Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 124
1.
Biomolecules ; 13(6)2023 06 06.
Article En | MEDLINE | ID: mdl-37371529

Recent developments in CFTR modulator drugs have had a significant transformational effect on the treatment of individuals with Cystic Fibrosis (CF) who carry the most frequent F508del-CFTR mutation in at least one allele. However, the clinical effects of these revolutionary drugs remain limited by their inability to fully restore the plasma membrane (PM) stability of the rescued mutant channels. Here, we shed new light on the molecular mechanisms behind the reduced half-life of rescued F508del-CFTR at the PM of airway cells. We describe that YES1 protein kinase is enriched in F508del-CFTR protein PM complexes, and that its interaction with rescued channels is mediated and dependent on the adaptor protein YAP1. Moreover, we show that interference with this complex, either by depletion of one of these components or inhibiting YES1 activity, is sufficient to significantly improve the abundance and stability of modulator-rescued F508del-CFTR at the surface of airway cells. In addition, we found that this effect was mediated by a decreased phosphorylation of the scaffold protein SHC1, a key regulator of MAPK pathway activity. In fact, we showed that depletion of SHC1 or inhibition of MAPK pathway signaling was sufficient to improve rescued F508del-CFTR surface levels, whereas an ectopic increase in pathway activation downstream of SHC1, through the use of a constitutively active H-RAS protein, abrogated the stabilizing effect of YES1 inhibition on rescued F508del-CFTR. Taken together, our findings not only provide new mechanistic insights into the regulation of modulator-rescued F508del-CFTR membrane stability, but also open exciting new avenues to be further explored in CF research and treatment.


Cystic Fibrosis Transmembrane Conductance Regulator , Cystic Fibrosis , Humans , Cell Line , Cell Membrane/metabolism , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Membranes , Mutation , Proto-Oncogene Proteins c-yes/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Epithelial Cells/metabolism
2.
Cell Mol Biol (Noisy-le-grand) ; 69(15): 270-276, 2023 Dec 31.
Article En | MEDLINE | ID: mdl-38279415

Prostatitis is one common male disease with a high prevalence. Traditional Chinese medicine (TCM) has been used as an alternative method for the treatment. However, the molecular mechanism of Prostatitis No.1 Traditional Chinese Medicine (P1TCM) on prostatitis is still unclear. For this purpose, the rat models were constructed and treated with PITCM of control, model, low (10 g/kg/d), medium (20 g/kg/d), and high (40 g/kg/d), as well as the transfections of medium dosage+NC mimic, and medium dosage+miR-205-5p mimic, medium dosage+NC mimic+pc-NC, medium dosage+miR-205-5p mimic+pc-NC, and medium dosage+miR-205-5p mimic+pc-v-YES-1 Yamaguchi sarcoma viral oncogene homolog 1 (YES1). Real-time quantitative PCR (qPCR) and western blotting analyses were carried out to evaluate the expression of miR-205-5p and YES1, respectively. The levels of interleukin-1ß (IL-1ß) and tumor necrosis factor-alpha (TNF-α) were assessed by enzyme-linked immunosorbent assay (ELISA). The targeting role of miR-205-5p on YES1 was predicted by StarBase and verified by a dual-luciferase reporter gene assay. Results showed that the optimal treatment of P1TCM relieved the damage of prostate tissue, decreased the immunity and inflammation factors, and reduced the expression level of miR-205-5p in prostate tissue and serum. miR-205-5p mimics significantly relieved tissue damage and reduced immunity and inflammatory functions. miR-205-5p targeted YES1. YES1 was significantly upregulated in medium dosage treatment compared with Control, while downregulated compared with the Model. YES1 was also upregulated in prostatitis patients. The pc-YES1 reversed the function of the miR-205-5p mimic. In conclusion, P1TCM significantly relieved the tissue damage and reduced prostate patients' inflammatory functions through miR-205-5p/YES1, which might be essential for clinical studies.


MicroRNAs , Prostatitis , Humans , Male , Rats , Animals , Prostatitis/drug therapy , Prostatitis/genetics , Medicine, Chinese Traditional , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Necrosis Factor-alpha , Blotting, Western , Anti-Inflammatory Agents , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism
3.
Mol Cancer Ther ; 21(9): 1371-1380, 2022 09 06.
Article En | MEDLINE | ID: mdl-35732509

YES1 is a nonreceptor tyrosine kinase that belongs to the SRC family of kinases (SFK) and controls multiple cancer signaling pathways. YES1 is amplified and overexpressed in many tumor types, where it promotes cell proliferation, survival, and invasiveness. Therefore, YES1 has been proposed as an emerging target in solid tumors. In addition, studies have shown that YES1 is a prognostic biomarker and a predictor of dasatinib activity. Several SFKs-targeting drugs have been developed, and some of them have reached clinical trials. However, these drugs have encountered challenges to their utilization in the clinical practice in unselected patients due to toxicity and lack of efficacy. In the case of YES1, novel specific inhibitors have been developed and tested in preclinical models, with impressive antitumor effects. In this review, we summarize the structure and activation of YES1 and describe its role in cancer as a target and prognostic and companion biomarker. We also address the efficacy of SFKs inhibitors that are currently in clinical trials, highlighting the main hindrances for their clinical use. Current available information strongly suggests that inhibiting YES1 in tumors with high expression of this protein is a promising strategy against cancer.


