Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
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
2.
Int Immunopharmacol ; 104: 108510, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34999393

ABSTRACT

Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis in young children, but there are few safe and effective treatments for this disease. Platycodonis Radix is widely used as an antitussive and expectorant drug for preventing various diseases in lower respiratory tract, in which the polysaccharides are one of the main bioactivity constituents. In this study, the protective effects of the P. Radix polysaccharides (PRP) against RSV-induced bronchiolitis in juvenile mice and RSV-induced apoptosis of epithelial HEp-2 cells were investigated. The results showed that PRP obviously decreased the levels of IL-1ß, IL-4, IL-6, TNF-α, IFN-γ and TSLP in lung tissues, and reduced the number of inflammatory cells in bronchoalveolar lavage fluid (BALF) of RSV-infected mice. Furthermore, it reduced the apoptosis of RSV-infected HEp-2 cells and remarkably inhibited the mRNA expressions of RSV L gene, which indicated that PRP affected transcription and replication of RSV in host cells. Compared with that in RSV-infected group, miR-181a-5p in the PRP-treated group presented the highest relative abundance and its expression was violently reduced by approximately 30%. Mechanistically, PRP had the similar effects as miR-181a-5p antagomir on RSV-induced apoptosis and inflammation in HEp-2 cells via upregulating BCL2, MLL3 and SIRT1, which could be reversed by miR-181a-5p mimic. Therefore, it demonstrated that PRP not only protected against RSV-induced lung inflammation in mice but also inhibited apoptosis of RSV-infected HEp-2 cells via suppressing miR-181a-5p and transcriptionally activating Hippo and SIRT1 pathways.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Plant Extracts , Platycodon , Polysaccharides/therapeutic use , Respiratory Hypersensitivity/drug therapy , Respiratory Syncytial Virus Infections/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/virology , Female , Hippo Signaling Pathway/drug effects , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice, Inbred BALB C , MicroRNAs , Polysaccharides/pharmacology , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus Infections/pathology , Respiratory Syncytial Viruses , Sirtuin 1/metabolism
3.
J Med Chem ; 65(2): 1352-1369, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34807584

ABSTRACT

Serine/threonine-protein kinases 3 and 4 (STK3 and STK4, respectively) are key components of the Hippo signaling pathway, which regulates cell proliferation and death and provides a potential therapeutic target for acute myeloid leukemia (AML). Herein, we report the structure-based design of a series of pyrrolopyrimidine derivatives as STK3 and STK4 inhibitors. In an initial screen, the compounds exhibited low nanomolar potency against both STK3 and STK4. Crystallization of compound 6 with STK4 revealed two-point hinge binding in the ATP-binding pocket. Further characterization and analysis demonstrated that compound 20 (SBP-3264) specifically inhibited the Hippo signaling pathway in cultured mammalian cells and possessed favorable pharmacokinetic and pharmacodynamic properties in mice. We show that genetic knockdown and pharmacological inhibition of STK3 and STK4 suppress the proliferation of AML cells in vitro. Thus, SBP-3264 is a valuable chemical probe for understanding the roles of STK3 and STK4 in AML and is a promising candidate for further advancement as a potential therapy.


Subject(s)
Hippo Signaling Pathway/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Serine-Threonine Kinase 3/antagonists & inhibitors , Animals , Female , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/chemistry
4.
J Mol Med (Berl) ; 100(1): 135-146, 2022 01.
Article in English | MEDLINE | ID: mdl-34689211

ABSTRACT

Fluoroscopy-induced chronic radiation dermatitis (FICRD) is a complication of fluoroscopy-guided intervention. Unlike acute radiation dermatitis, FICRD is different as delayed onset and usually appears without preexisting acute dermatitis. Unfortunately, the chronic and progressive pathology of FICRD makes it difficult to treat, and some patients need to receive wide excision and reconstruction surgery. Due to lack of standard treatment, investigating underlying mechanism is needed in order to develop an effective therapy. Herein, the Hippo pathway is specifically identified using an RNA-seq analysis in mild damaged skin specimens of patients with FICRD. Furthermore, specific increase of the Yes-associated protein (YAP1), an effector of the Hippo pathway, in skin region with mild damage plays a protective role for keratinocytes via positively regulating the numerous downstream genes involved in different biological processes. Interestingly, irradiated-keratinocytes inhibit activation of fibroblasts under TGF-ß1 treatment via remote control by an exosome containing YAP1. More importantly, targeting one of YAP1 downstream genes, nuclear receptor subfamily 3 group C member 1 (NR3C1), which encodes glucocorticoid receptor, has revealed its therapeutic potential to treat FICRD by inhibiting fibroblasts activation in vitro and preventing formation of radiation ulcers in a mouse model and in patients with FICRD. Taken together, this translational research demonstrates the critical role of YAP1 in FICRD and identification of a feasible, effective therapy for patients with FICRD. KEY MESSAGES: • YAP1 overexpression in skin specimens of radiation dermatitis from FICRD patient. • Radiation-induced YAP1 expression plays protective roles by promoting DNA damage repair and inhibiting fibrosis via remote control of exosomal YAP1. • YAP1 positively regulates NR3C1 which encodes glucocorticoid receptor expression. • Targeting glucocorticoid receptor by prednisolone has therapeutic potential for FICRD patient.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Fluoroscopy/adverse effects , Glucocorticoids/therapeutic use , Prednisolone/therapeutic use , Radiodermatitis/metabolism , Animals , Cell Line , Hippo Signaling Pathway/drug effects , Humans , Keratinocytes/metabolism , Mice, Inbred C57BL , Radiodermatitis/drug therapy , Radiodermatitis/genetics , Skin/drug effects , Skin/metabolism , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism
5.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166267, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34508829

ABSTRACT

Bromodomain and extraterminal (BET) proteins are promising therapeutic targets for hematological and solid tumors. However, BET inhibitor monotherapy did not show a significant therapeutic benefit for hepatocellular carcinoma (HCC) in preclinical trials. Here, we identified YAP/TAZ genes, as determinants for sensitivity to BET inhibitors. YAP/TAZ expression, especially TAZ, promote resistance to BET inhibitor. In addition, we analyzed that the mRNA level of PDE5 was positively correlated with YAP/TAZ based on TCGA database and demonstrated tadalafil, a PDE5 inhibitor, could block YAP/TAZ protein expression by activating Hippo pathway. Cotreatment with tadalafil and JQ-1 synergistically reduced YAP/TAZ protein expression, suppressed proliferation and induced G0-G1 arrest of cultured HCC cells. JQ-1 alone does not show significant benefits in a mouse model of HCC induced by c-Myc/N-Ras plasmids. In contrast, the combination, tadalafil and JQ-1, successfully suppressed tumor progression, enhanced antitumor immunity by improving the ratio of activated CD8 and extended the survival time of mice. Our data define the key role of YAP/TAZ in mediating resistance to BET inhibitor, described the PDE5/PKG/Hippo/YAP/TAZ axis and identified a common clinical drug that can be developed as an effective combined strategy to overcome BET inhibitor resistance in MYC/Ras-driven HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nerve Tissue Proteins/genetics , Receptors, Cell Surface/genetics , Tadalafil/pharmacology , YAP-Signaling Proteins/genetics , Animals , Azepines/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cyclic Nucleotide Phosphodiesterases, Type 5/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Hippo Signaling Pathway/drug effects , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Phosphodiesterase 5 Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/genetics , Receptors, Cell Surface/antagonists & inhibitors , Triazoles/pharmacology
6.
Clin Transl Med ; 11(7): e475, 2021 07.
Article in English | MEDLINE | ID: mdl-34323413

ABSTRACT

BACKGROUND: Uterine fibroids are highly prevalent, collagen-rich, mechanically stiff, fibrotic tumors for which new therapeutic options are needed. Increased extracellular matrix (ECM) stiffness activates mechanical signaling and Hippo/YAP promoting fibroid growth, but no prior studies have tested either as a therapeutic target. We tested the hypothesis that injection of a purified form of collagenase Clostridium histolyticum (CCH) that selectively digests type I and type III collagens would alter ECM stiffness, Hippo signaling, and selectively reduce fibroid cell growth. We also used two FDA-approved drugs, verteporfin and nintedanib, to elucidate the role of Hippo/YAP signaling in uterine fibroid and myometrial cells. METHODS: The clinical trial was registered (NCT02889848). Stiffness of samples was measured by rheometry. Protein expression in surgical samples was analyzed via immunofluorescence. Protein and gene expression in uterine fibroid or myometrial cell lines were measured by real time PCR and western blot, and immunofluorescence. RESULTS: Injection of CCH at high doses (0.1-0.2 mg/cm3 ) into fibroids resulted in a 46% reduction in stiffness in injected fibroids compared to controls after 60 days. Levels of the cell proliferation marker proliferative cell nuclear antigen (PCNA) were decreased in fibroids 60 days after injection at high doses of CCH. Key Hippo signaling factors, specifically the transcriptionally inactive phosphorylated YAP (p-YAP), was increased at high CCH doses, supporting the role of YAP in fibroid growth. Furthermore, inhibition of YAP via verteporfin (YAP inhibitor) decreased cell proliferation, gene and protein expression of key factors promoting fibrosis and mechanotransduction in fibroid cells. Additionally, the anti-fibrotic drug, nintedanib, inhibited YAP and showed anti-fibrotic effects. CONCLUSIONS: This is the first report that in vivo injection of collagenase into uterine fibroids led to a reduction in Hippo/YAP signaling and crucial genes and pathways involved in fibroid growth. These results indicate that targeting ECM stiffness and Hippo signaling might be an effective strategy for uterine fibroids.


Subject(s)
Antifibrotic Agents/pharmacology , Extracellular Matrix/metabolism , Hippo Signaling Pathway/drug effects , Microbial Collagenase/pharmacology , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Adult , Antifibrotic Agents/therapeutic use , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Indoles/pharmacology , Indoles/therapeutic use , Integrin beta1/genetics , Integrin beta1/metabolism , Leiomyoma/drug therapy , Leiomyoma/pathology , Microbial Collagenase/therapeutic use , Middle Aged , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Uterine Neoplasms/drug therapy , Uterine Neoplasms/pathology , Verteporfin/pharmacology
7.
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
8.
Pharmacol Res ; 170: 105723, 2021 08.
Article in English | MEDLINE | ID: mdl-34116210

ABSTRACT

FAT atypical cadherin 4 (FAT4) has been identified as a tumor suppressor in lung cancers. However, no agent for lung cancer treatment targeting FAT4 has been used in the clinic. Jujuboside A (JUA) is a major active compound in Semen Ziziphi Spinosae. Semen Ziziphi Spinosae is a traditional Chinese herbal medicine used clinically for tumor treatment to improve patients' quality of life. However, the anti-lung cancer activity and the underlying mechanisms of JUA are not yet fully understood. Here, we demonstrated the anti-lung cancer activity of JUA in two lung cancer mice models and three non-small cell lung cancer (NSCLC) cell lines, and further illustrated its underlying mechanisms. JUA suppressed the occurrence and development of lung cancer and extended mice survival in vivo, and suppressed NSCLC cell activities through cell cycle arrest, proliferation suppression, stemness inhibition and senescence promotion. Moreover, JUA directly bound with and activated FAT4, subsequently activating FAT4-HIPPO signaling and inhibiting YAP nuclear translocation. Knockdown of FAT4 diminished JUA's effects on HIPPO signaling, YAP nuclear translocation, cell proliferation and cellular senescence. In conclusion, JUA significantly suppressed NSCLC tumorigenesis by regulating FAT4-HIPPO-YAP signaling. Our findings suggest that JUA is a novel FAT4 activator that can be developed as a promising NSCLC therapeutic agent targeting the FAT4-HIPPO-YAP pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cadherins/agonists , Carcinoma, Non-Small-Cell Lung/drug therapy , Hippo Signaling Pathway/drug effects , Lung Neoplasms/drug therapy , Saponins/pharmacology , Tumor Suppressor Proteins/agonists , YAP-Signaling Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Tumor Suppressor Proteins/metabolism
9.
Br J Cancer ; 124(12): 2004-2016, 2021 06.
Article in English | MEDLINE | ID: mdl-33762722

ABSTRACT

BACKGROUND: Chemoresistance is one of the major factors for treatment failure in OSCC. Identifying key resistance triggering molecules will be useful strategy for developing novel treatment methods. METHODS: To identify the causative factors of chemoresistance, we performed RNA sequencing and global proteomic profiling of human OSCC lines presenting with sensitive, early and late cisplatin-resistance patterns. RESULTS: From the common set of dysregulated genes from both the analysis, RRBP1 was identified to be upregulated in both early and late cisplatin-resistant cells with respect to the sensitive counterpart. Analysis of OSCC patient sample indicates that RRBP1 expression is upregulated in chemotherapy-non-responder tumours as compared to chemotherapy-responder tumours. Genetic (knockout) or pharmacological (Radezolid, represses expression of RRBP1) inhibition of RRBP1 restores cisplatin-mediated cell death in chemo-resistant OSCC. Mechanistically, RRBP1 regulates Yes-associated protein1 (YAP1), a key protein in the Hippo pathway to induce chemoresistance. The PDC xenograft data suggests that knockout of RRBP1 induces cisplatin-mediated cell death and facilitates a significant reduction of tumour burden. CONCLUSION: Overall, our data suggests that (I) RRBP1 is a major driver of cisplatin-resistance in OSCC, (II) RRBP1 regulates YAP1 expression to mediate cisplatin-resistance, (III) Radezolid represses RRBP1 expression and (IV) targeting RRBP1 reverses cisplatin-induced chemoresistance in advanced OSCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carrier Proteins/physiology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Mouth Neoplasms/drug therapy , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carrier Proteins/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Knockout Techniques , HEK293 Cells , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Anticancer Agents Med Chem ; 21(12): 1520-1528, 2021.
Article in English | MEDLINE | ID: mdl-33121418

ABSTRACT

Hippo signalling pathway that is evolutionarily conserved affects diver's pathology and physiology processes, including tissue repair, wound healing, tissue size and tissue regeneration. Epigenetic changes are post-translational modifications in DNA proteins and histones. Epigenetic changes including histone acetylation and deacetylation, miRNAs dysregulation, and aberrant DNA methylation, inflammatory genes actives abnormal in Hippo signaling pathway. Using some treatments, including Histone Deacetylases (HDACs), herbal composition, siRNAs and long non-coding RNA (lncRNA) for suppressing cancer cells by targeting Hippo pathways, may open new views in cancer target therapy fields. The aim of this review study is firstly to demonstrate the importance of Hippo signalling and its association with epigenetic changes in cancer and then to demonstrate progress in targeting Hippo signalling in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Hippo Signaling Pathway/drug effects , Humans , Neoplasms/genetics , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL