p73. Together, these findings indicate p73-mediated DR5 induction as a potential tumor suppressive mechanism and a critical target engaged by different CDKIs in potentiating therapy-induced apoptosis in CRC cells. These findings help better understand the anticancer mechanisms of CDKIs and may help facilitate their clinical development and applications in CRC.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Humans , Cyclin-Dependent Kinases , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Fluorouracil/pharmacology , Fluorouracil/therapeutic use
4.
Proc Natl Acad Sci U S A ; 119(51): e2211775119, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36508676

ABSTRACT

Synthetic lethality is a powerful approach for targeting oncogenic drivers in cancer. Recent studies revealed that cancer cells with microsatellite instability (MSI) require Werner (WRN) helicase for survival; however, the underlying mechanism remains unclear. In this study, we found that WRN depletion strongly induced p53 and its downstream apoptotic target PUMA in MSI colorectal cancer (CRC) cells. p53 or PUMA deletion abolished apoptosis induced by WRN depletion in MSI CRC cells. Importantly, correction of MSI abrogated the activation of p53/PUMA and cell killing, while induction of MSI led to sensitivity in isogenic CRC cells. Rare p53-mutant MSI CRC cells are resistant to WRN depletion due to lack of PUMA induction, which could be restored by wildtype (WT) p53 knock in or reconstitution. WRN depletion or treatment with the RecQ helicase inhibitor ML216 suppressed in vitro and in vivo growth of MSI CRCs in a p53/PUMA-dependent manner. ML216 treatment was efficacious in MSI CRC patient-derived xenografts. Interestingly, p53 gene remains WT in the majority of MSI CRCs. These results indicate a critical role of p53/PUMA-mediated apoptosis in the vulnerability of MSI CRCs to WRN loss, and support WRN as a promising therapeutic target in p53-WT MSI CRCs.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Humans , Werner Syndrome Helicase/genetics , Tumor Suppressor Protein p53/genetics , Microsatellite Instability , Colorectal Neoplasms/genetics , RecQ Helicases/genetics
5.
Front Oncol ; 12: 1018775, 2022.
Article in English | MEDLINE | ID: mdl-36313707

ABSTRACT

Background: Colorectal cancer (CRC) frequently has a dysregulated epigenome causing aberrant up-regulation of oncogenes such as c-MYC. Bromodomain and extra-terminal domain (BET) proteins and histone acetyltransferases (HAT) are epigenetic regulatory proteins that create and maintain epigenetic states supporting oncogenesis. BET inhibitors and HAT inhibitors are currently being investigated as cancer therapeutics due to their ability to suppress cancer-promoting epigenetic modifiers. Due to the extensive molecular crosstalk between BET proteins and HAT proteins, we hypothesized that dual inhibition of BET and HAT could more potently inhibit CRC cells than inhibition of each individual protein. Methods: We investigated the activity and mechanisms of a dual BET and HAT inhibitor, NEO2734, in CRC cell lines and mouse xenografts. MTS, flow cytometry, and microscopy were used to assess cell viability. qPCR, Western blotting, and immunofluorescent staining were used to assess mechanisms of action. Results: We found that NEO2734 more potently suppresses CRC cell growth than first generation BET inhibitors, regardless of the status of common CRC driver mutations. We previously showed that BET inhibitors upregulate DR5 to induce extrinsic apoptosis. In the current study, we show that NEO2734 treatment induces CRC cell apoptosis via both the intrinsic and extrinsic apoptosis pathways. NEO2734 increases p53 expression and subsequently increased expression of the p53-upregulated mediator of apoptosis (PUMA), which is a critical mechanism for activating intrinsic apoptosis. We demonstrate that inhibition of either the intrinsic or extrinsic branches of apoptosis partially rescues CRC cells from NEO2734 treatment, while the dual inhibition of both branches of apoptosis more strongly rescues CRC cells from NEO2734 treatment. Finally, we show that NEO2734 monotherapy is able to suppress tumor growth in CRC xenografts by inducing apoptosis. Conclusions: Our study demonstrates NEO2734 potently suppresses CRC cells in vitro and in vivo by simultaneously upregulating PUMA and DR5 to induce cell death. Further studies of NEO2734 for treating CRC are warranted.

