Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Acta Pharmacol Sin ; 43(6): 1568-1580, 2022 Jun.
Article in English | MEDLINE | ID: mdl-34522004

ABSTRACT

Dysregulation of the Hippo signaling pathway seen in many types of cancer is usually associated with a poor prognosis. Paris saponin VII (PSVII) is a steroid saponin isolated from traditional Chinese herbs with therapeutic action against various human cancers. In this study we investigated the effects of PSVII on human breast cancer (BC) cells and its anticancer mechanisms. We showed that PSVII concentration-dependently inhibited the proliferation of MDA-MB-231, MDA-MB-436 and MCF-7 BC cell lines with IC50 values of 3.16, 3.45, and 2.86 µM, respectively, and suppressed their colony formation. PSVII (1.2-1.8 µM) induced caspase-dependent apoptosis in the BC cell lines. PSVII treatment also induced autophagy and promoted autophagic flux in the BC cell lines. PSVII treatment decreased the expression and nuclear translocation of Yes-associated protein (YAP), a downstream transcriptional effector in the Hippo signaling pathway; overexpression of YAP markedly attenuated PSVII-induced autophagy. PSVII-induced, YAP-mediated autophagy was associated with increased active form of LATS1, an upstream effector of YAP. The activation of LATS1 was involved the participation of multiple proteins (including MST2, MOB1, and LATS1 itself) in an MST2-dependent sequential activation cascade. We further revealed that PSVII promoted the binding of LATS1 with MST2 and MOB1, and activated LATS1 in the BC cell lines. Molecular docking showed that PSVII directly bound to the MST2-MOB1-LATS1 ternary complex. Microscale thermophoresis analysis and drug affinity responsive targeting stability assay confirmed the high affinity between PSVII and the MST2-MOB1-LATS1 ternary complex. In mice bearing MDA-MB-231 cell xenograft, administration of PSVII (1.5 mg/kg, ip, 4 times/week, for 4 weeks) significantly suppressed the tumor growth with increased pLATS1, LC3-II and Beclin 1 levels and decreased YAP, p62 and Ki67 levels in the tumor tissue. Overall, this study demonstrates that PSVII is a novel and direct Hippo activator that has great potential in the treatment of BC.


Subject(s)
Breast Neoplasms , Saponins , Animals , Autophagy , Breast Neoplasms/drug therapy , Cell Proliferation , Female , Hippo Signaling Pathway , Humans , Mice , Molecular Docking Simulation , Protein Serine-Threonine Kinases , Saponins/pharmacology , Saponins/therapeutic use
2.
BMC Pulm Med ; 21(1): 420, 2021 Dec 20.
Article in English | MEDLINE | ID: mdl-34923982

ABSTRACT

BACKGROUND: In recent years, immunotherapies and targeted therapies contribute to population-level improvement in NSCLC cancer-specific survival, however, the two novel therapeutic options have mainly benefit patients containing mutated driven genes. Thus, to explore other potential genes related with immunity or targeted therapies may provide novel options to improve survival of lung cancer patients without mutated driven genes. CTSF is unique in human cysteine proteinases. Presently, CTSF has been detected in several cell lines of lung cancer, but its role in progression and prognosis of lung cancer remains unclear. METHODS: CTSF expression and clinical datasets of lung cancer patients were obtained from GTEx, TIMER, CCLE, THPA, and TCGA, respectively. Association of CTSF expression with clinicopathological parameters and prognosis of lung cancer patients was analyzed using UALCAN and Kaplan-Meier Plotter, respectively. LinkedOmics were used to analyze correlation between CTSF and CTSF co-expressed genes. Protein-protein interaction and gene-gene interaction were analyzed using STRING and GeneMANIA, respectively. Association of CTSF with molecular markers of immune cells and immunomodulators was analyzed with Immunedeconv and TISIDB, respectively. RESULTS: CTSF expression was currently only available for patients with NSCLC. Compared to normal tissues, CTSF was downregulated in NSCLC samples and high expressed CTSF was correlated with favorable prognosis of NSCLC. Additionally, CTSF expression was correlated with that of immune cell molecular markers and immunomodulators both in LUAD and LUSC. Noticeably, high expression of CTSF-related CTLA-4 was found to be associated with better OS of LUAD patients. Increased expression of CTSF-related LAG-3 was related with poor prognosis of LUAD patients while there was no association between CTSF-related PD-1/PD-L1 and prognosis of LUAD patients. Moreover, increased expression of CTSF-related CD27 was related with poor prognosis of LUAD patients while favorable prognosis of LUSC patients. CONCLUSIONS: CTSF might play an anti-tumor effect via regulating immune response of NSCLC.