Neoplasms , src-Family Kinases , Biomarkers , Cell Line, Tumor , Cell Proliferation , Dasatinib/pharmacology , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-yes/metabolism
4.
Cell Death Dis ; 13(1): 51, 2022 01 11.
Article En | MEDLINE | ID: mdl-35017464

The long intergenic non-coding RNA linc01133 is reported to be oncogenic in various malignancies. However, the role and mechanism of linc01133 in regulating gastric cancer growth is still not clear. In the present study, we found that linc01133 was significantly upregulated in gastric cancer tissues compared to non-tumorous gastric tissues. Linc01133 over-expression significantly correlated with tumor size and tumor differentiation in gastric cancer patients. The expression of linc01133 was regulated by c-Jun and c-Fos collaboratively. In both in vitro and in vivo studies, linc01133 was shown to promote gastric cancer cell growth. Linc01133 localized in the cytoplasm and functioned as an endogenous competing RNA of miR-145-5p to upregulate the expression of YES1, which was proved to be the target gene of miR-145-5p. By promoting YES1-dependent YAP1 nuclear translocation, linc01133 upregulated the expression of the key cell cycle regulators CDK4, CDK6 and cyclin D1 to promote G1-S phase transition. Thus, our study unveiled the function and mechanism of linc01133 regulating cell cycle progression in gastric cancer.


MicroRNAs , RNA, Long Noncoding/genetics , Stomach Neoplasms , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oncogenes , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/genetics , YAP-Signaling Proteins
5.
Hypertension ; 79(2): 399-412, 2022 02.
Article En | MEDLINE | ID: mdl-34865521

Incomplete spiral artery remodeling, caused by impaired extravillous trophoblast invasion, is a fundamental pathogenic process associated with malplacentation and the development of preeclampsia. Nevertheless, the mechanisms controlling this regulation of trophoblast invasion are largely unknown. We report that sphingosine-1-phosphate synthesis and expression is abundant in healthy trophoblast, whereas in pregnancies complicated by preeclampsia the placentae are associated with reduced sphingosine-1-phosphate and lower SPHK1 (sphingosine kinase 1) expression and activity. In vivo inhibition of sphingosine kinase 1 activity during placentation in pregnant mice led to decreased placental sphingosine-1-phosphate production and defective placentation, resulting in a preeclampsia phenotype. Moreover, sphingosine-1-phosphate increased HTR8/SVneo (immortalized human trophoblst cells) cell invasion in a Hippo-signaling-dependent transcriptional coactivator YAP (Yes-associated protein) dependent manner, which is activated by S1PR2 (sphingosine-1-phosphate receptor-2) and downstream RhoA (Ras homolog gene family, member A)/ROCK (Rho-associated protein kinase) induced actin polymerization. Mutation-based YAP-5SA (S61A, S109A, S127A, S164A, S381A) demonstrated that sphingosine-1-phosphate activation of YAP could be either dependent or independent of Hippo signaling. Together, these findings suggest a novel pathogenic pathway of preeclampsia via disrupted sphingosine-1-phosphate metabolism and signaling-induced, interrupted actin dynamics and YAP deactivation; this may lead to potential novel intervention targets for the prevention and management of preeclampsia.


Actins/metabolism , Lysophospholipids/metabolism , Pre-Eclampsia/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Trophoblasts/metabolism , Animals , Female , Humans , Lysophospholipids/genetics , Mice , Placenta/metabolism , Placentation/physiology , Pregnancy , Proto-Oncogene Proteins c-yes/genetics , Signal Transduction/physiology , Sphingosine/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/genetics
6.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L392-L403, 2021 08 01.
Article En | MEDLINE | ID: mdl-34105991

