Advances in research on the carcinogenic mechanisms and therapeutic potential of YAP1 in bladder cancer (Review)
- Authors:
- Published online on: November 12, 2024 https://doi.org/10.3892/or.2024.8843
- Article Number: 10
-
Copyright: © Huang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
The Hippo signaling pathway is an evolutionarily conserved signal pathway that was first identified in Drosophila and plays an important role in the regulation of cell proliferation, apoptosis and tissue and organ growth (1,2). Yes-associated protein 1 (YAP1), a pivotal component of the Hippo signaling pathway, is encoded by the YAP1 gene located on human chromosome 11q22 and functions as an oncogenic protein (Fig. 1) (3). When the Hippo pathway is disrupted, mammalian STE20-like protein kinase 1/2 (Mst1/2) kinases become phosphorylated and activated, which in turn activates downstream large tumor suppressor kinase 1 and 2 ((LATS1/2) kinases. These activated LATS1/2 kinases phosphorylate YAP1, leading to its degradation in the cytoplasm (4). Conversely, dephosphorylated YAP1 translocates to the nucleus, binds to the TEA domain (TEAD) and promotes the transcription of downstream genes (for example, CTGF and Cyr61) (5,6).
YAP1/transcriptional co-activator with PDZ-binding motif (TAZ) play essential roles in regulating cell proliferation and are crucial for organ regeneration, development and stem cell self-renewal (7). They are influenced by various factors, including the microenvironment and extracellular signals. Additionally, YAP1 can activate survival pathways to inhibit apoptosis and interacts with multiple signaling networks (8), contributing to cancer development and progression (9). Elevated YAP1 expression is associated with malignant tumor recurrence, metastasis, reduced overall survival and chemotherapy resistance (10). In human cancers, YAP/TAZ target genes are associated with poor prognosis and increased treatment resistance (11). YAP1 is highly expressed in various cancers and linked to adverse outcomes, including bladder, colon, pancreatic, hepatocellular, gastric/esophageal, ovarian, brain, breast and lung cancers (12–17).
Bladder cancer is the most prevalent malignancy within the urinary system and is anticipated to rank as the ninth most common cancer in the United States by 2023 (18). Previous years have seen an increasing incidence of bladder cancer, with a trend toward younger patients (19). While the etiology remains incompletely understood, risk factors include smoking, aging and occupational exposures (20). Genetic mutations and altered RNA expression levels also contribute to bladder cancer risk. YAP1 is notably upregulated in bladder cancer and is associated with poor prognosis (21). Research indicates that single nucleotide polymorphisms (SNPs) in YAP1, a key molecule in the Hippo pathway, are linked to bladder cancer development (22). BCa is a cancer that frequently exhibits mutations. A study has indicated that in human bladder smooth muscle, Gene Ontology annotation analysis shows that differential genes are enriched in positive regulation of cellular processes, metabolism, protein binding and signal transduction processes. Kyoto Encyclopedia of Genes and Genomes enrichment analysis highlights genes in the MAPK and PI3K/Akt signaling pathways (23). Additionally, a next-generation sequencing study suggests that BCa is primarily associated with a high mutation burden in genes related to the cell cycle and chromatin regulation. Dysfunctional Hippo signaling may contribute to the development of bladder cancer, although it is not an independent driver (24). The present study reviews the carcinogenic mechanisms of YAP1 in bladder cancer and evaluates its potential as a therapeutic target.
Regulation of YAP1 activity in bladder cancer
YAP1 activity is controlled by its gene expression levels. Knockdown of this gene in bladder cancer cell lines significantly reduces YAP1 protein activity (21). Furthermore, the primary method of regulating YAP1 activity involves modulation of its phosphorylation status. When phosphorylated, YAP1 is retained in the cytoplasm and thus is unable to translocate to the nucleus to perform its transcriptional activation functions. 4-hydroxynonenal induces phosphorylation of YAP1 at serine residues 127 and 387, leading to reduced YAP1 expression and consequently decreases bladder cancer cell proliferation (25). Ubiquitination is regulated by deubiquitinases (DUBs), which are specialized Ub proteases that process Ub chains in various ways to regulate ubiquitination (26). MINDY1, a DUB specific to YAP1, removes the K48-linked ubiquitin chain from YAP1, thereby preventing its proteasomal degradation. This action enhances the stability and abundance of the YAP1 protein, consequently boosting its transcriptional activation capability and facilitating the progression of bladder cancer (Fig. 2) (27).
YAP1 participates in the progression of bladder cancer
Tumor cell proliferation and migration is a dynamic and intricate process that contributes to the advancement of numerous diseases. Abnormal cell proliferation often leads to tumor formation. Once tumor cells acquire the capability to migrate and detach from the primary site, they can significantly accelerate tumor dissemination and metastasis. Phosphorylated YAP1 not only directly influences tumor proliferation but also collaborates with various molecules to promote both tumor proliferation and migration. For instance, The drosophile Mask (multiple ankyrin repeats single KH domain) whose mammalian homologues MASK1 (also known as ANKHD1) and MASK2 (also known as GTAR or ANKRD17) are a novel cofactor of YAP1. In bladder cancer cell lines overexpressing YAP1, MASK2interacts with YAP1 to modulate its downstream target genes, consequently promoting the proliferation and migration of bladder cancer cells (28).
The MAPK pathway, ubiquitously present in organisms, potentially facilitates the onset and progression of cancer when its signaling becomes dysregulated (29). YAP1 serves as both an upstream regulator and an activator of the MAPK pathway. Its stimulation of the MAPK/ERK pathway synergistically promotes the proliferation and invasion of bladder cancer cells (30). Research demonstrates that YAP1 modulates the transcription of both long non-coding RNA (lncRNA) and mRNA (31). Specifically, lncRNA H19, a downstream target of YAP1, is upregulated by YAP1, influencing the proliferation and migration of bladder cancer cells (32). Furthermore, in bladder cancer, lncRNA H19 interacts with microRNA (miRNA/miR)-29b-3p, impacting epithelial-mesenchymal transition (EMT) and thereby facilitating cancer metastasis (33).