Subject(s)
CTLA-4 Antigen , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cathepsin F , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Biomarkers, Tumor , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cathepsin F/genetics , Cathepsin F/immunology , Computational Biology , Databases, Genetic , Down-Regulation , Epistasis, Genetic , Humans , Lung Neoplasms/pathology , Prognosis
3.
Arch Biochem Biophys ; 687: 108285, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32074500

ABSTRACT

Polyphyllin I (PPI), a bioactive constituent extracted from the rhizomes of Paris polyphylla, is cytotoxic to several cancer types. This study was designed to explore whether PPI prevents non-small-cell lung cancer (NSCLC) growth and to investigate the molecular mechanism. AMP-activated protein kinase (AMPK) has been implicated in the activation of autophagy in distinct tissues. In cultured human NSCLC cell lines, PPI induces autophagy by activating AMPK and then inhibiting mTOR signaling in a concentration-dependent manner. Furthermore, the activation of autophagy induced by PPI was reversed by the AMPK inhibitor compound C. Computational docking showed that PPI directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. Microscale thermophoresis and Drug Affinity Responsive Targeting Stability (DARTS) assay further confirmed the high affinity between PPI and AMPK. In vivo studies indicated that PPI suppressed the growth of NSCLC and increased the levels of LC3-II and phosphorylated AMPK in tumors isolated from a xenograft model of NSCLC in mice. Moreover, PPI exhibited favorable pharmacokinetics in rats. In summary, PPI conclusively acts as a direct AMPK activator to induce cell autophagy which inhibits the growth of NSCLC cells. In the future, PPI therapy should be applied to treat patients with NSCLC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Diosgenin/analogs & derivatives , Enzyme Activators/therapeutic use , Lung Neoplasms/drug therapy , AMP-Activated Protein Kinases/chemistry , Allosteric Site , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Diosgenin/metabolism , Diosgenin/pharmacokinetics , Diosgenin/therapeutic use , Enzyme Activators/metabolism , Enzyme Activators/pharmacokinetics , Female , Humans , Male , Mice, Nude , Molecular Docking Simulation , Protein Binding , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
4.
J Pharmacol Sci ; 139(4): 304-310, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30852180

ABSTRACT

Acute myeloid leukemia (AML) is the most common subtype of hematological malignancy in humans, and its incidence increases with age. The treatment of AML still faces challenges. Therefore, there is an urgent need to develop more effective targeted therapies. The receptor tyrosine kinase C-KIT confers critical proliferative signals to AML. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an endogenous inhibitor of protein phosphatase 2A (PP2A), which promotes the growth and transformation of various solid tumors. These actions make CIP2A a promising target for tumor treatment. Here, we reported the effects and underlying mechanisms of a natural compound, cucurbitacin B (CuB), on AML. We reported that CuB suppressed growth and induced apoptosis in AML cells. The inhibition of growth and activation of apoptosis were mediated through CuB-induced downregulation of the CIP2A/PP2A/C-KIT signal pathway. Furthermore, CuB inactivated the JAK2 and STAT3 molecules downstream of C-KIT via the downregulation of CIP2A. These results advance our understanding of CuB-induced growth inhibition and apoptosis and support further investigation of CuB as a CIP2A inhibitor for AML therapies.


Subject(s)
Autoantigens/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Membrane Proteins/metabolism , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Triterpenes/pharmacology , Animals , Autoantigens/genetics , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid, Acute/pathology , Male , Membrane Proteins/genetics , Mice, Nude , Molecular Targeted Therapy , Protein Phosphatase 2/genetics , Proto-Oncogene Proteins c-kit/genetics , Triterpenes/therapeutic use , Tumor Cells, Cultured
5.
Molecules ; 24(3)2019 Feb 12.
Article in English | MEDLINE | ID: mdl-30759826

ABSTRACT

Non-small cell lung cancer (NSCLC) patients carrying an epidermal growth factor receptor (EGFR) mutation are initially sensitive to EGFR-tyrosine kinase inhibitors (TKIs) treatment, but soon develop an acquired resistance. The treatment effect of EGFR-TKIs-resistant NSCLC patients still faces challenges. Cucurbitacin B (CuB), a triterpene hydrocarbon compound isolated from plants of various families and genera, elicits anticancer effects in a variety of cancer types. However, whether CuB is a viable treatment option for gefitinib-resistant (GR) NSCLC remains unclear. Here, we investigated the anticancer effects and underlying mechanisms of CuB. We report that CuB inhibited the growth and invasion of GR NSCLC cells and induced apoptosis. The inhibitory effect of CuB occurred through its promotion of the lysosomal degradation of EGFR and the downregulation of the cancerous inhibitor of protein phosphatase 2A/protein phosphatase 2A/Akt (CIP2A/PP2A/Akt) signaling axis. CuB and cisplatin synergistically inhibited tumor growth. A xenograft tumor model indicated that CuB inhibited tumor growth in vivo. Immunohistochemistry results further demonstrated that CuB decreased EGFR and CIP2A levels in vivo. These findings suggested that CuB could suppress the growth and invasion of GR NSCLC cells by inducing the lysosomal degradation of EGFR and by downregulating the CIP2A/PP2A/Akt signaling axis. Thus, CuB may be a new drug candidate for the treatment of GR NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Gefitinib/pharmacology , Lung Neoplasms/drug therapy , Lysosomes/drug effects , Signal Transduction/drug effects , Triterpenes/pharmacology , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autoantigens/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Down-Regulation/drug effects , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lysosomes/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism
6.
Front Pharmacol ; 10: 1503, 2019.
Article in English | MEDLINE | ID: mdl-31969821