Endothelial cell apoptosis is an early event in the development of acute lung injury (ALI). We have previously found that the Src family tyrosine kinase (STK) Yes activates caspase-3, whereas the STK Fyn inhibits caspase-3 activation in cultured pulmonary endothelial cells. We hypothesized that deficiency in Yes or Fyn in mice would have differential effects on lipopolysaccharide (LPS)-induced ALI. Mice were treated with LPS (10 mg/kg ip) for 24 h. Histological evidence of lung injury was greater in LPS-treated wild-type mice than in vehicle-treated wild-type mice, and the LPS-induced histological evidence of lung injury was attenuated in yes-/- mice and enhanced in fyn-/- mice. In wild-type or fyn-/- mice, LPS resulted in greater lung wet-to-dry weight ratios than in controls, whereas in yes-/- mice lung, wet-to-dry weight was similar between LPS and controls. LPS-exposed fyn-/- mice had greater respiratory system resistance and lower respiratory system compliance than did LPS-exposed wild-type mice. TUNEL positive cells in the lung following LPS treatment were greater in the fyn-/- mice and lower in the yes-/- mice compared with that in the wild-type mice. Following LPS treatment lung protein levels of PECAM-1 were lower in fyn-/- mice than in controls or yes-/- mice. LPS treatment increased cleaved caspase-3 protein levels in wild-type mice, whereas LPS-induced caspase-3 activation was attenuated in yes-/- mice and enhanced in fyn-/- mice. These results indicate that LPS-induced ALI is positively mediated via Yes-related mechanisms and negatively mediated by Fyn-related mechanisms.


Acute Lung Injury , Lipopolysaccharides/toxicity , Proto-Oncogene Proteins c-fyn , Proto-Oncogene Proteins c-yes , Acute Lung Injury/chemically induced , Acute Lung Injury/enzymology , Acute Lung Injury/genetics , Acute Lung Injury/pathology , Animals , Mice , Mice, Knockout , Proto-Oncogene Proteins c-fyn/genetics , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism
7.
Oncogene ; 40(20): 3610-3623, 2021 05.
Article En | MEDLINE | ID: mdl-33941853

Erythropoietin-producing hepatocellular receptor A2 (EphA2) is a key member of the receptor tyrosine kinase (RTK) family, while YES Proto-Oncogene 1 (YES1) is a non-receptor tyrosine kinase (nRTK) and annexin A2 (ANXA2) belongs to the calcium-dependent phospholipid-binding protein family annexins. Here, we show that EphA2, YES1, and ANXA2 form a signal axis, in which YES1 activated by EphA2 phosphorylates ANXA2 at Tyr24 site, leading to ANXA2 activation and increased ANXA2 nuclear distribution in gastric cancer (GC) cells. Overexpression (OE) of YES1 increases, while knockdown (KD) of YES1 or ANXA2 decreases GC cell invasion and migration in vitro and tumor growth in mouse models. Reexpression of wildtype (WT) rather than mutant ANXA2 (Tyr24F) in ANXA2 knockdown (ANXA2-KD) GC cells restores YES1-induced cell invasion and migration, while neither WT nor mutant ANXA2 (Tyr24F) can restore cell invasion and migration in YES1-KD GC cells. In addition, the activation of EphA2-YES1-ANXA2 pathway is correlated with poor prognosis. Thus, our results establish EphA2-YES1-ANXA2 axis as a novel pathway that drives GC invasion and metastasis, targeting this pathway would be an efficient way for the treatment of GC.


Annexin A2/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Receptor, EphA2/metabolism , Stomach Neoplasms/pathology , Animals , Annexin A2/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis , Phosphorylation , Proto-Oncogene Proteins c-yes/genetics , Receptor, EphA2/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Survival Rate , Xenograft Model Antitumor Assays
8.
Science ; 372(6540)2021 04 23.
Article En | MEDLINE | ID: mdl-33888614

Skin scarring, the end result of adult wound healing, is detrimental to tissue form and function. Engrailed-1 lineage-positive fibroblasts (EPFs) are known to function in scarring, but Engrailed-1 lineage-negative fibroblasts (ENFs) remain poorly characterized. Using cell transplantation and transgenic mouse models, we identified a dermal ENF subpopulation that gives rise to postnatally derived EPFs by activating Engrailed-1 expression during adult wound healing. By studying ENF responses to substrate mechanics, we found that mechanical tension drives Engrailed-1 activation via canonical mechanotransduction signaling. Finally, we showed that blocking mechanotransduction signaling with either verteporfin, an inhibitor of Yes-associated protein (YAP), or fibroblast-specific transgenic YAP knockout prevents Engrailed-1 activation and promotes wound regeneration by ENFs, with recovery of skin appendages, ultrastructure, and mechanical strength. This finding suggests that there are two possible outcomes to postnatal wound healing: a fibrotic response (EPF-mediated) and a regenerative response (ENF-mediated).