Furthermore, YAP1 can collaborate synergistically with miRNA to facilitate the progression of bladder cancer. MiRNAs, serving as critical regulatory factors in tumor development, are intricately linked to tumor formation, metastasis, cellular apoptosis and chemoresistance when expressed abnormally (34). A study by Luo (35) identified a cross-network interaction between the Hippo and TGF-β signaling pathways, where the YAP1-Smad3 complex plays a pivotal role. MiR-497 and miR-195 disrupt the formation of the YAP1/SMAD3 complex, thereby interfering with the interactions between the Hippo/YAP1 and TGF-β/Smad pathways. This disruption diminishes their influence on the shared target gene, connective tissue growth factor (CTGF), ultimately reducing bladder cancer cell proliferation (36). Furthermore, the overexpression of hypermethylated in cancer 1 suppresses CTGF activity, limits interactions between YAP1 and the TEAD, and inhibits proliferation, migration and invasion of bladder cancer cells (37). Additionally, miR-599 curtails proliferation and invasion, while enhancing apoptosis in bladder urothelial carcinoma cells by modulating YAP1 expression (38).
Exosomes, a type of extracellular vesicle, are secreted by virtually all cell types. These vesicles support a wide array of cellular functions, encompassing intercellular communication, cell differentiation and proliferation, angiogenesis, stress responses and immune signaling (39,40). Research demonstrates a distinct relationship between exosomes and YAP1, where exosomal miR-217 from normal human bladder stromal cells is significantly elevated in bladder cancer cell lines, and by modulating YAP1 and its downstream targets, cysteine-rich angiogenic inducer 61 (CYR61), CTGF and ankyrin repeat domain-containing protein 1 miR-217 mimics or inhibitors can notably influence the proliferation, migration, and apoptosis of bladder cancer cells (41).
EMT is a critical process in tumor metastasis and a hallmark of malignant tumors. Additionally, EMT is integral to embryogenesis and wound healing (42,43). In a study, high-glucose culture media induces YAP1 and TAZ expression in bladder cancer cells, resulting in the upregulation of wave protein, N-cadherin, and fibronectin, alongside the downregulation of E-cadherin to promote EMT, while also regulating glucose metabolism through glucose transporter type 1 activity, thereby highlighting the Hippo signaling pathway as a crucial factor in the dysregulation of glucose metabolism in bladder cancer (Table I) (44).
Table I.In bladder cancer cell lines, YAP1 and its downstream signaling pathways influence various biological behaviors of bladder cancer. |
Stem cells (SCs) are multipotent cells with the capacity for self-renewal. Provided specific conditions, these cells can differentiate into a variety of functional cell types (45,46). Cancer SCs (CSCs) are characterized by their capacity for self-renewal and differentiation. These cells significantly contribute to tumor progression, treatment resistance and disease recurrence by continuously proliferating, invading normal tissues and exhibiting resistance to conventional cancer therapies (47,48).
YAP1 is pivotal in regulating several key processes in CSCs, including self-renewal, multilineage differentiation, tumorigenesis, treatment resistance and metastatic invasion (49,50). In embryonic, neural and hematopoietic stem cells, elevated expression of the YAP1-TEAD2 complex sustains stem cell potency and serves as an indicator of stemness (51). YAP1, a regulator of stem cell functions, is notably upregulated in bladder cancer stem cells. The overexpression of YAP significantly enhances the self-renewal ability of these cells (52). YAP1 collaborates with the pro-inflammatory cyclooxygenase 2 (COX2)/prostaglandin E2 (PGE2) signaling pathway to enhance the proliferation of bladder urothelial carcinoma stem-like cells and influence tumor chemoresistance. As the COX2/PGE2 pathway induces methylation of the let-7 promoter, this results in reduced let-7 expression and subsequent upregulation of SRY-box transcription factor 2 (SOX2), which is further amplified by YAP1′s binding to the SOX2 enhancer, thereby increasing SOX2 expression (53). The interaction between the COX2 and YAP1 pathways may significantly influence the recurrence of bladder cancer.
Furthermore, the miR-146a rs2910164 SNP genotype C>G significantly affects the prognosis of bladder cancer patients by modulating key targets within the Hippo and COX2 signaling pathways, leading to the suppression of YAP1 and COX2 at both mRNA and protein levels, which results in the downregulation of YAP1 and aldehyde dehydrogenase 1 family, member A1, along with reduced expression of COX2 and SOX2, all of which are closely associated with bladder cancer recurrence (54). Simultaneously, a study has demonstrated that YAP1 is involved in its own recurrence process. During the postoperative wound healing phase in bladder cancer, it has been observed that the extracellular matrix regulates the transport of YAP1 to the nucleus through integrin-FAK-YAP1 signaling. This regulation contributes to the progression and recurrence of bladder cancer (55).
This implies that YAP1 overexpression not only affects bladder cancer progression through interactions with multiple signaling pathways but also contributes to this progression by regulating the stemness of tumor stem cells. Consequently, targeting YAP1 and its associated regulatory network could provide novel therapeutic approaches for bladder cancer.
Therapeutic potential of YAP1-targeting in bladder cancer
Bladder urothelial carcinoma is classified into two main categories based on clinical staging, each with different clinical outcomes and treatment options: Muscle invasive bladder cancer (MIBC) and non-MIBC (NMIBC). Treatment decisions are primarily based on clinical factors. Besides surgery, common treatments currently include cisplatin chemotherapy, immunotherapy and targeted therapy. Despite these options, the treatment outlook for bladder cancer remains unsatisfactory, with a high risk of recurrence. Although the targeting of cancer treatment signaling pathways has increased over the past few decades, bladder cancer requires the exploration of potentially more effective treatments (Table II).
YAP1 and chemotherapy resistance in bladder cancer
Chemotherapy remains a cornerstone of treatment for malignant tumors, with cisplatin used across a spectrum of cancers, including bladder cancer (56–58). Yet, the emergence of cisplatin resistance has notably diminished its therapeutic effectiveness (59,60). Moreover, the redox state, crucial in the adaptation of cancer cells to therapeutic stresses, significantly influences chemoresistance (61). Research demonstrates that YAP1 activation plays a pivotal role in conferring resistance across targeted therapy, chemotherapy, radiotherapy and possibly immunotherapy, thus establishing it as a critical mediator of chemoresistance in diverse cancers (62,63).