ABSTRACT

Ethoxysanguinarine (Eth) is a benzophenanthridine alkaloid extracted from Macleaya cordata (Willd) R. Br. It possesses antibacterial and antiviral activities and offers therapeutic benefits for the treatment of respiratory syndrome virus-induced cytopathic effects. However, the effect of Eth on human tumors and its pharmacological effects remain to be elucidated, together with its cellular target. Here, we examined the effects of Eth on breast cancer (BC) cells. We found that at low doses, Eth strongly inhibited the viability of BC cell lines and induced autophagy. Mechanistic studies showed that Eth induced autophagy by upregulating the activity of the AMP-activated protein kinase (AMPK). The AMPK inhibitor compound C significantly attenuated Eth-induced autophagy and inhibited proliferation. Meanwhile, the AMPK activator metformin significantly enhanced Eth-induced autophagy and inhibited proliferation. Computational docking and affinity assays showed that Eth directly interacted with the allosteric drug and metabolite site of AMPK to stabilize its activation. AMPK was less activated in tumor samples compared to normal breast tissues and was inversely associated with the prognosis of the patients. Moreover, Eth exhibited potent anti-BC activity in nude mice and favorable pharmacokinetics in rats. These characteristics render Eth as a promising candidate drug for further development and for designing new effective AMPK activators.

7.
J Cell Physiol ; 234(7): 10360-10371, 2019 07.
Article in English | MEDLINE | ID: mdl-30417376

ABSTRACT

Patients with non-small-cell lung cancer (NSCLC) are routinely treated with the platinum-based chemotherapeutics such as cisplatin. The drug exerts anticancer effects via multiple mechanisms, including DNA double-strand breaks (DSBs). Enhanced DNA DSB repair capacity would be associated with innate or acquired drug resistance. However, despite strong evidence for the role of the chromokinesin kinesin family member 4A (KIF4A) in DSB repair, the relationship between the chromokinesin and cisplatin sensitivity of human NSCLC cells remains unknown. Furthermore, little is known regarding the effect of targeting KIF4A on the function of DSB repair-related proteins in these cells. In the current study, we demonstrated that cisplatin treatment stimulated the expression of KIF4A protein in human NSCLC cells. Depletion of KIF4A by small interfering RNA significantly enhanced cisplatin-induced cell cycle arrest in S and G2/M phases and cytotoxicity in human NSCLC cells. Furthermore, we found that KIF4A inhibition suppressed the ability of cisplatin to induce BRCA2 and Rad51 focus formation and limits the further increase in poly(ADP-ribose) polymerase 1 activity induced by cisplatin treatment in human NSCLC cells. These studies thus identify the chromokinesin KIF4A as a novel modulator of cisplatin sensitivity that is significantly enhanced by the chromokinesin in human NSCLC cells via multiple mechanisms.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA/drug effects , Kinesins/metabolism , Lung Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , BRCA2 Protein/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Rad51 Recombinase/metabolism
8.
Cancer Lett ; 408: 60-70, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28844714

ABSTRACT

Metastatic spread of cancer cells is the most life-threatening aspect of breast cancer and involves multiple steps including cell migration. We recently found that the TBC1D3 oncogene promotes the migration of breast cancer cells, and its interaction with CaM enhances the effects of TBC1D3. However, little is known regarding the mechanism by which TBC1D3 induces the migration of cancer cells. Here, we demonstrated that TBC1D3 stimulated the expression of oxidized low density lipoprotein receptor 1 (OLR1), a stimulator of cell migration, in breast cancer cells at the transcriptional level. Depletion of OLR1 by siRNAs or down-regulation of OLR1 expression using pomalidomide, a TNFα inhibitor, significantly decreased TBC1D3-induced migration of these cells. Notably, TBC1D3 overexpression activated NF-κB, a major effector of TNFα signaling, while inhibition of TNFα signaling suppressed the effects of TBC1D3. Consistent with this, NF-κB inhibition using its specific inhibitor caffeic acid phenethyl ester decreased both TBC1D3-induced OLR1 expression and cell migration, suggesting a critical role for TNFα/NF-κB signaling in TBC1D3-induced migration of breast cancer cells. Mechanistically, TBC1D3 induced activation of this signaling pathway on multiple levels, including by increasing the release of TNFα, elevating the transcription of TNFR1, TRAF1, TRAF5 and TRAF6, and decreasing the degradation of TNFR1. In summary, these studies identify the TBC1D3 oncogene as a novel regulator of TNFα/NF-κB signaling that mediates this oncogene-induced migration of human breast cancer cells by up-regulating OLR1.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement , GTPase-Activating Proteins/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , Scavenger Receptors, Class E/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Proliferation , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/genetics , Scavenger Receptors, Class E/antagonists & inhibitors , Scavenger Receptors, Class E/genetics , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
9.
Oncotarget ; 8(22): 36383-36398, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28422741