Cicatrix/pathology , Fibroblasts/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Regeneration , Skin/injuries , Wound Healing , Animals , Cicatrix/prevention & control , Fibroblasts/transplantation , Gene Expression Regulation , Gene Knockout Techniques , Mechanotransduction, Cellular , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-yes/antagonists & inhibitors , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , Signal Transduction , Stress, Mechanical , Transcriptional Activation , Transcriptome , Verteporfin/pharmacology
9.
J Immunother Cancer ; 9(3)2021 03.
Article En | MEDLINE | ID: mdl-33658304

INTRODUCTION: The use of immune-checkpoint inhibitors has drastically improved the management of patients with non-small cell lung cancer (NSCLC), but innate and acquired resistances are hurdles needed to be solved. Immunomodulatory drugs that can reinvigorate the immune cytotoxic activity, in combination with antiprogrammed cell death 1 (PD-1) antibody, are a great promise to overcome resistance. We evaluated the impact of the SRC family kinases (SFKs) on NSCLC prognosis, and the immunomodulatory effect of the SFK inhibitor dasatinib, in combination with anti-PD-1, in clinically relevant mouse models of NSCLC. METHODS: A cohort of patients from University Clinic of Navarra (n=116) was used to study immune infiltrates by multiplex immunofluorescence (mIF) and YES1 protein expression in tumor samples. Publicly available resources (TCGA, Km Plotter, and CIBERSORT) were used to study patient's survival based on expression of SFKs and tumor infiltrates. Syngeneic NSCLC mouse models 393P and UNSCC680AJ were used for in vivo drug testing. RESULTS: Among the SFK members, YES1 expression showed the highest association with poor prognosis. Patients with high YES1 tumor levels also showed high infiltration of CD4+/FOXP3+ cells (regulatory T cells (Tregs)), suggesting an immunosuppressive phenotype. After testing for YES1 expression in a panel of murine cell lines, 393P and UNSCC680AJ were selected for in vivo studies. In the 393P model, dasatinib+anti-PD-1 treatment resulted in synergistic activity, with 87% tumor regressions and development of immunological memory that impeded tumor growth when mice were rechallenged. In vivo depletion experiments further showed that CD8+ and CD4+ cells are necessary for the therapeutic effect of the combination. The antitumor activity was accompanied by a very significant decrease in the number of Tregs, which was validated by mIF in tumor sections. In the UNSCC680AJ model, the antitumor effects of dasatinib+anti-PD-1 were milder but similar to the 393P model. In in vitro assays, we demonstrated that dasatinib blocks proliferation and transforming growth factor beta-driven conversion of effector CD4+ cells into Tregs through targeting of phospholymphocyte-specific protein tyrosine kinase and downstream effectors pSTAT5 and pSMAD3. CONCLUSIONS: YES1 protein expression is associated with increased numbers of Tregs in patients with NSCLC. Dasatinib synergizes with anti-PD-1 to impair tumor growth in NSCLC experimental models. This study provides the preclinical rationale for the combined use of dasatinib and PD-1/programmed death-ligand 1 blockade to improve outcomes of patients with NSCLC.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Proliferation/drug effects , Dasatinib/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-yes/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/enzymology , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, 129 Strain , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment
10.
Cell Chem Biol ; 28(8): 1235-1241.e5, 2021 08 19.
Article En | MEDLINE | ID: mdl-33730553

The multi-domain scaffolding protein Scribble (Scrib) regulates cell polarity and growth signaling at cell-cell junctions. In epithelial cancers, Scrib mislocalization and overexpression paradoxically transform Scrib from a basolateral tumor suppressor to a cytosolic driver of tumorigenicity. To address the function of Scrib (mis)localization, a Scrib-HaloTag fusion was genome engineered in polarized epithelial cells. Expression of the epithelial to mesenchymal transcription factor Snail displaced Scrib-HaloTag from cell junctions, mirroring the mislocalization observed in cancers. Interestingly, Snail expression promotes Yes-associated protein-1 (YAP1) nuclear localization independent of hippo pathway-regulated YAP-S127 phosphorylation. Furthermore, Scrib HaloPROTAC degradation attenuates YAP1-Y357 phosphorylation. Halo-ligand affinity purification mass spectrometry analysis identified the Src family kinase YES1 as a mislocalized Scrib interaction partner, preferentially recruiting the kinase active and open global conformation (αC helix in). Altogether, mislocalized Scrib enhances YAP1 phosphorylation by scaffolding active YES1.


Proto-Oncogene Proteins c-yes/metabolism , YAP-Signaling Proteins/metabolism , Animals , Cells, Cultured , Dogs , Female , Humans , Male , Phosphorylation , Proto-Oncogene Proteins c-yes/genetics , YAP-Signaling Proteins/genetics
11.
J Dermatol Sci ; 101(3): 202-209, 2021 Mar.
Article En | MEDLINE | ID: mdl-33509657

BACKGROUND: Lesions of atopic dermatitis have fewer Th17 cells than those of psoriasis, resulting in frequent skin infections. Expression of CCL20, a chemokine that is important for recruiting Th17 cells, is suppressed in the lesions of atopic dermatitis. We previously reported that IL-4 induces the expression of cytokine-inducible SH2-containing protein 1 (CIS1), a member of the CIS/SOCS family, in epidermal keratinocytes. OBJECTIVE: To investigate whether CIS1 influences CCL20 production in epidermal keratinocytes. METHODS: Expression of CIS1 was examined in atopic dermatitis skin and in cultured keratinocytes. The effects of overexpression of CIS1 on CCL20 production by IL-17A, and on signaling pathways inhibited by CIS1, were assessed in vitro. RESULTS: Expression of CIS1 was enhanced in the basal layer of the lesional epidermis of skin with atopic dermatitis. When CIS1 was expressed in keratinocytes using adenoviral vectors, IL-17A-induced CCL20 expression, but not HBD2 or S100A7 expression, was significantly suppressed. TNF-α/IL-1-induced CCL20 production was not altered by CIS1. Overexpression of CIS1 attenuated IL-17A-induced ERK phosphorylation. ERK phosphorylation was mediated by the Act1 and Src family kinase pathways. CIS1 overexpression suppressed Src phosphorylation. Among the Src family kinases, the Yes kinase may have an important role because knockdown of Yes in epidermal keratinocytes resulted in suppression of ERK phosphorylation and CCL20 mRNA expression by IL-17A. CONCLUSION: CIS1 induced by Th2 cytokines has the ability to change the response of epidermal keratinocytes to IL-17A by suppression of Src family kinases.


Chemokine CCL20/metabolism , Dermatitis, Atopic/immunology , Epidermis/pathology , Suppressor of Cytokine Signaling 1 Protein/metabolism , Cells, Cultured , Dermatitis, Atopic/pathology , Epidermis/immunology , Gene Expression Regulation/immunology , Gene Knockdown Techniques , Humans , Interleukin-17/metabolism , Keratinocytes/immunology , Keratinocytes/pathology , Phosphorylation/genetics , Phosphorylation/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-yes/genetics , Proto-Oncogene Proteins c-yes/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
12.
Am J Pathol ; 191(3): 527-544, 2021 03.
Article En | MEDLINE | ID: mdl-33307038

The growth and spread of malignant tumors, such as ovarian carcinomas, are governed in part by complex interconnected signaling cascades occurring between stromal and tumor cells. These reciprocal cross-talk signaling networks operating within the local tissue microenvironment may enhance malignant tumor progression. Understanding how novel bioactive molecules generated within the tumor microenvironment regulate signaling pathways in distinct cellular compartments is critical for the development of more effective treatment paradigms. Herein, we provide evidence that blocking cellular interactions with an RGDKGE-containing collagen peptide that selectively binds integrin ß3 on ovarian tumor cells enhances the phosphorylation of the hippo effector kinase large tumor suppressor kinase-1 and reduces nuclear accumulation of yes-associated protein and its target gene c-Myc. Selectively targeting this RGDKGE-containing collagen fragment inhibited ovarian tumor growth and the development of ascites fluid in vivo. These findings suggest that this bioactive collagen fragment may represent a previously unknown regulator of the hippo effector kinase large tumor suppressor kinase-1 and regulate ovarian tumor growth by a yes-associated protein-dependent mechanism. Taken together, these data not only provide new mechanistic insight into how a unique collagen fragment may regulate ovarian cancer, but in addition may help provide a useful new alternative strategy to control ovarian tumor progression based on selectively disrupting a previously unappreciated signaling cascade.


Biomarkers, Tumor/metabolism , Collagen/metabolism , Ovarian Neoplasms/pathology , Peptide Fragments/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-yes/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred C57BL , Mice, Nude , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-yes/genetics , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
13.
Biomed Pharmacother ; 133: 110956, 2021 Jan.
Article En | MEDLINE | ID: mdl-33189066

Breast cancer is the most commonly diagnosed cancer among women. Although routine and targeted therapies have improved the survival rate, there are still considerable challenges in the treatment of breast cancer. Metastasis is the leading cause of death in patients diagnosed with breast cancer. Yes-associated protein (YAP) and/or PDZ binding motif (TAZ) are usually abnormally activated in breast cancer leading to a variety of effects on tumour promotion, such as epithelial-mesenchymal transition, cancer stem cell production and drug-resistance. The abnormal activation of YAP/TAZ can affect metastasis-related processes and promote cancer progression and metastasis by interacting with some metastasis-related factors and pathways. In this article, we summarise the evidence that YAP/TAZ regulates breast cancer metastasis, its post-translational modification mechanisms, and the latest advances in the treatment of YAP/TAZ-related breast cancer metastasis, besides providing a new strategy of YAP/TAZ-based treatment of human breast cancer.


Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-yes/metabolism , Trans-Activators/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , Molecular Targeted Therapy , Neoplasm Metastasis , Protein Domains , Proto-Oncogene Proteins c-yes/chemistry , Signal Transduction , Structure-Activity Relationship , Trans-Activators/chemistry , Transcriptional Coactivator with PDZ-Binding Motif Proteins
14.
Anticancer Drugs ; 32(3): 257-268, 2021 03 01.
Article En | MEDLINE | ID: mdl-33186139

Non-small cell lung cancer (NSCLC) is a major type of lung cancer, leading to a high fatality rate. The role of circular RNAs (circRNAs) in cancer has been increasingly emphasized and studied. However, the function of circ-ZNF124 in NSCLC is largely unclear, and associated regulatory mechanism is not studied. Here, we examined the expression pattern of circ-ZNF124 using quantitative real-time PCR. For functional analysis, cell proliferation, cell apoptosis/cycle and cell invasion were investigated using MTT [3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide] assay, flow cytometry assay and transwell assay, respectively. As results, we found that the expression of circ-ZNF124 was elevated in NSCLC tissues and cells. Functionally, circ-ZNF124 downregulation inhibited NSCLC cell proliferation and invasion but induced apoptosis and cycle arrest in vitro, and blocked tumor growth in vivo by animal experiments. Mechanistically, we identified that miR-498 was a target of circ-ZNF124, and miR-498 directly bound to YES proto-oncogene 1 (YES1). Besides, rescue experiments discovered that the cellular effects caused by circ-ZNF124 downregulation could be reversed by miR-498 inhibition or YES1 overexpression. Moreover, we discovered that circ-ZNF124 downregulation inactivated the expression of ß-catenin and c-Myc by mediating the miR-498/YES axis. In conclusion, these findings supported that circ-ZNF124 regulated the expression of YES1 by acting as a sponge of miR-498, thus restraining NSCLC development by inactivating the Wnt/ß-catenin signaling pathway, which provided a novel strategy to treat NSCLC.


Carcinoma, Non-Small-Cell Lung/pathology , DNA-Binding Proteins/metabolism , Lung Neoplasms/pathology , MicroRNAs/metabolism , RNA, Circular/metabolism , Wnt Signaling Pathway/physiology , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Proto-Oncogene Mas , Proto-Oncogene Proteins c-yes/metabolism , beta Catenin/metabolism
15.
Cells ; 9(11)2020 11 04.
Article En | MEDLINE | ID: mdl-33158300

The multifunctional HSP70 co-chaperone BAG3 (BCL-2-associated athanogene 3) represents a key player in the quality control of the cellular proteostasis network. In response to stress, BAG3 specifically targets aggregation-prone proteins to the perinuclear aggresome and promotes their degradation via BAG3-mediated selective macroautophagy. To adapt cellular homeostasis to stress, BAG3 modulates and functions in various cellular processes and signaling pathways. Noteworthy, dysfunction and deregulation of BAG3 and its pathway are pathophysiologically linked to myopathies, cancer, and neurodegenerative disorders. Here, we report a BAG3 proteomic signature under proteostasis stress. To elucidate the dynamic and multifunctional action of BAG3 in response to stress, we established BAG3 interactomes under basal and proteostasis stress conditions by employing affinity purification combined with quantitative mass spectrometry. In addition to the identification of novel potential BAG3 interactors, we defined proteins whose interaction with BAG3 was altered upon stress. By functional annotation and protein-protein interaction enrichment analysis of the identified potential BAG3 interactors, we confirmed the multifunctionality of BAG3 and highlighted its crucial role in diverse cellular signaling pathways and processes, ensuring cellular proteostasis and cell viability. These include protein folding and degradation, gene expression, cytoskeleton dynamics (including cell cycle and transport), as well as granulostasis, in particular.


Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Proteomics , Proteostasis , Stress, Physiological , Gene Ontology , HEK293 Cells , Humans , Molecular Sequence Annotation , Multivariate Analysis , Proteasome Inhibitors/pharmacology , Protein Binding/drug effects , Protein Interaction Maps/drug effects , Proteostasis/drug effects , Proto-Oncogene Proteins c-yes/metabolism , Stress, Physiological/drug effects
16.
Int J Mol Sci ; 21(20)2020 Oct 14.
Article En | MEDLINE | ID: mdl-33066548

Cholangiocarcinoma (CCA) is associated with high mortality rates because of its resistance to conventional gemcitabine-based chemotherapy. Hydroxy-methyl-glutaryl-coenzyme A reductase inhibitors (statins) reportedly exert anti-cancer effects in CCA and lower the risk of CCA; however, the underlying mechanism of these effects remains unclear. The proliferative and oncogenic activities of the transcriptional co-activator Yes-associated protein (YAP) are driven by its association with the TEA domain (TEAD) of transcription factors; thereby, upregulating genes that promote cell growth, inhibit apoptosis, and confer chemoresistance. This study investigated the effects of atorvastatin in combination with gemcitabine on the progression of human CCA associated with YAP oncogenic regulation. Both atorvastatin and gemcitabine concentration-dependently suppressed the proliferation of HuCCT-1 and KKU-M213 human CCA cells. Moreover, both agents induced cellular apoptosis by upregulating the pro-apoptotic marker BAX and downregulating the anti-apoptotic markers MCL1 and BCL2. Atorvastatin also significantly decreased the mRNA expression of the TEAD target genes CTGF, CYR61, ANKRD1, and MFAP5 in both CCA cell lines. A xenograft tumor growth assay indicated that atorvastatin and gemcitabine potently repressed human CCA cell-derived subcutaneous tumor growth by inhibiting YAP nuclear translocation and TEAD transcriptional activation. Notably, the anti-cancer effects of the individual agents were significantly enhanced in combination. These results indicate that gemcitabine plus atorvastatin could serve as a potential novel treatment option for CCA.


Anticholesteremic Agents/pharmacology , Antimetabolites, Antineoplastic/pharmacology , Atorvastatin/pharmacology , Bile Duct Neoplasms/drug therapy , Cholangiocarcinoma/drug therapy , Deoxycytidine/analogs & derivatives , Proto-Oncogene Proteins c-yes/metabolism , Animals , Anticholesteremic Agents/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis/drug effects , Atorvastatin/administration & dosage , Bile Duct Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cholangiocarcinoma/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Contractile Proteins/genetics , Contractile Proteins/metabolism , Cysteine-Rich Protein 61/genetics , Cysteine-Rich Protein 61/metabolism , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Combinations , Drug Interactions , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Muscle Proteins/genetics , Muscle Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gemcitabine
17.
Biomolecules ; 10(8)2020 08 05.
Article En | MEDLINE | ID: mdl-32764330

Cells are continuously sensing their microenvironment and subsequently respond to different physicochemical cues by the activation or inhibition of different signaling pathways. To study a very complex cellular response, it is necessary to diminish background environmental influences and highlight the particular event. However, surface-driven nonspecific interactions of the abundant biomolecules from the environment influence the targeted cell response significantly. Yes-associated protein (YAP) translocation may serve as a marker of human hepatocellular carcinoma (Huh7) cell responses to the extracellular matrix and surface-mediated stresses. Here, we propose a platform of tunable functionable antifouling poly(carboxybetain) (pCB)-based brushes to achieve a molecularly clean background for studying arginine, glycine, and aspartic acid (RGD)-induced YAP-connected mechanotransduction. Using two different sets of RGD-functionalized zwitterionic antifouling coatings with varying compositions of the antifouling layer, a clear correlation of YAP distribution with RGD functionalization concentrations was observed. On the other hand, commonly used surface passivation by the oligo(ethylene glycol)-based self-assembled monolayer (SAM) shows no potential to induce dependency of the YAP distribution on RGD concentrations. The results indicate that the antifouling background is a crucial component of surface-based cellular response studies, and pCB-based zwitterionic antifouling brush architectures may serve as a potential next-generation easily functionable surface platform for the monitoring and quantification of cellular processes.


Biofouling/prevention & control , Coated Materials, Biocompatible/chemistry , Mechanotransduction, Cellular , Acrylamides/chemistry , Cell Line, Tumor , Extracellular Matrix/metabolism , Humans , Oligopeptides/chemistry , Proto-Oncogene Proteins c-yes/metabolism , Stress, Mechanical
18.
Thorac Cancer ; 11(9): 2736-2739, 2020 09.
Article En | MEDLINE | ID: mdl-32744377

Epidermal growth factor receptor (EGFR) mutation-positive non-small cell lung cancer (NSCLC) patients benefit from EGFR tyrosine kinase inhibitors (TKIs), while some patients demonstrate a resistance to EGFR-TKIs. In the case reported here, the NSCLC patient harboring an EGFR-sensitive mutation and YES1 amplification was treated with afatinib as first-line therapy, but was found to have progressive disease four weeks later. During subsequent chemotherapy, this patient's disease progressed rapidly. Mechanisms of primary resistance to EGFR-TKIs remain unclear. This case suggested that YES1 amplification might be associated with primary resistance to EGFR-TKIs and YES1 amplification might be a negative predictor of EGFR-TKI treatment in NSCLC patients harboring EGFR sensitive mutations.