6.
Cancer Res ; 82(7): 1340-1352, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35149588

ABSTRACT

Targeting cyclin-dependent kinases 4 and 6 (CDK4/6) is a successful therapeutic approach against breast and other solid tumors. Inhibition of CDK4/6 halts cell cycle progression and promotes antitumor immunity. However, the mechanisms underlying the antitumor activity of CDK4/6 inhibitors are not fully understood. We found that CDK4/6 bind and phosphorylate the p53 family member p73 at threonine 86, which sequesters p73 in the cytoplasm. Inhibition of CDK4/6 led to dephosphorylation and nuclear translocation of p73, which transcriptionally activated death receptor 5 (DR5), a cytokine receptor and key component of the extrinsic apoptotic pathway. p73-mediated induction of DR5 by CDK4/6 inhibitors promoted immunogenic cell death of cancer cells. Deletion of DR5 in cancer cells in vitro and in vivo abrogated the potentiating effects of CDK4/6 inhibitors on immune cytokine TRAIL, 5-fluorouracil chemotherapy, and anti-PD-1 immunotherapy. Together, these results reveal a previously unrecognized consequence of CDK4/6 inhibition, which may be critical for potentiating the killing and immunogenic effects on cancer cells. SIGNIFICANCE: This work demonstrates how inhibition of CDK4/6 sensitizes cancer cells to chemotherapy and immune checkpoint blockade and may provide a new molecular marker for improving CDK4/6-targeted cancer therapies. See related commentary by Frank, p. 1170.


Subject(s)
Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Immune Checkpoint Inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand , TNF-Related Apoptosis-Inducing Ligand , Tumor Protein p73 , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Fluorouracil/pharmacology , Humans , Phosphorylation , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Protein p73/metabolism
7.
Oncogene ; 40(48): 6566-6578, 2021 12.
Article in English | MEDLINE | ID: mdl-34615996

ABSTRACT

Bromodomain and extra-terminal domain (BET) family proteins are epigenetic readers that play a critical role in oncogenesis by controlling the expression of oncogenes such as c-Myc. Targeting BET family proteins has recently emerged as a promising anticancer strategy. However, the molecular mechanisms by which cancer cells respond to BET inhibition are not well understood. In this study, we found that inducing the degradation of BET proteins by the proteolysis targeting chimeras (PROTAC) approach potently suppressed the growth of colorectal cancer (CRC) including patient-derived tumors. Mechanistically, BET degradation transcriptionally activates Death Receptor 5 (DR5) to trigger immunogenic cell death (ICD) in CRC cells. Enhanced DR5 induction further sensitizes CRC cells with a mutation in Speckle-type POZ protein (SPOP). Furthermore, DR5 is indispensable for a striking antitumor effect of combining BET degradation and anti-PD-1 antibody, which was well tolerated in mice and almost eradicated syngeneic tumors. Our results demonstrate that BET degradation triggers DR5-mediated ICD to potently suppress CRC and potentiate immune checkpoint blockade. These results provide a rationale, mechanistic insights, and potential biomarkers for developing a precision CRC therapy by inducing BET protein degradation.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic , Immune Checkpoint Inhibitors/pharmacology , Immunogenic Cell Death , Proteins/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Proteins/genetics , Proteolysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Oncogene ; 40(11): 2035-2050, 2021 03.
Article in English | MEDLINE | ID: mdl-33603166

ABSTRACT

Use of non-steroidal anti-inflammatory drugs (NSAIDs) is associated with reduced risk of colorectal cancer (CRC). However, the mechanism by which NSAIDs suppress colorectal tumorigenesis remains unclear. We previously showed that NSAIDs selectively kill emerging tumor cells via death receptor (DR) signaling and a synthetic lethal interaction mediated by the proapoptotic Bcl-2 family protein BID. In this study, we found NSAIDs induce endoplasmic reticulum (ER) stress to activate DR signaling and BID in tumor suppression. Importantly, our results unveiled an ER stress- and BID-dependent immunogenic effect of NSAIDs, which may be critical for tumor suppression. NSAID treatment induced hallmarks of immunogenic cell death (ICD) in CRC cells and colonic epithelial cells upon loss of APC tumor suppressor, and elevated tumor-infiltrating lymphocytes (TILs) in the polyps of APCMin/+ mice. ER stress inhibition or BID deletion abrogated the antitumor and immunogenic effects of NSAIDs. Furthermore, increased ER stress and TILs were detected in human advanced adenomas from NSAID-treated patients. Together, our results suggest that NSAIDs induce ER stress- and BID-mediated ICD to restore immunosurveillance and suppress colorectal tumor formation.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carcinogenesis/genetics , Colorectal Neoplasms/drug therapy , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Stress/drug effects , Humans , Immunogenic Cell Death/drug effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Signal Transduction/drug effects
9.
Cell Death Dis ; 11(12): 1061, 2020 12 12.
Article in English | MEDLINE | ID: mdl-33311453

