Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
Cell Rep ; 43(4): 114002, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38547126

ABSTRACT

The dysfunction of matriptase, a membrane-anchored protease, is highly related to the progression of skin and breast cancers. Epidermal growth factor (EGF)-induced matriptase activation and cancer invasion are known but with obscure mechanisms. Here, we demonstrate a vesicular-trafficking-mediated interplay between matriptase and EGF signaling in cancer promotion. We found that EGF induces matriptase to undergo endocytosis together with the EGF receptor, followed by acid-induced activation in endosomes. Activated matriptase is then secreted extracellularly on exosomes to catalyze hepatocyte growth factor precursor (pro-HGF) cleavage, resulting in autocrine HGF/c-Met signaling. Matriptase-induced HGF/c-Met signaling represents the second signal wave of EGF, which promotes cancer cell scattering, migration, and invasion. These findings demonstrate a role of vesicular trafficking in efficient activation and secretion of membrane matriptase and a reciprocal regulation of matriptase and EGF signaling in cancer promotion, providing insights into the physiological functions of vesicular trafficking and the molecular pathological mechanisms of skin and breast cancers.


Subject(s)
Breast Neoplasms , Neoplasm Invasiveness , Serine Endopeptidases , Signal Transduction , Animals , Female , Humans , Mice , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Endocytosis , Endosomes/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Exosomes/metabolism , Hepatocyte Growth Factor/metabolism , Protein Precursors , Proto-Oncogene Proteins c-met/metabolism , Serine Endopeptidases/metabolism , Skin Neoplasms/pathology , Skin Neoplasms/metabolism
2.
Adv Healthc Mater ; : e2303445, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38290499

ABSTRACT

The application of oncolytic peptides has become a powerful approach to induce complete and long-lasting remission in multiple types of carcinomas, as affirmed by the appearance of tumor-associated antigens and adenosine triphosphate (ATP) in large quantities, which jumpstarts the cancer-immunity cycle. However, the ATP breakdown product adenosine is a significant contributor to forming the immunosuppressive tumor microenvironment, which substantially weakens peptide-driven oncolytic immunotherapy. In this study, a lipid-coated micelle (CA@TLM) loaded with a stapled oncolytic peptide (PalAno) and an adenosine 2A receptor (A2AR) inhibitor (CPI-444) is devised to enact tumor-targeted oncolytic immunotherapy and to overcome adenosine-mediated immune suppression simultaneously. The CA@TLM micelle accumulates in tumors with high efficiency, and the acidic lysosomal environment prompts the rapid release of PalAno and CPI-444. Subsequently, PalAno induces swift membrane lysis of tumor cells and the release of antigenic materials. Meanwhile, CPI-444 blocks activation of the immunosuppressive adenosine-A2AR signaling pathway. This combined approach exhibit pronounced synergy at stalling tumor growth and metastasis in animal models for triple-negative breast cancer (TNBC) and melanoma, providing a novel strategy for enhanced oncolytic immunotherapy. This article is protected by copyright. All rights reserved.

