Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biomed Pharmacother ; 168: 115772, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37879209

ABSTRACT

The involvement of chondrocyte ferroptosis in the development of osteoarthritis (OA) has been observed, and Sarsasapogenin (Sar) has therapeutic promise in a variety of inflammatory diseases. This study investigates the potential influence of Sar on the mechanism of chondrocyte ferroptotic cell death in the progression of osteoarthritic cartilage degradation. An in vivo medial meniscus destabilization (DMM)-induced OA animal model as well as an in vitro examination of chondrocytes in an OA microenvironment induced by interleukin-1ß (IL-1ß) exposure were employed. Histology, immunofluorescence, quantitative RT-PCR, Western blot, cell viability, and Micro-CT analysis were utilized in conjunction with gene overexpression and knockdown to evaluate the chondroprotective effects of Sar in OA progression and the role of Yes-associated protein 1 (YAP1) in Sar-induced ferroptosis resistance of chondrocytes. In this study we found Sar reduced chondrocyte ferroptosis and OA progression. And Sar-induced chondrocyte ferroptosis resistance was mediated by YAP1. Furthermore, infection of siRNA specific to YAP1 in chondrocytes reduced Sar's chondroprotective and ferroptosis-suppressing effects during OA development. The findings suggest that Sar mitigates the progression of osteoarthritis by decreasing the sensitivity of chondrocytes to ferroptosis through the promotion of YAP1, indicating that Sar has the potential to serve as a therapeutic approach for diseases associated with ferroptosis.


Subject(s)
Chondrocytes , Ferroptosis , Osteoarthritis , YAP-Signaling Proteins , Animals , Cartilage/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Ferroptosis/drug effects , Interleukin-1beta/metabolism , Osteoarthritis/drug therapy , Transcription Factors/metabolism , YAP-Signaling Proteins/antagonists & inhibitors
2.
Eur J Med Chem ; 244: 114847, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36265280

ABSTRACT

The Hippo pathway is an evolutionarily conserved signaling pathway that plays critical roles in the tumorigenesis and progression of breast cancer, oral cancer, rectal cancer, colloid cancer, and so on. YAP/TAZ-TEAD complex is a key knot in the Hippo pathway regulating cell proliferation and stem cell functions. Activation or overexpression of this complex has been proved to lead to cell transformation, proliferation and eventually cancerization. In this review, the association between the alterations of hippo pathway and tumorigenesis of various cancer had been elucidated. The structural basis of YAP/TAZ-TEAD complex is analyzed, and the targeting inhibitors are summarized within the medicinal chemistry classification. Moreover, we have also discussed the clinical status and current challenges of these drug candidates, and provide guidance for the future development of inhibitors targeting this pathway, especially YAP/TAZ-TEAD complex.


Subject(s)
Antineoplastic Agents , Carcinogenesis , Hippo Signaling Pathway , Neoplasms , TEA Domain Transcription Factors , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Humans , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Hippo Signaling Pathway/drug effects , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Transcriptional Coactivator with PDZ-Binding Motif Proteins/antagonists & inhibitors , Transcriptional Coactivator with PDZ-Binding Motif Proteins/chemistry , TEA Domain Transcription Factors/antagonists & inhibitors , TEA Domain Transcription Factors/chemistry , Protein Conformation , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/chemistry
3.
Expert Opin Ther Pat ; 32(8): 899-912, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35768160

ABSTRACT

INTRODUCTION: The Hippo pathway represents a new opportunity for the treatment of cancer. Overexpression of Yes-associated protein (YAP) or transcriptional coactivator with PDZ-binding motif (TAZ) or TEAD has been demonstrated in cancers and YAP mediates resistance to cancer drugs. Since 2018, the potential of this pathway has been illustrated by numerous articles and patents and the first drugs entering in clinical trial phase 1. AREAS COVERED: This review is limited to published patent applications that have disclosed direct small-molecule inhibitors of the YAP/TAZ-TEAD interaction. EXPERT OPINION: The YAP/TAZ-TEAD transcriptional complex is a promising target for the treatment of cancer. Approximately 30 international patents (used database: Sci-finder, query: TEAD; documents: patents; period: from 2017-January 2022) that disclose TEAD transcriptional inhibitors have been filled since 2018. The mechanism of action is not always described in the patents, we can divide the drugs into three different categories: (i) external TEAD ligands; (ii) non-covalent TEAD ligands of the palmitate pocket; (iii) covalent TEAD ligands, which bind into the palmitate pocket. The first molecules in clinical trial phase 1 are non-covalent TEAD ligands. The selective TEAD ligand have also been patented, published and selectivity could be of great interest for personalized medicine.