ABSTRACT

Although the survival rate of patients with cancer have increased due to the use of current chemotherapeutic agents, adverse effects of cancer therapy remain a concern. The reversal of drug resistance, reduction in harmful side effects and accelerated increase in efficiency have often been addressed in the development of combination therapeutics. Tazemetostat (EPZ-6438), a histone methyltransferase EZH2 selective inhibitor, was approved by the FDA for the treatment of advanced epithelioid sarcoma. However, the effect of tazemetostat on colorectal cancer (CRC) and 5-FU sensitivity remains unclear. In this study, the enhancement of tazemetostat on 5-FU sensitivity was examined in CRC cells. Our findings demonstrated that tazemetostat combined with 5-FU exhibits synergistic antitumor function in vitro and in vivo in CRC cells. In addition, tazemetostat promotes PUMA induction through the ROS/ER stress/CHOP axis. PUMA depletion attenuates the antitumor effect of the combination therapy. Therefore, tazemetostat may be a novel treatment to improve the sensitivity of tumors to 5-FU in CRC therapy. In conclusion, the combination of 5-FU and tazemetostat shows high therapeutic possibility with reduced unfavorable effects.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Fluorouracil/therapeutic use , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins/genetics , Up-Regulation/genetics , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Benzamides/pharmacology , Benzamides/therapeutic use , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Line, Tumor , Colorectal Neoplasms/pathology , Endoplasmic Reticulum Stress/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorouracil/pharmacology , Humans , Mice, Inbred BALB C , Mice, Nude , Morpholines/pharmacology , Morpholines/therapeutic use , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Proto-Oncogene Proteins/metabolism , Pyridines/pharmacology , Pyridines/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Xenograft Model Antitumor Assays
10.
Elife ; 92020 11 02.
Article in English | MEDLINE | ID: mdl-33135632

ABSTRACT

To better understand a role of eIF4E S209 in oncogenic translation, we generated EIF4ES209A/+ heterozygous knockin (4EKI) HCT 116 human colorectal cancer (CRC) cells. 4EKI had little impact on total eIF4E levels, cap binding or global translation, but markedly reduced HCT 116 cell growth in spheroids and mice, and CRC organoid growth. 4EKI strongly inhibited Myc and ATF4 translation, the integrated stress response (ISR)-dependent glutamine metabolic signature, AKT activation and proliferation in vivo. 4EKI inhibited polyposis in ApcMin/+ mice by suppressing Myc protein and AKT activation. Furthermore, p-eIF4E was highly elevated in CRC precursor lesions in mouse and human. p-eIF4E cooperated with mutant KRAS to promote Myc and ISR-dependent glutamine addiction in various CRC cell lines, characterized by increased cell death, transcriptomic heterogeneity and immune suppression upon deprivation. These findings demonstrate a critical role of eIF4E S209-dependent translation in Myc and stress-driven oncogenesis and as a potential therapeutic vulnerability.


Subject(s)
Carcinogenesis/genetics , Eukaryotic Initiation Factor-4E/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adenomatous Polyposis Coli Protein , Animals , Cell Death , Cell Line, Tumor , Cell Proliferation , Cell Survival , Eukaryotic Initiation Factor-4E/genetics , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Stress, Physiological , Xenograft Model Antitumor Assays
11.
Cell Death Dis ; 11(10): 902, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097691