3.
Zhongguo Zhong Yao Za Zhi ; 48(18): 4981-4992, 2023 Sep.
Article in Chinese | MEDLINE | ID: mdl-37802840

ABSTRACT

This study constructed a nano-drug delivery system, A3@GMH, by co-delivering the stapled anoplin peptide(Ano-3, A3) with the light-harvesting material graphene oxide(GO), and evaluated its oncolytic immunotherapy effect on triple-negative breast cancer(TNBC). A3@GMH was prepared using an emulsion template method and its physicochemical properties were characterized. The in vivo and in vitro photothermal conversion abilities of A3@GMH were investigated using an infrared thermal imager. The oncoly-tic activity of A3@GMH against TNBC 4T1 cells was evaluated through cell counting kit-8(CCK-8), lactate dehydrogenase(LDH) release, live/dead cell staining, and super-resolution microscopy. The targeting properties of A3@GMH on 4T1 cells were assessed using a high-content imaging system and flow cytometry. In vitro and in vivo studies were conducted to investigate the antitumor mechanism of A3@GMH in combination with photothermal therapy(PTT) through inducing immunogenic cell death(ICD) in 4T1 cells. The results showed that the prepared A3@GMH exhibited distinct mesoporous and coated structures with an average particle size of(308.9±7.5) nm and a surface potential of(-6.79±0.58) mV. The encapsulation efficiency and drug loading of A3 were 23.9%±0.6% and 20.5%±0.5%, respectively. A3@GMH demonstrated excellent photothermal conversion ability and biological safety. A3@GMH actively mediated oncolytic features such as 4T1 cell lysis and LDH release, as well as ICD effects, and showed enhanced in vitro antitumor activity when combined with PTT. In vivo, A3@GMH efficiently induced ICD effects with two rounds of PTT, activated the host's antitumor immune response, and effectively suppressed tumor growth in 4T1 tumor-bearing mice, achieving an 88.9% tumor inhibition rate with no apparent toxic side effects. This study suggests that the combination of stapled anoplin peptide and PTT significantly enhances the oncolytic immunotherapy for TNBC and provides a basis for the innovative application of anti-tumor peptides derived from TCM in TNBC treatment.


Subject(s)
Nanoparticles , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Photothermal Therapy , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Antimicrobial Cationic Peptides , Immunotherapy/methods , Cell Line, Tumor , Phototherapy/methods , Nanoparticles/chemistry
4.
BMC Biol ; 21(1): 197, 2023 09 21.
Article in English | MEDLINE | ID: mdl-37735649

ABSTRACT

BACKGROUND: The maturation of microRNAs (miRNAs) successively undergoes Drosha, Dicer, and Argonaute -mediated processing, however, the intricate regulations of the individual miRNA maturation are largely unknown. Retinoid x receptor alpha (RXRα) belongs to nuclear receptors that regulate gene transcription by binding to DNA elements, however, whether RXRα binds to miRNAs to exert physiological functions is not known. RESULTS: In this work, we found that RXRα directly binds to the precursor of miR-103 (pre-miR-103a-2) via its DNA-binding domain with a preferred binding sequence of AGGUCA. The binding of RXRα inhibits the processing of miR-103 maturation from pre-miR-103a-2. Mechanistically, RXRα prevents the nuclear export of pre-miR-103a-2 for further processing by inhibiting the association of exportin-5 with pre-miR-103a-2. Pathophysiologically, the negative effect of RXRα on miR-103 maturation correlates to the positive effects of RXRα on the expression of Dicer, a target of miR-103, and on the inhibition of breast cancer. CONCLUSIONS: Our findings unravel an unexpected role of transcription factor RXRα in specific miRNA maturation at post-transcriptional level through pre-miRNA binding, and present a mechanistic insight regarding RXRα role in breast cancer progression.


Subject(s)
MicroRNAs , Receptors, Cytoplasmic and Nuclear , Transcription Factors , Argonaute Proteins , MicroRNAs/genetics
5.
Molecules ; 28(3)2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36770929

ABSTRACT

Prostate adenocarcinoma (PRAD) is the most frequent malignancy, and is the second leading cause of death due to cancer in men. Thus, new prognostic biomarkers and drug targets for PRAD are urgently needed. As we know, nuclear receptor Nur77 is important in cancer development and changes in the tumor microenvironment; whereas, the function of Nur77 in PRAD remains to be elucidated. The TCGA database was used to explore the Nur77 expression and its role in the prognosis of PRAD. It was shown that Nur77 was down regulated in PRAD, and low Nur77 expression was correlated with advanced clinical pathologic characteristics (high grade, histological type, age) and poor prognosis. Furthermore, key genes screening was examined by univariate Cox analysis and Kaplan-Meier survival. Additionally, Nur77 was closely related to immune infiltration and some anti-tumor immune functions. The differentially expressed genes (DEGs) were presented by protein-protein interaction (PPI) network analysis. Therefore, the expression level of Nur77 might help predict the survival of PRAD cases, which presents a new insight and a new target for the treatment of PRAD. In vitro experiments verified that natural product malayoside targeting Nur77 exhibited significant therapeutic effects on PRAD and largely induced cell apoptosis by up-regulating the expression of Nur77 and its mitochondrial localization. Taken together, Nur77 is a prognostic biomarker for patients with PRAD, which may refresh the profound understanding of PRAD individualized treatment.