Subject(s)
Neoplasms , Patents as Topic , TEA Domain Transcription Factors , YAP-Signaling Proteins , Humans , Ligands , Neoplasms/drug therapy , Palmitates , TEA Domain Transcription Factors/antagonists & inhibitors , YAP-Signaling Proteins/antagonists & inhibitors
4.
Nature ; 607(7920): 790-798, 2022 07.
Article in English | MEDLINE | ID: mdl-35768505

ABSTRACT

Ageing is intimately connected to the induction of cell senescence1,2, but why this is so remains poorly understood. A key challenge is the identification of pathways that normally suppress senescence, are lost during ageing and are functionally relevant to oppose ageing3. Here we connected the structural and functional decline of ageing tissues to attenuated function of the master effectors of cellular mechanosignalling YAP and TAZ. YAP/TAZ activity declines during physiological ageing in stromal cells, and mimicking such decline through genetic inactivation of YAP/TAZ in these cells leads to accelerated ageing. Conversely, sustaining YAP function rejuvenates old cells and opposes the emergence of ageing-related traits associated with either physiological ageing or accelerated ageing triggered by a mechano-defective extracellular matrix. Ageing traits induced by inactivation of YAP/TAZ are preceded by induction of tissue senescence. This occurs because YAP/TAZ mechanotransduction suppresses cGAS-STING signalling, to the extent that inhibition of STING prevents tissue senescence and premature ageing-related tissue degeneration after YAP/TAZ inactivation. Mechanistically, YAP/TAZ-mediated control of cGAS-STING signalling relies on the unexpected role of YAP/TAZ in preserving nuclear envelope integrity, at least in part through direct transcriptional regulation of lamin B1 and ACTR2, the latter of which is involved in building the peri-nuclear actin cap. The findings demonstrate that declining YAP/TAZ mechanotransduction drives ageing by unleashing cGAS-STING signalling, a pillar of innate immunity. Thus, sustaining YAP/TAZ mechanosignalling or inhibiting STING may represent promising approaches for limiting senescence-associated inflammation and improving healthy ageing.


Subject(s)
Aging , Membrane Proteins , Nucleotidyltransferases , Stromal Cells , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Actin-Related Protein 2/metabolism , Aging/metabolism , Cellular Senescence , Extracellular Matrix , Healthy Aging , Immunity, Innate , Lamin Type B/metabolism , Mechanotransduction, Cellular/genetics , Membrane Proteins/metabolism , Nuclear Envelope/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction , Stromal Cells/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/antagonists & inhibitors , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/metabolism
5.
Mol Pharmacol ; 101(1): 1-12, 2022 01.
Article in English | MEDLINE | ID: mdl-34732527

ABSTRACT

Most B-Raf proto-oncogene (BRAF)-mutant melanoma tumors respond initially to BRAF inhibitor (BRAFi)/mitogen-activated protein kinase kinase 1 inhibitor (MEKi) therapy, although few patients have durable long-term responses to these agents. The goal of this study was to use an unbiased computational approach to identify inhibitors that reverse an experimentally derived BRAFi resistance gene expression signature. Using this approach, we found that ibrutinib effectively reverses this signature, and we demonstrate experimentally that ibrutinib resensitizes a subset of BRAFi-resistant melanoma cells to vemurafenib. Ibrutinib is used clinically as an inhibitor of the Src family kinase Bruton tyrosine kinase (BTK); however, neither BTK deletion nor treatment with acalabrutinib, another BTK inhibitor with reduced off-target activity, resensitized cells to vemurafenib. These data suggest that ibrutinib acts through a BTK-independent mechanism in vemurafenib resensitization. To better understand this mechanism, we analyzed the transcriptional profile of ibrutinib-treated BRAFi-resistant melanoma cells and found that the transcriptional profile of ibrutinib was highly similar to that of multiple Src proto-oncogene kinase inhibitors. Since ibrutinib, but not acalabrutinib, has appreciable off-target activity against multiple Src family kinases, it suggests that ibrutinib may be acting through this mechanism. Furthermore, genes that are differentially expressed in ibrutinib-treated cells are enriched in Yes1-associated transcriptional regulator (YAP1) target genes, and we showed that ibrutinib, but not acalabrutinib, reduces YAP1 activity in BRAFi-resistant melanoma cells. Taken together, these data suggest that ibrutinib, or other Src family kinase inhibitors, may be useful for treating some BRAFi/MEKi-refractory melanoma tumors. SIGNIFICANCE STATEMENT: MAPK-targeted therapies provide dramatic initial responses, but resistance develops rapidly; a subset of these tumors may be rendered sensitive again by treatment with an approved Src family kinase inhibitor-ibrutinub-potentially providing improved clinical outcomes.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Melanoma/metabolism , Piperidines/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , YAP-Signaling Proteins/metabolism , Adenine/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/physiology , HEK293 Cells , Humans , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Vemurafenib/pharmacology , YAP-Signaling Proteins/antagonists & inhibitors
6.
J Hepatol ; 76(2): 394-406, 2022 02.
Article in English | MEDLINE | ID: mdl-34648896

