Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Sci Immunol ; 9(95): eadl2171, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38820140

ABSTRACT

Tumors evade attacks from the immune system through various mechanisms. Here, we identify a component of tumor immune evasion mediated by YTH domain-containing family protein 2 (YTHDF2), a reader protein that usually destabilizes m6A-modified mRNA. Loss of tumoral YTHDF2 inhibits tumor growth and prolongs survival in immunocompetent tumor models. Mechanistically, tumoral YTHDF2 deficiency promotes the recruitment of macrophages via CX3CL1 and enhances mitochondrial respiration of CD8+ T cells by impairing tumor glycolysis metabolism. Tumoral YTHDF2 deficiency promotes inflammatory macrophage polarization and antigen presentation in the presence of IFN-γ. In addition, IFN-γ induces autophagic degradation of tumoral YTHDF2, thereby sensitizing tumor cells to CD8+ T cell-mediated cytotoxicity. Last, we identified a small molecule compound that preferentially induces YTHDF2 degradation, which shows a potent antitumor effect alone but a better effect when combined with anti-PD-L1 or anti-PD-1 antibodies. Collectively, YTHDF2 appears to be a tumor-intrinsic regulator that orchestrates immune evasion, representing a promising target for enhancing cancer immunotherapy.


Subject(s)
Mice, Inbred C57BL , RNA-Binding Proteins , Animals , RNA-Binding Proteins/immunology , RNA-Binding Proteins/genetics , Mice , Humans , Immune Evasion , Tumor Escape/immunology , Mice, Knockout , Neoplasms/immunology , Neoplasms/genetics , Cell Line, Tumor , CD8-Positive T-Lymphocytes/immunology , Female
2.
Int J Oral Sci ; 14(1): 40, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35927232

ABSTRACT

Osteoarthritis (OA) is a prevalent degenerative joint disease characterized by cartilage loss and accounts for a major source of pain and disability worldwide. However, effective strategies for cartilage repair are lacking, and patients with advanced OA usually need joint replacement. Better comprehending OA pathogenesis may lead to transformative therapeutics. Recently studies have reported that exosomes act as a new means of cell-to-cell communication by delivering multiple bioactive molecules to create a particular microenvironment that tunes cartilage behavior. Specifically, exosome cargos, such as noncoding RNAs (ncRNAs) and proteins, play a crucial role in OA progression by regulating the proliferation, apoptosis, autophagy, and inflammatory response of joint cells, rendering them promising candidates for OA monitoring and treatment. This review systematically summarizes the current insight regarding the biogenesis and function of exosomes and their potential as therapeutic tools targeting cell-to-cell communication in OA, suggesting new realms to improve OA management.


Subject(s)
Cartilage, Articular , Exosomes , Osteoarthritis , Apoptosis , Cartilage/metabolism , Cartilage/pathology , Cartilage, Articular/metabolism , Cell Communication , Chondrocytes/metabolism , Exosomes/metabolism , Exosomes/pathology , Humans , Osteoarthritis/metabolism , Osteoarthritis/therapy
3.
iScience ; 25(5): 104349, 2022 May 20.
Article in English | MEDLINE | ID: mdl-35602963

ABSTRACT

Glycyrrhetinic acid (GA) is a natural product of licorice with mitochondria targeting properties and shows broad anticancer activities, but its targets and underlying mechanisms remain elusive. Here, we identified the mitochondrial enzyme serine hydroxymethyltransferase 2 (SHMT2) as a target of GA by using chemical proteomics. Binding to and inhibiting the activity of SHMT2 by GA were validated in vitro and in vivo. Knockout of SHMT2 or inhibiting SHMT2 with GA restricts mitochondrial energy supplies by downregulating mitochondrial oxidative phosphorylation (OXPHOS) and fatty acid ß-oxidation, and consequently suppresses cancer cell proliferation and tumor growth. Crystal structures of GA derivatives indicate that GA occupies SHMT2 folate-binding pocket and regulates SHMT2 activity. Modifications at GA carboxylic group with diamines significantly improved its anticancer potency, demonstrating GA as a decent structural template for SHMT2 inhibitor development.