Subject(s)
Adenocarcinoma , Nuclear Receptor Subfamily 4, Group A, Member 1 , Prostatic Neoplasms , Humans , Male , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Biomarkers , Prognosis , Prostate , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Tumor Microenvironment/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
6.
Mar Drugs ; 20(12)2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36547883

ABSTRACT

Six new citreoviridins (citreoviridins J-O, 1-6) and twenty-two known compounds (7-28) were isolated from the deep-sea-derived Penicillium citreonigrum MCCC 3A00169. The structures of the new compounds were determined by spectroscopic methods, including the HRESIMS, NMR, ECD calculations, and dimolybdenum tetraacetate-induced CD (ICD) experiments. Citreoviridins J-O (1-6) are diastereomers of 6,7-epoxycitreoviridin with different chiral centers at C-2-C-7. Pyrenocine A (7), terrein (14), and citreoviridin (20) significantly induced apoptosis for HeLa cells with IC50 values of 5.4 µM, 11.3 µM, and 0.7 µM, respectively. To be specific, pyrenocine A could induce S phase arrest, while terrein and citreoviridin could obviously induce G0-G1 phase arrest. Citreoviridin could inhibit mTOR activity in HeLa cells.


Subject(s)
Penicillium , Humans , HeLa Cells , Cell Line, Tumor , Penicillium/chemistry , Magnetic Resonance Spectroscopy/methods , Molecular Structure
7.
J Biol Chem ; 298(12): 102665, 2022 12.
Article in English | MEDLINE | ID: mdl-36334626

ABSTRACT

The glucagon receptor (GCGR) is a potential target for diabetes therapy. Several emerging GCGR antagonism-based therapies are under preclinical and clinical development. However, GCGR antagonism, as well as genetically engineered GCGR deficiency in animal models, are accompanied by α-cell hyperplasia and hyperglucagonemia, which may limit the application of GCGR antagonism. To better understand the physiological changes in α cells following GCGR disruption, we performed single cell sequencing of α cells isolated from control and gcgr-/- (glucagon receptor deficient) zebrafish. Interestingly, beyond the α-cell hyperplasia, we also found that the expression of gcga, gcgb, pnoca, and several glucagon-regulatory transcription factors were dramatically increased in one cluster of gcgr-/- α cells. We further confirmed that glucagon mRNA was upregulated in gcgr-/- animals by in situ hybridization and that glucagon promoter activity was increased in gcgr-/-;Tg(gcga:GFP) reporter zebrafish. We also demonstrated that gcgr-/- α cells had increased glucagon protein levels and increased granules after GCGR disruption. Intriguingly, the increased mRNA and protein levels could be suppressed by treatment with high-level glucose or knockdown of the pnoca gene. In conclusion, these data demonstrated that GCGR deficiency not only induced α-cell hyperplasia but also increased glucagon expression in α cells, findings which provide more information about physiological changes in α-cells when the GCGR is disrupted.