ABSTRACT

The hominoid oncoprotein TBC1D3 enhances growth factor (GF) signaling and GF signaling, conversely, induces the ubiquitination and subsequent degradation of TBC1D3. However, little is known regarding the regulation of this degradation, and the role of TBC1D3 in the progression of tumors has also not been defined. In the present study, we demonstrated that calmodulin (CaM), a ubiquitous cellular calcium sensor, specifically interacted with TBC1D3 in a Ca2+-dependent manner and inhibited GF signaling-induced ubiquitination and degradation of the oncoprotein in both cytoplasm and nucleus of human breast cancer cells. The CaM-interacting site of TBC1D3 was mapped to amino acids 157~171, which comprises two 1-14 hydrophobic motifs and one lysine residue (K166). Deletion of these motifs was shown to abolish interaction between TBC1D3 and CaM. Surprisingly, this deletion mutation caused inability of GF signaling to induce the ubiquitination and subsequent degradation of TBC1D3. In agreement with this, we identified lysine residue 166 within the CaM-interacting motifs of TBC1D3 as the actual site for the GF signaling-induced ubiquitination using mutational analysis. Point mutation of this lysine residue exhibited the same effect on TBC1D3 as the deletion mutant, suggesting that CaM inhibits GF signaling-induced degradation of TBC1D3 by occluding its ubiquitination at K166. Notably, we found that TBC1D3 promoted the expression and activation of MMP-9 and the migration of MCF-7 cells. Furthermore, interaction with CaM considerably enhanced such effect of TBC1D3. Taken together, our work reveals a novel model by which CaM promotes cell migration through inhibiting the ubiquitination and degradation of TBC1D3.


Subject(s)
Breast Neoplasms/metabolism , Calmodulin/metabolism , GTPase-Activating Proteins/metabolism , Matrix Metalloproteinase 9/metabolism , Proto-Oncogene Proteins/metabolism , Binding Sites , Breast Neoplasms/genetics , Calcium/metabolism , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinase 9/genetics , Mutation , Protein Binding , Proteolysis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Signal Transduction , Ubiquitination
10.
PLoS One ; 9(4): e94134, 2014.
Article in English | MEDLINE | ID: mdl-24714105

ABSTRACT

The hominoid oncogene TBC1D3 enhances epidermal growth factor receptor (EGFR) signaling and induces cell transformation. However, little is known regarding its spatio-temporal regulation and mechanism of tumorigenesis. In the current study, we identified the microtubule subunit ß-tubulin as a potential interaction partner for TBC1D3 using affinity purification combined with mass spectrometry analysis. The interaction between TBC1D3 and ß-tubulin was confirmed by co-immunoprecipitation. Using the same method, we also revealed that TBC1D3 co-precipitated with endogenous α-tubulin, another subunit of the microtubule. In agreement with these results, microtubule cosedimentation assays showed that TBC1D3 associated with the microtubule network. The ß-tubulin-interacting site of TBC1D3 was mapped to amino acids 286∼353 near the C-terminus of the TBC domain. Deletion mutation within these amino acids was shown to abolish the interaction of TBC1D3 with ß-tubulin. Interestingly, the deletion mutation caused a complete loss of TBC1D3 from the cytoplasmic filamentous and punctate structures, and TBC1D3 instead appeared in the nucleus. Consistent with this, wild-type TBC1D3 exhibited the same nucleocytoplasmic distribution in cells treated with the microtubule depolymerizing agent nocodazole, suggesting that the microtubule network associates with and retains TBC1D3 in the cytoplasm. We further found that deficiency in ß-tubulin-interacting resulted in TBC1D3's inability to inhibit c-Cbl recruitment and EGFR ubiquitination, ultimately leading to dysregulation of EGFR degradation and signaling. Taken together, these studies indicate a novel model by which the microtubule network regulates EGFR stability and signaling through tubulin dimer/oligomer interaction with the nucleocytoplasmic protein TBC1D3.


Subject(s)
Cytoplasm/metabolism , ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , Microtubules/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Humans , Tubulin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...