4.
Mol Cancer ; 21(1): 99, 2022 04 11.
Article in English | MEDLINE | ID: mdl-35410300

ABSTRACT

Proteolysis-targeting chimeras (PROTACs) are engineered techniques for targeted protein degradation. A bifunctional PROTAC molecule with two covalently-linked ligands recruits target protein and E3 ubiquitin ligase together to trigger proteasomal degradation of target protein by the ubiquitin-proteasome system. PROTAC has emerged as a promising approach for targeted therapy in various diseases, particularly in cancers. In this review, we introduce the principle and development of PROTAC technology, as well as the advantages of PROTACs over traditional anti-cancer therapies. Moreover, we summarize the application of PROTACs in targeting critical oncoproteins, provide the guidelines for the molecular design of PROTACs and discuss the challenges in the targeted degradation by PROTACs.


Subject(s)
Neoplasms , Humans , Ligands , Neoplasms/drug therapy , Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ubiquitin-Protein Ligases/metabolism
5.
J Oral Microbiol ; 14(1): 2015130, 2022.
Article in English | MEDLINE | ID: mdl-34992737

ABSTRACT

BACKGROUND: Microtubule dynamics plays a crucial role in the spatial arrangement of cell organelles and activation of the NLRP3 inflammasome. PURPOSE: This study aimed to explore whether microtubule affinity regulating kinase 4 (MARK4) can be a therapeutic target of periodontitis by affecting microtubule dynamics and NLRP3 inflammasome-mediated pyroptosis in macrophages. MATERIALS AND METHODS: The NLRP3 inflammasome-related genes and MARK4 were measured in the healthy and inflamed human gingival tissues. Bone marrow-derived macrophages (BMDMs) were infected with Porphyromonas gingivalis, while the MARK4 inhibitors (OTSSP167 and Compound 50) and small interference RNA were utilized to restrain MARK4. Apoptosis-associated speck-like protein (ASC) speck was detected by confocal, and levels of interleukin-1ß (IL-1ß), as well as IL-18, were assessed by ELISA. RESULTS: Increased staining and transcription of MARK4, NLRP3, ASC, and Caspase-1 were observed in the inflamed gingiva. P. gingivalis infection promoted MARK4 expression and the NLRP3 inflammasome in BMDMs. Inhibition of MARK4 decreased LDH release, IL-1ß and IL-18 production, ASC speck formation, and the pyroptosis-related genes transcription. Furthermore, MARK4 inhibition reduced microtubule polymerization and acetylation in P. gingivalis-infected BMDMs. CONCLUSIONS: MARK4 promoted NLRP3 inflammasome activation and pyroptosis in P. gingivalis-infected BMDMs by affecting microtubule dynamics. MARK4 inhibition might be a potential target in regulating the NLRP3 inflammasome during periodontitis progress.

6.
Nat Prod Res ; 36(5): 1205-1214, 2022 Mar.
Article in English | MEDLINE | ID: mdl-33375884

ABSTRACT

Five 3-formyl-2-arylbenzofuran derivatives, including three new compounds (1-3) and two known analogues (4-5), were identified from the 95% EtOH extract of Itea yunnanensis. Extensive spectroscopic analyses were performed for the structure elucidation of all new benzofurans, and single-crystal X-ray diffraction analyses were further employed for the structure verification of iteafuranals C (1) and D (2). In MTT assay, iteafuranal E (3) and iteafuranal A (4) displayed significant growth inhibition effect on SK-Hep-1 cells with IC50 values of 5.365 µM and 6.013 µM, respectively. The colony formation assay of 3 and 4 further confirmed their remarkable inhibitory effect on cell growth. Preliminary mechanism study demonstrated that 3 remarkably down-regulated the phosphorylation level of ERK, which suggested 3 could inhibit cell growth and induce apoptosis of SK-Hep-1 cells by blocking RAS/RAF/MEK/ERK signaling pathway. This study highlighted the potential of 3-fomyl-2-benzofuran derivatives as novel lead compounds to treat Hepatocellular carcinoma.[Formula: see text].


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Apoptosis , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , MAP Kinase Signaling System , Signal Transduction
7.
Cell Mol Gastroenterol Hepatol ; 13(4): 1223-1242, 2022.
Article in English | MEDLINE | ID: mdl-34952201

ABSTRACT

BACKGROUND & AIMS: Latent metastasis of colorectal cancer (CRC) frequently develops months or years after primary surgery, followed by adjuvant therapies, and may progress rapidly even with targeted therapy administered, but the underlying mechanism remains unclear. Here, we aim to explore the molecular basis for the aggressive behavior of latent metastasis in CRC. METHODS: Transcriptional profiling and pathway enrichment analysis of paired primary and metastatic tumor samples were performed. The underlying mechanisms of pleckstrin homology-like domain, family B, member 2 (PHLDB2) in CRC were investigated by RNA immunoprecipitation assay, immunohistochemistry, mass spectrometry analysis, and Duolink in situ proximity ligation assay (Sigma-Aldrich, Shanghai, China). The efficacy of targeting PHLDB2 in cetuximab treatment was elucidated in CRC cell lines and mouse models. RESULTS: Based on the transcriptional profile of paired primary and metastatic tumor samples, we identified PHLDB2 as a potential regulator in latent liver metastasis. A detailed mechanistic study showed that chemotherapeutic agent-induced oxidative stress promotes methyltransferase-like 14 (METTL14)-mediated N6-methyladenosine modification of PHLDB2 messenger RNA, facilitating its protein expression. Up-regulated PHLDB2 stabilizes epidermal growth factor receptor (EGFR) and promotes its nuclear translocation, which in turn results in EGFR signaling activation and consequent cetuximab resistance. Moreover, Arg1163 (R1163) of PHLDB2 is crucial for interaction with EGFR, and the R1163A mutation abrogates its regulatory function in EGFR signaling. CONCLUSIONS: PHLDB2 plays a crucial role in cetuximab resistance and is proposed to be a potential target for the treatment of CRC.


Subject(s)
Antineoplastic Agents , Carrier Proteins/metabolism , Colorectal Neoplasms , Membrane Proteins/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cetuximab/pharmacology , Cetuximab/therapeutic use , China , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Mice
10.
Front Cell Dev Biol ; 8: 168, 2020.
Article in English | MEDLINE | ID: mdl-32296699

ABSTRACT

Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) is an evolutionally conserved and unique enzyme that specifically catalyzes the cis-trans isomerization of phosphorylated serine/threonine-proline (pSer/Thr-Pro) motif and, subsequently, induces the conformational change of its substrates. Mounting evidence has demonstrated that Pin1 is widely overexpressed and/or overactivated in cancer, exerting a critical influence on tumor initiation and progression via regulation of the biological activity, protein degradation, or nucleus-cytoplasmic distribution of its substrates. Moreover, Pin1 participates in the cancer hallmarks through activating some oncogenes and growth enhancers, or inactivating some tumor suppressors and growth inhibitors, suggesting that Pin1 could be an attractive target for cancer therapy. In this review, we summarize the findings on the dysregulation, mechanisms, and biological functions of Pin1 in cancer cells, and also discuss the significance and potential applications of Pin1 dysregulation in human cancer.

11.
Trends Cancer ; 6(4): 319-336, 2020 04.
Article in English | MEDLINE | ID: mdl-32209446

ABSTRACT

Circular RNA (circRNA) is a class of single-stranded molecules with tissue/development-specific expression patterns. Unlike linear RNA, circRNA forms a covalently closed loop produced from 'back-splicing' of primary transcripts, conferring on them inherent resistance to exonucleolytic RNA decay. Increasing evidence demonstrates that many circRNAs exert important biological functions by acting as miRNA inhibitors ('sponges'), protein 'decoys', or by encoding small peptides. Importantly, circRNAs are aberrantly expressed in cancer and play indispensable oncogenic or tumor suppressive roles during tumor development and progression. In this review, we summarize the biogenesis, turnover, and involvements of circRNAs in cancer and also discuss their potential as diagnostic biomarkers or therapeutic targets.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Neoplasms/genetics , RNA, Circular/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/agonists , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Disease Progression , Genes, Tumor Suppressor , Humans , Mice , MicroRNAs/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Oncogenes/genetics , RNA Precursors/genetics , RNA Splicing , RNA Stability , RNA, Circular/chemistry , RNA, Circular/genetics , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , Xenograft Model Antitumor Assays
12.
Hepatology ; 71(5): 1643-1659, 2020 05.
Article in English | MEDLINE | ID: mdl-31509262

ABSTRACT

BACKGROUND AND AIMS: Tumor metastasis is a major factor of high recurrence and mortality in hepatocellular carcinoma (HCC), but its underlying mechanism remains elusive. We report that PDZ and LIM domain protein 1 (PDLIM1) is significantly down-regulated in metastatic human HCC tissues, which predicts unfavorable prognosis, suggesting that PDLIM1 may play an important inhibitory role during HCC metastasis. APPROACH AND RESULTS: Functional studies indicate that PDLIM1 knockdown induces epithelial-to-mesenchymal transition (EMT) of HCC cells, elevates their invasive capacity, and promotes metastasis in vitro and in vivo, whereas overexpression of PDLIM1 exhibits opposite phenotypes. Mechanistically, PDLIM1 competitively binds to the cytoskeleton cross-linking protein alpha-actinin 4 (ACTN4), leading to the disassociation of ACTN4 from F-actin, thus preventing F-actin overgrowth. In contrast, loss of PDLIM1 induces excessive F-actin formation, resulting in dephosphorylation of large tumor suppressor kinase 1 and activation of Yes-associated protein, thereby promoting HCC metastasis. Moreover, Asn145 (N145) of PDLIM1 is critical for its interaction with ACTN4, and N145A mutation abolishes its regulatory function in Hippo signaling and HCC metastasis. CONCLUSIONS: Our findings indicate that PDLIM1 suppresses HCC metastasis by modulating Hippo signaling, suggesting that PDLIM1 may be a potential prognostic marker for metastatic HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , LIM Domain Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Transcription Factors/metabolism , Actinin/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Hippo Signaling Pathway , Humans , LIM Domain Proteins/genetics , Liver Neoplasms/genetics , Neoplasm Metastasis , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transcription Factors/genetics , YAP-Signaling Proteins
13.
Anal Chem ; 91(24): 15818-15825, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31743002

ABSTRACT

Activity-based chemical proteomics approaches used for identifying cellular targets of drugs are mainly dependent on the availability of probes derived from drugs. However, all chemical probes are structurally different from the drugs themselves and cannot fully mimic the real actions of drugs in cells. Here we present a concise and unbiased immunoaffinity-based strategy for identifying covalent drug targets in vivo. By using the specific antibody, we not only confirm the well-known ibrutinib-binding target BTK, but also identify some previously undescribed strongly binding proteins, such as CKAP4 in human cell lines and TAP1 in mouse organs. The observed target profiles between species may partially explain why certain drug candidates are very effective in mice but not in humans. This approach avoids the chemical modification of drugs, eliminates the nonspecific bindings of chemical probes, and allows to unbiasedly decode the underlying mechanisms of action of covalent drugs.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Peptides/chemistry , Proteomics , Pyrazoles/chemistry , Pyrimidines/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 2/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 2/metabolism , Adenine/analogs & derivatives , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Amino Acid Sequence , Animals , Antibodies/immunology , Cell Line , Humans , Liver/chemistry , Liver/metabolism , Liver/pathology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Peptides/analysis , Piperidines , Protein Binding , Pyrazoles/immunology , Pyrazoles/metabolism , Pyrimidines/immunology , Pyrimidines/metabolism , Spleen/chemistry , Spleen/metabolism , Spleen/pathology
14.
Theranostics ; 9(16): 4704-4716, 2019.
Article in English | MEDLINE | ID: mdl-31367251

ABSTRACT

Hepatocellular carcinoma (HCC) is in an urgent need of new, effective therapies to reduce morbidity and mortality. We have previously demonstrated that peptidyl-prolyl cis/trans isomerase Pin1 is a potential target for HCC therapy, due to its pivotal role in HCC development through regulating miRNA biogenesis, and discovered the small molecule API-1 as a novel and specific Pin1 inhibitor. Despite its significant anti-HCC activity, the low water solubility and in vivo bioavailability of API-1 limit its clinical application. To address these issues, we herein developed a liposomal formulation of API-1 to improve API-1 delivery and enhance its anti-HCC efficacy. Methods: We designed and developed a nanoscale liposomal formulation of API-1, named as API-LP. Subsequently, the mean diameter, polydispersity, zeta potential, encapsulation efficiency and thermal properties of the optimization API-LP were characterized. The enhanced anti-HCC activity and the molecular mechanism of API-LP were investigated both in vitro and in vivo. Finally, the safety and pharmacokinetic property of API-LP were evaluated systematically. Results: API-LP had good formulation characteristics and exhibited an enhanced in vitro activity of suppressing proliferation and migration of HCC cells when compared with free API-1. The mechanism study showed that API-LP upregulated miRNA biogenesis via inhibiting Pin1 activity followed by restoring the nucleus-to-cytoplasm export of XPO5. Because of the increased delivery efficiency, API-LP displayed a stronger ability to promote miRNA biogenesis than free API-1. Importantly, API-LP displayed higher systemic exposure than free API-1 in mice without apparent toxicity, resulting in an enhanced tumor inhibition in xenograft mice. Conclusion: The development and assessment of API-LP provide an attractive and safe anti-HCC agent, highlighting the miRNA-based treatment for human cancers.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Enzyme Inhibitors/administration & dosage , Liver Neoplasms/drug therapy , MicroRNAs/genetics , Animals , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Compounding , Enzyme Inhibitors/chemistry , Gene Expression Regulation, Neoplastic , Humans , Liposomes/chemistry , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism
15.
Front Chem ; 7: 366, 2019.
Article in English | MEDLINE | ID: mdl-31179271