The YAP1/TEAD complex is known to regulate Forkhead box protein M1(FOXM1) expression, which in turn affects nuclear factor erythroid 2-related factor 2 (Nrf2) expression (64). Both Nrf2 and YAP1 contribute to the antioxidative capacity of bladder cancer cells, with a positive association observed between the expression levels of YAP1 and Nrf2. Targeted inhibition of YAP1 and Nrf2, aimed at decreasing cell viability, inducing apoptosis and curbing cancer cell migration, has been shown to substantially enhance the sensitivity of these cells to cisplatin (66). The transcription factor Nrf2 is a pivotal regulator of antioxidant defenses and cellular protection mechanisms, playing a significant role in mediating cisplatin resistance in bladder cancer (66). Emerging research based on the interaction between YAP1 and Nrf2 indicates that Ailanthone (Aila) can potentially serve as an effective therapeutic for cisplatin-resistant bladder cancer. Notably, Aila consistently suppresses Nrf2 expression while concurrently inhibiting YAP1 and c-Myc, thereby sustaining a robust anti-proliferative effect on cisplatin-resistant cells (67). Additionally, Aila markedly decreases the expression of the Nrf2 target glutathione S-transferase A4 and the YAP1/TEAD target survivin. In cisplatin-resistant bladder cancer cells, levels of intracellular oxidative stress are notably lower compared with those in cisplatin-sensitive cells. The post-translational downregulation of Nrf2 and YAP1 proteins can effectively reduce cancer cell proliferation and migration, while simultaneously increasing oxidative stress in chemotherapy-resistant bladder cancer cell lines (68). Decitabine, a DNA methyltransferase inhibitor, demonstrates cytotoxic effects at high doses by directly inducing cell death. At lower doses, it reactivates silenced genes via demethylation of promoter regions and is extensively employed in the treatment of diseases such as leukemia (69).
In bladder cancer cells, decitabine reactivates the Hippo signaling pathway through the restoration of Ras association domain-containing protein 1, concurrently inhibiting YAP1 expression and reducing the expression of its oncogenic downstream targets, including CTGF and CYR61. Furthermore, decitabine enhances the cytotoxic effects of cisplatin and doxorubicin in these cells (70). YAP1 expression and activation are inversely correlated with cisplatin sensitivity both in vitro and in vivo. The YAP1 inhibitor verteporfin has been demonstrated to inhibit tumor cell proliferation and enhance cisplatin sensitivity in bladder cancer cells. Inhibition of YAP1 expression increases DNA damage and promotes apoptosis accumulation, thereby sensitizing urothelial carcinoma cells to radiochemotherapy. Notably, nuclear YAP1 expression is linked to adverse outcomes in patients with urothelial carcinoma undergoing perioperative chemotherapy (71). The YAP1/TEAD1/PDGF-BB/PDGFR autocrine loop plays a pivotal role in sustaining the characteristics of tumor stem-like cells. Inhibition of YAP1 has been shown to significantly reduce cisplatin resistance in OV6-positive bladder cancer stem-like cells, a finding corroborated by animal model studies.
YAP1 and bladder cancer immunotherapy
Immunotherapy is a treatment method aimed at enhancing the body's immune system or modifying the ongoing cancer defense mechanisms to combat cancer (72,73). In recent years, immunotherapy has gained prominence in cancer treatment and has undergone rapid development (74). In bladder cancer, Bacillus Calmette-Guérin (BCG) remains the preferred immunotherapy for NMIBC, effectively stimulating immune responses and modulating the immune system to produce antitumor effects, thereby significantly reducing recurrence rates and prolonging the interval between recurrences (75,76). Nevertheless, ~1/3 of patients with NMIBC do not respond to BCG therapy. A previous study indicates that combining programmed cell death protein 1 (PD-1) inhibitors with BCG treatment is a safe and potentially effective strategy for these non-responders (77). Additionally, immune checkpoint inhibitors (ICIs) such as PD-1 and programmed death-ligand 1 (PD-L1) antibodies are approved for bladder cancer treatment. These ICIs can directly interact with the urothelium or malignant cells on the bladder mucosa, initiating active immune responses (78). Baek et al (79) revealed three molecular subtypes of advanced urothelial carcinoma through gene expression analysis, which have potential clinical application value in predicting responses to immune checkpoint inhibitor therapy and patient prognosis. Among these, the inactivation of the YAP/TAZ pathway in the third subtype is associated with the activation of cell cycle and DNA damage response pathways. This suggests that in patients with the third subtype, the inactivation of the YAP/TAZ pathway might be related to a good response to ICI therapy and improved prognosis (79). Further research by Baek et al (80) categorized YAP1 activation into three distinct subtypes based on differential gene expression and integrated this classification with immunotherapy prognostic analysis for patients. This analysis suggests that NMIBC subgroups with YAP1 activation might derive significant benefit from ICI treatments. Additionally, the activation status of YAP1 in NMIBC is not only associated with the biological behavior of the disease but also has the potential to serve as a crucial biomarker for predicting treatment response and patient prognosis (80).
YAP1 and targeted therapy for bladder cancer
Based on platinum-based chemotherapy, which serves as the first-line treatment for patients with NMIBC, survival rates are improved but still remain relatively low (81). Advances in targeted therapies, which have transformed treatment paradigms in various cancers, are only beginning to make inroads into bladder cancer (82,83). Approved treatments such as PD-1/PD-L1 inhibitors, the fibroblast growth factor receptor inhibitor Erdafitinib, and Enfortumab vedotin (an antibody-drug conjugate directed at Nectin-4 that comprises a fully human, monoclonal antibody and the microtubule-disrupting agent, monomethyl auristatin E) have shown promise (84). Notably, Enfortumab vedotin has achieved a remission rate of ~8% in patients with locally advanced or metastatic bladder cancer previously treated with platinum-based and anti-PD-1/PD-L1 therapies (85). Furthermore, the YAP1-specific inhibitor, Verteporfin (VP), demonstrates potential as a targeted therapeutic by inhibiting YAP1-induced gene expression within the Hippo pathway and reducing the proliferation and invasion of bladder cancer cells in a dose-dependent manner (86). Dual treatment involving Crizotinib and VP in canine bladder cancer organoids significantly reduce cell vitality by inducing apoptosis, underscoring the potential of combining Crizotinib with VP (87). An additional study has explored the synergistic effects of Verteporfin and the RhoA inhibitor Simvastatin, which counteract the proliferative and metastatic activities induced by PLAGL2. Protein kinase membrane-associated tyrosine/threonine 1, another direct target of YAP1/TEAD1, is implicated in activating the YAP1/TAZ pathway, further highlighting the complex regulatory mechanisms in bladder cancer pathogenesis (88).