Subject(s)
Glucagon , Receptors, Glucagon , Animals , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Hyperplasia , RNA, Messenger
8.
Biomed Pharmacother ; 151: 113076, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35550529

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is the most common liver disease that may progress to nonalcoholic steatohepatitis (NASH), hepatic tissue fibrosis, liver cirrhosis, and hepatocellular carcinoma. In this study, we investigated the effects of Pien Tze Huang (PTH), a well-known traditional Chinese herbal formula with liver protective effect, in methionine-choline deficient diet (MCD)- and high-fat diet (HFD)-induced NASH mouse models. Our results showed that PTH could exert hepatoprotective effects by improving liver weight and steatosis and reducing the fibrosis and serum levels of alanine transaminase (ALT) and aspartate transaminase (AST) in both animal models. The effects of PTH was accompanied with the reduction of infiltrated macrophages, the inhibition of the expression of cytokines, and the induction of adiponectin expression. Mechanistically, we found that PTH could inhibit the activation of proinflammatory transcription factor nuclear factor-κB (NF-κB) by preventing the degradation of inhibitor of κBα (IκBα). These results demonstrate that PTH can improve NAFLD largely due to its suppression of the NF-κB inflammatory pathway.


Subject(s)
Drugs, Chinese Herbal , Non-alcoholic Fatty Liver Disease , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Disease Models, Animal , Drugs, Chinese Herbal/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Fibrosis , Liver , Liver Cirrhosis/metabolism , Methionine/pharmacology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Non-alcoholic Fatty Liver Disease/metabolism
9.
Biochem Biophys Res Commun ; 591: 118-123, 2022 02 05.
Article in English | MEDLINE | ID: mdl-35007835

ABSTRACT

3-chyomotrypsin like protease (3CLpro) has been considered as a promising target for developing anti-SARS-CoV-2 drugs. Herein, about 6000 compounds were analyzed by high-throughput screening using enzyme activity model, and Merbromin, an antibacterial agent, was identified as a potent inhibitor of 3CLpro. Merbromin strongly inhibited the proteolytic activity of 3CLpro but not the other three proteases Proteinase K, Trypsin and Papain. Michaelis-Menten kinetic analysis showed that Merbromin was a mixed-type inhibitor of 3CLpro, due to its ability of increasing the KM and decreasing the Kcat of 3CLpro. The binding assays and molecular docking suggested that 3CLpro possessed two binding sites for Merbromin. Consistently, Merbromin showed a weak binding to the other three proteases. Together, these findings demonstrated that Merbromin is a selective inhibitor of 3CLpro and provided a scaffold to design effective inhibitors of SARS-CoV-2.


Subject(s)
Coronavirus 3C Proteases/antagonists & inhibitors , Merbromin/pharmacology , Molecular Docking Simulation , Protease Inhibitors/pharmacology , SARS-CoV-2/drug effects , Binding Sites , COVID-19/prevention & control , COVID-19/virology , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , High-Throughput Screening Assays/methods , Humans , Kinetics , Merbromin/chemistry , Merbromin/metabolism , Models, Molecular , Molecular Structure , Protease Inhibitors/chemistry , Protease Inhibitors/metabolism , Protein Binding , Protein Domains , SARS-CoV-2/enzymology , SARS-CoV-2/physiology , Surface Plasmon Resonance/methods
10.
Psychol Health Med ; 27(2): 403-408, 2022 02.
Article in English | MEDLINE | ID: mdl-33971765

ABSTRACT

This study aimed to explore which age group out of the patients in quarantine wards with novel coronavirus pneumonia is the most susceptible to anxiety. The data of 32 Covid-19 patients isolated in the quarantine wards of the second Infectious Diseases Department of Baoding Hospital and 71 Covid-19 patients in Tangshan City Infectious Disease Hospital from January 24th to March 5th, 2020, a total of 103 patients, were analyzed. Among these patients, 97 isolated patients were scored with a self-rating anxiety scale (SAS) score seven days after quarantine, and the correlation between age and score was analyzed. These 97 isolated patients were then divided into three groups according to age: group A (up to 35 years old), group B (36-60 years), and group C (over 60 years). One-way analysis of variance was used to compare the scores among groups. The Q-test was used for pairwise comparison.P < 0.05 was considered statistically significant.There was a negative correlation between age and SAS score in isolated Covid-19 patients, and the differences in the score among groups were statistically significant. Patients under 35 years old were more prone to anxiety when they were isolated for seven days. Isolated patients aged up to 35 years old need more attention from quarantine medical staff, communication should be strengthened, and psychological intervention from psychotherapists should be given if necessary.


Subject(s)
COVID-19 , Quarantine , Adult , Aged , Anxiety/epidemiology , Anxiety/psychology , COVID-19/epidemiology , Humans , Quarantine/psychology , SARS-CoV-2 , Surveys and Questionnaires
11.
Nat Commun ; 12(1): 5989, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645818

ABSTRACT

Liquid-liquid phase separation promotes the formation of membraneless condensates that mediate diverse cellular functions, including autophagy of misfolded proteins. However, how phase separation participates in autophagy of dysfunctional mitochondria (mitophagy) remains obscure. We previously discovered that nuclear receptor Nur77 (also called TR3, NGFI-B, or NR4A1) translocates from the nucleus to mitochondria to mediate celastrol-induced mitophagy through interaction with p62/SQSTM1. Here, we show that the ubiquitinated mitochondrial Nur77 forms membraneless condensates capable of sequestrating damaged mitochondria by interacting with the UBA domain of p62/SQSTM1. However, tethering clustered mitochondria to the autophagy machinery requires an additional interaction mediated by the N-terminal intrinsically disordered region (IDR) of Nur77 and the N-terminal PB1 domain of p62/SQSTM1, which confers Nur77-p62/SQSTM1 condensates with the magnitude and liquidity. Our results demonstrate how composite multivalent interaction between Nur77 and p62/SQSTM1 coordinates to sequester damaged mitochondria and to connect targeted cargo mitochondria for autophagy, providing mechanistic insight into mitophagy.


Subject(s)
Mitochondria/drug effects , Mitophagy/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Pentacyclic Triterpenes/pharmacology , Sequestosome-1 Protein/genetics , Animals , Electron Transport Complex IV , Female , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mitochondria/metabolism , Mitophagy/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Protein Binding , Protein Transport , Recombinant Fusion Proteins , Rheology , Sequestosome-1 Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Red Fluorescent Protein
12.
Nanoscale ; 13(41): 17648-17654, 2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34664606

ABSTRACT

Liposome-based immunoassay (LIA) is an attractive protocol for amplifying the detection signals because of the excellent ability of liposomes to encapsulate signal marker compounds. The antigen-binding activity of the conjugated antibodies on the liposomal surface is crucial for the specificity and sensitivity of LIA. We present here a general platform to ensure that antibodies can conjugate onto the surface of liposomes in a site-specific and oriented manner. A His-handle-modified antibody with Fc region-specific and covalent conjugation was first fabricated using a photoactivatable ZBpa-His tag that was engineered using the aminoacyl-tRNA synthetase/suppressor tRNA technique. Based on the high affinity between the His tag and divalent metal ions, the novel His-modified antibody was oriented onto the surface of nickel ion-modified liposomes encapsulating horseradish peroxidase. With the prostate-specific antigen as a model, the detection efficiency of the new immunoliposomes was evaluated by chemiluminescence immunoassay. The immunoliposomes exhibited a limit of detection of 0.2 pg mL-1, which was a six time improvement compared with that of the chemical-coupled antibody-liposome conjugates. Thus, the proposed immunoliposomes are expected to hold potential applications for the sensitive detection of various biomarkers in complicated serum samples.


Subject(s)
Immunoconjugates , Liposomes , Antibodies , Antigens , Humans , Immunoassay , Male
13.
STAR Protoc ; 2(3): 100495, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34195669

ABSTRACT

During eukaryotic cell mitosis, the nuclear envelope disintegrates and transcription factors are dissociated from condensed chromosomes. Here, we describe a protocol to study centrosomal translocation of nuclear receptor RXRα. We detail procedures for HeLa cell synchronization followed by immunofluorescence, in situ proximity ligation assay, and centrosome isolation. This protocol can be used to identify other transcription factors associated with the centrosome or other subcellular structures during mitotic progression. For complete details on the use and execution of this protocol, please refer to Xie et al. (2020).