Afatinib/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-yes/metabolism , Afatinib/pharmacology , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Humans , Lung Neoplasms/pathology , Male , Mutation
19.
J Hepatol ; 73(5): 1155-1169, 2020 11.
Article En | MEDLINE | ID: mdl-32461121

BACKGROUND & AIMS: Growing evidence shows that some non-coding RNAs (ncRNAs) contain small open reading frames (smORFs) that are translated into short peptides. Herein, we aimed to determine where and how these short peptides might promote hepatocellular carcinoma (HCC) development. METHODS: We performed an RNA-immunoprecipitation followed by high-throughput sequencing (RIP-seq) assay with an antibody against ribosomal protein S6 (RPS6) on 4 cancer cell lines. Focusing on 1 long non-coding RNA (lncRNA), LINC00998, we used qPCR and public databases to evaluate its expression level in patients with HCC. Special vectors were constructed to confirm its coding potential. We also explored the function and mechanism of LINC00998-encoded peptide in tumor growth and metastasis. RESULTS: We discovered that many lncRNAs bind to RPS6 in cancer cells. One of these lncRNAs, LINC00998, encoded a small endogenous peptide, termed SMIM30. SMIM30, rather than the RNA itself, promoted HCC tumorigenesis by modulating cell proliferation and migration, and its level was correlated with poor survival in patients with HCC. Furthermore, SMIM30 was transcribed by c-Myc and then drove the membrane anchoring of the non-receptor tyrosine kinases SRC/YES1. Moreover, the downstream MAPK signaling pathway was activated by SRC/YES1. CONCLUSIONS: Our results not only unravel a new mechanism of HCC tumorigenesis promoted by ncRNA-encoded peptides, but also suggest that these peptides can serve as a new target for HCC cancer therapy and a new biomarker for HCC diagnosis and prognosis. LAY SUMMARY: Very little is known about how peptides activate signaling pathways that play a crucial role in diseases such as cancer. Specifically, we reported on a conserved peptide encoded by LINC00998, SMIM30. This peptide promoted the tumorigenesis of hepatocellular carcinoma (HCC) by modulating cell proliferation and migration. Of note, it bound the non-receptor tyrosine kinases, SRC/YES1, to drive their membrane anchoring and phosphorylation, activating the downstream MAPK signaling pathway. Our work not only unravels a new mechanism of HCC tumorigenesis promoted by peptides, but also demonstrates how the peptide works to activate a signaling pathway.


Carcinoma, Hepatocellular , Liver Neoplasms , Proto-Oncogene Proteins c-yes/metabolism , RNA, Long Noncoding/metabolism , src-Family Kinases/metabolism , Animals , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , High-Throughput Nucleotide Sequencing , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , MAP Kinase Signaling System , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/metabolism , Prognosis
20.
Technol Cancer Res Treat ; 19: 1533033820927011, 2020.
Article En | MEDLINE | ID: mdl-32462982

Triple-negative breast cancer shows worse outcome compared with other subtypes of breast cancer. The discovery of dysregulated microRNAs and their roles in the progression of triple-negative breast cancer provide novel strategies for the treatment of patients with triple-negative breast cancer. In this study, we identified the significant reduction of miR-133 in triple-negative breast cancer tissues and cell lines. Ectopic overexpression of miR-133 suppressed the proliferation, colony formation, and upregulated the apoptosis of triple-negative breast cancer cells. Mechanism study revealed that the YES Proto-Oncogene 1 was a target of miR-133. miR-133 bound the 3'-untranslated region of YES Proto-Oncogene 1 and decreased the level of YES Proto-Oncogene 1 in triple-negative breast cancer cells. Consistent with miR-133 downregulation, YES1 was significantly increased in triple-negative breast cancer, which was inversely correlated with the level of miR-133. Restoration of YES Proto-Oncogene 1 attenuated the inhibitory effects of miR-133 on the proliferation and colony formation of triple-negative breast cancer cells. Consistent with the decreased expression of YES Proto-Oncogene 1, overexpression of miR-133 suppressed the phosphorylation of YAP1 in triple-negative breast cancer cells. Our results provided novel evidence for the role of miR-133/YES1 axis in the development of triple-negative breast cancer, which indicated miR-133 might be a potential therapeutic strategy for triple-negative breast cancer.


Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-yes/metabolism , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Female , Humans , Middle Aged , Phosphorylation , Prognosis , Proto-Oncogene Mas , Proto-Oncogene Proteins c-yes/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/surgery , Tumor Cells, Cultured , YAP-Signaling Proteins
...