ABSTRACT

BACKGROUND & AIMS: Currently there is no effective treatment for liver fibrosis, which is one of the main histological determinants of non-alcoholic steatohepatitis (NASH). While Hippo/YAP (Yes-associated protein) signaling is essential for liver regeneration, its aberrant activation frequently leads to fibrosis and tumorigenesis. Unravelling "context-specific" contributions of YAP in liver repair might help selectively bypass fibrosis and preserve the pro-regenerative YAP function in hepatic diseases. METHODS: We used murine liver fibrosis and minipig NASH models, and liver biopsies from patients with cirrhosis. Single-cell RNA-sequencing (scRNA-Seq) was performed, and a G-protein-coupled receptor (GPCR) ligand screening system was used to identify cell-selective YAP inhibitors. RESULTS: YAP levels in macrophages are increased in the livers of humans and mice with liver fibrosis. The increase in type I interferon and attenuation of hepatic fibrosis observed in mice specifically lacking Yap1 in myeloid cells provided further evidence for the fibrogenic role of macrophage YAP. ScRNA-Seq further showed that defective YAP pathway signaling in macrophages diminished a fibrogenic vascular endothelial cell subset that exhibited profibrotic molecular signatures such as angiocrine CTGF and VCAM1 expression. To specifically target fibrogenic YAP in macrophages, we utilized a GPCR ligand screening system and identified a dopamine receptor D2 (DRD2) antagonist that selectively blocked YAP in macrophages but not hepatocytes. Genetic and pharmacological targeting of macrophage DRD2 attenuated liver fibrosis. In a large animal (minipig) NASH model recapitulating human pathology, the DRD2 antagonist blocked fibrosis and restored hepatic architecture. CONCLUSIONS: DRD2 antagonism selectively targets YAP-dependent fibrogenic crosstalk between macrophages and CTGF+VCAM1+ vascular niche, promoting liver regeneration over fibrosis in both rodent and large animal models. LAY SUMMARY: Fibrosis in the liver is one of the main histological determinants of non-alcoholic steatohepatitis (NASH), a disease paralleling a worldwide surge in metabolic syndromes. Our study demonstrates that a macrophage-specific deficiency in Yes-associated protein (YAP) attenuates liver fibrosis. Dopamine receptor D2 (DRD2) antagonism selectively blocks YAP in macrophages and thwarts liver fibrosis in both rodent and large animal models, and thus holds potential for the treatment of NASH.


Subject(s)
Liver Cirrhosis/genetics , Non-alcoholic Fatty Liver Disease/genetics , Receptors, Dopamine D2/metabolism , Animals , Disease Models, Animal , Liver/drug effects , Liver/pathology , Liver Cirrhosis/drug therapy , Macrophages/drug effects , Macrophages/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Swine , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/therapeutic use
7.
Cells ; 10(11)2021 10 24.
Article in English | MEDLINE | ID: mdl-34831091

ABSTRACT

Around 70% of breast cancers express the estrogen receptor alpha (ERα). This receptor is of central importance for breast cancer development and estrogen-dependent tumor growth. However, the molecular mechanisms that are responsible for the control of ERα expression and function in the context of breast carcinogenesis are complex and not fully understood. In previous work, we have demonstrated that the tumor suppressor RASSF1A suppresses estrogen-dependent growth of breast cancer cells through a complex network that keeps ERα expression and function under control. We observed that RASSF1A mediates the suppression of ERα expression through modulation of the Hippo effector Yes-associated protein 1 (YAP1) activity. Here we report that RASSF1A-mediated alteration of YAP1 depends on the Hippo-kinases LATS1 and LATS2. Based on these results, we conclude that inactivation of RASSF1A causes changes in the function of the Hippo signaling pathway and altered activation of YAP1, and as a consequence, increased expression and function of ERα. Thus, the inactivation of RASSF1A might constitute a fundamental event that supports the initiation of ERα-dependent breast cancer. Furthermore, our results support the notion that the Hippo pathway is important for the suppression of luminal breast cancers, and that the tumor-suppressor function of RASSF1A depends on LATS1 and LATS2.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor alpha/metabolism , Hippo Signaling Pathway , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cellular Senescence/drug effects , Dasatinib/pharmacology , Female , Forkhead Box Protein M1/metabolism , HEK293 Cells , Humans , Models, Biological , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/metabolism
8.
Nat Cell Biol ; 23(11): 1187-1198, 2021 11.
Article in English | MEDLINE | ID: mdl-34737445

ABSTRACT

How cancer cells adapt to evade the therapeutic effects of drugs targeting oncogenic drivers is poorly understood. Here we report an epigenetic mechanism leading to the adaptive resistance of triple-negative breast cancer (TNBC) to fibroblast growth factor receptor (FGFR) inhibitors. Prolonged FGFR inhibition suppresses the function of BRG1-dependent chromatin remodelling, leading to an epigenetic state that derepresses YAP-associated enhancers. These chromatin changes induce the expression of several amino acid transporters, resulting in increased intracellular levels of specific amino acids that reactivate mTORC1. Consistent with this mechanism, addition of mTORC1 or YAP inhibitors to FGFR blockade synergistically attenuated the growth of TNBC patient-derived xenograft models. Collectively, these findings reveal a feedback loop involving an epigenetic state transition and metabolic reprogramming that leads to adaptive therapeutic resistance and provides potential therapeutic strategies to overcome this mechanism of resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/metabolism , Drug Resistance, Neoplasm , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/drug therapy , YAP-Signaling Proteins/metabolism , Amino Acids/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/genetics , DNA Helicases/metabolism , Drug Resistance, Neoplasm/genetics , Drug Synergism , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Molecular Targeted Therapy , Multiprotein Complexes , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction , Transcription Factors/genetics , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays , YAP-Signaling Proteins/antagonists & inhibitors , YAP-Signaling Proteins/genetics
9.
Oncogene ; 40(41): 6049-6056, 2021 10.
Article in English | MEDLINE | ID: mdl-34471235

ABSTRACT

Yes-associated protein 1 (YAP1), a key player in the Hippo pathway, has been shown to play a critical role in tumor progression. However, the role of YAP1 in prostate cancer cell invasion, migration, and metastasis is not well defined. Through functional, transcriptomic, epigenomic, and proteomic analyses, we showed that prolyl hydroxylation of YAP1 plays a critical role in the suppression of cell migration, invasion, and metastasis in prostate cancer. Knockdown (KD) or knockout (KO) of YAP1 led to an increase in cell migration, invasion, and metastasis in prostate cancer cells. Microarray analysis showed that the EMT pathway was activated in Yap1-KD cells. ChIP-seq analysis showed that YAP1 target genes are enriched in pathways regulating cell migration. Mass spectrometry analysis identified P4H prolyl hydroxylase in the YAP1 complex and YAP1 was hydroxylated at multiple proline residues. Proline-to-alanine mutations of YAP1 isoform 3 identified proline 174 as a critical residue, and its hydroxylation suppressed cell migration, invasion, and metastasis. KO of P4ha2 led to an increase in cell migration and invasion, which was reversed upon Yap1 KD. Our study identified a novel regulatory mechanism of YAP1 by which P4HA2-dependent prolyl hydroxylation of YAP1 determines its transcriptional activities and its function in prostate cancer metastasis.