Subject(s)
Centrosome/metabolism , Mitosis , Transcription Factors/metabolism , HeLa Cells , Humans , Subcellular Fractions/metabolism
14.
Eur J Med Chem ; 222: 113542, 2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34118723

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a valuable drug target for diabetic treatment and ligands of PPARγ have shown potent anti-diabetic efficacy. However, to overcome the severe side effects of current PPARγ-targeted drugs, novel PPARγ ligands need to be developed. Sulindac, an identified ligand of PPARγ, is widely used in clinic as a non-steroidal anti-inflammatory drug. To explore its potential application for diabetes, we designed and synthesized a series of sulindac derivatives to investigate their structure-activity relationship as PPARγ ligand and potential anti-diabetic effect. We found that meta-substitution in sulindac's benzylidene moiety was beneficial to PPARγ binding and transactivation. Z rather than E configuration of the benzylidene double bond endowed derivatives with the selectivity of PPARγ activation. The indene fluorine is essential for binding and regulating PPARγ. Compared with rosiglitazone, compound 6b with benzyloxyl meta-substitution and Z benzylidene double bond weakly induced adipogenesis and PPARγ-targeted gene expression. However, 6b potently improved glucose tolerance in a diabetic mice model. Unlike rosiglitazone, 6b was devoid of apparent toxicity to osteoblastic formation. Thus, we provided some useful guidelines for PPARγ-based optimization of sulindac and an anti-diabetic lead compound with less side effects.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Drug Design , Hypoglycemic Agents/pharmacology , PPAR gamma/agonists , Sulindac/pharmacology , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Mice , Mice, Inbred C57BL , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Sulindac/chemical synthesis , Sulindac/chemistry
15.
Biochem Pharmacol ; 190: 114622, 2021 08.
Article in English | MEDLINE | ID: mdl-34043967

ABSTRACT

Lung cancer is the leading cause of cancer deaths in the world. Non-small cell lung cancer (NSCLC), with poor prognosis and resistance to chemoradiotherapy, is the most common histological type of lung cancer. Therefore, it is necessary to develop new and more effective treatment strategy for NSCLC. Nur77, an orphan member of the nuclear receptor superfamily, induces apoptosis in cancer cells including NSCLC cells, by high expression and translocation to mitochondria. Small molecules trigger expression and mitochondrial localization of Nur77 may be an ideal anti-cancer drug candidate. Here, we report malayoside, a cardiac glycoside in the extract of Antiaris toxicaria Lesch., had different sensitivities to NSCLC cells. Malayoside induced apoptosis in NCI-H460 cells. Meanwhile, malayoside induced Nur77 expression and mitochondrial localization, and its induction of apoptosis was Nur77-dependent. To investigate the molecular mechanism of malayoside inducing Nur77 and apoptosis, we found that malayoside activated MAPK signaling pathway, including both ERK and p38 phosphorylation. The suppression of MAPK signaling activation inhibited the expression of Nur77 and apoptosis induced by malayoside. Our studies in nude mice showed that malayside potently inhibited the growth of tumor cells in vivo. Furthermore, the anti-cancer effect of malayosidwas in vivo was also related to the elevated expression of Nur77, p-ERK, and p-p38 proteins. Our results suggest that malayoside possesses an anti-NSCLC activity in vitro and in vivo mainly via activation of MAPK-Nur77 signaling pathway, indicating that malayoside is a promising chemotherapeutic candidate for NSCLC.