Limitations and prospects
YAP1, recognized as an oncogene (89), plays critical roles in cell proliferation, migration, epithelial-mesenchymal transition, stemness and chemotherapy resistance across various malignancies, including bladder cancer. However, research on YAP1 in bladder cancer mainly focuses on bladder transitional cell carcinoma, and there is a lack of exploration regarding the expression and mechanisms of YAP1 in other subtypes of bladder cancer (such as bladder squamous cell carcinoma, bladder adenocarcinoma and bladder sarcoma). Despite its established significance in the molecular mechanisms underlying bladder cancer, in vivo experimental validation remains limited, and the regulatory mechanisms of the upstream factors of YAP1 are poorly understood. Enhanced exploration of the HIPPO signaling pathway's interactions in bladder cancer could revolutionize diagnostic and therapeutic approaches. Currently, post-translational modifications of YAP1 are extensively studied in various human solid tumors (90–92). Ubiquitination modification of YAP1 has been confirmed to be of significant importance for tumor progression in multiple tumors (14,93).
Additionally, glycosylation modification of YAP1 can increase its stability, promoting the malignant development of chronic kidney disease (94). Previous research findings suggest that metabolic pathways play a role in regulating YAP/TAZ activity. Glycolysis, energy stress and mevalonate biosynthesis have been identified as regulators of YAP/TAZ activity (95). Currently, glucose metabolism is a hotspot in tumor research. At present, metabolic diagnostics can improve accuracy and affordability (96). YAP1 is also involved in the tumor glycolysis process and is considered a glucose metabolism regulator. YAP1 mediates cardiac hypertrophy induced by pressure overload through aerobic glycolysis (97). In thyroid tumors and liver tumors, high glucose levels regulate YAP1 activity and promote tumor progression (98,99). In bladder tumors, research has shown that extracellular glucose levels regulate YAP1 and promote the progression of bladder cancer EMT (44). In bladder cancer, YAP1 is not only related to glucose metabolism but also acts as an oncogene and glycoprotein, making it a promising tumor marker for bladder cancer. Currently, research combines composite materials with metabolic analysis to screen for potential biomarkers (100). YAP1 is expected to become a biomarker in bladder cancer; however, extensive clinical data is still needed to clarify the sensitivity and specificity of YAP1.
YAP1, known as a cancer stem cell marker, contributes to the progression of bladder cancer. However, while a previous study has examined YAP1′s interactions with other molecular pathways affecting cancer stem cell dynamics, the exact mechanisms of YAP1′s involvement in these processes remain poorly defined (49). Research indicates that YAP1/TAZ may be a new decisive factor in ferroptosis (101). Activation of ferroptosis can specifically target and kill certain cancer cells, leading to improved survival rates. Ferroptosis is currently a major focus in tumor research. Meanwhile, a single study suggests that nanomaterials and copper ions could offer new approaches for tumor treatment (102). However, there are currently no reports on the interaction between YAP1 and ferroptosis in bladder cancer.
Although YAP1-targeted inhibitors have demonstrated efficacy in various cancers, research specifically addressing these inhibitors in bladder cancer is sparse, and their molecular mechanisms of action are not well understood. Additionally, YAP1 expression is closely linked to the occurrence, size and stage of bladder cancer, highlighting its potential as a biomarker. Despite the high recurrence rate of bladder cancer, research into the fundamental mechanisms driving this recurrence is inadequate. Notably, the expression of YAP1 is associated with recurrence rates, but detailed investigations into how YAP1 contributes to bladder cancer recurrence are still required.
Conclusion
In summary, there is a critical need for comprehensive studies in bladder cancer focusing on several key areas: i) The interactions of the Hippo signaling pathway with YAP1, particularly its post-translational modifications and their impact on tumor progression; ii) the dynamics between YAP1 and glucose metabolism; iii) the role of YAP1 in bladder cancer stem cell behavior; iv) the effectiveness of YAP1-targeted inhibitors in clinical treatment; and v) the specific mechanisms through which YAP1 contributes to bladder cancer recurrence. Given its significant influence on various cancer-related processes, YAP1 holds great promise as a novel biomarker and therapeutic target for bladder cancer.
Acknowledgements
Not applicable.
Funding
This study was supported by the National Natural Science Foundation of China (grant no. 81860524).
Availability of data and materials
Not applicable.
Authors' contributions
TH contributed to the creation of figures, drafted the manuscript and conducted research. LF contributed to table creation and drafted the manuscript. JT contributed to revision of the manuscript. SC contributed to drafting the original manuscript. GD contributed to drafting manuscript. ZN contributed to the conceptual design, provided resources, oversaw the project and reviewed and edited the manuscript.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
Misra JR and Irvine KD: The Hippo signaling network and its biological functions. Annu Rev Genet. 52:65–87. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ma S, Meng Z, Chen R and Guan KL: The Hippo pathway: Biology and pathophysiology. Annu Rev Biochem. 88:577–604. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lorenzetto E, Brenca M, Boeri M, Verri C, Piccinin E, Gasparini P, Facchinetti F, Rossi S, Salvatore G, Massimino M, et al: YAP1 acts as oncogenic target of 11q22 amplification in multiple cancer subtypes. Oncotarget. 5:2608–2621. 2014. View Article : Google Scholar : PubMed/NCBI | |
Wu Z and Guan KL: Hippo signaling in embryogenesis and development. Trends Biochem Sci. 46:51–63. 2021. View Article : Google Scholar : PubMed/NCBI | |
Pocaterra A, Romani P and Dupont S: YAP/TAZ functions and their regulation at a glance. J Cell Sci. 133:jcs2304252020. View Article : Google Scholar : PubMed/NCBI | |
Zanconato F, Cordenonsi M and Piccolo S: YAP/TAZ at the roots of cancer. Cancer Cell. 29:783–803. 2016. View Article : Google Scholar : PubMed/NCBI | |
Zhong Z, Jiao Z and Yu FX: The Hippo signaling pathway in development and regeneration. Cell Rep. 43:1139262024. View Article : Google Scholar : PubMed/NCBI | |
Morciano G, Vezzani B, Missiroli S, Boncompagni C, Pinton P and Giorgi C: An updated understanding of the role of YAP in driving oncogenic responses. Cancers (Basel). 13:31002021. View Article : Google Scholar : PubMed/NCBI | |
Abylkassov R and Xie Y: Role of Yes-associated protein in cancer: An update. Oncol Lett. 12:2277–2282. 2016. View Article : Google Scholar : PubMed/NCBI | |
Galsky MD, Stensland KD, Moshier E, Sfakianos JP, McBride RB, Tsao CK, Casey M, Boffetta P, Oh WK, Mazumdar M and Wisnivesky JP: Effectiveness of adjuvant chemotherapy for locally advanced bladder cancer. J Clin Oncol. 34:825–832. 2016. View Article : Google Scholar : PubMed/NCBI | |
Luo J, Deng L, Zou H, Guo Y, Tong T, Huang M, Ling G and Li P: New insights into the ambivalent role of YAP/TAZ in human cancers. J Exp Clin Cancer Res. 42:1302023. View Article : Google Scholar : PubMed/NCBI | |
Fu M, Hu Y, Lan T, Guan KL, Luo T and Luo M: The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther. 7:3762022. View Article : Google Scholar : PubMed/NCBI | |
Zhao X, Fu J, Hu B, Chen L, Wang J, Fang J, Ge C, Lin H, Pan K, Fu L, et al: Serine metabolism regulates YAP activity through USP7 in colon cancer. Front Cell Dev Biol. 9:6391112021. View Article : Google Scholar : PubMed/NCBI | |
Zhu H, Yan F, Yuan T, Qian M, Zhou T, Dai X, Cao J, Ying M, Dong X, He Q and Yang B: USP10 promotes proliferation of hepatocellular carcinoma by deubiquitinating and stabilizing YAP/TAZ. Cancer Res. 80:2204–2216. 2020. View Article : Google Scholar : PubMed/NCBI | |
Eibl G and Rozengurt E: KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol. 54:50–62. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lee JY, Chang JK, Dominguez AA, Lee HP, Nam S, Chang J, Varma S, Qi LS, West RB and Chaudhuri O: YAP-independent mechanotransduction drives breast cancer progression. Nat Commun. 10:18482019. View Article : Google Scholar : PubMed/NCBI | |
Driskill JH and Pan D: The Hippo pathway in liver homeostasis and pathophysiology. Annu Rev Pathol. 16:299–322. 2021. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI | |
Antoni S, Ferlay J, Soerjomataram I, Znaor A, Jemal A and Bray F: Bladder cancer incidence and mortality: A global overview and recent trends. Eur Urol. 71:96–108. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lenis AT, Lec PM, Chamie K and Mshs MD: Bladder cancer: A review. JAMA. 324:1980–1991. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liu JY, Li YH, Lin HX, Liao YJ, Mai SJ, Liu ZW, Zhang ZL, Jiang LJ, Zhang JX, Kung HF, et al: Overexpression of YAP 1 contributes to progressive features and poor prognosis of human urothelial carcinoma of the bladder. BMC Cancer. 13:3492013. View Article : Google Scholar : PubMed/NCBI | |
Huang Z, Wang X, Ma L, Guo Z, Liu H, Du M, Chu H, Wang M, Wang Z and Zhang Z: Genetic variations in Hippo pathway genes influence bladder cancer risk in a Chinese population. Arch Toxicol. 94:785–794. 2020. View Article : Google Scholar : PubMed/NCBI | |
Di X, Xiang L and Jian Z: YAP-mediated mechanotransduction in urinary bladder remodeling: Based on RNA-seq and CUT&Tag. Front Genet. 14:11069272023. View Article : Google Scholar : PubMed/NCBI | |
Tong T, Huang M, Yan B, Lin B, Yu J, Teng Q, Li P and Pang J: Hippo signaling modulation and its biological implications in urological malignancies. Mol Aspects Med. 98:1012802024. View Article : Google Scholar : PubMed/NCBI | |
Cucci MA, Compagnone A, Daga M, Grattarola M, Ullio C, Roetto A, Palmieri A, Rosa AC, Argenziano M, Cavalli R, et al: Post-translational inhibition of YAP oncogene expression by 4-hydroxynonenal in bladder cancer cells. Free Radic Biol Med. 141:205–219. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang Q and Jiang J: Regulation of hedgehog signal transduction by ubiquitination and deubiquitination. Int J Mol Sci. 22:133382021. View Article : Google Scholar : PubMed/NCBI | |
Luo Y, Zhou J, Tang J, Zhou F, He Z and Liu T and Liu T: MINDY1 promotes bladder cancer progression by stabilizing YAP. Cancer Cell Int. 21:3952021. View Article : Google Scholar : PubMed/NCBI | |
Dong L, Lin F, Wu W, Huang W and Cai Z: Transcriptional cofactor Mask2 is required for YAP-induced cell growth and migration in bladder cancer cell. J Cancer. 7:2132–2138. 2016. View Article : Google Scholar : PubMed/NCBI | |
Rovida E and Tusa I: Targeting MAPK in cancer 2.0. Int J Mol Sci. 23:57022022. View Article : Google Scholar : PubMed/NCBI | |
Qiu D, Zhu Y and Cong Z: YAP triggers bladder cancer proliferation by affecting the MAPK pathway. Cancer Manag Res. 12:12205–12214. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang J, Wang H, Zhang Y, Zhen N, Zhang L, Qiao Y, Weng W, Liu X, Ma L, Xiao W, et al: Mutual inhibition between YAP and SRSF1 maintains long non-coding RNA, Malat1-induced tumourigenesis in liver cancer. Cell Signal. 26:1048–1059. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li S, Yu Z, Chen SS, Li F, Lei CY, Chen XX, Bao JM, Luo Y, Lin GZ, Pang SY and Tan WL: The YAP1 oncogene contributes to bladder cancer cell proliferation and migration by regulating the H19 long noncoding RNA. Urol Oncol. 33:427.e1–e10. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lv M, Zhong Z, Huang M, Tian Q, Jiang R and Chen J: lncRNA H19 regulates epithelial-mesenchymal transition and metastasis of bladder cancer by miR-29b-3p as competing endogenous RNA. Biochim Biophys Acta Mol Cell Res. 1864:1887–1899. 2017. View Article : Google Scholar : PubMed/NCBI | |
Jafri I: MiRNA a new insight in metabolic and human diseases: A review. Cell Mol Biol (Noisy-le-grand). 69:102–110. 2023. View Article : Google Scholar : PubMed/NCBI | |
Luo K: Signaling cross talk between TGF-β/Smad and other signaling pathways. Cold Spring Harb Perspect Biol. 9:a0221372017. View Article : Google Scholar : PubMed/NCBI | |
Zhuang C, Liu Y, Fu S, Yuan C, Luo J, Huang X, Yang W, Xie W and Zhuang C: Silencing of lncRNA MIR497HG via CRISPR/Cas13d induces bladder cancer progression through promoting the crosstalk between Hippo/Yap and TGF-β/Smad signaling. Front Mol Biosci. 7:6167682020. View Article : Google Scholar : PubMed/NCBI | |
Zhou X, Zhang P, Han H, Lei H and Zhang X: Hypermethylated in cancer 1 (HIC1) suppresses bladder cancer progression by targeting yes-associated protein (YAP) pathway. J Cell Biochem. 120:6471–6481. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhang J and Deng X: Effects of miR-599 targeting YAP1 on proliferation, invasion and apoptosis of bladder urothelial carcinoma cells. Exp Mol Pathol. 118:1045992021. View Article : Google Scholar : PubMed/NCBI | |
Hade MD, Suire CN and Suo Z: Mesenchymal stem cell-derived exosomes: Applications in regenerative medicine. Cells. 10:19592021. View Article : Google Scholar : PubMed/NCBI | |
Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020. View Article : Google Scholar : PubMed/NCBI | |
Huang ZM, Wang H and Ji ZG: Bladder mesenchymal stromal cell-derived exosomal miRNA-217 modulates bladder cancer cell survival through Hippo-YAP pathway. Inflamm Res. 70:959–969. 2021. View Article : Google Scholar : PubMed/NCBI | |
Schmitz-Dräger BJ, Droller M, Lokeshwar VB, Lotan Y, Hudson MA, van Rhijn BW, Marberger MJ, Fradet Y, Hemstreet GP, Malmstrom PU, et al: Molecular markers for bladder cancer screening, early diagnosis, and surveillance: The WHO/ICUD consensus. Urol Int. 94:1–24. 2015. View Article : Google Scholar : PubMed/NCBI | |
Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li S, Zhu H, Chen H, Xia J, Zhang F, Xu R and Lin Q: Glucose promotes epithelial-mesenchymal transitions in bladder cancer by regulating the functions of YAP1 and TAZ. J Cell Mol Med. 24:10391–10401. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zakrzewski W, Dobrzyński M, Szymonowicz M and Rybak Z: Stem cells: Past, present, and future. Stem Cell Res Ther. 10:682019. View Article : Google Scholar : PubMed/NCBI | |
Fuchs E and Blau HM: Tissue stem cells: Architects of their niches. Cell Stem Cell. 27:532–556. 2020. View Article : Google Scholar : PubMed/NCBI | |
Huang T, Song X, Xu D, Tiek D, Goenka A, Wu B, Sastry N, Hu B and Cheng SY: Stem cell programs in cancer initiation, progression, and therapy resistance. Theranostics. 10:8721–8743. 2020. View Article : Google Scholar : PubMed/NCBI | |
Nassar D and Blanpain C: Cancer stem cells: Basic concepts and therapeutic implications. Annu Rev Pathol. 11:47–76. 2016. View Article : Google Scholar : PubMed/NCBI | |
Driskill JH and Pan D: Control of stem cell renewal and fate by YAP and TAZ. Nat Rev Mol Cell Biol. 24:895–911. 2023. View Article : Google Scholar : PubMed/NCBI | |
Luo J and Li P: Context-dependent transcriptional regulations of YAP/TAZ in stem cell and differentiation. Stem Cell Res Ther. 13:102022. View Article : Google Scholar : PubMed/NCBI | |
Damkham N, Issaragrisil S and Lorthongpanich C: Role of YAP as a mechanosensing molecule in stem cells and stem cell-derived hematopoietic cells. Int J Mol Sci. 23:146342022. View Article : Google Scholar : PubMed/NCBI | |
Zhao AY, Dai YJ, Lian JF, Huang Y, Lin JG, Dai YB and Xu TW: YAP regulates ALDH1A1 expression and stem cell property of bladder cancer cells. Onco Targets Ther. 11:6657–6663. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ooki A, Del Carmen Rodriguez Pena M, Marchionni L, Dinalankara W, Begum A, Hahn NM, VandenBussche CJ, Rasheed ZA, Mao S, Netto GJ, et al: YAP1 and COX2 coordinately regulate urothelial cancer stem-like cells. Cancer Res. 78:168–181. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wang T, Yang Y, Wang Z, Zhang X, Li D and Wei J: A SNP of miR-146a is involved in bladder cancer relapse by affecting the function of bladder cancer stem cells via the miR-146a signallings. J Cell Mol Med. 24:8545–8556. 2020. View Article : Google Scholar : PubMed/NCBI | |
Ghasemi H, Mousavibahar SH, Hashemnia M, Karimi J, Khodadadi I, Mirzaei F and Tavilani H: Tissue stiffness contributes to YAP activation in bladder cancer patients undergoing transurethral resection. Ann N Y Acad Sci. 1473:48–61. 2020. View Article : Google Scholar : PubMed/NCBI | |
Gwak HS, Youn SM, Kwon AH, Lee SH, Kim JH and Rhee CH: ACNU-cisplatin continuous infusion chemotherapy as salvage therapy for recurrent glioblastomas: Phase II study. J Neurooncol. 75:173–180. 2005. View Article : Google Scholar : PubMed/NCBI | |
Gentilin E: New advancements in cisplatin-based treatments. Int J Mol Sci. 24:59202023. View Article : Google Scholar : PubMed/NCBI | |
Gómez-Ruiz S, Maksimović-Ivanić D, Mijatović S and Kaluđerović GN: On the discovery, biological effects, and use of Cisplatin and metallocenes in anticancer chemotherapy. Bioinorg Chem Appl. 2012:1402842012. View Article : Google Scholar : PubMed/NCBI | |
Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M and Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 31:1869–1883. 2012. View Article : Google Scholar : PubMed/NCBI | |
Wenmaekers S, Viergever BJ, Kumar G, Kranenburg O, Black PC, Daugaard M and Meijer RP: A potential role for HUWE1 in modulating cisplatin sensitivity. Cells. 10:12622021. View Article : Google Scholar : PubMed/NCBI | |
Rebucci M and Michiels C: Molecular aspects of cancer cell resistance to chemotherapy. Biochem Pharmacol. 85:1219–1226. 2013. View Article : Google Scholar : PubMed/NCBI | |
Nguyen CDK and Yi C: YAP/TAZ Signaling and resistance to cancer therapy. Trends Cancer. 5:283–296. 2019. View Article : Google Scholar : PubMed/NCBI | |
Kim MH and Kim J: Role of YAP/TAZ transcriptional regulators in resistance toanti-cancer therapies. Cell Mol Life Sci. 74:1457–1474. 2017. View Article : Google Scholar : PubMed/NCBI | |
Fan Q, Cai Q and Xu Y: FOXM1 is a downstream target of LPA and YAP oncogenic signaling pathways in high grade serous ovarian cancer. Oncotarget. 6:27688–27699. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ciamporcero E, Daga M, Pizzimenti S, Roetto A, Dianzani C, Compagnone A, Palmieri A, Ullio C, Cangemi L, Pili R and Barrera G: Crosstalk between Nrf2 and YAP contributes to maintaining the antioxidant potential and chemoresistance in bladder cancer. Free Radic Biol Med. 115:447–457. 2018. View Article : Google Scholar : PubMed/NCBI | |
Hayden A, Douglas J, Sommerlad M, Andrews L, Gould K, Hussain S, Thomas GJ, Packham G and Crabb SJ: The Nrf2 transcription factor contributes to resistance to cisplatin in bladder cancer. Urol Oncol. 32:806–814. 2014. View Article : Google Scholar : PubMed/NCBI | |
Daga M, Pizzimenti S, Dianzani C, Cucci MA, Cavalli R, Grattarola M, Ferrara B, Scariot V, Trotta F and Barrera G: Ailanthone inhibits cell growth and migration of cisplatin resistant bladder cancer cells through down-regulation of Nrf2, YAP, and c-Myc expression. Phytomedicine. 56:156–164. 2019. View Article : Google Scholar : PubMed/NCBI | |
Cucci MA, Grattarola M, Dianzani C, Damia G, Ricci F, Roetto A, Trotta F, Barrera G and Pizzimenti S: Ailanthone increases oxidative stress in CDDP-resistant ovarian and bladder cancer cells by inhibiting of Nrf2 and YAP expression through a post-translational mechanism. Free Radic Biol Med. 150:125–135. 2020. View Article : Google Scholar : PubMed/NCBI | |
Dhillon S: Decitabine/cedazuridine: First approval. Drugs. 80:1373–1378. 2020. View Article : Google Scholar : PubMed/NCBI | |
Khandelwal M, Anand V, Appunni S, Seth A, Singh P, Mathur S and Sharma A: Decitabine augments cytotoxicity of cisplatin and doxorubicin to bladder cancer cells by activating hippo pathway through RASSF1A. Mol Cell Biochem. 446:105–114. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ciamporcero E, Shen H, Ramakrishnan S, Yu Ku S, Chintala S, Shen L, Adelaiye R, Miles KM, Ullio C, Pizzimenti S, et al: YAP activation protects urothelial cell carcinoma from treatment-induced DNA damage. Oncogene. 35:1541–1553. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hong WX, Haebe S, Lee AS, Westphalen CB, Norton JA, Jiang W and Levy R: Intratumoral immunotherapy for early-stage solid tumors. Clin Cancer Res. 26:3091–3099. 2020. View Article : Google Scholar : PubMed/NCBI | |
Demaria O, Gauthier L, Debroas G and Vivier E: Natural killer cell engagers in cancer immunotherapy: Next generation of immuno-oncology treatments. Eur J Immunol. 51:1934–1942. 2021. View Article : Google Scholar : PubMed/NCBI | |
Rui R, Zhou L and He S: Cancer immunotherapies: Advances and bottlenecks. Front Immunol. 14:12124762023. View Article : Google Scholar : PubMed/NCBI | |
Han J, Gu X, Li Y and Wu Q: Mechanisms of BCG in the treatment of bladder cancer-current understanding and the prospect. Biomed Pharmacother. 129:1103932020. View Article : Google Scholar : PubMed/NCBI | |
Donin NM, Lenis AT, Holden S, Drakaki A, Pantuck A, Belldegrun A and Chamie K: Immunotherapy for the treatment of urothelial carcinoma. J Urol. 197:14–22. 2017. View Article : Google Scholar : PubMed/NCBI | |
Gill J and Prasad V: Pembrolizumab for non-muscle-invasive bladder cancer-a costly therapy in search of evidence. JAMA Oncol. 7:501–502. 2021. View Article : Google Scholar : PubMed/NCBI | |
Song D, Powles T, Shi L, Zhang L, Ingersoll MA and Lu YJ: Bladder cancer, a unique model to understand cancer immunity and develop immunotherapy approaches. J Pathol. 249:151–165. 2019. View Article : Google Scholar : PubMed/NCBI | |
Baek SW, Jang IH, Kim SK, Nam JK, Leem SH and Chu IS: Transcriptional profiling of advanced urothelial cancer predicts prognosis and response to immunotherapy. Int J Mol Sci. 21:18502020. View Article : Google Scholar : PubMed/NCBI | |
Baek SW, Mun JY, Jang IH, Yang GE, Jeong MS, Kim SK, Nam JK, Chu IS and Leem SH: YAP1 activation is associated with the progression and response to immunotherapy of non-muscle invasive bladder cancer. EBioMedicine. 81:1040922022. View Article : Google Scholar : PubMed/NCBI | |
Grossman H, Natale RB, Tangen CM, Speights VO, Vogelzang NJ, Trump DL, deVere White RW, Sarosdy MF, Wood DP Jr, Raghavan D and Crawford ED: Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med. 349:859–866. 2003. View Article : Google Scholar : PubMed/NCBI | |
Joo WD, Visintin I and Mor G: Targeted cancer therapy-are the days of systemic chemotherapy numbered? Maturitas. 76:308–314. 2013. View Article : Google Scholar : PubMed/NCBI | |
Pérez-Herrero E and Fernández-Medarde A: Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm. 93:52–79. 2015. View Article : Google Scholar : PubMed/NCBI | |
Rouprêt M, Seisen T, Birtle AJ, Capoun O, Compérat EM, Dominguez-Escrig JL, Gürses Andersson I, Liedberg F, Mariappan P, Hugh Mostafid A, et al: European association of urology guidelines on upper urinary tract urothelial carcinoma: 2023 Update. Eur Urol. 84:49–64. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yu EY, Petrylak DP, O'Donnell PH, Lee JL, van der Heijden MS, Loriot Y, Stein MN, Necchi A, Kojima T, Harrison MR, et al: Enfortumab vedotin after PD-1 or PD-L1 inhibitors in cisplatin-ineligible patients with advanced urothelial carcinoma (EV-201): A multicentre, single-arm, phase 2 trial. Lancet Oncol. 22:872–882. 2021. View Article : Google Scholar : PubMed/NCBI | |
Dong L, Lin F, Wu W, Liu Y and Huang W: Verteporfin inhibits YAP-induced bladder cancer cell growth and invasion via Hippo signaling pathway. Int J Med Sci. 15:645–652. 2018. View Article : Google Scholar : PubMed/NCBI | |
Elbadawy M, Sato Y, Mori T, Goto Y, Hayashi K, Yamanaka M, Azakami D, Uchide T, Fukushima R, Yoshida T, et al: Anti-tumor effect of trametinib in bladder cancer organoid and the underlying mechanism. Cancer Biol Ther. 22:357–371. 2021. View Article : Google Scholar : PubMed/NCBI | |
Chen H, Yang W, Li Y and Ji Z: PLAGL2 promotes bladder cancer progression via RACGAP1/RhoA GTPase/YAP1 signaling. Cell Death Dis. 14:4332023. View Article : Google Scholar : PubMed/NCBI | |
Sudol M: YAP1 oncogene and its eight isoforms. Oncogene. 32:39222013. View Article : Google Scholar : PubMed/NCBI | |
Fang L, Teng H, Wang Y, Liao G, Weng L, Li Y, Wang X, Jin J, Jiao C, Chen L, et al: SET1A-mediated mono-methylation at K342 regulates YAP activation by blocking its nuclear export and promotes tumorigenesis. Cancer Cell. 34:103–118.e9. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mao B, Hu F, Cheng J, Wang P, Xu M, Yuan F, Meng S, Wang Y, Yuan Z and Bi W: SIRT1 regulates YAP2-mediated cell proliferation and chemoresistance in hepatocellular carcinoma. Oncogene. 33:1468–1474. 2014. View Article : Google Scholar : PubMed/NCBI | |
Yan F, Qian M, He Q, Zhu H and Yang B: The posttranslational modifications of Hippo-YAP pathway in cancer. Biochim Biophys Acta Gen Subj. 1864:1293972020. View Article : Google Scholar : PubMed/NCBI | |
Wu L, Ou Z, Liu P, Zhao C, Tong S, Wang R, Li Y, Yuan J, Chen M, Fan B, et al: ATXN3 promotes prostate cancer progression by stabilizing YAP. Cell Commun Signal. 21:1522023. View Article : Google Scholar : PubMed/NCBI | |
Xu TH, Sheng Z, Li Y, Qiu X, Tian B and Yao L: OGT knockdown counteracts high phosphate-induced vascular calcification in chronic kidney disease through autophagy activation by downregulating YAP. Life Sci. 261:1181212020. View Article : Google Scholar : PubMed/NCBI | |
Santinon G, Pocaterra A and Dupont S: Control of YAP/TAZ activity by metabolic and nutrient-sensing pathways. Trends Cell Biol. 26:289–299. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang R, Yang S, Wang M, Zhou Y, Li X, Chen W, Liu W, Huang Y, Wu J, Cao J, et al: A sustainable approach to universal metabolic cancer diagnosis. Nat Sustain. 7:602–615. 2024. View Article : Google Scholar | |
Kashihara T, Mukai R, Oka SI, Zhai P, Nakada Y, Yang Z, Mizushima W, Nakahara T, Warren JS, Abdellatif M and Sadoshima J: YAP mediates compensatory cardiac hypertrophy through aerobic glycolysis in response to pressure overload. J Clin Invest. 132:e1505952022. View Article : Google Scholar : PubMed/NCBI | |
Li X, Wu Z, He J, Jin Y, Chu C, Cao Y, Gu F, Wang H, Hou C, Liu X and Zou Q: OGT regulated O-GlcNAcylation promotes papillary thyroid cancer malignancy via activating YAP. Oncogene. 40:4859–4871. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhang X, Qiao Y, Wu Q, Chen Y, Zou S, Liu X, Zhu G, Zhao Y, Chen Y, Yu Y, et al: The essential role of YAP O-GlcNAcylation in high-glucose-stimulated liver tumorigenesis. Nat Commun. 8:152802017. View Article : Google Scholar : PubMed/NCBI | |
Pei C, Wang Y, Ding Y, Li R, Shu W, Zeng Y, Yin X and Wan J: Designed concave octahedron heterostructures decode distinct metabolic patterns of epithelial ovarian tumors. Adv Mater. 35:22090832023. View Article : Google Scholar | |
Sun T and Chi JT: Regulation of ferroptosis in cancer cells by YAP/TAZ and Hippo pathways: The therapeutic implications. Genes Dis. 8:241–249. 2021. View Article : Google Scholar : PubMed/NCBI | |
Wei C and Fu Q: Cell death mediated by nanotechnology via the cuproptosis pathway: A novel horizon for cancer therapy. VIEW. 4:202300012023. View Article : Google Scholar |