Subject(s)
Prolyl Hydroxylases/metabolism , Prostatic Neoplasms/metabolism , YAP-Signaling Proteins/metabolism , Animals , Cell Movement/physiology , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Prostatic Neoplasms/pathology , YAP-Signaling Proteins/antagonists & inhibitors
10.
Cell Death Dis ; 12(8): 754, 2021 07 30.
Article in English | MEDLINE | ID: mdl-34330891

ABSTRACT

Acute kidney injury (AKI) is associated with significant morbidity and its chronic inflammation contributes to subsequent chronic kidney disease (CKD) development. Yes-associated protein (YAP), the major transcriptional coactivator of the Hippo pathway, has been shown associated with chronic inflammation, but its role and mechanism in AKI-CKD transition remain unclear. Here we aimed to investigate the role of YAP in AKI-induced chronic inflammation. Renal ischemia/reperfusion (I/R) was used to induce a mouse model of AKI-CKD transition. We used verteporfin (VP), a pharmacological inhibitor of YAP, to treat post-IRI mice for a period, and evaluated the influence of YAP inhibition on long-term outcomes of AKI. In our results, severe IRI led to maladaptive tubular repair, macrophages infiltration, and progressive fibrosis. Following AKI, the Hippo pathway was found significantly altered with YAP persistent activation. Besides, tubular YAP activation was associated with the maladaptive repair, also correlated with interstitial macrophage infiltration. Monocyte chemoattractant protein 1 (MCP-1) was found notably upregulated with YAP activation. Of note, pharmacological inhibition of YAP in vivo attenuated renal inflammation, including macrophage infiltration and MCP-1 overexpression. Consistently, in vitro oxygen-glucose deprivation and reoxygenation (OGD/R) induced YAP activation and MCP-1 overproduction whereas these could be inhibited by VP. In addition, we modulated YAP activity by RNA interference, which further confirmed YAP activation enhances MCP-1 expression. Together, we concluded tubular YAP activation with maladaptive repair exacerbates renal inflammation probably via promoting MCP-1 production, which contributes to AKI-CKD transition.


Subject(s)
Acute Kidney Injury/metabolism , Hippo Signaling Pathway , Ischemia/metabolism , Monocyte Chemoattractant Proteins/metabolism , YAP-Signaling Proteins/metabolism , Acute Kidney Injury/blood , Acute Kidney Injury/complications , Acute Kidney Injury/pathology , Animals , Blood Urea Nitrogen , Cell Line , Creatinine/blood , Fibrosis , Glucose/deficiency , Humans , Inflammation/pathology , Ischemia/blood , Ischemia/complications , Ischemia/pathology , Kidney Tubules/drug effects , Kidney Tubules/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice, Inbred C57BL , Models, Biological , Oxygen , Protein Binding/drug effects , TEA Domain Transcription Factors/metabolism , Up-Regulation/drug effects , Verteporfin/pharmacology , YAP-Signaling Proteins/antagonists & inhibitors
11.
Lipids Health Dis ; 20(1): 74, 2021 Jul 25.
Article in English | MEDLINE | ID: mdl-34304741

ABSTRACT

BACKGROUND: This study aimed to explore the effect of inhibiting the Hippo/Yes-associated protein (YAP) signaling pathway on the outcomes of transcatheter arterial chemoembolization (TACE) in treating transplanted hepatocellular carcinoma (HCC). METHODS: A transplanted HCC rat model was established. Then, rats were randomly divided into four groups: Sham, TACE, verteporfin (inhibitor of Hippo/YAP), and TACE+verteporfin. Lent-OE-YAP was transfected into rats to overexpress YAP in vivo. After treatments, morphological changes, tumor weight, and the overall survival of rats in different groups were analyzed. Real-time PCR, immunohistochemistry staining, and Western blotting were used to determine the expression of factors related to the Hippo/YAP signaling pathway. RESULTS: Tumor weight and tissue lesions in the TACE and verteporfin groups were significantly reduced compared with the Sham group. Verteporfin significantly decreased tumor weight after TACE treatment. In addition, verteporfin significantly improved the overall survival of rats with transplanted HCC after TACE treatment. Compared with the Sham group, both TACE and verteporfin groups exhibited significantly decreased expression of macrophage-stimulating (MST)1, MST2, long-acting thyroid stimulator 1, transcriptional co-activator with PDZ-binding motif (TAZ), Yes-associated protein (YAP), TEA domain transcription factor (TEAD)1, TEAD2, TEAD3, and TEAD4. TACE plus verteporfin significantly enhanced the downregulation of effectors in the Hippo/YAP signaling pathway and decreased tumor size, while the overexpression of YAP exerted opposite effects. CONCLUSION: The inhibition of the Hippo/YAP signaling pathway via verteporfin significantly improved the outcomes of TACE in treating transplanted HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic/methods , Hippo Signaling Pathway/drug effects , Liver Neoplasms/therapy , Verteporfin/therapeutic use , Animals , Blotting, Western , Carcinoma 256, Walker , Carcinoma, Hepatocellular/mortality , Combined Modality Therapy , Liver Neoplasms/mortality , Male , Neoplasm Transplantation , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Serine-Threonine Kinase 3/antagonists & inhibitors , YAP-Signaling Proteins/antagonists & inhibitors
12.
Drug Des Devel Ther ; 15: 2445-2456, 2021.
Article in English | MEDLINE | ID: mdl-34140763

ABSTRACT

Gastric cancer (GC) is one of the most common cancers globally, threatening global health. The deregulation of the Hippo signaling pathway has been discovered in GC and may be related to cancer development, proliferation, metastasis, and drug resistance. Yes-associated protein (YAP), as a downstream effector of the Hippo signaling pathway and a crucial co-transcription factor in the nucleus, is a promising and vital potential drug target for the treatment of GC. A series of drugs or compounds that inhibit YAP has been developed or confirmed. Therefore, this review will focus on summarizing the drugs and small-molecule inhibitors that have been reported to inhibit YAP and discuss the clinical prospects of YAP inhibitors in GC.


Subject(s)
Antineoplastic Agents/pharmacology , Stomach Neoplasms/drug therapy , YAP-Signaling Proteins/antagonists & inhibitors , Animals , Drug Development , Hippo Signaling Pathway/drug effects , Humans , Molecular Targeted Therapy , Stomach Neoplasms/pathology , YAP-Signaling Proteins/metabolism
13.
ChemMedChem ; 16(18): 2823-2844, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34032019

ABSTRACT

Starting from our previously reported hit, a series of 1,5-diaryl-1,2,3-triazole-4-carbohydrazones were synthesized and evaluated as inhibitors of the YAP/TAZ-TEAD complex. Their binding to hTEAD2 was confirmed by nanodifferential scanning fluorimetry, and some of the compounds were also found to moderately disrupt the YAP-TEAD interaction, as assessed by a fluorescence polarization assay. A TEAD luciferase gene reporter assay performed in HEK293T cells and RTqPCR measurements in MDA-MB231 cells showed that these compounds inhibit YAP/TAZ-TEAD activity to cells in the micromolar range. In spite of the cytotoxic effects displayed by some of the compounds of this series, they are still good starting points and can be suitably modified into an effective and viable YAP-TEAD disruptor in the future.


Subject(s)
Antineoplastic Agents/pharmacology , Hydrazones/pharmacology , TEA Domain Transcription Factors/antagonists & inhibitors , Transcriptional Coactivator with PDZ-Binding Motif Proteins/antagonists & inhibitors , Triazoles/pharmacology , YAP-Signaling Proteins/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Hydrazones/chemical synthesis , Hydrazones/chemistry , Molecular Structure , Structure-Activity Relationship , TEA Domain Transcription Factors/metabolism , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism , Triazoles/chemical synthesis , Triazoles/chemistry , YAP-Signaling Proteins/metabolism
14.
Acta Pharmacol Sin ; 42(11): 1847-1859, 2021 11.
Article in English | MEDLINE | ID: mdl-33879841

ABSTRACT

Metastasis is the main cause of mortality in patients with cancer. Epithelial-mesenchymal transition (EMT), a crucial process in cancer metastasis, is an established target for antimetastatic drug development. LFG-500, a novel synthetic flavonoid, has been revealed as a potential antitumor agent owing to its various activities, including modulation of EMT in the inflammatory microenvironment. Here, using a transforming growth factor beta (TGF-ß)-induced EMT models, we found that LFG-500 inhibited EMT-associated migration and invasion in human breast cancer, MCF-7, and lung adenocarcinoma, A549, cell lines, consistent with the observed downregulation of YAP activity. Further studies demonstrated that LGF-500-induced suppression of YAP activation was mediated by integrin-linked kinase (ILK), suggesting that the ILK/YAP axis might be feasible target for anti-EMT and antimetastatic treatments, which was verified by a correlation analysis with clinical data and tumor specimens. Hence, our data support the use of LGF-500 as an antimetastatic drug in cancer therapy and provide evidence that the ILK/YAP axis is a feasible biomarker of cancer progression and a promising target for repression of EMT and metastasis in cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Epithelial-Mesenchymal Transition/drug effects , Flavonoids/administration & dosage , Neoplasms/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , YAP-Signaling Proteins/antagonists & inhibitors , A549 Cells , Animals , Cell Movement/drug effects , Cell Movement/physiology , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , Epithelial-Mesenchymal Transition/physiology , Female , Hep G2 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Transgenic , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology , YAP-Signaling Proteins/metabolism
15.
Can J Physiol Pharmacol ; 99(10): 1000-1006, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33852804

ABSTRACT

The Hippo/YAP (yes-associated protein) pathway is an important signaling pathway to control organ development and tissue homeostasis. YAP is a downstream effector of the Hippo pathway and a critical mediator of mechanic stress. Hypertensive nephropathy is characterized with glomerular sclerosis stiffness and renal fibrosis. The present study investigated the role of YAP pathway in angiotensin (Ang) II hypertensive renal injury by using YAP activation inhibitor verteporfin. Ang II increased the protein expression of YAP in renal nucleus fraction, decreased phospho-YAP, and phospho-LATS1/2 (large tumor suppressors 1 and 2) expressions in renal cytoplasmic fraction, suggesting Ang II activation of renal YAP. Ang II significantly increased systolic blood pressure (SBP), proteinuria, glomerular sclerosis, and fibrosis; treatment with verteporfin attenuated Ang II-induced proteinuria and renal injury with a mild reduction in SBP. Moreover, Ang II increased the protein expressions of inflammatory factors including tumor necrosis factor α, interleukin 1ß, and monocyte chemoattractant protein-1, and profibrotic factors including transforming growth factor ß, phospho-Smad3 and fibronectin. Verteporfin reversed abovementioned Ang II-induced molecule expressions. Our results for the first time demonstrate that the activation of the YAP pathway promotes hypertensive renal inflammation and fibrosis, which may promote hypertensive renal injury. YAP may be a new target for prevention and treatment of hypertensive renal diseases.


Subject(s)
Acute Kidney Injury/drug therapy , Angiotensin II/toxicity , Hypertension, Renal/drug therapy , Hypertension/metabolism , Nephritis/drug therapy , Verteporfin/pharmacology , YAP-Signaling Proteins/antagonists & inhibitors , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , Blood Pressure , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Fibrosis , Hypertension/chemically induced , Hypertension/pathology , Hypertension, Renal/etiology , Hypertension, Renal/metabolism , Hypertension, Renal/pathology , Male , Mice , Mice, Inbred C57BL , Nephritis/etiology , Nephritis/metabolism , Nephritis/pathology , Photosensitizing Agents/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vasoconstrictor Agents/toxicity
16.
Sci China Life Sci ; 64(3): 404-418, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32804340

ABSTRACT

Targeting the white-to-brown fat conversion is important for developing potential strategies to counteract metabolic diseases; yet the mechanisms are not fully understood. Yes-associated-protein (YAP), a transcription co-activator, was demonstrated to regulate adipose tissue functions; however, its effects on browning of subcutaneous white adipose tissue (sWAT) are unclear. We demonstrated that YAP was highly expressed in cold-induced beige fat. Mechanistically, YAP was found as a target gene of miR-429, which downregulated YAP expression in vivo and in vitro. In addition, miR-429 level was decreased in cold-induced beige fat. Additionally, pharmacological inhibition of the interaction between YAP and transcriptional enhanced associate domains by verteporfin dampened the browning of sWAT. Although adipose tissue-specific YAP overexpression increased energy expenditure with increased basal uncoupling protein 1 expression, it had no additional effects on the browning of sWAT in young mice. However, we found age-related impairment of sWAT browning along with decreased YAP expression. Under these circumstances, YAP overexpression significantly improved the impaired WAT browning in middle-aged mice. In conclusion, YAP as a regulator of sWAT browning, was upregulated by lowering miR-429 level in cold-induced beige fat. Targeting the miR-429-YAP pathway could be exploited for therapeutic strategies for age-related impairment of sWAT browning.


Subject(s)
Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Cold Temperature , MicroRNAs/metabolism , YAP-Signaling Proteins/metabolism , 3T3-L1 Cells , Adipose Tissue, White/drug effects , Aging/metabolism , Animals , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protein Domains , Transcription, Genetic , Uncoupling Protein 1/metabolism , YAP-Signaling Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...