Subject(s)
Antiaris/chemistry , Apoptosis/drug effects , Cardiac Glycosides/pharmacology , Mitogen-Activated Protein Kinase Kinases/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Animals , Carcinoma, Non-Small-Cell Lung , Cardiac Glycosides/chemistry , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phytotherapy , Protein Transport/drug effects
16.
Nat Commun ; 12(1): 2809, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990575

ABSTRACT

The paradoxical roles of transforming growth factor-ß (TGFß) signaling and nuclear receptor Nur77 in colon cancer development are known but the underlying mechanisms remain obscure. Inhibitor of differentiation 1 (ID1) is a target gene of TGFß and a key promoter for colon cancer progression. Here, we show that Nur77 enhances TGFß/Smad3-induced ID1 mRNA expression through hindering Smurf2-mediated Smad3 mono-ubiquitylation, resulting in ID1 upregulation. In the absence of TGFß, however, Nur77 destabilizes ID1 protein by promoting Smurf2-mediated ID1 poly-ubiquitylation, resulting in ID1 downregulation. Interestingly, TGFß stabilizes ID1 protein by switching Nur77 interaction partners to inhibit ID1 ubiquitylation. This also endows TGFß with an active pro-tumorigenic action in Smad4-deficient colon cancers. Thus, TGFß converts Nur77's role from destabilizing ID1 protein and cancer inhibition to inducing ID1 mRNA expression and cancer promotion, which is highly relevant to colon cancer stemness, metastasis and oxaliplatin resistance. Our data therefore define the integrated duality of Nur77 and TGFß signaling in regulating ID1 expression and provide mechanistic insights into the paradoxical roles of TGFß and Nur77 in colon cancer progression.


Subject(s)
Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Inhibitor of Differentiation Protein 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Transforming Growth Factor beta/metabolism , Animals , Carcinogenesis , Cell Line, Tumor , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Inhibitor of Differentiation Protein 1/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Models, Biological , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Smad3 Protein/metabolism , Smad4 Protein/deficiency , Smad4 Protein/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
17.
Acta Pharm Sin B ; 11(2): 442-455, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33643823

ABSTRACT

Although different types of drugs are available for postmenopausal osteoporosis, the limitations of the current therapies including drug resistances and adverse effects require identification of novel anti-osteoporosis agents. Here, we defined that norlichexanthone (NOR), a natural product, is a ligand of estrogen receptor-alpha (ERα) and revealed its therapeutic potential for postmenopausal osteoporosis. We used mammalian-one hybrid assay to screen for ERα modulators from crude extracts of several plant endophytes. As a result, NOR purified from the extract of endophyte ARL-13 was identified as a selective ERα modulator. NOR directly bound to ERα with an affinity in nanomolar range, revealing that it is a natural ligand of ERα. NOR induced osteoblast formation in MC3T3-E1 precursor cells. Conversely, NOR inhibited receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclast formation in both RAW264.7 macrophages and mouse primary monocytes. Mechanistically, NOR inhibited RANKL-induced association of ERα and TRAF6 to prevent ERα-mediated TRAF6 activation via Lys63-linked ubiquitination. Importantly, NOR exhibited potent anti-osteoporosis efficacy in an ovariectomized mouse model. Comparing to estrogen, NOR was of much less capability in stimulating endometrial hyperplasia and promoting mammalian cancer cell proliferation. Taken together, our study identified NOR as a natural and high affinity ligand of ERα with substantial anti-osteoporosis but less estrogenic activity.

18.
Anal Bioanal Chem ; 413(3): 945-953, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33210177

ABSTRACT

Fluorophore-antibody conjugates with high photobleaching resistance, high chemical stability, and Fc-specific attachment is a great advantage for immunofluorescence imaging. Here, an Fc-binding protein (Z-domain) carrying a photo-cross-linker (p-benzoylphenylalanine, Bpa) fused with enhanced green fluorescent protein (EGFP), namely photoactivatable ZBpa-EGFP recombinant, was directly generated using the aminoacyl-tRNA synthetase/suppressor tRNA technique without any further modification. By employing the photoactivatable ZBpa-EGFP, an optimal approach was successfully developed which enabled EGFP to site-selectively and covalently attach to native antibody (IgG) with approximately 90% conjugation efficiency. After characterizing the Fc-specific and covalent manner of the EGFP-photoconjugated antibody, its excellent photobleaching resistance for immunofluorescence imaging was demonstrated in a model study by monitoring the toll-like receptor 4 (TLR4) expression in HepG2 cells. The proposed approach here for the preparation of a novel fluorescent antibody is available and reliable, which would play an important role in fluorescence immunoassay, and is expected to be extended to the generation of other biomolecule-photoconjugated antibodies, such as other fluorescent proteins for multiplex immunofluorescence imaging or reporter enzymes for highly sensitive enzyme immunoassays.Graphical abstract.