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the most prevalent cancers worldwide. Nowadays, pharmacological therapy for HCC is in urgent needs. Paclitaxel is an effective drug against diverse solid tumors, but commonly resisted in HCC patients. We recently have disclosed that microtubule affinity-regulating kinase 4 (MARK4) increases the microtubule dynamics and confers paclitaxel resistance in HCC, suggesting MARK4 as an attractive target to overcome paclitaxel resistance. Herein, we synthesized and identified coumarin derivatives 50 as a novel MARK4 inhibitor. Biological evaluation indicated compound 50 directly interacted with MARK4 and inhibited its activity in vitro, suppressed cell viability and induced apoptosis of HCC cells in a MARK4-dependent manner. Importantly, compound 50 significantly increased the drug response of paclitaxel treatment to HCC cells, providing a promise strategy to HCC treatment and broadening the application of paclitaxel in cancer therapy.

16.
Mol Cancer ; 18(1): 98, 2019 05 22.
Article in English | MEDLINE | ID: mdl-31118036

ABSTRACT

Cancer-associated chromosomal translocations are reported to generate oncogenic circular RNA (circRNA), contributing to tumorigenesis. The fusion gene SLC34A2-ROS1 (solute carrier family 34 member 2 and ROS proto-oncogene 1) plays an important role in non-small cell lung cancer (NSCLC) progression. However, whether SLC34A2-ROS1 gene can produce circRNA remains unknown. Here, we identified two novel circRNAs (F-circSR1 and F-circSR2) generated from SLC34A2-ROS1 fusion gene, while F-circSR1 has higher expression than F-circSR2. Functional studies through gain- and loss-of-function strategies showed that both F-circSRs promote cell migration in lung cancer cells, whereas they have little effect on cell proliferation. Using the minigene GFP reporter assay, we verified that the flanking complementary sequences with canonical splicing sites are essential for F-circSR biogenesis. Therefore, our findings demonstrate the oncogenic role of F-circSR in NSCLC and highlight its therapeutic potential.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , RNA, Circular/genetics , A549 Cells , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Humans , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Translocation, Genetic , Up-Regulation
17.
Cancer Lett ; 458: 76-85, 2019 08 28.
Article in English | MEDLINE | ID: mdl-31125641

ABSTRACT

Limited drug response and severe drug resistance confer the high mortality of non-small-cell lung cancer (NSCLC), a leading cause of cancer death worldwide. There is an urgent need for novel treatment against NSCLC. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is aberrantly overexpressed and participats in NSCLC development and EGFR-TKIs-induced drug resistance. Increasing evidences indicate that oncogenic ROR1 is a potential target for NSCLC therapy. However, nearly no ROR1 inhibitor was reported until now. Here, combining the computer-aided drug design and cell-based activity screening, we discover (R)-5,7-bis(methoxymethoxy)-2-(4-methoxyphenyl)chroman-4-one (ARI-1) as a novel ROR1 inhibitor. Biological evaluation demonstrates that ARI-1 specifically targets the extracellular frizzled domain of ROR1 and potently suppresses NSCLC cell proliferation and migration by regulating PI3K/AKT/mTOR signaling in a ROR1-dependent manner. Moreover, ARI-1 significantly inhibits tumor growth in vivo without obvious toxicity. Intriguingly, ARI-1 is effective to EGFR-TKIs-resistant NSCLC cells with high ROR1 expression. Therefore, our work suggests that the ROR1 inhibitor ARI-1 is a novel drug candidate for NSCLC treatment, especially for EGFR-TKIs-resisted NSCLC with high ROR1 expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Chromones/pharmacology , Lung Neoplasms/drug therapy , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Random Allocation , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Xenograft Model Antitumor Assays
18.
Bioorg Med Chem ; 27(11): 2235-2244, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31027708