ABSTRACT

HCC (hepatocellular carcinoma) is a major health threat for the Chinese population and has poor prognosis because of strong resistance to chemotherapy in patients. For instance, a considerable challenge for the treatment of HCC is sorafenib resistance. The aberrant glucose metabolism in cancer cells aerobic glycolysis is associated with resistance to chemotherapeutic agents. Drug-resistance cells and tumors were exposed to sorafenib to establish sorafenib-resistance cell lines and tumors. Western blotting and real-time PCR or IHC staining were used to analyze the level of CLCF1 in the sorafenib resistance cell lines or tumors. The aerobic glycolysis was analyzed by ECAR assay. The mechanism mediating the high expression of CLCF1 in sorafenib-resistant cells and its relationships with miR-130-5p was determined by bioinformatic analysis, dual luciferase reporter assays, real-time PCR, and western blotting. The in vivo effect was evaluated by xenografted with nude mice. The relation of CLCF1 and miR-30a-5p was determined in patients' samples. In this study, we report the relationship between sorafenib resistance and increased glycolysis in HCC cells. We also show the vital role of CLCF1 in promoting glycolysis by activating PI3K/AKT signaling and its downstream genes, thus participating in glycolysis in sorafenib-resistant HCC cells. Furthermore, we also show that miR-30a-5p directly targets CLCF1 and that sorafenib-mediated suppression of miR-30a-5p results in the upregulation of CLCF1 in HCC cells resistant to sorafenib. We also found that when a cholesterol modified agomiR-30a-5p was delivered systemically to mice harboring sorafenib-resistant HCC tumors, tumor growth decreased significantly. There is an uncharacterized mechanism of biochemical resistance to hormone therapies orchestrated by the miR-30a-5p/CLCF1 axis to mediate sorafenib resistance and aerobic glycolysis in HCC. Therefore, this study indicates that targeting the miR-30a-5p/CLCF1 axis may hold promise for therapeutic intervention in HCC sorafenib resistance patients.


Subject(s)
Carcinoma, Hepatocellular/genetics , Drug Resistance, Neoplasm/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Sorafenib , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Mice , Signal Transduction/drug effects , Sorafenib/metabolism , Sorafenib/pharmacology
12.
J Cell Mol Med ; 24(18): 10830-10841, 2020 09.
Article in English | MEDLINE | ID: mdl-32735377

ABSTRACT

Overexpression of P2X7R has been observed in several tumours and is related to cancer advancement and metastasis. However, the role of P2X7R in colorectal cancer (CRC) patients is not well understood. In the current study, overexpression of P2X7R and the effects at the molecular and functional levels in CRC were assessed in a mouse orthotopic model. Functional assays, such as the CCK-8 assay, wound healing and transwell assay, were used to determine the biological role of P2X7R in CRC cells. CSC-related genes and properties were detected via sphere formation and real-time PCR assays. The underlying mechanisms were explored by Western blotting, real-time PCR and Flow cytometry. In this study, we found that overexpression of P2X7R increases in the in vivo growth of tumours. P2X7R overexpression also increased CD31, VEGF and concurrent angiogenesis. P2X7R up-regulates aldehyde dehydrogenase-1 (ALDH1) and CSC characteristics. Transplanted tumour cells with P2X7R overexpression stimulated cytokines to recruit tumour-associated macrophage (TAMs) to increase the growth of tumours. We also found that the NF-κB signalling pathway is involved in P2X7R-induced cytokine up-regulation. P2X7R promotes NF-κB-dependent cytokine induction, which leads to TAM recruitment to control tumour growth and advancement and remodelling of the stroma. Our findings demonstrate that P2X7R plays a key role in TAM recruitment, which may be a therapeutic target for CRC patients.


Subject(s)
Adenocarcinoma/physiopathology , Colorectal Neoplasms/physiopathology , NF-kappa B/metabolism , Neoplasm Proteins/physiology , Neovascularization, Pathologic/physiopathology , Receptors, Purinergic P2X7/physiology , Tumor-Associated Macrophages/physiology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytokines/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Receptors, Purinergic P2X7/biosynthesis , Receptors, Purinergic P2X7/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology
13.
Theranostics ; 10(18): 8098-8110, 2020.
Article in English | MEDLINE | ID: mdl-32724460