Subject(s)
Green Fluorescent Proteins/chemistry , Immunoglobulin Fc Fragments/chemistry , Microscopy, Fluorescence/methods , Antibodies, Monoclonal/chemistry , Flow Cytometry , Hep G2 Cells , Humans , Recombinant Fusion Proteins/chemistry
19.
Dev Cell ; 55(6): 707-722.e9, 2020 12 21.
Article in English | MEDLINE | ID: mdl-33321102

ABSTRACT

Retinoid X receptor alpha (RXRα), a nuclear receptor of transcription factor, controls various physiological and pathological pathways including cellular growth, proliferation, differentiation, and apoptosis. Here, we report that RXRα is phosphorylated at its N-terminal A/B domain by cyclin-dependent kinase 1 (Cdk1) at the onset of mitosis, triggering its translocation to the centrosome, where phosphorylated-RXRα (p-RXRα) interacts with polo-like kinase 1 (PLK1) through its N-terminal A/B domain by a unique mechanism. The interaction promotes PLK1 activation, centrosome maturation, and mitotic progression. Levels of p-RXRα are abnormally elevated in cancer cell lines, during carcinogenesis in animals, and in clinical tumor tissues. An RXRα ligand XS060, which specifically inhibits p-RXRα/PLK1 interaction but not RXRα heterodimerization, promotes mitotic arrest and catastrophe in a tumor-specific manner. These findings unravel a transcription-independent action of RXRα at the centrosome during mitosis and identify p-RXRα as a tumor-specific vulnerability for developing mitotic drugs with improved therapeutic index.


Subject(s)
Carcinogenesis/metabolism , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Mitosis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Retinoid X Receptor alpha/metabolism , Animals , Binding Sites , CDC2 Protein Kinase/metabolism , Carcinogenesis/pathology , Cell Cycle Proteins/chemistry , Female , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Protein Binding , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Retinoid X Receptor alpha/chemistry , Polo-Like Kinase 1
20.
Biochem Biophys Res Commun ; 530(1): 160-166, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32828280

ABSTRACT

Rosiglitazone is a ligand of peroxisome proliferation-activated receptor gamma (PPARγ). However, it exerts biological activities and therapeutic effects through both PPARγ-dependent and independent mechanisms. In this study, we defined that rosiglitazone was also a ligand of retinoid X receptor alpha (RXRα) and displayed RXRα-dependent activities. We found that rosiglitazone directly bound to the ligand binding domain (LBD) of RXRα and induced RXRα/LBD tetramerization. Rosiglitazone inhibited the agonist-induced transcriptional activity of RXRα homodimers and heterodimers likely through inhibiting RXRα homo- and hetero-dimerization. In acute promyelocytic leukemia (APL) NB4 cells, rosiglitazone inhibited cell proliferation and induced cell differentiation, resulting from inhibiting RXRα/PML-RARα complex formation and down-regulating PML-RARα. Together, our study identified RXRα as a novel target of rosiglitazone and RXRα mediating the anti-APL activity of rosiglitazone.


Subject(s)
Cell Differentiation/drug effects , Hypoglycemic Agents/pharmacology , Protein Multimerization/drug effects , Retinoid X Receptor alpha/metabolism , Rosiglitazone/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , HEK293 Cells , Humans , Leukemia, Promyelocytic, Acute/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...