ABSTRACT

Peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) participates in diverse cancer-associated signaling pathways, playing an oncogenic role in multiple human cancers, including hepatocellular carcinoma (HCC). Our recent works clarify that Pin1 modulates miRNAs biogenesis by interacting with ERK-phosphorylated exportin-5 (XPO5) and changing XPO5 conformation, giving a potential target for HCC treatment. Herein, we discover 4,6-bis(benzyloxy)-3-phenylbenzofuran (TAB29) as a novel Pin1 inhibitor that targets Pin1 PPIase domain. TAB29 potently inhibits Pin1 activity with the IC50 value of 874 nM and displays an excellent selectivity toward Pin1 in vitro. Cell-based biological evaluation reveals that TAB29 significantly suppresses cell proliferation of HCC cells through restoring the nucleus-to-cytoplasm export of XPO5 and upregulating mature miRNAs expression. Collectively, this work provides a promising small molecule lead compound for Pin1 inhibition, highlighting the therapeutic potential of miRNA-based treatment for human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Benzofurans/pharmacology , Carcinoma, Hepatocellular/drug therapy , Enzyme Inhibitors/pharmacology , Liver Neoplasms/drug therapy , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Benzofurans/chemical synthesis , Benzofurans/metabolism , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Karyopherins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Docking Simulation , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Protein Binding , Up-Regulation/drug effects
19.
Chem Asian J ; 14(1): 130-134, 2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30474357

ABSTRACT

Peptidyl-prolyl cis-trans isomerase Pin1 plays a crucial role in the development of human cancers. Recently, we have disclosed that Pin1 regulates the biogenesis of miRNA, which is aberrantly expressed in HCC and promotes HCC progression, indicating the therapeutic role of Pin1 in HCC therapy. Here, 7-(benzyloxy)-3,5-dihydroxy-2-(4-methoxyphenyl)-8-(3-methylbut-2-en-1-yl)-4H-chromen-4-one (AF-39) was identified as a novel Pin1 inhibitor. Biochemical tests indicate that AF-39 potently inhibits Pin1 activity with an IC50 values of 1.008 µm, and also displays high selectivity for Pin1 among peptidyl prolyl isomerases. Furthermore, AF-39 significantly suppresses cell proliferation of HCC cells in a dose- and time-dependent manner. Mechanistically, AF-39 regulates the subcellular distribution of XPO5 and increases miRNAs biogenesis in HCC cells. This work provides a promising lead compound for HCC treatment, highlighting the therapeutic potential of miRNA-based therapy against human cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Discovery , Flavonols/pharmacology , Liver Neoplasms/drug therapy , MicroRNAs/genetics , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Computer-Aided Design , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Flavonols/chemical synthesis , Flavonols/chemistry , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Structure , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Real-Time Polymerase Chain Reaction , Structure-Activity Relationship , Time Factors
20.
Cell Mol Life Sci ; 75(24): 4667-4681, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30293090

ABSTRACT

Long non-coding RNA (lncRNA) plays an important role in tumor progression and metastasis. Emerging evidence indicates that lncRNA actin filament-associated protein 1-antisense RNA 1 (AFAP1-AS1) is dysregulated in certain tumors. However, the function of AFAP1-AS1 in non-small cell lung cancer (NSCLC) remains elusive. In this study, we conducted global lncRNA profiling and identified that AFAP1-AS1 is significantly upregulated in NSCLC, suggesting that AFAP1-AS1 may be important for lung cancer development. For the first time, the transcription initiation and termination sites of AFAP1-AS1 were identified by rapid amplification of cDNA ends technology, and the sequencing data indicated that AFAP1-AS1 in lung cancer cells is a novel transcript variant. Through gain- and loss-of-function studies, AFAP1-AS1 was demonstrated to promote cell migration and invasion. Mechanistically, AFAP1-AS1 functions through positively regulating the expression of AFAP1 protein. On the other hand, the expression of lncRNA AFAP1-AS1 negatively correlates with CpG methylation status of its gene promoter, identified in both lung cancer cells and patient tissues, and treatment with DNA methyltransferase inhibitor decitabine significantly activates AFAP1-AS1 expression, strongly supporting that AFAP1-AS1 expression is tightly regulated by DNA methylation. Taken together, this study demonstrates that AFAP1-AS1 acts as an oncogene in NSCLC to promote cell migration partly by upregulating AFAP1 expression, while its own expression is controlled by DNA methylation, and highlights its diagnostic and therapeutic values for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Neoplasm Invasiveness/genetics , RNA, Long Noncoding/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , DNA Methylation , Humans , Lung Neoplasms/pathology , Microfilament Proteins/genetics , Neoplasm Invasiveness/pathology , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...