ABSTRACT

Intrinsic and acquired resistance to targeted therapies is a significant clinical problem in cancer. We previously showed that resistance to regorafenib, a multi-kinase inhibitor for treating colorectal cancer (CRC) patients, can be caused by mutations in the tumor suppressor FBW7, which block degradation of the pro-survival Bcl-2 family protein Mcl-1. We tested if Mcl-1 inhibition can be used to develop a precision combination therapy for overcoming regorafenib resistance. METHODS: Small-molecule Mcl-1 inhibitors were tested on CRC cells with knock-in (KI) of a non-degradable Mcl-1. Effects of Mcl-1 inhibitors on regorafenib sensitivity were determined in FBW7-mutant and -wild-type (WT) CRC cells and tumors, and in those with acquired regorafenib resistance due to enriched FBW7 mutations. Furthermore, translational potential was explored by establishing and analyzing FBW7-mutant and -WT patient-derived organoid (PDO) and xenograft (PDX) tumor models. RESULTS: We found that highly potent and specific Mcl-1 inhibitors such as S63845 overcame regorafenib resistance by restoring apoptosis in multiple regorafenib-resistant CRC models. Mcl-1 inhibition re-sensitized CRC tumors with intrinsic and acquired regorafenib resistance in vitro and in vivo, including those with FBW7 mutations. Importantly, Mcl-1 inhibition also sensitized FBW7-mutant PDO and PDX models to regorafenib. In contrast, Mcl-1 inhibition had no effect in FBW7-WT CRCs. CONCLUSIONS: Our results demonstrate that Mcl-1 inhibitors can overcome intrinsic and acquired regorafenib resistance in CRCs by restoring apoptotic response. FBW7 mutations might be a potential biomarker predicting for response to the regorafenib/Mcl-1 inhibitor combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , Aged , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Drug Synergism , F-Box-WD Repeat-Containing Protein 7/genetics , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Organoids , Phenylurea Compounds/therapeutic use , Precision Medicine/methods , Primary Cell Culture , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Thiophenes/pharmacology , Thiophenes/therapeutic use , Xenograft Model Antitumor Assays
14.
J Cell Mol Med ; 24(13): 7600-7608, 2020 07.
Article in English | MEDLINE | ID: mdl-32410348

ABSTRACT

MicroRNAs (miRNAs) have been found to play a key role in drug resistance. In the current study, we aimed to explore the potential role of miR-126 in trastuzumab resistance in breast cancer cells. We found that the trastuzumab-resistant cell lines SKBR3/TR and BT474/TR had low expression of miR-126 and increased ability to migrate and invade. The resistance, invasion and mobilization abilities of the cells resistant to trastuzumab were reduced by ectopic expression of miR-126 mimics. In comparison, inhibition of miR-126 in SKBR3 parental cells had the opposite effect of an increased resistance to trastuzumab as well as invasion and migration. It was also found that miR-126 directly targets PIK3R2 in breast cancer cells. PIK3R2-knockdown cells showed decreased resistance to trastuzumab, while overexpression of PIK3R2 increased trastuzumab resistance. In addition, our finding showed that overexpression of miR-126 reduced resistance to trastuzumab in the trastuzumab-resistant cells and that inhibition of the PIK3R2/PI3K/AKT/mTOR signalling pathway was involved in this effect. SKBR3/TR cells also showed increased sensitivity to trastuzumab mediated by miR-126 in vivo. In conclusion, the above findings demonstrated that overexpression of miR-126 or down-regulation of its target gene may be a potential approach to overcome trastuzumab resistance in breast cancer cells.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Trastuzumab/therapeutic use , Animals , Base Sequence , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/drug effects , Trastuzumab/pharmacology
15.
Cancer Res ; 79(6): 1191-1203, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30674532

ABSTRACT

Bromodomain and extraterminal domain (BET) family proteins such as BRD4 are epigenetic readers that control expression of a number of oncogenic proteins. Targeting this family of proteins has recently emerged as a promising anticancer approach. BET inhibitors (BETi), either alone or in combination with other anticancer agents, have exhibited efficacy in a variety of tumors. However, the molecular mechanisms underlying differential response to BETi are not well understood. In this study, we report that death receptor 5 (DR5), a key component of the extrinsic apoptotic pathway, is markedly induced in response to BRD4 depletion and BETi treatment in colorectal cancer cells. Induction of DR5, following BET inhibition, was mediated by endoplasmic reticulum stress and CHOP-dependent transcriptional activation. Enhanced DR5 induction was necessary for the chemosensitization and apoptotic effects of BETi and was responsible for increased BETi sensitivity in colorectal cancer cells containing a mutation in speckle-type POZ protein (SPOP), a subunit of BRD4 E3 ubiquitin ligase. In a colorectal cancer xenograft model, BETi combined with chemotherapy suppressed the tumor growth in a DR5-dependent manner and potently inhibited patient-derived xenograft tumor growth with enhanced DR5 induction and apoptosis. These findings suggest that BETi alone or in combination with chemotherapy is effective against colorectal cancer due to enhanced DR5 induction and apoptosis. DR5 induction may also serve as a useful marker for designing personalized treatment and improved colorectal cancer combination therapies.Significance: These findings reveal how BET inhibition sensitizes chemotherapy and kills a subset of colon cancer cells with specific genetic alterations and may provide a new molecular marker for improving colon cancer therapies.


Subject(s)
Azepines/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Drug Synergism , Fluorouracil/pharmacology , Mutation , Nuclear Proteins/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Repressor Proteins/genetics , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Cancer Res ; 78(16): 4704-4715, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29895675

ABSTRACT

Mcl-1, a prosurvival Bcl-2 family protein, is frequently overexpressed in cancer cells and plays a critical role in therapeutic resistance. It is well known that anticancer agents induce phosphorylation of Mcl-1, which promotes its binding to E3 ubiquitin ligases and subsequent proteasomal degradation and apoptosis. However, other functions of Mcl-1 phosphorylation in cancer cell death have not been well characterized. In this study, we show in colon cancer cells that histone deacetylase inhibitors (HDACi) induce GSK3ß-dependent Mcl-1 phosphorylation, but not degradation or downregulation. The in vitro and in vivo anticancer effects of HDACi were dependent on Mcl-1 phosphorylation and were blocked by genetic knock-in of a Mcl-1 phosphorylation site mutant. Phosphorylation-dead Mcl-1 maintained cell survival by binding and sequestering BH3-only Bcl-2 family proteins PUMA, Bim, and Noxa, which were upregulated and necessary for apoptosis induction by HDACi. Resistance to HDACi mediated by phosphorylation-dead Mcl-1 was reversed by small-molecule Mcl-1 inhibitors that liberated BH3-only proteins. These results demonstrate a critical role of Mcl-1 phosphorylation in mediating HDACi sensitivity through a novel and degradation-independent mechanism. These results provide new mechanistic insights on how Mcl-1 maintains cancer cell survival and suggest that Mcl-1-targeting agents are broadly useful for overcoming therapeutic resistance in cancer cells.Significance: These findings present a novel degradation-independent function of Mcl-1 phosphorylation in anticancer therapy that could be useful for developing new Mcl-1-targeting agents to overcome therapeutic resistance. Cancer Res; 78(16); 4704-15. ©2018 AACR.


Subject(s)
Colonic Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Histone Deacetylases/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Phosphorylation/drug effects , Proteolysis/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Small Molecule Libraries/pharmacology
17.
Oncogene ; 37(33): 4599-4610, 2018 08.
Article in English | MEDLINE | ID: mdl-29755130

ABSTRACT

Intrinsic and acquired resistance to anti-EGFR antibody therapy, frequently mediated by a mutant or amplified KRAS oncogene, is a significant challenge in the treatment of colorectal cancer (CRC). However, the mechanism of KRAS-mediated therapeutic resistance is not well understood. In this study, we demonstrate that clinically used anti-EGFR antibodies, including cetuximab and panitumumab, induce killing of sensitive CRC cells through p73-dependent transcriptional activation of the pro-apoptotic Bcl-2 family protein PUMA. PUMA induction and p73 activation are abrogated in CRC cells with acquired resistance to anti-EGFR antibodies due to KRAS alterations. Inhibition of aurora kinases preferentially kills mutant KRAS CRC cells and overcomes KRAS-mediated resistance to anti-EGFR antibodies in vitro and in vivo by restoring PUMA induction. Our results suggest that PUMA plays a critical role in meditating the sensitivity of CRC cells to anti-EGFR antibodies, and that restoration of PUMA-mediated apoptosis is a promising approach to improve the efficacy of EGFR-targeted therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis Regulatory Proteins/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cetuximab/pharmacology , ErbB Receptors/metabolism , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Panitumumab/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism
18.
Proc Natl Acad Sci U S A ; 115(15): 3930-3935, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29581256

ABSTRACT

Necroptosis, a form of regulated necrotic cell death, is governed by RIP1/RIP3-mediated activation of MLKL. However, the signaling process leading to necroptotic death remains to be elucidated. In this study, we found that PUMA, a proapoptotic BH3-only Bcl-2 family member, is transcriptionally activated in an RIP3/MLKL-dependent manner following induction of necroptosis. The induction of PUMA, which is mediated by autocrine TNF-α and enhanced NF-κB activity, contributes to necroptotic death in RIP3-expressing cells with caspases inhibited. On induction, PUMA promotes the cytosolic release of mitochondrial DNA and activation of the DNA sensors DAI/Zbp1 and STING, leading to enhanced RIP3 and MLKL phosphorylation in a positive feedback loop. Furthermore, deletion of PUMA partially rescues necroptosis-mediated developmental defects in FADD-deficient embryos. Collectively, our results reveal a signal amplification mechanism mediated by PUMA and cytosolic DNA sensors that is involved in TNF-driven necroptotic death in vitro and in vivo.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , DNA-Binding Proteins/metabolism , Glycoproteins/metabolism , Membrane Proteins/metabolism , Necrosis/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Cytosol/metabolism , DNA/genetics , DNA/metabolism , DNA-Binding Proteins/genetics , Glycoproteins/genetics , Humans , Membrane Proteins/genetics , Mice , Mitochondria/genetics , Mitochondria/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis/genetics , Necrosis/physiopathology , Phosphorylation , Protein Kinases/genetics , Protein Kinases/metabolism , Proto-Oncogene Proteins/genetics , RNA-Binding Proteins , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Proteins/genetics
19.
Mol Cancer Ther ; 16(9): 1979-1988, 2017 09.
Article in English | MEDLINE | ID: mdl-28619760

ABSTRACT

Heat shock protein 90 (Hsp90) is widely overexpressed in cancer cells and necessary for maintenance of malignant phenotypes. Hsp90 inhibition induces tumor cell death through degradation of its client oncoproteins and has shown promises in preclinical studies. However, the mechanism by which Hsp90 inhibitors kill tumor cells is not well-understood. Biomarkers associated with differential sensitivity and resistance to Hsp90 inhibitors remain to be identified. In this study, we found that colorectal cancer cells containing inactivating mutations of FBW7, a tumor suppressor and E3 ubiquitin ligase, are intrinsically insensitive to Hsp90 inhibitors. The insensitive colorectal cancer cells lack degradation of Mcl-1, a prosurvival Bcl-2 family protein. Hsp90 inhibition promotes GSK3ß-dependent phosphorylation of Mcl-1, which subsequently binds to FBW7 and undergoes ubiquitination and proteasomal degradation. Specifically blocking Mcl-1 phosphorylation by genetic knock-in abrogates its degradation and renders in vitro and in vivo resistance to Hsp90 inhibitors, which can be overcame by Mcl-1-selective small-molecule inhibitors. Collectively, our findings demonstrate a key role of GSK3ß/FBW7-dependent Mcl-1 degradation in killing of colorectal cancer cells by Hsp90 inhibitors and suggest FBW7 mutational status as a biomarker for Hsp90-targeted therapy. Mol Cancer Ther; 16(9); 1979-88. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , F-Box-WD Repeat-Containing Protein 7/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasms/metabolism , Animals , Apoptosis/genetics , Cell Line, Tumor , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Heterografts , Humans , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , RNA, Small Interfering/genetics
20.
Cancer Res ; 77(9): 2512-2521, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28202514

ABSTRACT

The Bcl-2 family protein Mcl-1 is often degraded in cancer cells subjected to effective therapeutic treatment, and defective Mcl-1 degradation has been associated with intrinsic and acquired drug resistance. However, a causal relationship between Mcl-1 degradation and anticancer drug responses has not been directly established, especially in solid tumor cells where Mcl-1 inhibition alone is insufficient to trigger cell death. In this study, we present evidence that Mcl-1 participates directly in determining effective therapeutic responses in colon cancer cells. In this setting, Mcl-1 degradation was induced by a variety of multikinase inhibitor drugs, where it relied upon GSK3ß phosphorylation and FBW7-dependent ubiquitination. Specific blockade by genetic knock-in (KI) abolished apoptotic responses and conferred resistance to kinase inhibitors. Mcl-1-KI also suppressed the antiangiogenic and anti-hypoxic effects of kinase inhibitors in the tumor microenvironment. Interestingly, these same inhibitors also induced the BH3-only Bcl-2 family protein PUMA, which is required for apoptosis. Degradation-resistant Mcl-1 bound and sequestered PUMA from other prosurvival proteins to maintain cell survival, which was abolished by small-molecule Mcl-1 inhibitors. Our findings establish a pivotal role for Mcl-1 degradation in the response of colon cancer cells to targeted therapeutics, and they provide a useful rational platform to develop Mcl-1-targeting agents that can overcome drug resistance. Cancer Res; 77(9); 2512-21. ©2017 AACR.


Subject(s)
Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Knock-In Techniques , Glycogen Synthase Kinase 3 beta/genetics , Humans , Mice , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Proteolysis/drug effects , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL