1
|
Uttagomol J, Wongviriya A, Chantaramanee A, Prasitsak T. YAP Expression is Related to the Aggressive Behavior of Odontogenic Cysts. Eur J Dent 2025. [PMID: 40311635 DOI: 10.1055/s-0044-1801275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025] Open
Abstract
OBJECTIVE Yes-associated protein (YAP) and Ki-67 are known to be involved in cell proliferation. While their overexpression is associated with aggressive behaviors in oral squamous cell carcinoma (OSCC), their roles in odontogenic cysts have not been thoroughly investigated. Therefore, this study aimed to evaluate the immunohistochemical expression of YAP and Ki-67 in odontogenic cysts. MATERIAL AND METHODS Tissue samples included 12 radicular cysts, 10 dentigerous cysts, 9 odontogenic keratocysts, and 9 normal oral mucosa specimens. Immunohistochemical analysis was conducted both manually and using ImageJ software for comparison. STATISTICAL ANALYSIS The Kruskal-Wallis, followed by the Mann-Whitney U-test was used to assess the differences in the YAP and Ki-67 immunoexpression levels among various group lesions. The correlation between those two proteins in individual groups was tested by Spearman correlation. A p-value of less than 0.05 was considered statistically significant. SPSS software version 25.0 was used. RESULTS There was a strong nuclear staining of YAP in basal and superficial cells and an intense cytoplasmic YAP positivity in odontogenic keratocysts, whereas a weaker YAP staining in both the nucleus and cytoplasm throughout the epithelial thickness was observed in dentigerous cyst, radicular cyst, and normal oral mucosa, respectively. YAP expression differed significantly in all odontogenic cysts compared with normal oral mucosa (p < 0.05). Ki-67 expression was notably higher in odontogenic keratocysts relative to other cysts and normal oral mucosa. Although no statistically significant correlation was found between YAP and Ki-67 across the groups, both proteins displayed similar positive trends in odontogenic keratocysts. CONCLUSION These findings suggest that YAP activation may be related to the proliferative behavior of odontogenic cysts, especially in more aggressive lesions, but less likely to influence the inflammatory cysts. This insight could improve understanding of their pathogenesis and pave the way for new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Jutamas Uttagomol
- Department of Oral Diagnosis, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Adjabhak Wongviriya
- Department of Oral Diagnosis, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Ariya Chantaramanee
- Department of Preventive Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| | - Thanit Prasitsak
- Department of Oral Biology, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
2
|
Sun Y, Wei H, Yu W, Gao H, Li J, Li X, Zhang H, Zhang H, Miao S, Zhao L, Yang R, Xu J, Lu Y, Wei F, Zhou H, Gao D, Jin Y, Zhang L. The actin-binding protein drebrin disrupts NF2-LATS kinases complex assembly to facilitate liver tumorigenesis. Hepatology 2025; 81:1433-1451. [PMID: 39325963 DOI: 10.1097/hep.0000000000001063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/17/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS The Hippo signaling has emerged as a crucial regulator of tissue homeostasis, regeneration, and tumorigenesis, representing a promising therapeutic target. Neurofibromin 2 (NF2), a component of Hippo signaling, is directly linked to human cancers but has been overlooked as a target for cancer therapy. APPROACH AND RESULTS Through a high-content RNA interference genome-wide screen, the actin-binding protein Drebrin (DBN1) has been identified as a novel modulator of YAP localization. Further investigations have revealed that DBN1 directly interacts with NF2, disrupting the activation of large tumor suppressor kinases (LATS1/2) by competing with LATS kinases for NF2 binding. Consequently, DBN1 knockout considerably promotes YAP nuclear exclusion and repression of target gene expression, thereby preventing cell proliferation and liver tumorigenesis. We identified three lysine residues (K238, K248, and K252) essential for DBN1-NF2 interaction and developed a mutant DBN1 (DBN1-3K mut ) that is defective in NF2 binding and incompetent to trigger NF2-dependent YAP activation and tumorigenesis both in vitro and in vivo. Furthermore, BTP2, a DBN1 inhibitor, successfully restored NF2-LATS kinase binding and elicited potent antitumor activity. The combination of sorafenib and BTP2 exerted synergistic inhibitory effects against HCC. CONCLUSIONS Our study identifies a novel DBN1-NF2-LATS axis, and pharmacological inhibition of DBN1 represents a promising alternative intervention targeting the Hippo pathway in cancer treatment.
Collapse
Affiliation(s)
- Yang Sun
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Henan Wei
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Wentao Yu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haoran Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jinhui Li
- HuidaGene Therapeutics Co., Ltd., Shanghai, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Haijiao Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Haoen Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Sen Miao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Lihua Zhao
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Ruizeng Yang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Jinjin Xu
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Fang Wei
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Department of Analytical Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Daming Gao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yunyun Jin
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- Department of Emergency and Critical Care Medicine, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Lei Zhang
- Sheng Yushou Center of Cell Biology and Immunology, Department of Genetics and Developmental Science, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Minghang, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
3
|
Noser AA, Salem MM, ElSafty EM, Baren MH, Selim AI, Mandour HSA. Synthesis, molecular docking, and biological investigations of new pyrazolone chalcones. RSC Adv 2025; 15:13214-13224. [PMID: 40290744 PMCID: PMC12023741 DOI: 10.1039/d5ra01233c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Heterocyclic compounds are essential to the drug development and discovery processes. Herein, we synthesized new pyrazolone chalcones (3a-g) through the reaction of azopyrazolone (2) with different aromatic aldehydes in a basic medium. Numerous techniques including elemental analysis, 1H-NMR, 13C-NMR, and FT-IR spectroscopies, were used to characterize pyrazolone chalcone derivatives. Compound 3b exhibited the highest binding energy towards YAP/TEAD protein with a value of -8.45 kcal mol-1 in in silico studies. This observation suggested that compound 3b inhibits the YAP/TEAD Hippo signaling pathway. In addition, compound 3b offered a prospective anticancer effect against various cancer cell lines, such as HepG-2, MCF-7, and HCT-116, among the other synthesized compounds, with IC50 values equal to 5.03 ± 0.4, 3.92 ± 0.2, and 6.34 ± 0.5 μM, respectively. These results validated our findings regarding the in silico suppression of the YAP/TEAD protein. Its pharmacokinetic properties were theoretically observed using ADMET. Additionally, compound 3b demonstrated a potent antioxidant scavenging action (in vitro) against DPPH free radicals. Thus, based on our findings, compound 3b may act as a potential anticancer scaffold owing to its inhibitory impact towards the YAP/TEAD-mediated Hippo signaling pathway with a safe toxic profile on normal cells.
Collapse
Affiliation(s)
- Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Esraa M ElSafty
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Mohamed H Baren
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Adel I Selim
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| | - Hamada S A Mandour
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University Tanta 31527 Egypt
| |
Collapse
|
4
|
Dalal K, McAnany C, Weilert M, McKinney MC, Krueger S, Zeitlinger J. Interpreting regulatory mechanisms of Hippo signaling through a deep learning sequence model. CELL GENOMICS 2025; 5:100821. [PMID: 40174587 PMCID: PMC12008814 DOI: 10.1016/j.xgen.2025.100821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/23/2024] [Accepted: 03/05/2025] [Indexed: 04/04/2025]
Abstract
Signaling pathway components are well studied, but how they mediate cell-type-specific transcription responses is an unresolved problem. Using the Hippo pathway in mouse trophoblast stem cells as a model, we show that the DNA binding of signaling effectors is driven by cell-type-specific sequence rules that can be learned genome wide by deep learning models. Through model interpretation and experimental validation, we show that motifs for the cell-type-specific transcription factor TFAP2C enhance TEAD4/YAP1 binding in a nucleosome-range and distance-dependent manner, driving synergistic enhancer activation. We also discovered that Tead double motifs are widespread, highly active canonical response elements. Molecular dynamics simulations suggest that TEAD4 binds them cooperatively through surprisingly labile protein-protein interactions that depend on the DNA template. These results show that the response to signaling pathways is encoded in the cis-regulatory sequences and that interpreting the rules reveals insights into the mechanisms by which signaling effectors influence cell-type-specific enhancer activity.
Collapse
Affiliation(s)
- Khyati Dalal
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Charles McAnany
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Melanie Weilert
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | | | - Sabrina Krueger
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Julia Zeitlinger
- Stowers Institute for Medical Research, Kansas City, MO, USA; Department of Pathology & Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
5
|
Guo P, Song S, Niu Y, Kuang X, Zhou D, Zhou Z, Zhang Y, Ma X. Alternative splicing of bunched confers a dual role in hippo pathway-dependent growth and tumorigenesis. Oncogene 2025:10.1038/s41388-025-03348-6. [PMID: 40175650 DOI: 10.1038/s41388-025-03348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 04/04/2025]
Abstract
Alternative splicing is a fundamental mechanism that generates functionally distinct proteins from individual genes, contributing to gene regulation and proteomic diversity. In Drosophila, the bunched (bun) gene, a member of the TSC-22 domain gene family, undergoes alternative splicing, yielding diverse protein isoforms involved in crucial biological processes. Nevertheless, the specific roles and regulatory mechanisms of each isoform remain elusive. Here, we employed CRISPR/Cas9 technology to introduce targeted deletions within the endogenous locus of the bun gene, resulting in the removal of either long or short isoforms. We discovered that the short isoforms demonstrated a growth-suppressive role, whereas the long isoforms exhibited a growth-promoting effect. Surprisingly, the long isoforms exhibited a remarkable dual functionality, as both deletion and amplification of long isoform expression impede the excess growth induced by Hippo pathway inactivation. Mechanistically, ectopically expressed Bun long isoforms act as the transcriptional suppressor by competitively binding to targets' promoter regions in conjunction with Yorkie/Scalloped (Yki/Sd), thereby inhibiting its transcriptional outputs and ultimately leading to the growth suppression. These findings unveil the intricate interaction between distinct spliced isoforms of Bun and oncogenic outcomes, highlighting Bun long isoforms as the critical transcription suppressor regulating Hippo pathway inactivation-mediated growth and tumorigenesis in Drosophila.
Collapse
Affiliation(s)
- Pengjuan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Yuxiao Niu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Xiaoyu Kuang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Dafa Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Zizhang Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an, Shandong, 271018, China
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
| |
Collapse
|
6
|
Li X, Cho YS, Han Y, Zhou M, Liu Y, Yang Y, Zhuo S, Jiang J. The Hippo pathway effector YAP inhibits NF-κB signaling and ccRCC growth by opposing ZHX2. J Biol Chem 2025; 301:108430. [PMID: 40120683 PMCID: PMC12018991 DOI: 10.1016/j.jbc.2025.108430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/07/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
The prevailing view in the cancer field is that Hippo (Hpo) signaling pathway functions as a tumor suppressor pathway by blocking the oncogenic potential of the pathway effectors Yes1-associated transcriptional regulator (YAP)/transcriptional coactivator with PDZ-binding motif. However, YAP can also function as a context-dependent tumor suppressor in several types of cancer including clear cell renal cell carcinomas (ccRCCs). We find that, in addition to inhibiting hypoxia-inducible factor 2α, a major oncogenic driver in Von Hippel-Lindau-/- ccRCC, YAP also blocks nuclear factor κB (NF-κB) signaling in ccRCC to inhibit cancer cell growth under conditions where hypoxia-inducible factor 2α is dispensable. Mechanistically, YAP inhibits the expression of Zinc fingers and homeoboxes 2 (ZHX2), a Von Hippel-Lindau substrate and critical cofactor of NF-κB in ccRCC. Furthermore, YAP competes with ZHX2 for binding to the NF-κB subunit p65. Consequently, elevated nuclear YAP blocks the cooperativity between ZHX2 and the NF-κB subunit p65, leading to diminished NF-κB target gene expression. Pharmacological inhibition of Hpo kinase blocked NF-κB transcriptional program and suppressed ccRCC cell growth, which can be rescued by overexpression of ZHX2 or p65. Our study uncovers a crosstalk between the Hpo and NF-κB/ZHX2 pathways and its involvement in ccRCC growth inhibition, suggesting that targeting the Hpo pathway may provide a therapeutical opportunity for ccRCC treatment.
Collapse
Affiliation(s)
- Xu Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Math and Sciences, Tarrant County College-NE Campus, Hurst, Texas, USA
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mengmeng Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Boston, Massachusetts, USA; Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, USA; Harvard Stem Cell Institute, Boston, Massachusetts, USA; Dana-Farber/Harvard Cancer Center, Boston, Massachusetts, USA
| | - Shu Zhuo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
7
|
Guo P, Wan S, Guan KL. The Hippo pathway: Organ size control and beyond. Pharmacol Rev 2025; 77:100031. [PMID: 40148032 DOI: 10.1016/j.pharmr.2024.100031] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
The Hippo signaling pathway is a highly conserved signaling network for controlling organ size, tissue homeostasis, and regeneration. It integrates a wide range of intracellular and extracellular signals, such as cellular energy status, cell density, hormonal signals, and mechanical cues, to modulate the activity of YAP/TAZ transcriptional coactivators. A key aspect of Hippo pathway regulation involves its spatial organization at the plasma membrane, where upstream regulators localize to specific membrane subdomains to regulate the assembly and activation of the pathway components. This spatial organization is critical for the precise control of Hippo signaling, as it dictates the dynamic interactions between pathway components and their regulators. Recent studies have also uncovered the role of biomolecular condensation in regulating Hippo signaling, adding complexity to its control mechanisms. Dysregulation of the Hippo pathway is implicated in various pathological conditions, particularly cancer, where alterations in YAP/TAZ activity contribute to tumorigenesis and drug resistance. Therapeutic strategies targeting the Hippo pathway have shown promise in both cancer treatment, by inhibiting YAP/TAZ signaling, and regenerative medicine, by enhancing YAP/TAZ activity to promote tissue repair. The development of small molecule inhibitors targeting the YAP-TEAD interaction and other upstream regulators offers new avenues for therapeutic intervention. SIGNIFICANCE STATEMENT: The Hippo signaling pathway is a key regulator of organ size, tissue homeostasis, and regeneration, with its dysregulation linked to diseases such as cancer. Understanding this pathway opens new possibilities for therapeutic approaches in regenerative medicine and oncology, with the potential to translate basic research into improved clinical outcomes.
Collapse
Affiliation(s)
- Pengfei Guo
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| | - Sicheng Wan
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Kun-Liang Guan
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
8
|
Taroc EZM, Amato E, Semon A, Dolphin N, Beck B, Belin S, Poitelon Y, Forni PE. Shared Lineage, Distinct Outcomes: Yap and Taz Loss Differentially Impact Schwann and Olfactory Ensheathing Cell Development Without Disrupting GnRH-1 Migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638196. [PMID: 40027653 PMCID: PMC11870449 DOI: 10.1101/2025.02.13.638196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Olfactory Ensheathing Cells (OECs) are glial cells originating from the neural crest, critical for bundling olfactory axons to the brain. Their development is crucial for the migration of Gonadotropin-Releasing Hormone-1 (GnRH-1) neurons, which are essential for puberty and fertility. OECs have garnered interest as potential therapeutic targets for central nervous system lesions, although their development is not fully understood. Our single-cell RNA sequencing of mouse embryonic nasal tissues suggests that OECs and Schwann cells share a common origin from Schwann cell precursors yet exhibit significant genetic differences. The transcription factors Yap and Taz have previously been shown to play a crucial role in Schwann cell development. We used Sox10 -Cre mice to conditionally ablate Yap and Taz in migrating the neural crest and its derivatives. Our analyses showed reduced Sox10+ glial cells along nerves in the nasal region, altered gene expression of SCs, melanocytes, and OECs, and a significant reduction in olfactory sensory neurons and vascularization in the vomeronasal organ. However, despite these changes, GnRH-1 neuronal migration remained unaffected. Our findings highlight the importance of the Hippo pathway in OEC development and how changes in cranial neural crest derivatives indirectly impact the development of olfactory epithelia.
Collapse
|
9
|
Li KP, Wan S, Wang CY, Chen SY, Wang L, Liu SH, Yang L. Multi-omics analysis reveals the impact of YAP/TEAD4-mediated EIF5A1 expression on mitochondrial apoptosis and bladder cancer progression. BMC Cancer 2025; 25:234. [PMID: 39934701 PMCID: PMC11817321 DOI: 10.1186/s12885-025-13522-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Eukaryotic Initiation Factor 5A1 (EIF5A1) is a translation factor, and its pro-tumorigenic role has been extensively documented across various cancer types. However, its specific function in bladder cancer (BLCA) remains unclear. METHODS We integrated proteomics and transcriptomics data with clinical data from BLCA patients to investigate the correlation between EIF5A1 expression and BLCA, as well as its potential clinical applications. Transcriptomic data were employed to explore the downstream signaling pathways regulated by EIF5A1. Furthermore, ChIP analysis and luciferase reporter assays were conducted to identify the upstream transcription factors regulating EIF5A1. RESULTS EIF5A1 expression is significantly upregulated in cancer tissues and cells and is strongly associated with poor prognosis. Silencing EIF5A1 in BLCA cells significantly reduced invasiveness, and proliferative capacity. Mechanistic studies identified YAP/TEAD4 as a transcription factor that regulates EIF5A1, influencing mitochondrial-mediated apoptosis by activating the JAK2/STAT3 signaling pathway, thereby promoting BLCA progression. CONCLUSION Our research demonstrates that EIF5A1 is upregulated in BLCA and associated with poor prognosis. We identified TEAD4 as a potential transcriptional regulator of EIF5A1 and showed that EIF5A1 expression is associated with changes in JAK2/STAT3 signaling and mitochondrial apoptosis in BLCA.
Collapse
Affiliation(s)
- Kun-Peng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Shun Wan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Chen-Yang Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Si-Yu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Li Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China
| | - Shan-Hui Liu
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China.
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China.
- Institute of Urology, Clinical Research Center for Urology in Gansu Province, Lanzhou, China.
| |
Collapse
|
10
|
Chen H, Xu Y, Chen D, Xiao D, Yang B, Wang W, Han H. The Hippo pathway promotes platinum-based chemotherapy by inhibiting MTF1-dependent heavy metal response. BMC Cancer 2025; 25:223. [PMID: 39920630 PMCID: PMC11806854 DOI: 10.1186/s12885-025-13661-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/05/2025] [Indexed: 02/09/2025] Open
Abstract
The platinum-based compounds are widely used in treating various types of cancer through their heavy metal component platinum. However, the development of chemoresistance often limits their clinical effectiveness. In this study, we report the roles of heavy metal response and its associated Hippo pathway in regulating platinum-based chemotherapy. Our data show that the MTF1-dependent heavy metal response induces cancer cell resistance to platinum-based compounds both in vitro and in vivo. This resistance is mitigated by Hippo pathway-mediated phosphorylation of MTF1. Moreover, pharmacological activation of the Hippo pathway sensitizes cancer cells to platinum-based compounds. Clinically, lung adenocarcinoma (LUAD) patients with high MTF1 activity exhibit poor overall survival rates, and Hippo pathway inactivation is positively correlated with elevated MTF1 transcriptional activity in platinum-treated LUAD patients. Collectively, our findings not only unveil a critical role of the Hippo-MTF1 pathway in regulating the response to platinum-based chemotherapy, but also suggest new strategies to enhance its efficacy by targeting the heavy metal response.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Yue Xu
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Dingshan Chen
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Di Xiao
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China
| | - Bing Yang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA
| | - Wenqi Wang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
| | - Han Han
- Department of Pathophysiology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, Hubei, China.
| |
Collapse
|
11
|
Li X, Cho YS, Han Y, Zhou M, Liu Y, Yang Y, Zhuo S, Jiang J. The Hippo pathway effector YAP inhibits NF-κB signaling and ccRCC growth by opposing ZHX2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.21.600079. [PMID: 38979373 PMCID: PMC11230290 DOI: 10.1101/2024.06.21.600079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
The prevailing view in the cancer field is that Hippo signaling pathway functions as a tumor suppressor pathway by blocking the oncogenic potential of the pathway effectors Yes1 associated transcriptional regulator (YAP)/transcriptional coactivator with PDZ-binding motif (TAZ). However, YAP can also function as a context-dependent tumor suppressor in several types of cancer including clear cell renal cell carcinomas (ccRCC). We find that, in additional to inhibiting hypoxia-inducible factor 2α (HIF2α), a major oncogenic driver in Von Hippel-Lindau (VHL)-/- ccRCC, YAP also blocks nuclear factor κB (NF-κB ) signaling in ccRCC to inhibit cancer cell growth under conditions where HIF2α is dispensable. Mechanistically, YAP inhibits the expression of Zinc fingers and homeoboxes 2 (ZHX2), a VHL substrate and critical co-factor of NF-κB in ccRCC. Furthermore, YAP competes with ZHX2 for binding to the NF-κB subunit p65. Consequently, elevated nuclear YAP blocks the cooperativity between ZHX2 and the NF-κB subunit p65, leading to diminished NF-κB target gene expression. Pharmacological inhibition of Hippo kinase blocked NF-κB transcriptional program and suppressed ccRCC cancer cell growth, which can be rescued by overexpression of ZHX2 or p65. Our study uncovers a crosstalk between the Hippo and NF-κB/ZHX2 pathways and its involvement in ccRCC growth inhibition, suggesting that targeting the Hippo pathway may provide a therapeutical opportunity for ccRCC treatment.
Collapse
Affiliation(s)
- Xu Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yong Suk Cho
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Math and Sciences, Tarrant County College-NE Campus, 828 W Harwood Rd, Hurst, TX 76054, USA
| | - Yuhong Han
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mengmeng Zhou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA
- Harvard Stem Cell Institute, 188 Longwood Ave. Boston, MA 02215, USA
- Dana-Farber/Harvard Cancer Center, 188 Longwood Ave. Boston, MA 02215, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Ave. Boston, MA 02215, USA
- Harvard Stem Cell Institute, 188 Longwood Ave. Boston, MA 02215, USA
- Dana-Farber/Harvard Cancer Center, 188 Longwood Ave. Boston, MA 02215, USA
| | - Shu Zhuo
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jin Jiang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
12
|
Jiang D, Li P, Lu Y, Tao J, Hao X, Wang X, Wu W, Xu J, Zhang H, Li X, Chen Y, Jin Y, Zhang L. A feedback loop between Paxillin and Yorkie sustains Drosophila intestinal homeostasis and regeneration. Nat Commun 2025; 16:570. [PMID: 39794306 PMCID: PMC11724037 DOI: 10.1038/s41467-024-55255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/04/2024] [Indexed: 01/13/2025] Open
Abstract
Balanced self-renewal and differentiation of stem cells are crucial for maintaining tissue homeostasis, but the underlying mechanisms of this process remain poorly understood. Here, from an RNA interference (RNAi) screen in adult Drosophila intestinal stem cells (ISCs), we identify a factor, Pax, which is orthologous to mammalian PXN, coordinates the proliferation and differentiation of ISCs during both normal homeostasis and injury-induced midgut regeneration in Drosophila. Loss of Pax promotes ISC proliferation while suppressing its differentiation into absorptive enterocytes (ECs). Mechanistically, our findings demonstrate that Pax is a conserved target gene of the Hippo signaling pathway in both Drosophila and mammals. Subsequent investigations have revealed Pax interacts with Yki and enhances its cytoplasmic localization, thereby establishing a feedback regulatory mechanism that attenuates Yki activity and ultimately inhibits ISCs proliferation. Additionally, Pax induces the differentiation of ISCs into ECs by activating Notch expression, thus facilitating the differentiation process. Overall, our study highlights Pax as a pivotal component of the Hippo and Notch pathways in regulating midgut homeostasis, shedding light on this growth-related pathway in tissue maintenance and intestinal function.
Collapse
Affiliation(s)
- Dan Jiang
- The Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Pengyue Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yi Lu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiaxin Tao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xue Hao
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaodong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Wei Wu
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jinjin Xu
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China
| | - Haoen Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaoyu Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yixing Chen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yunyun Jin
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China.
| | - Lei Zhang
- The Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, 200233, China.
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minghang, Shanghai, 200240, China.
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
13
|
Liu M, Xie XJ, Li X, Ren X, Sun JL, Lin Z, Hemba-Waduge RUS, Ji JY. Transcriptional coupling of telomeric retrotransposons with the cell cycle. SCIENCE ADVANCES 2025; 11:eadr2299. [PMID: 39752503 PMCID: PMC11698117 DOI: 10.1126/sciadv.adr2299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025]
Abstract
Unlike most species that use telomerase for telomere maintenance, many dipterans, including Drosophila, rely on three telomere-specific retrotransposons (TRs)-HeT-A, TART, and TAHRE-to form tandem repeats at chromosome ends. Although TR transcription is crucial in their life cycle, its regulation remains poorly understood. This study identifies the Mediator complex, E2F1-Dp, and Scalloped/dTEAD as key regulators of TR transcription. Reducing the activity of the Mediator or Sd/dTEAD increases TR expression and telomere length, while overexpressing E2F1-Dp or depleting Rbf1 stimulates TR transcription. The Mediator and Sd/dTEAD regulate this process through E2F1-Dp. CUT&RUN (Cleavage under targets and release using nuclease) analysis shows direct binding of CDK8, Dp, and Sd/dTEAD to telomeric repeats, with motif enrichment revealing E2F- and TEAD-binding sites. These findings uncover the Mediator complex's role in controlling TR transcription and telomere length through E2F1-Dp and Sd, coupling the transcriptional regulation of the TR life cycle with host cell-cycle machinery to protect chromosome ends in Drosophila.
Collapse
Affiliation(s)
- Mengmeng Liu
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Xiao-Jun Xie
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Xiao Li
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Xingjie Ren
- Institute for Human Genetics and Department of Neurology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jasmine L. Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Zhen Lin
- Department of Pathology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Rajitha-Udakara-Sampath Hemba-Waduge
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
| | - Jun-Yuan Ji
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, 1700 Tulane Avenue, New Orleans, LA 70112, USA
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
14
|
Zhu R, Jiao Z, Yu FX. Advances towards potential cancer therapeutics targeting Hippo signaling. Biochem Soc Trans 2024; 52:2399-2413. [PMID: 39641583 DOI: 10.1042/bst20240244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024]
Abstract
Decades of research into the Hippo signaling pathway have greatly advanced our understanding of its roles in organ growth, tissue regeneration, and tumorigenesis. The Hippo pathway is frequently dysregulated in human cancers and is recognized as a prominent cancer signaling pathway. Hence, the Hippo pathway represents an ideal molecular target for cancer therapies. This review will highlight recent advancements in targeting the Hippo pathway for cancer treatment and discuss the potential opportunities for developing new therapeutic modalities.
Collapse
Affiliation(s)
- Rui Zhu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Wang X, Bao H, Huang YC, Barua A, Lai CM, Sun J, Zhou Y, Cong F, Gong S, Chang CH, Deng WM. Sex-dimorphic tumor growth is regulated by tumor microenvironmental and systemic signals. SCIENCE ADVANCES 2024; 10:eads4229. [PMID: 39642218 PMCID: PMC11623276 DOI: 10.1126/sciadv.ads4229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/01/2024] [Indexed: 12/08/2024]
Abstract
Tumor growth and progression involve coordinated regulation by internal, microenvironmental, and systemic signals and often display conspicuous sexual dimorphism. The mechanisms governing the integration and coordination of these signals, along with their sex-based differences, remain largely unknown. Using a Drosophila tumor model originating from nonreproductive tissue, we show that female-biased tumor growth involves multifaceted communications among tumor cells, hemocytes, and neuroendocrine insulin-producing cells (IPCs). Notch-active tumor cells recruit hemocytes carrying the tumor necrosis factor-α (TNF-α) homolog Eiger to the tumor microenvironment (TME), activating the c-Jun N-terminal kinase (JNK) pathway in tumor cells, instigating the sexually dimorphic up-regulation of cytokine Unpaired 2 (Upd2). Upd2, in turn, exerts a distal influence by modulating the release of a Drosophila insulin-like peptide (Dilp2) from IPCs. Dilp2 then activates the insulin signaling in the tumor, thereby fostering sexual-dimorphic tumor growth. Together, these findings reveal a relay mechanism involving the TME and systemic signals that collectively control the sexual dimorphism of tumor growth.
Collapse
Affiliation(s)
- Xianfeng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Hongcun Bao
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Anindita Barua
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | - Jie Sun
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Youfang Zhou
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | - Fei Cong
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| | | | | | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Louisiana Cancer Research Center, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
Xi Y, Li J, Wu Z, Ma Y, Li J, Yang Z, Wang F, Yang D, Jiang Y, Yi Q, Huang S. Yorkie negatively regulates the Crustin expression during molting in Chinese mitten crab, Eriocheir sinensis. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 161:105242. [PMID: 39128619 DOI: 10.1016/j.dci.2024.105242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
Molting is a key biological process of crustaceans, which is mainly regulated by 20-hydroxyecdyone (20E). The molting cycle could be divided into three main stages including pre-molt, post-molt and inter-molt stages. The mechanism of immune regulation during molting process still requires further exploration. Yorkie (Yki) is a pivotal transcription factor in the Hippo signaling pathway, and it plays an essential role in regulating cell growth and immune response. In the present study, a Yki gene was identified from Eriocheir sinensis (designed as EsYki), and the regulatory role of EsYki in controlling the expression of antimicrobial peptide genes throughout the molting process was investigated. The mRNA expression level of EsYki was higher at the pre-molt stage compared to the post-molt stage and inter-molt stage. Following the injection of 20E, there was a notable and consistent rise in the EsYki mRNA expression in haemocytes. The increase was observed from 3 h to 48 h with the maximum level at 12 h. And the phosphorylation of Yki in the haemocytes was also significantly up-regulated at 3 h post 20E injection. Moreover, the levels of EsYki mRNA expression at three molting stages were significantly increased post Aeromonas hydrophila stimulation. The maximum level was detected at post-molt stage following A. hydrophila stimulation, while the lowest level was observed at inter-molt stage. The expression pattern of EsCrus was in contrast to EsCrus. After EsYki mRNA transcripts were inhibited by Yki inhibitor (CA3), the mRNA expression levels of EsCrus1 and EsCrus2 following A. hydrophila stimulation were significantly elevated. Furthermore, the phosphorylation level of NF-κB was also increased following the inhibition of Yki. Collectively, our findings indicated that EsYki could be induced by 20E and has a suppressive effect on the expression of EsCrus via inhibiting NF-κB during molting process. This research contributes to the understanding of the immunological regulation mechanism during molting process in crustaceans.
Collapse
Affiliation(s)
- Yuting Xi
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Jialin Li
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Zihao Wu
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Yuhan Ma
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Jiaming Li
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Zhichao Yang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Fengchi Wang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China
| | - Dazuo Yang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Key Laboratory of Marine Bio-Resources Restoration and Habitat Reparation in Liaoning Province, Dalian, 116023, China
| | - Yusheng Jiang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Dalian Key Laboratory of Breeding, Reproduction and Aquaculture of Crustaceans, Dalian, 116023, China
| | - Qilin Yi
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China.
| | - Shu Huang
- College of Aquaculture and Life Science, Dalian Ocean University, Dalian, 11026, China; Key Laboratory of Marine Bio-Resources Restoration and Habitat Reparation in Liaoning Province, Dalian, 116023, China; Dalian Key Laboratory of Breeding, Reproduction and Aquaculture of Crustaceans, Dalian, 116023, China.
| |
Collapse
|
17
|
Wang X, Guo Y, Lin P, Yu M, Song S, Xu W, Kong D, Wang Y, Zhang Y, Lu F, Xie Q, Ma X. Nuclear receptor E75/NR1D2 promotes tumor malignant transformation by integrating Hippo and Notch pathways. EMBO J 2024; 43:6336-6363. [PMID: 39516282 PMCID: PMC11649922 DOI: 10.1038/s44318-024-00290-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Hormone therapy resistance and the ensuing aggressive tumor progression present a significant clinical challenge. However, the mechanisms underlying the induction of tumor malignancy upon inhibition of steroid hormone signaling remain poorly understood. Here, we demonstrate that Drosophila malignant epithelial tumors show a similar reduction in ecdysone signaling, the main steroid hormone pathway. Our analysis of ecdysone-induced downstream targets reveals that overexpression of the nuclear receptor E75, particularly facilitates the malignant transformation of benign tumors. Genome-wide DNA binding profiles and biochemistry data reveal that E75 not only binds to the transcription factors of both Hippo and Notch pathways, but also exhibits widespread co-binding to their target genes, thus contributing to tumor malignancy. We further validated these findings by demonstrating that depletion of NR1D2, the mammalian homolog of E75, inhibits the activation of Hippo and Notch target genes, impeding glioblastoma progression. Together, our study unveils a novel mechanism by which hormone inhibition promotes tumor malignancy, and describes an evolutionarily conserved role of the oncogene E75/NR1D2 in integration of Hippo and Notch pathway activity during tumor progression.
Collapse
Affiliation(s)
- Xianping Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Yifan Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Peng Lin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Min Yu
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Sha Song
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Wenyan Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Du Kong
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Yin Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
- Department of Diabetes & Cancer Metabolism, Beckman Research Institute of City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Yanxiao Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China
| | - Fei Lu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Qi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
18
|
Noser AA, Salem MM, Baren MH, Selim AI, ElSafty EM. Discovering the inhibition of YAP/TEAD Hippo signaling pathway via new pyrazolone derivatives: synthesis, molecular docking and biological investigations. Sci Rep 2024; 14:28859. [PMID: 39572674 PMCID: PMC11582634 DOI: 10.1038/s41598-024-79992-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
Heterocyclic compounds play a crucial role in the drug discovery process and development due to their significant presence and importance. Here, we report a comprehensive analysis of new pyrazolone derivatives, prepared according to a clear-cut, uncomplicated procedure. The pyrazolone derivatives were thoroughly characterized using various methods, such as elemental analysis, NMR, and FT-IR. The molecular docking interactions between the new pyrazolone derivatives and YAP/TEAD target protein observed that compound 4 had the top-ranked binding energy towards YAP/TEAD with a value equal to - 9.670 kcal/mol and this theoretically proves its inhibitory efficacy against YAP/TEAD Hippo signaling pathway. Besides, compound 4 showed the best IC50 against HCT-116, HepG-2, and MCF-7 (in-vitro) with IC50 7.67 ± 0.5, 5.85 ± 0.4, and 6.97 ± 0.5 μM, respectively which confirmed our results towards suppressing YAP/TEAD protein (in-silico) compared with the IC50 of Sorafenib (SOR) reference chemotherapeutic drug 5.47 ± 0.3, 9.18 ± 0.6 and 7.26 ± 0.3 μM, respectively. Also, compound 4 showed no cytotoxic effects against human lung fibroblast normal cell line (WI-38) and its pharmacokinetics were elucidated theoretically using ADMET compared with SOR which observed highly toxic effects on normal cells with IC50 equal to 20.27 ± 0.45 μM. Additionally, compound 4 clarified a powerful antioxidant scavenging activity against DPPH free radicals (in-vitro). Conclusively, newly synthesized pyrazolone derivative 4 could be used as anticancer candidate via inhibiting the YAP/TEAD mediated Hippo signaling pathway.
Collapse
Affiliation(s)
- Ahmed A Noser
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed H Baren
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Adel I Selim
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Esraa M ElSafty
- Organic Chemistry, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
19
|
Tang Y, Chen F, Fang G, Zhang H, Zhang Y, Zhu H, Zhang X, Han Y, Cao Z, Guo F, Wang W, Ye D, Ju J, Tan L, Li C, Zhao Y, Zhou Z, An L, Jiao S. A cofactor-induced repressive type of transcription factor condensation can be induced by synthetic peptides to suppress tumorigenesis. EMBO J 2024; 43:5586-5612. [PMID: 39358623 PMCID: PMC11574045 DOI: 10.1038/s44318-024-00257-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 08/23/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
Transcriptional factors (TFs) act as key determinants of cell death and survival by differentially modulating gene expression. Here, we identified many TFs, including TEAD4, that form condensates in stressed cells. In contrast to YAP-induced transcription-activating condensates of TEAD4, we found that co-factors such as VGLL4 and RFXANK alternatively induced repressive TEAD4 condensates to trigger cell death upon glucose starvation. Focusing on VGLL4, we demonstrated that heterotypic interactions between TEAD4 and VGLL4 favor the oligomerization and assembly of large TEAD4 condensates with a nonclassical inhibitory function, i.e., causing DNA/chromatin to be aggregated and entangled, which eventually impede gene expression. Based on these findings, we engineered a peptide derived from the TEAD4-binding motif of VGLL4 to selectively induce TEAD4 repressive condensation. This "glue" peptide displayed a strong antitumor effect in genetic and xenograft mouse models of gastric cancer via inhibition of TEAD4-related gene transcription. This new type of repressive TF phase separation exemplifies how cofactors can orchestrate opposite functions of a given TF, and offers potential new antitumor strategies via artificial induction of repressive condensation.
Collapse
Grants
- 2020YFA0803200 MOST | National Key Research and Development Program of China (NKPs)
- 2023YFC2505903 MOST | National Key Research and Development Program of China (NKPs)
- 32270747,92168116, 22077002, 82222052 MOST | National Natural Science Foundation of China (NSFC)
- 32200567, 31930026, 82150112 MOST | National Natural Science Foundation of China (NSFC)
- 32070710, 82372613 MOST | National Natural Science Foundation of China (NSFC)
- 82361168638, 32170706, 82002493 MOST | National Natural Science Foundation of China (NSFC)
- 22ZR1448100, 22QA1407200, 23ZR1448900 STCSM | Natural Science Foundation of Shanghai Municipality ()
- 22QA1407300, 23ZR1480400, 23YF1432900 STCSM | Natural Science Foundation of Shanghai Municipality ()
- STCSM | Natural Science Foundation of Shanghai Municipality (上海市自然科学基金)
Collapse
Affiliation(s)
- Yang Tang
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Fan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Gemin Fang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Hui Zhang
- Department of General Surgery, Hua'shan Hospital, Fudan University Shanghai Medical College, Shanghai, 200040, China
| | - Yanni Zhang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Hanying Zhu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, China
| | - Xinru Zhang
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Yi Han
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Zhifa Cao
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Fenghua Guo
- Department of General Surgery, Hua'shan Hospital, Fudan University Shanghai Medical College, Shanghai, 200040, China
| | - Wenjia Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Dan Ye
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Junyi Ju
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China
| | - Lijie Tan
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Chuanchuan Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China
| | - Yun Zhao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhaocai Zhou
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
- Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, 211166, China.
| | - Liwei An
- Department of Medical Ultrasound, Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Shi Jiao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
20
|
Umegaki T, Moriizumi H, Ogushi F, Takekawa M, Suzuki T. Molecular dynamics simulations of a multicellular model with cell-cell interactions and Hippo signaling pathway. PLoS Comput Biol 2024; 20:e1012536. [PMID: 39527559 PMCID: PMC11554158 DOI: 10.1371/journal.pcbi.1012536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The transcriptional coactivator Yes-associated protein (YAP)/transcriptional co-activator with PDZ binding motif (TAZ) induces cell proliferation through nuclear localization at low cell density. Conversely, at extremely high cell density, the Hippo pathway, which regulates YAP/TAZ, is activated. This activation leads to the translocation of YAP/TAZ into the cytoplasm, resulting in cell cycle arrest. Various cancer cells have several times more YAP/TAZ than normal cells. However, it is not entirely clear whether this several-fold increase in YAP/TAZ alone is sufficient to overcome proliferation inhibition (contact inhibition) under high-density conditions, thereby allowing continuous proliferation. In this study, we construct a three-dimensional (3D) mathematical model of cell proliferation incorporating the Hippo-YAP/TAZ pathway. Herein, a significant innovation in our approach is the introduction of a novel modeling component that inputs cell density, which reflects cell dynamics, into the Hippo pathway and enables the simulation of cell proliferation as the output response. We assume such 3D model with cell-cell interactions by solving reaction and molecular dynamics (MD) equations by applying adhesion and repulsive forces that act between cells and frictional forces acting on each cell. We assume Lennard-Jones (12-6) potential with a softcore character so that each cell secures its exclusive domain. We set cell cycles composed of mitotic and cell growth phases in which cells divide and grow under the influence of cell kinetics. We perform mathematical simulations at various YAP/TAZ levels to investigate the extent of YAP/TAZ increase required for sustained proliferation at high density. The results show that a twofold increase in YAP/TAZ levels of cancer cells was sufficient to evade cell cycle arrest compared to normal cells, enabling cells to continue proliferating even under high-density conditions. Finally, this mathematical model, which incorporates cell-cell interactions and the Hippo-YAP/TAZ pathway, may be applicable for evaluating cancer malignancy based on YAP/TAZ levels, developing drugs to suppress the abnormal proliferation of cancer cells, and determining appropriate drug dosages. The source codes are freely available.
Collapse
Affiliation(s)
- Toshihito Umegaki
- The Center for Mathematical Modeling and Data Science, Osaka University, Osaka, Japan
| | - Hisashi Moriizumi
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Fumiko Ogushi
- Faculty of Mathematical Informatics, Meiji Gakuin University, Tokyo, Japan
| | | | - Takashi Suzuki
- The Center for Mathematical Modeling and Data Science, Osaka University, Osaka, Japan
| |
Collapse
|
21
|
Gao Y, Tan YS, Lin J, Chew LY, Aung HY, Palliyana B, Gujar MR, Lin KY, Kondo S, Wang H. SUMOylation of Warts kinase promotes neural stem cell reactivation. Nat Commun 2024; 15:8557. [PMID: 39419973 PMCID: PMC11487185 DOI: 10.1038/s41467-024-52569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
A delicate balance between neural stem cell (NSC) quiescence and proliferation is important for adult neurogenesis and homeostasis. Small ubiquitin-related modifier (SUMO)-dependent post-translational modifications cause rapid and reversible changes in protein functions. However, the role of the SUMO pathway during NSC reactivation and brain development is not established. Here, we show that the key components of the SUMO pathway play an important role in NSC reactivation and brain development in Drosophila. Depletion of SUMO/Smt3 or SUMO conjugating enzyme Ubc9 results in notable defects in NSC reactivation and brain development, while their overexpression leads to premature NSC reactivation. Smt3 protein levels increase with NSC reactivation, which is promoted by the Ser/Thr kinase Akt. Warts/Lats, the core protein kinase of the Hippo pathway, can undergo SUMO- and Ubc9-dependent SUMOylation at Lys766. This modification attenuates Wts phosphorylation by Hippo, leading to the inhibition of the Hippo pathway, and consequently, initiation of NSC reactivation. Moreover, inhibiting Hippo pathway effectively restores the NSC reactivation defects induced by SUMO pathway inhibition. Overall, our study uncovered an important role for the SUMO-Hippo pathway during Drosophila NSC reactivation and brain development.
Collapse
Affiliation(s)
- Yang Gao
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Ye Sing Tan
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Jiaen Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Liang Yuh Chew
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Htet Yamin Aung
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Brinda Palliyana
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Mahekta R Gujar
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Kun-Yang Lin
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore
| | - Shu Kondo
- Department of Biological Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Niijuku, Katsushika-ku, Tokyo, Japan
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Programme, Duke-NUS Medical School, Singapore, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Graduate School - Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
22
|
Hikasa H, Kawahara K, Inui M, Yasuki Y, Yamashita K, Otsubo K, Kitajima S, Nishio M, Arima K, Endo M, Taira M, Suzuki A. A highly sensitive reporter system to monitor endogenous YAP1/TAZ activity and its application in various human cells. Cancer Sci 2024; 115:3370-3383. [PMID: 39155534 PMCID: PMC11447953 DOI: 10.1111/cas.16316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/22/2024] [Accepted: 08/01/2024] [Indexed: 08/20/2024] Open
Abstract
The activation of yes-associated protein 1 (YAP1) and transcriptional co-activator with PDZ-binding motif (TAZ) has been implicated in both regeneration and tumorigenesis, thus representing a double-edged sword in tissue homeostasis. However, how the activity of YAP1/TAZ is regulated or what leads to its dysregulation in these processes remains unknown. To explore the upstream stimuli modulating the cellular activity of YAP1/TAZ, we developed a highly sensitive YAP1/TAZ/TEAD-responsive DNA element (YRE) and incorporated it into a lentivirus-based reporter cell system to allow for sensitive and specific monitoring of the endogenous activity of YAP1/TAZ in terms of luciferase activity in vitro and Venus fluorescence in vivo. Furthermore, by replacing YRE with TCF- and NF-κB-binding DNA elements, we demonstrated the applicability of this reporter system to other pathways such as Wnt/β-catenin/TCF- and IL-1β/NF-κB-mediated signaling, respectively. The practicality of this system was evaluated by performing cell-based reporter screening of a chemical compound library consisting of 364 known inhibitors, using reporter-introduced cells capable of quantifying YAP1/TAZ- and β-catenin-mediated transcription activities, which led to the identification of multiple inhibitors, including previously known as well as novel modulators of these signaling pathways. We further confirmed that novel YAP1/TAZ modulators, such as potassium ionophores, Janus kinase inhibitors, platelet-derived growth factor receptor inhibitors, and genotoxic stress inducers, alter the protein level or phosphorylation of endogenous YAP1/TAZ and the expression of their target genes. Thus, this reporter system provides a powerful tool to monitor endogenous signaling activities of interest (even in living cells) and search for modulators in various cellular contexts.
Collapse
Affiliation(s)
- Hiroki Hikasa
- Department of Biochemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Kohichi Kawahara
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masako Inui
- Department of Biochemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Yukichika Yasuki
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Chemistry and Bioscience, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Keita Yamashita
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Chemistry and Bioscience, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Kohei Otsubo
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shojiro Kitajima
- Department of Biochemistry, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Miki Nishio
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kazunari Arima
- Department of Chemistry and Bioscience, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Motoyoshi Endo
- Department of Molecular Biology, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Masanori Taira
- Faculty of Science and Engineering, Chuo University, Tokyo, Japan
| | - Akira Suzuki
- Division of Cancer Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Division of Molecular and Cellular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
23
|
Bhattacharya R, Kumari J, Banerjee S, Tripathi J, Parihar SS, Mohan N, Sinha P. Hippo effector, Yorkie, is a tumor suppressor in select Drosophila squamous epithelia. Proc Natl Acad Sci U S A 2024; 121:e2319666121. [PMID: 39288176 PMCID: PMC11441523 DOI: 10.1073/pnas.2319666121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Mammalian Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) and Drosophila Yorkie (Yki) are transcription cofactors of the highly conserved Hippo signaling pathway. It has been long assumed that the YAP/TAZ/Yki signaling drives cell proliferation during organ growth. However, its instructive role in regulating developmentally programmed organ growth, if any, remains elusive. Out-of-context gain of YAP/TAZ/Yki signaling often turns oncogenic. Paradoxically, mechanically strained, and differentiated squamous epithelia display developmentally programmed constitutive nuclear YAP/TAZ/Yki signaling. The unknown, therefore, is how a growth-promoting YAP/TAZ/Yki signaling restricts proliferation in differentiated squamous epithelia. Here, we show that reminiscent of a tumor suppressor, Yki negatively regulates the cell growth-promoting PI3K/Akt/TOR signaling in the squamous epithelia of Drosophila tubular organs. Thus, downregulation of Yki signaling in the squamous epithelium of the adult male accessory gland (MAG) up-regulates PI3K/Akt/TOR signaling, inducing cell hypertrophy, exit from their cell cycle arrest, and, finally, culminating in squamous cell carcinoma (SCC). Thus, blocking PI3K/Akt/TOR signaling arrests Yki loss-induced MAG-SCC. Further, MAG-SCCs, like other lethal carcinomas, secrete a cachectin, Impl2-the Drosophila homolog of mammalian IGFBP7-inducing cachexia and shortening the lifespan of adult males. Moreover, in the squamous epithelium of other tubular organs, like the dorsal trunk of larval tracheal airways or adult Malpighian tubules, downregulation of Yki signaling triggers PI3K/Akt/TOR-induced cell hypertrophy. Our results reveal that Yki signaling plays an instructive, antiproliferative role in the squamous epithelia of tubular organs.
Collapse
Affiliation(s)
- Rachita Bhattacharya
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jaya Kumari
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Shweta Banerjee
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jyoti Tripathi
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Saurabh Singh Parihar
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Nitin Mohan
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Pradip Sinha
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| |
Collapse
|
24
|
Mitchell KA, Vissers JHA, Pojer JM, Brooks E, Hilmi AJS, Papenfuss AT, Schröder J, Harvey KF. The JNK and Hippo pathways control epithelial integrity and prevent tumor initiation by regulating an overlapping transcriptome. Curr Biol 2024; 34:3966-3982.e7. [PMID: 39146938 DOI: 10.1016/j.cub.2024.07.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 06/07/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024]
Abstract
Epithelial organs maintain their integrity and prevent tumor initiation by actively removing defective cells, such as those that have lost apicobasal polarity. Here, we identify how transcription factors of two key signaling pathways-Jun-N-terminal kinase (JNK) and Hippo-regulate epithelial integrity by controlling transcription of an overlapping set of target genes. Targeted DamID experiments reveal that, in proliferating cells of the Drosophila melanogaster eye, the AP-1 transcription factor Jun and the Hippo pathway transcription regulators Yorkie and Scalloped bind to a common suite of target genes that promote organ growth. In defective neoplastic cells, AP-1 transcription factors repress transcription of growth genes together with the C-terminal binding protein (CtBP) co-repressor. If gene repression by AP-1/CtBP fails, neoplastic tumor growth ensues, driven by Yorkie/Scalloped. Thus, AP-1/CtBP eliminates defective cells and prevents tumor initiation by acting in parallel to Yorkie/Scalloped to repress expression of a shared transcriptome. These findings shed new light on the maintenance of epithelial integrity and tumor suppression.
Collapse
Affiliation(s)
- Katrina A Mitchell
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Joseph H A Vissers
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Jonathan M Pojer
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Elliot Brooks
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Abdul Jabbar Saiful Hilmi
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony T Papenfuss
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Walter and Eliza Hall Institute, Parkville, VIC 3010, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jan Schröder
- Walter and Eliza Hall Institute, Parkville, VIC 3010, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Kieran F Harvey
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
25
|
Sun X, Zhou D, Sun Y, Zhao Y, Deng Y, Pang X, Liu Q, Zhou Z. Oxidative stress reprograms the transcriptional coactivator Yki to suppress cell proliferation. Cell Rep 2024; 43:114584. [PMID: 39106181 DOI: 10.1016/j.celrep.2024.114584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/09/2024] Open
Abstract
The transcriptional coactivator Yorkie (Yki) regulates organ size by promoting cell proliferation. It is unclear how cells control Yki activity when exposed to harmful stimuli such as oxidative stress. In this study, we show that oxidative stress inhibits the binding of Yki to Scalloped (Sd) but promotes the interaction of Yki with another transcription factor, forkhead box O (Foxo), ultimately leading to a halt in cell proliferation. Mechanistically, Foxo normally exhibits a low binding affinity for Yki, allowing Yki to form a complex with Sd and activate proliferative genes. Under oxidative stress, Usp7 deubiquitinates Foxo to promote its interaction with Yki, thereby activating the expression of proliferation suppressors. Finally, we show that Yki is essential for Drosophila survival under oxidative stress. In summary, these findings suggest that oxidative stress reprograms Yki from a proliferation-promoting factor to a proliferation suppressor, forming a self-protective mechanism.
Collapse
Affiliation(s)
- Xiaohan Sun
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang 330022, China; School of Life Sciences and Medicine, Shandong University of Technology, Zibo 255000, China
| | - Dafa Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Yuanfei Sun
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Yunhe Zhao
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Yanran Deng
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang 330022, China
| | - Xiaolin Pang
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Qingxin Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China
| | - Zizhang Zhou
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang 330022, China; College of Life Sciences, Shandong Agricultural University, Tai'an 271018, China.
| |
Collapse
|
26
|
Manning SA, Kroeger B, Deng Q, Brooks E, Fonseka Y, Hinde E, Harvey KF. The Drosophila Hippo pathway transcription factor Scalloped and its co-factors alter each other's chromatin binding dynamics and transcription in vivo. Dev Cell 2024; 59:1640-1654.e5. [PMID: 38670104 DOI: 10.1016/j.devcel.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/12/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
The Hippo pathway is an important regulator of organ growth and cell fate. The major mechanism by which Hippo is known to control transcription is by dictating the nucleo-cytoplasmic shuttling rate of Yorkie, a transcription co-activator, which promotes transcription with the DNA binding protein Scalloped. The nuclear biophysical behavior of Yorkie and Scalloped, and whether this is regulated by the Hippo pathway, remains unexplored. Using multiple live-imaging modalities on Drosophila tissues, we found that Scalloped interacts with DNA on a broad range of timescales, and enrichment of Scalloped at sites of active transcription is mediated by longer DNA dwell times. Further, Yorkie increased Scalloped's DNA dwell time, whereas the repressors Nervous fingers 1 (Nerfin-1) and Tondu-domain-containing growth inhibitor (Tgi) decreased it. Therefore, the Hippo pathway influences transcription not only by controlling nuclear abundance of Yorkie but also by modifying the DNA binding kinetics of the transcription factor Scalloped.
Collapse
Affiliation(s)
- Samuel A Manning
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Benjamin Kroeger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Qiji Deng
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Elliot Brooks
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Yoshana Fonseka
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia
| | - Elizabeth Hinde
- School of Physics, University of Melbourne, Parkville, VIC 3010, Australia; Department of Biochemistry and Pharmacology, Bio21 Institute, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Kieran F Harvey
- Department of Anatomy and Developmental Biology, Monash University, Clayton, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
27
|
Li Z, Su P, Yu M, Zhang X, Xu Y, Jia T, Yang P, Zhang C, Sun Y, Li X, Yang H, Ding Y, Zhuang T, Guo H, Zhu J. YAP represses the TEAD-NF-κB complex and inhibits the growth of clear cell renal cell carcinoma. Sci Signal 2024; 17:eadk0231. [PMID: 38954637 DOI: 10.1126/scisignal.adk0231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 06/11/2024] [Indexed: 07/04/2024]
Abstract
The Hippo pathway is generally understood to inhibit tumor growth by phosphorylating the transcriptional cofactor YAP to sequester it to the cytoplasm and reduce the formation of YAP-TEAD transcriptional complexes. Aberrant activation of YAP occurs in various cancers. However, we found a tumor-suppressive function of YAP in clear cell renal cell carcinoma (ccRCC). Using cell cultures, xenografts, and patient-derived explant models, we found that the inhibition of upstream Hippo-pathway kinases MST1 and MST2 or expression of a constitutively active YAP mutant impeded ccRCC proliferation and decreased gene expression mediated by the transcription factor NF-κB. Mechanistically, the NF-κB subunit p65 bound to the transcriptional cofactor TEAD to facilitate NF-κB-target gene expression that promoted cell proliferation. However, by competing for TEAD, YAP disrupted its interaction with NF-κB and prompted the dissociation of p65 from target gene promoters, thereby inhibiting NF-κB transcriptional programs. This cross-talk between the Hippo and NF-κB pathways in ccRCC suggests that targeting the Hippo-YAP axis in an atypical manner-that is, by activating YAP-may be a strategy for slowing tumor growth in patients.
Collapse
Affiliation(s)
- Zhongbo Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Peng Su
- Department of Pathology, Shandong University Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, P.R. China
| | - Miao Yu
- Department of General Surgery, Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, P.R. China
| | - Xufeng Zhang
- Kidney Transplantation, Second Hospital, Cheloo College of Medicine, Shandong University, Jinan 250033, Shandong Province, P.R. China
| | - Yaning Xu
- Department of Clinical Laboratory, Second Hospital, Cheloo College of Medicine, Shandong University, Jinan 250033, Shandong Province, P.R. China
| | - Tianwei Jia
- Department of Clinical Laboratory, Second Hospital, Cheloo College of Medicine, Shandong University, Jinan 250033, Shandong Province, P.R. China
| | - Penghe Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Chenmiao Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Yanan Sun
- Department of Pathology, Shandong University Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, P.R. China
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Huijie Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Yinlu Ding
- Department of General Surgery, Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, P.R. China
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, P.R. China
| | - Haiyang Guo
- Department of Clinical Laboratory, Second Hospital, Cheloo College of Medicine, Shandong University, Jinan 250033, Shandong Province, P.R. China
| | - Jian Zhu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110000, Liaoning Province, PR China
| |
Collapse
|
28
|
Xue Y, Ruan Y, Wang Y, Xiao P, Xu J. Signaling pathways in liver cancer: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:20. [PMID: 38816668 PMCID: PMC11139849 DOI: 10.1186/s43556-024-00184-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/23/2024] [Indexed: 06/01/2024] Open
Abstract
Liver cancer remains one of the most prevalent malignancies worldwide with high incidence and mortality rates. Due to its subtle onset, liver cancer is commonly diagnosed at a late stage when surgical interventions are no longer feasible. This situation highlights the critical role of systemic treatments, including targeted therapies, in bettering patient outcomes. Despite numerous studies on the mechanisms underlying liver cancer, tyrosine kinase inhibitors (TKIs) are the only widely used clinical inhibitors, represented by sorafenib, whose clinical application is greatly limited by the phenomenon of drug resistance. Here we show an in-depth discussion of the signaling pathways frequently implicated in liver cancer pathogenesis and the inhibitors targeting these pathways under investigation or already in use in the management of advanced liver cancer. We elucidate the oncogenic roles of these pathways in liver cancer especially hepatocellular carcinoma (HCC), as well as the current state of research on inhibitors respectively. Given that TKIs represent the sole class of targeted therapeutics for liver cancer employed in clinical practice, we have particularly focused on TKIs and the mechanisms of the commonly encountered phenomena of its resistance during HCC treatment. This necessitates the imperative development of innovative targeted strategies and the urgency of overcoming the existing limitations. This review endeavors to shed light on the utilization of targeted therapy in advanced liver cancer, with a vision to improve the unsatisfactory prognostic outlook for those patients.
Collapse
Affiliation(s)
- Yangtao Xue
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yeling Ruan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Yali Wang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China
- Zhejiang University Cancer Center, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Peng Xiao
- Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Hangzhou, 310016, China.
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, Hangzhou, 310016, China.
- Zhejiang University Cancer Center, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
29
|
Li M, Ding W, Deng Y, Zhao Y, Liu Q, Zhou Z. The AAA-ATPase Ter94 regulates wing size in Drosophila by suppressing the Hippo pathway. Commun Biol 2024; 7:533. [PMID: 38710747 PMCID: PMC11074327 DOI: 10.1038/s42003-024-06246-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Insect wing development is a fascinating and intricate process that involves the regulation of wing size through cell proliferation and apoptosis. In this study, we find that Ter94, an AAA-ATPase, is essential for proper wing size dependently on its ATPase activity. Loss of Ter94 enables the suppression of Hippo target genes. When Ter94 is depleted, it results in reduced wing size and increased apoptosis, which can be rescued by inhibiting the Hippo pathway. Biochemical experiments reveal that Ter94 reciprocally binds to Mer, a critical upstream component of the Hippo pathway, and disrupts its interaction with Ex and Kib. This disruption prevents the formation of the Ex-Mer-Kib complex, ultimately leading to the inactivation of the Hippo pathway and promoting proper wing development. Finally, we show that hVCP, the human homolog of Ter94, is able to substitute for Ter94 in modulating Drosophila wing size, underscoring their functional conservation. In conclusion, Ter94 plays a positive role in regulating wing size by interfering with the Ex-Mer-Kib complex, which results in the suppression of the Hippo pathway.
Collapse
Affiliation(s)
- Mingming Li
- College of Life Sciences, Shandong Agricultural University, Tai'an, China
| | - Wenhao Ding
- College of Life Sciences, Shandong Agricultural University, Tai'an, China
| | - Yanran Deng
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang, China
| | - Yunhe Zhao
- College of Life Sciences, Shandong Agricultural University, Tai'an, China
| | - Qingxin Liu
- College of Life Sciences, Shandong Agricultural University, Tai'an, China.
| | - Zizhang Zhou
- College of Life Sciences, Shandong Agricultural University, Tai'an, China.
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang, China.
| |
Collapse
|
30
|
Waghmare I, Gangwani K, Rai A, Singh A, Kango-Singh M. A Tumor-Specific Molecular Network Promotes Tumor Growth in Drosophila by Enforcing a Jun N-Terminal Kinase-Yorkie Feedforward Loop. Cancers (Basel) 2024; 16:1768. [PMID: 38730720 PMCID: PMC11083887 DOI: 10.3390/cancers16091768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer cells expand rapidly in response to altered intercellular and signaling interactions to achieve the hallmarks of cancer. Impaired cell polarity combined with activated oncogenes is known to promote several hallmarks of cancer, e.g., activating invasion by increased activity of Jun N-terminal kinase (JNK) and sustained proliferative signaling by increased activity of Hippo effector Yorkie (Yki). Thus, JNK, Yki, and their downstream transcription factors have emerged as synergistic drivers of tumor growth through pro-tumor signaling and intercellular interactions like cell competition. However, little is known about the signals that converge onto JNK and Yki in tumor cells and enable tumor cells to achieve the hallmarks of cancer. Here, using mosaic models of cooperative oncogenesis (RasV12,scrib-) in Drosophila, we show that RasV12,scrib- tumor cells grow through the activation of a previously unidentified network comprising Wingless (Wg), Dronc, JNK, and Yki. We show that RasV12,scrib- cells show increased Wg, Dronc, JNK, and Yki signaling, and all these signals are required for the growth of RasV12,scrib- tumors. We report that Wg and Dronc converge onto a JNK-Yki self-reinforcing positive feedback signal-amplification loop that promotes tumor growth. We found that the Wg-Dronc-Yki-JNK molecular network is specifically activated in polarity-impaired tumor cells and not in normal cells, in which apical-basal polarity remains intact. Our findings suggest that the identification of molecular networks may provide significant insights into the key biologically meaningful changes in signaling pathways and paradoxical signals that promote tumorigenesis.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; (I.W.); (A.R.); (A.S.)
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Karishma Gangwani
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; (I.W.); (A.R.); (A.S.)
- Computational Biology Department, St Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Arushi Rai
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; (I.W.); (A.R.); (A.S.)
| | - Amit Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; (I.W.); (A.R.); (A.S.)
- Premedical Programs, University of Dayton, Dayton, OH 45469, USA
- Integrative Science and Engineering Centre (ISE), University of Dayton, Dayton, OH 45469, USA
| | - Madhuri Kango-Singh
- Department of Biology, University of Dayton, Dayton, OH 45469, USA; (I.W.); (A.R.); (A.S.)
- Premedical Programs, University of Dayton, Dayton, OH 45469, USA
- Integrative Science and Engineering Centre (ISE), University of Dayton, Dayton, OH 45469, USA
| |
Collapse
|
31
|
Wu M, Hu L, He L, Yuan L, Yang L, Zhao B, Zhang L, He X. The tumor suppressor NF2 modulates TEAD4 stability and activity in Hippo signaling via direct interaction. J Biol Chem 2024; 300:107212. [PMID: 38522513 PMCID: PMC11046300 DOI: 10.1016/j.jbc.2024.107212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/10/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024] Open
Abstract
As an output effector of the Hippo signaling pathway, the TEAD transcription factor and co-activator YAP play crucial functions in promoting cell proliferation and organ size. The tumor suppressor NF2 has been shown to activate LATS1/2 kinases and interplay with the Hippo pathway to suppress the YAP-TEAD complex. However, whether and how NF2 could directly regulate TEAD remains unknown. We identified a direct link and physical interaction between NF2 and TEAD4. NF2 interacted with TEAD4 through its FERM domain and C-terminal tail and decreased the protein stability of TEAD4 independently of LATS1/2 and YAP. Furthermore, NF2 inhibited TEAD4 palmitoylation and induced the cytoplasmic translocation of TEAD4, resulting in ubiquitination and dysfunction of TEAD4. Moreover, the interaction with TEAD4 is required for NF2 function to suppress cell proliferation. These findings reveal an unanticipated role of NF2 as a binding partner and inhibitor of the transcription factor TEAD, shedding light on an alternative mechanism of how NF2 functions as a tumor suppressor through the Hippo signaling cascade.
Collapse
Affiliation(s)
- Mengying Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Liqiao Hu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| | - Lingli He
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Liang Yuan
- College of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lingling Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhao
- The MOE Key Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China; College of Life Science and Technology, ShanghaiTech University, Shanghai, China; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou, China
| | - Xiaojing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
32
|
Rodríguez A, Foronda D, Córdoba S, Felipe-Cordero D, Baonza A, Miguez DG, Estella C. Cell proliferation and Notch signaling coordinate the formation of epithelial folds in the Drosophila leg. Development 2024; 151:dev202384. [PMID: 38512712 PMCID: PMC11058088 DOI: 10.1242/dev.202384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 03/12/2024] [Indexed: 03/23/2024]
Abstract
The formation of complex three-dimensional organs during development requires precise coordination between patterning networks and mechanical forces. In particular, tissue folding is a crucial process that relies on a combination of local and tissue-wide mechanical forces. Here, we investigate the contribution of cell proliferation to epithelial morphogenesis using the Drosophila leg tarsal folds as a model. We reveal that tissue-wide compression forces generated by cell proliferation, in coordination with the Notch signaling pathway, are essential for the formation of epithelial folds in precise locations along the proximo-distal axis of the leg. As cell numbers increase, compressive stresses arise, promoting the folding of the epithelium and reinforcing the apical constriction of invaginating cells. Additionally, the Notch target dysfusion plays a key function specifying the location of the folds, through the apical accumulation of F-actin and the apico-basal shortening of invaginating cells. These findings provide new insights into the intricate mechanisms involved in epithelial morphogenesis, highlighting the crucial role of tissue-wide forces in shaping a three-dimensional organ in a reproducible manner.
Collapse
Affiliation(s)
- Alonso Rodríguez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - David Foronda
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
- Departamento de Medicina, Facultad de Ciencias Biomédicas y de la Salud, Universidad Europea de Madrid, Madrid 28670, Spain
| | - Sergio Córdoba
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Daniel Felipe-Cordero
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Antonio Baonza
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - David G. Miguez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
- Departmento de Física de la Materia Condensada, Instituto de Física de la Materia Condensada (IFIMAC), Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Carlos Estella
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
33
|
Nita A, Moroishi T. Hippo pathway in cell-cell communication: emerging roles in development and regeneration. Inflamm Regen 2024; 44:18. [PMID: 38566194 PMCID: PMC10986044 DOI: 10.1186/s41232-024-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024] Open
Abstract
The Hippo pathway is a central regulator of tissue growth that has been widely studied in mammalian organ development, regeneration, and cancer biology. Although previous studies have convincingly revealed its cell-autonomous functions in controlling cell fate, such as cell proliferation, survival, and differentiation, accumulating evidence in recent years has revealed its non-cell-autonomous functions. This pathway regulates cell-cell communication through direct interactions, soluble factors, extracellular vesicles, and the extracellular matrix, providing a range of options for controlling diverse biological processes. Consequently, the Hippo pathway not only dictates the fate of individual cells but also triggers multicellular responses involving both tissue-resident cells and infiltrating immune cells. Here, we have highlighted the recent understanding of the molecular mechanisms by which the Hippo pathway controls cell-cell communication and discuss its importance in tissue homeostasis, especially in development and regeneration.
Collapse
Affiliation(s)
- Akihiro Nita
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Toshiro Moroishi
- Department of Molecular and Medical Pharmacology, Faculty of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
- Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
34
|
Zhong Z, Jiao Z, Yu FX. The Hippo signaling pathway in development and regeneration. Cell Rep 2024; 43:113926. [PMID: 38457338 DOI: 10.1016/j.celrep.2024.113926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/10/2024] Open
Abstract
The Hippo signaling pathway is a central growth control mechanism in multicellular organisms. By integrating diverse mechanical, biochemical, and stress cues, the Hippo pathway orchestrates proliferation, survival, differentiation, and mechanics of cells, which in turn regulate organ development, homeostasis, and regeneration. A deep understanding of the regulation and function of the Hippo pathway therefore holds great promise for developing novel therapeutics in regenerative medicine. Here, we provide updates on the molecular organization of the mammalian Hippo signaling network, review the regulatory signals and functional outputs of the pathway, and discuss the roles of Hippo signaling in development and regeneration.
Collapse
Affiliation(s)
- Zhenxing Zhong
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhihan Jiao
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, and the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
35
|
Mannion AJ, Zhao H, Zhang Y, von Wright Y, Bergman O, Roy J, Saharinen P, Holmgren L. Regulation of YAP Promotor Accessibility in Endothelial Mechanotransduction. Arterioscler Thromb Vasc Biol 2024; 44:666-689. [PMID: 38299356 PMCID: PMC10880945 DOI: 10.1161/atvbaha.123.320300] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/11/2024] [Indexed: 02/02/2024]
Abstract
BACKGROUND Endothelial cells are constantly exposed to mechanical forces in the form of fluid shear stress, extracellular stiffness, and cyclic strain. The mechanoresponsive activity of YAP (Yes-associated protein) and its role in vascular development are well described; however, whether changes to transcription or epigenetic regulation of YAP are involved in these processes remains unanswered. Furthermore, how mechanical forces are transduced to the nucleus to drive transcriptional reprogramming in endothelial cells is poorly understood. The YAP target gene, AmotL2 (angiomotin-like 2), is a junctional mechanotransducer that connects cell-cell junctions to the nuclear membrane via the actin cytoskeleton. METHODS We applied mechanical manipulations including shear flow, stretching, and substrate stiffness to endothelial cells to investigate the role of mechanical forces in modulating YAP transcription. Using in vitro and in vivo endothelial depletion of AmotL2, we assess nuclear morphology, chromatin organization (using transposase-accessible chromatin sequencing), and whole-mount immunofluorescent staining of the aorta to determine the regulation and functionality of YAP. Finally, we use genetic and chemical inhibition to uncouple the nuclear-cytoskeletal connection to investigate the role of this pathway on YAP transcription. RESULTS Our results reveal that mechanical forces sensed at cell-cell junctions by the YAP target gene AmotL2 are directly involved in changes in global chromatin accessibility and activity of the histone methyltransferase EZH2, leading to modulation of YAP promotor activity. Functionally, shear stress-induced proliferation of endothelial cells in vivo was reliant on AmotL2 and YAP/TAZ (transcriptional coactivator with PDZ-binding motif) expression. Mechanistically, uncoupling of the nuclear-cytoskeletal connection from junctions and focal adhesions led to altered nuclear morphology, chromatin accessibility, and YAP promotor activity. CONCLUSIONS Our findings reveal a role for AmotL2 and nuclear-cytoskeletal force transmission in modulating the epigenetic and transcriptional regulation of YAP to maintain a mechano-enforced positive feedback loop of vascular homeostasis. These findings may offer an explanation as to the proinflammatory phenotype that leads to aneurysm formation observed in AmotL2 endothelial deletion models.
Collapse
Affiliation(s)
- Aarren J. Mannion
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
- Department of Cell and Tissue Dynamics, Max Planck Institute of Molecular Biomedicine, Münster, Germany (A.J.M.)
| | - Honglei Zhao
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Yuanyuan Zhang
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| | - Ylva von Wright
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
| | - Otto Bergman
- Medicine (O.B.), Karolinska Institute, Stockholm, Sweden
| | - Joy Roy
- Molecular Medicine and Surgery (J.R.), Karolinska Institute, Stockholm, Sweden
- Department of Vascular Surgery, Karolinska University Hospital, Stockholm, Sweden (J.R.)
| | - Pipsa Saharinen
- Wihuri Research Institute, Biomedicum Helsinki, Finland (Y.v.W., P.S.)
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Finland (P.S.)
| | - Lars Holmgren
- Departments of Oncology-Pathology (A.J.M., H.Z., Y.Z., L.H.), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
36
|
Li Y, Zheng Z, Xiao L, Chen Y, Liu X, Long D, Chai L, Li Y, Tan C. Dinaciclib exerts a tumor-suppressing effect via β-catenin/YAP axis in pancreatic ductal adenocarcinoma. Anticancer Drugs 2024; 35:140-154. [PMID: 37694833 DOI: 10.1097/cad.0000000000001545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Dinaciclib, a cyclin-dependent kinase-5 (CDK5) inhibitor, has significant anti-tumor properties. However, the precise mechanism of dinaciclib requires further investigation. Herein, we investigated the anti-tumor functions and molecular basis of dinaciclib in pancreatic ductal adenocarcinoma (PDAC). PDAC and matched para-carcinoma specimens were collected from the patients who underwent radical resection. Immunohistochemistry was performed to assess CDK5 expression. Cell proliferation ability, migration, and invasion were measured using Cell Counting Kit-8, wound healing, and transwell assay, respectively. The cell cycle and apoptosis were assessed using flow cytometry. Gene expression was examined using RNA-seq and quantitative real-time PCR. Protein expression of proteins was measured by western blot analysis and immunofluorescence microscopy. Tumor-bearing mice were intraperitoneally injected with dinaciclib. CDK5 is highly expressed in PDAC. The expression level of CDK5 was significantly related to tumor size, T stage, and the American Joint Committee on Cancer stage. High CDK5 expression can predict poor survival in PDAC patients. In addition, the expression level of CDK5 might be an independent prognostic factor for PDAC patients. Dinaciclib inhibits the growth and motility of PDAC cells and induces apoptosis and cell cycle arrest in the G2/M phase. Mechanistically, dinaciclib down-regulated yes-associated protein (YAP) mRNA and protein expression by reducing β-catenin expression. Moreover, dinaciclib significantly inhibited PDAC cell growth in vivo . Our findings reveal a novel anti-tumor mechanism of dinaciclib in which it decreases YAP expression by down-regulating β-catenin at the transcriptional level rather than by activating Hippo pathway-mediated phosphorylation-dependent degradation.
Collapse
Affiliation(s)
- Yichen Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Li Xiao
- Department of Traditional Chinese Medicine, Chengdu Third People's Hospital
| | - Yonghua Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Xubao Liu
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| | - Dan Long
- Key Laboratory of Transplant Engineering and Immunology, National Clinical Research Center for Geriatrics, Frontiers Science Center for Diseaserelated Molecular Network, West China Hospital, Sichuan University
| | - Li Chai
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University
| |
Collapse
|
37
|
Kroeger B, Manning SA, Fonseka Y, Oorschot V, Crawford SA, Ramm G, Harvey KF. Basal spot junctions of Drosophila epithelial tissues respond to morphogenetic forces and regulate Hippo signaling. Dev Cell 2024; 59:262-279.e6. [PMID: 38134928 DOI: 10.1016/j.devcel.2023.11.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/08/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023]
Abstract
Organ size is controlled by numerous factors including mechanical forces, which are mediated in part by the Hippo pathway. In growing Drosophila epithelial tissues, cytoskeletal tension influences Hippo signaling by modulating the localization of key pathway proteins to different apical domains. Here, we discovered a Hippo signaling hub at basal spot junctions, which form at the basal-most point of the lateral membranes and resemble adherens junctions in protein composition. Basal spot junctions recruit the central kinase Warts via Ajuba and E-cadherin, which prevent Warts activation by segregating it from upstream Hippo pathway proteins. Basal spot junctions are prominent when tissues undergo morphogenesis and are highly sensitive to fluctuations in cytoskeletal tension. They are distinct from focal adhesions, but the latter profoundly influences basal spot junction abundance by modulating the basal-medial actomyosin network and tension experienced by spot junctions. Thus, basal spot junctions couple morphogenetic forces to Hippo pathway activity and organ growth.
Collapse
Affiliation(s)
- Benjamin Kroeger
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Samuel A Manning
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia
| | - Yoshana Fonseka
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, VIC 3800, Australia
| | - Viola Oorschot
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Melbourne, VIC 3168, Australia
| | - Simon A Crawford
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Melbourne, VIC 3168, Australia
| | - Georg Ramm
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Melbourne, VIC 3168, Australia
| | - Kieran F Harvey
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Melbourne, VIC 3800, Australia; Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia.
| |
Collapse
|
38
|
Zhang T, Lin H, Ren T, He M, Zheng W, Tong Y, Jin B, Xie K, Deng A, Liu S, Chen Y, Xu G, Chen T, Pan W, Xiao Z. ROCK1 is a multifunctional factor maintaining the primordial follicle reserve and follicular development in mice. Am J Physiol Cell Physiol 2024; 326:C27-C39. [PMID: 37661919 PMCID: PMC11192470 DOI: 10.1152/ajpcell.00019.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 08/14/2023] [Accepted: 08/15/2023] [Indexed: 09/05/2023]
Abstract
The follicle is the basic structural and functional unit of the ovary in female mammals. The excessive depletion of follicles will lead to diminished ovarian reserve or even premature ovarian failure, resulting in diminished ovarian oogenesis and endocrine function. Excessive follicular depletion is mainly due to loss of primordial follicles. Our analysis of published human ovarian single-cell sequencing results by others revealed a significant increase in rho-associated protein kinase 1 (ROCK1) expression during primordial follicle development. However, the role of ROCK1 in primordial follicle development and maintenance is not clear. This study revealed a gradual increase in ROCK1 expression during primordial follicle activation. Inhibition of ROCK1 resulted in reduced primordial follicle activation, decreased follicular reserve, and delayed development of growing follicles. This effect may be achieved through the HIPPO pathway. The present study indicates that ROCK1 is a key molecule for primordial follicular reserve and follicular development.NEW & NOTEWORTHY ROCK1, one of the Rho GTPases, plays an important role in primordial follicle reserve and follicular development. ROCK1 was primarily expressed in the cytoplasm of oocytes and granulosa cell in mice. Inhibition of ROCK1 significantly reduced the primordial follicle reserve and delayed growing follicle development. ROCK1 regulates primordial follicular reserve and follicle development through the HIPPO signaling pathway. These findings shed new lights on the physiology of sustaining female reproduction.
Collapse
Affiliation(s)
- Tuo Zhang
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, People's Republic of China
| | - Huan Lin
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Tianhe Ren
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Meina He
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Wenying Zheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yuntong Tong
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Bangming Jin
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, People's Republic of China
| | - Kaiyun Xie
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Ankang Deng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Shiyu Liu
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Yuqian Chen
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Guoqiang Xu
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
| | - Tengxiang Chen
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, People's Republic of China
| | - Wei Pan
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Ziwen Xiao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| |
Collapse
|
39
|
Rashid F, Xie Z, Li M, Xie Z, Luo S, Xie L. Roles and functions of IAV proteins in host immune evasion. Front Immunol 2023; 14:1323560. [PMID: 38152399 PMCID: PMC10751371 DOI: 10.3389/fimmu.2023.1323560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023] Open
Abstract
Influenza A viruses (IAVs) evade the immune system of the host by several regulatory mechanisms. Their genomes consist of eight single-stranded segments, including nonstructural proteins (NS), basic polymerase 1 (PB1), basic polymerase 2 (PB2), hemagglutinin (HA), acidic polymerase (PA), matrix (M), neuraminidase (NA), and nucleoprotein (NP). Some of these proteins are known to suppress host immune responses. In this review, we discuss the roles, functions and underlying strategies adopted by IAV proteins to escape the host immune system by targeting different proteins in the interferon (IFN) signaling pathway, such as tripartite motif containing 25 (TRIM25), inhibitor of nuclear factor κB kinase (IKK), mitochondrial antiviral signaling protein (MAVS), Janus kinase 1 (JAK1), type I interferon receptor (IFNAR1), interferon regulatory factor 3 (IRF3), IRF7, and nuclear factor-κB (NF-κB). To date, the IAV proteins NS1, NS2, PB1, PB1-F2, PB2, HA, and PA have been well studied in terms of their roles in evading the host immune system. However, the detailed mechanisms of NS3, PB1-N40, PA-N155, PA-N182, PA-X, M42, NA, and NP have not been well studied with respect to their roles in immune evasion. Moreover, we also highlight the future perspectives of research on IAV proteins.
Collapse
Affiliation(s)
- Farooq Rashid
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Zhixun Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Meng Li
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Zhiqin Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Sisi Luo
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| | - Liji Xie
- Department of Biotechnology, Guangxi Veterinary Research Institute, Nanning, China
- Guangxi Key Laboratory of Veterinary Biotechnology, Nanning, China
- Key Laboratory of China (Guangxi)-ASEAN Cross-border Animal Disease Prevention and Control, Ministry of Agriculture and Rural Affairs of China, Nanning, China
| |
Collapse
|
40
|
Chailertrit V, Panthum T, Kongkaew L, Chalermwong P, Singchat W, Ahmad SF, Kraichak E, Muangmai N, Duengkae P, Peyachoknagul S, Han K, Srikulnath K. Genome-wide SNP analysis provides insights into the XX/XY sex-determination system in silver barb (Barbonymus gonionotus). Genomics Inform 2023; 21:e47. [PMID: 38224714 PMCID: PMC10788355 DOI: 10.5808/gi.23075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/17/2024] Open
Abstract
Silver barb (Barbonymus gonionotus) is among the most economically important freshwater fish species in Thailand. It ranks fourth in economic value and third in production weight for fisheries and culture in Thailand. An XX/XY sex-determination system based on gynogenesis was previously reported for this fish. In this study, the molecular basis underlying the sex-determination system was further investigated. Genome-wide single-nucleotide polymorphism data were generated for 32 captive-bred silver barb individuals, previously scored by phenotypic sex, to identify sex-linked regions associated with sex determination. Sixty-three male-linked loci, indicating putative XY chromosomes, were identified. Male-specific loci were not observed, which indicates that the putative Y chromosome is young and the sex determination region is cryptic. A homology search revealed that most male-linked loci were homologous to the Mariner/Tc1 and Gypsy transposable elements and are probably the remnants of an initial accumulation of repeats on the Y chromosome from the early stages of sex chromosome differentiation. This research provides convincing insights into the mechanism of sex determination and reveals the potential sex determination regions in silver barb. The study provides the basic data necessary for increasing the commercial value of silver barbs through genetic improvements.
Collapse
Affiliation(s)
- Visarut Chailertrit
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Pathum Thani Aquatic Animal Genetics Research and Development Center, Aquatic Animal Genetics Research and Development Division, Department of Fisheries, Pathum Thani 12120, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
| | - Thitipong Panthum
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
| | - Lalida Kongkaew
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
| | - Piangjai Chalermwong
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Sciences for Industry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Worapong Singchat
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
| | - Syed Farhan Ahmad
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Ekaphan Kraichak
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Department of Botany, Kasetsart University, Bangkok 10900, Thailand
| | - Narongrit Muangmai
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Department of Fishery Biology, Faculty of Fisheries, Kasetsart University, Bangkok 10900, Thailand
| | - Prateep Duengkae
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
| | - Surin Peyachoknagul
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
| | - Kyudong Han
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Department of Microbiology, Dankook University, Cheonan 31116, Korea
- Bio-Medical Engineering Core Facility Research Center, Dankook University, Cheonan 31116, Korea
| | - Kornsorn Srikulnath
- Animal Genomics and Bioresource Research Unit (AGB Research Unit), Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Laboratory of Animal Cytogenetics and Comparative Genomics (ACCG), Department of Genetics, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Special Research Unit for Wildlife Genomics (SRUWG), Department of Forest Biology, Faculty of Forestry, Kasetsart University, Bangkok 10900, Thailand
- Sciences for Industry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand
- Center for Advanced Studies in Tropical Natural Resources (CASTNAR), National Research University-Kasetsart University (NRU-KU), Kasetsart University, Bangkok 10900, Thailand
| |
Collapse
|
41
|
Driskill JH, Pan D. Control of stem cell renewal and fate by YAP and TAZ. Nat Rev Mol Cell Biol 2023; 24:895-911. [PMID: 37626124 DOI: 10.1038/s41580-023-00644-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2023] [Indexed: 08/27/2023]
Abstract
Complex physiological processes control whether stem cells self-renew, differentiate or remain quiescent. Two decades of research have placed the Hippo pathway, a highly conserved kinase signalling cascade, and its downstream molecular effectors YAP and TAZ at the nexus of this decision. YAP and TAZ translate complex biological cues acting on stem cells - from mechanical forces to cellular metabolism - into genome-wide effects to mediate stem cell functions. While aberrant YAP/TAZ activity drives stem cell dysfunction in ageing, tumorigenesis and disease, therapeutic targeting of Hippo signalling and YAP/TAZ can boost stem cell activity to enhance regeneration. In this Review, we discuss how YAP/TAZ control the self-renewal, fate and plasticity of stem cells in different contexts, how dysregulation of YAP/TAZ in stem cells leads to disease, and how therapeutic modalities targeting YAP/TAZ may benefit regenerative medicine and cancer therapy.
Collapse
Affiliation(s)
- Jordan H Driskill
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Duojia Pan
- Department of Physiology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
42
|
Cachoux VML, Balakireva M, Gracia M, Bosveld F, López-Gay JM, Maugarny A, Gaugué I, di Pietro F, Rigaud SU, Noiret L, Guirao B, Bellaïche Y. Epithelial apoptotic pattern emerges from global and local regulation by cell apical area. Curr Biol 2023; 33:4807-4826.e6. [PMID: 37827152 PMCID: PMC10681125 DOI: 10.1016/j.cub.2023.09.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023]
Abstract
Geometry is a fundamental attribute of biological systems, and it underlies cell and tissue dynamics. Cell geometry controls cell-cycle progression and mitosis and thus modulates tissue development and homeostasis. In sharp contrast and despite the extensive characterization of the genetic mechanisms of caspase activation, we know little about whether and how cell geometry controls apoptosis commitment in developing tissues. Here, we combined multiscale time-lapse microscopy of developing Drosophila epithelium, quantitative characterization of cell behaviors, and genetic and mechanical perturbations to determine how apoptosis is controlled during epithelial tissue development. We found that early in cell lives and well before extrusion, apoptosis commitment is linked to two distinct geometric features: a small apical area compared with other cells within the tissue and a small relative apical area with respect to the immediate neighboring cells. We showed that these global and local geometric characteristics are sufficient to recapitulate the tissue-scale apoptotic pattern. Furthermore, we established that the coupling between these two geometric features and apoptotic cells is dependent on the Hippo/YAP and Notch pathways. Overall, by exploring the links between cell geometry and apoptosis commitment, our work provides important insights into the spatial regulation of cell death in tissues and improves our understanding of the mechanisms that control cell number and tissue size.
Collapse
Affiliation(s)
- Victoire M L Cachoux
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Maria Balakireva
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Mélanie Gracia
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Floris Bosveld
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Jesús M López-Gay
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Aude Maugarny
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Isabelle Gaugué
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Florencia di Pietro
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Stéphane U Rigaud
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Lorette Noiret
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France
| | - Boris Guirao
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| | - Yohanns Bellaïche
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR3215, INSERM U934, Genetics and Developmental Biology, 75005 Paris, France.
| |
Collapse
|
43
|
Wang T, Wang D, Sun Y, Zhuang T, Li X, Yang H, Zang Y, Liu Z, Yang P, Zhang C, Cui J, Fu M, Zhang S, Su P, Li Z, Zhu J, Ding Y. Regulation of the Hippo/YAP axis by CXCR7 in the tumorigenesis of gastric cancer. J Exp Clin Cancer Res 2023; 42:297. [PMID: 37950281 PMCID: PMC10636825 DOI: 10.1186/s13046-023-02870-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/18/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The Hippo pathway is crucial in organ size control and tumorigenesis. Dysregulation of the Hippo/YAP axis is commonly observed in gastric cancer, while effective therapeutic targets for the Hippo/YAP axis are lacking. Identification of reliable drug targets and the underlying mechanisms that could inhibit the activity of the Hippo/YAP axis and gastric cancer progression is urgently needed. METHODS We used several gastric cancer cell lines and xenograft models and performed immunoblotting, qPCR, and in vivo studies to investigate the function of CXCR7 in gastric cancer progression. RESULTS In our current study, we demonstrate that the membrane receptor CXCR7 (C-X-C chemokine receptor 7) is an important modulator of the Hippo/YAP axis. The activation of CXCR7 could stimulate gastric cancer cell progression through the Hippo/YAP axis in vitro and in vivo, while pharmaceutical inhibition of CXCR7 via ACT-1004-1239 could block tumorigenesis in gastric cancer. Molecular studies revealed that the activation of CXCR7 could dephosphorylate YAP and facilitate YAP nuclear accumulation and transcriptional activation in gastric cancer. CXCR7 functions via G-protein Gαq/11 and Rho GTPase to activate YAP activity. Interestingly, ChIP assays showed that YAP could bind to the promoter region of CXCR7 and facilitate its gene transcription, which indicates that CXCR7 is both the upstream signalling and downstream target of the Hippo/YAP axis in gastric cancer. CONCLUSION In general, we identified a novel positive feedback loop between CXCR7 and the Hippo/YAP axis, and blockade of CXCR7 could be a plausible strategy for gastric cancer.
Collapse
Affiliation(s)
- Tianshi Wang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Dehai Wang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Yanan Sun
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Ting Zhuang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, P. R. China
| | - Xin Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Huijie Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Yifeng Zang
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Ziping Liu
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Penghe Yang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Chenmiao Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Jiayao Cui
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Mingxi Fu
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Shuqing Zhang
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China
| | - Peng Su
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| | - Zhongbo Li
- Xinxiang Key Laboratory of Tumor Migration and Invasion Precision Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, Henan Province, P. R. China.
| | - Jian Zhu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yinlu Ding
- Department of General Surgery, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China.
| |
Collapse
|
44
|
Rader AE, Bayarmagnai B, Frolov MV. Combined inactivation of RB and Hippo converts differentiating Drosophila photoreceptors into eye progenitor cells through derepression of homothorax. Dev Cell 2023; 58:2261-2274.e6. [PMID: 37848027 PMCID: PMC10842633 DOI: 10.1016/j.devcel.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 08/06/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
The retinoblastoma (RB) and Hippo pathways interact to regulate cell proliferation and differentiation. However, the mechanism of interaction is not fully understood. Drosophila photoreceptors with inactivated RB and Hippo pathways specify normally but fail to maintain their neuronal identity and dedifferentiate. We performed single-cell RNA sequencing to elucidate the cause of dedifferentiation and to determine the fate of these cells. We find that dedifferentiated cells adopt a progenitor-like fate due to inappropriate activation of the retinal differentiation suppressor homothorax (hth) by Yki/Sd. This results in the activation of a distinct Yki/Hth transcriptional program, driving photoreceptor dedifferentiation. We show that Rbf physically interacts with Yki and, together with the GAGA factor, inhibits the hth expression. Thus, RB and Hippo pathways cooperate to maintain photoreceptor differentiation by preventing inappropriate expression of hth in differentiating photoreceptors. Our work highlights the importance of both RB and Hippo pathway activities for maintaining the state of terminal differentiation.
Collapse
Affiliation(s)
- Alexandra E Rader
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Battuya Bayarmagnai
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Maxim V Frolov
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
45
|
Wang H, Yu H, Huang T, Wang B, Xiang L. Hippo-YAP/TAZ signaling in osteogenesis and macrophage polarization: Therapeutic implications in bone defect repair. Genes Dis 2023; 10:2528-2539. [PMID: 37554194 PMCID: PMC10404961 DOI: 10.1016/j.gendis.2022.12.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 01/18/2023] Open
Abstract
Bone defects caused by diseases or surgery are a common clinical problem. Researchers are devoted to finding biological mechanisms that accelerate bone defect repair, which is a complex and continuous process controlled by many factors. As members of transcriptional costimulatory molecules, Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) play an important regulatory role in osteogenesis, and they affect cell function by regulating the expression of osteogenic genes in osteogenesis-related cells. Macrophages are an important group of cells whose function is regulated by YAP/TAZ. Currently, the relationship between YAP/TAZ and macrophage polarization has attracted increasing attention. In bone tissue, YAP/TAZ can realize diverse osteogenic regulation by mediating macrophage polarization. Macrophages polarize into M1 and M2 phenotypes under different stimuli. M1 macrophages dominate the inflammatory response by releasing a number of inflammatory mediators in the early phase of bone defect repair, while massive aggregation of M2 macrophages is beneficial for inflammation resolution and tissue repair, as they secrete many anti-inflammatory and osteogenesis-related cytokines. The mechanism of YAP/TAZ-mediated macrophage polarization during osteogenesis warrants further study and it is likely to be a promising strategy for bone defect repair. In this article, we review the effect of Hippo-YAP/TAZ signaling and macrophage polarization on bone defect repair, and highlight the regulation of macrophage polarization by YAP/TAZ.
Collapse
Affiliation(s)
- Haochen Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tianyu Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Xiang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
46
|
Waghmare I, Gangwani K, Rai A, Singh A, Kango-Singh M. A Tumour-Specific Molecular Network Promotes Tumour Growth in Drosophila by Enforcing a JNK-YKI Feedforward Loop. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.561369. [PMID: 37904920 PMCID: PMC10614921 DOI: 10.1101/2023.10.18.561369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Cancer cells expand rapidly in response to altered intercellular and signalling interactions to achieve hallmarks of cancer. Impaired cell polarity combined with activated oncogenes is known to promote several hallmarks of cancer e.g., activating invasion by increased activity of Jun N-terminal kinase (JNK), and sustained proliferative signalling by increased activity of Hippo effector Yorkie (Yki). Thus, JNK, Yki, and their downstream transcription factors have emerged as synergistic drivers of tumour growth through pro-tumour signalling and intercellular interactions like cell-competition. However, little is known about the signals that converge onto JNK and Yki in tumour cells that enable the tumour cells to achieve hallmarks of cancer. Here, using mosaic models of cooperative oncogenesis ( Ras V12 , scrib - ) in Drosophila , we show that Ras V12 , scrib - tumour cells grow by activation of a previously unidentified network comprising Wingless (Wg), Dronc, JNK and Yki. We show that Ras V12 , scrib - cells show increased Wg, Dronc, JNK, and Yki signalling, and all of these signals are required for the growth of Ras V12 , scrib - tumours. We report that Wg and Dronc converge onto a JNK-Yki self-reinforcing positive feedback signal-amplification loop that promotes tumour growth. We found that Wg-Dronc-Yki-JNK molecular network is specifically activated in polarity-impaired tumour cells and not in normal cells where apical basal polarity is intact. Our findings suggest that identification of molecular networks may provide significant insights about the key biologically meaningful changes in signalling pathways, and paradoxical signals that promote Tumourigenesis.
Collapse
|
47
|
Zhou L, Guo H, Liao Q, Zou J, Le Y, Fang Z, Xiong J, Huang S, Deng J, Xiang X. miR-3133 inhibits gastrointestinal cancer progression through activation of Hippo and p53 signalling pathways via multi-targets. J Cell Mol Med 2023; 27:3090-3106. [PMID: 37555915 PMCID: PMC10568676 DOI: 10.1111/jcmm.17880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Malignant cell growth and chemoresistance, the main obstacles in treating gastrointestinal cancer (GIC), rely on the Hippo and p53 signalling pathways. However, the upstream regulatory mechanisms of these pathways remain complex and poorly understood. METHODS Immunohistochemistry (IHC), western blot and RT-qPCR were used to analyse the expression of RNF146, miR-3133 and key components of Hippo and p53 pathway. CCK-8, colony formation, drug sensitivity assays and murine xenograft models were used to investigate the effect of RNF146 and miR-3133 in GIC. Further exploration of the upstream regulatory mechanism was performed using bioinformatics analysis, dual-luciferase reporter gene, immunoprecipitation assays and bisulfite sequencing PCR (BSP). RESULTS Clinical samples, in vitro and in vivo experiments demonstrated that RNF146 exerts oncogenic effects in GIC by regulating the Hippo pathway. Bioinformatics analysis identified a novel miRNA, miR-3133, as an upstream regulatory factor of RNF146. fluorescence in situ hybridization and RT-qPCR assays revealed that miR-3133 was less expressed in gastrointestinal tumour tissues and was associated with adverse pathological features. Functional assays and animal models showed that miR-3133 promoted the proliferation and chemotherapy sensitivity of GIC cells. miR-3133 affected YAP1 protein expression by targeting RNF146, AGK and CUL4A, thus activating the Hippo pathway. miR-3133 inhibited p53 protein degradation and extended p53's half-life by targeting USP15, SPIN1. BSP experiments confirmed that miR-3133 promoter methylation is an important reason for its low expression. CONCLUSION miR-3133 inhibits GIC progression by activating the Hippo and p53 signalling pathways via multi-targets, including RNF146, thereby providing prognostic factors and valuable potential therapeutic targets for GIC.
Collapse
Affiliation(s)
- Ling Zhou
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Hui Guo
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Quan Liao
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jianping Zou
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Yi Le
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Ziling Fang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jianping Xiong
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Shanshan Huang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jun Deng
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Xiaojun Xiang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| |
Collapse
|
48
|
Liu M, Xie XJ, Li X, Ren X, Sun J, Lin Z, Hemba-Waduge RUS, Ji JY. Transcriptional coupling of telomeric retrotransposons with the cell cycle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.30.560321. [PMID: 37808851 PMCID: PMC10557779 DOI: 10.1101/2023.09.30.560321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Instead of employing telomerases to safeguard chromosome ends, dipteran species maintain their telomeres by transposition of telomeric-specific retrotransposons (TRs): in Drosophila , these are HeT-A , TART , and TAHRE . Previous studies have shown how these TRs create tandem repeats at chromosome ends, but the exact mechanism controlling TR transcription has remained unclear. Here we report the identification of multiple subunits of the transcription cofactor Mediator complex and transcriptional factors Scalloped (Sd, the TEAD homolog in flies) and E2F1-Dp as novel regulators of TR transcription and telomere length in Drosophila . Depletion of multiple Mediator subunits, Dp, or Sd increased TR expression and telomere length, while over-expressing E2F1-Dp or knocking down the E2F1 regulator Rbf1 (Retinoblastoma-family protein 1) stimulated TR transcription, with Mediator and Sd affecting TR expression through E2F1-Dp. The CUT&RUN analysis revealed direct binding of CDK8, Dp, and Sd to telomeric repeats. These findings highlight the essential role of the Mediator complex in maintaining telomere homeostasis by regulating TR transcription through E2F1-Dp and Sd, revealing the intricate coupling of TR transcription with the host cell-cycle machinery, thereby ensuring chromosome end protection and genomic stability during cell division.
Collapse
|
49
|
Yamashita T, Ohde T, Nakamura T, Ishimaru Y, Watanabe T, Tomonari S, Nakamura Y, Noji S, Mito T. Involvement of the scalloped gene in morphogenesis of the wing margin via regulating cell growth in a hemimetabolous insect Gryllus bimaculatus. Dev Growth Differ 2023; 65:348-359. [PMID: 37310211 DOI: 10.1111/dgd.12869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/14/2023]
Abstract
The acquisition of wings was a key event in insect evolution. As hemimetabolous insects were the first group to acquire functional wings, establishing the mechanisms of wing formation in this group could provide useful insights into their evolution. In this study, we aimed to elucidate the expression and function of the gene scalloped (sd), which is involved in wing formation in Drosophila melanogaster, and in Gryllus bimaculatus mainly during postembryonic development. Expression analysis showed that sd is expressed in the tergal edge, legs, antennae, labrum, and cerci during embryogenesis and in the distal margin of the wing pads from at least the sixth instar in the mid to late stages. Because sd knockout caused early lethality, nymphal RNA interference experiments were performed. Malformations were observed in the wings, ovipositor, and antennae. By analyzing the effects on wing morphology, it was revealed that sd is mainly involved in the formation of the margin, possibly through the regulation of cell proliferation. In conclusion, sd might regulate the local growth of wing pads and influence wing margin morphology in Gryllus.
Collapse
Grants
- 17H03945 Ministry of Education, Culture, Sports, Science and Technology
- 19H02970 Ministry of Education, Culture, Sports, Science and Technology
- 19K06691 Ministry of Education, Culture, Sports, Science and Technology
- 20K21436 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Takahisa Yamashita
- Graduate School of Advanced Technology and Science, Tokushima University, Tokushima, Japan
| | - Takahiro Ohde
- Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Taro Nakamura
- Division of Evolutionary Developmental Biology, National Institute for Basic Biology, Okazaki, Japan
| | - Yoshiyasu Ishimaru
- Graduate School of Sciences and Technology for Innovation, Tokushima University, Tokushima, Japan
| | - Takahito Watanabe
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Sayuri Tomonari
- Technical Support Department, Tokushima University, Tokushima, Japan
| | - Yuki Nakamura
- Graduate School of Advanced Technology and Science, Tokushima University, Tokushima, Japan
| | - Sumihare Noji
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| | - Taro Mito
- Bio-Innovation Research Center, Tokushima University, Tokushima, Japan
| |
Collapse
|
50
|
Yang L, Han D, Wang ZA, Chen N, Zuo H, Guo Z, Xu M, Weng S, He J, Xu X. The Hippo-Yki pathway downstream transcription factor Scalloped negatively regulates immune defense against Vibrio parahaemolyticus infection in shrimp. FISH & SHELLFISH IMMUNOLOGY 2023:108917. [PMID: 37355218 DOI: 10.1016/j.fsi.2023.108917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 06/26/2023]
Abstract
The Hippo-Yki signaling pathway plays a crucial role in numerous biological processes. Previous studies have demonstrated the significance of signal transduction components of the Hippo pathway in the immune response of shrimp. In this study, the downstream transcription factor of the Hippo signaling pathway, Scalloped, was analyzed in the context of Vibrio parahaemolyticus infection in Pacific white shrimp, Penaeus vannamei. Upon bacterial and fungal infections, the expression of Scalloped was upregulated in hemocytes. Scalloped was found to localize in the nucleus and interact with the Hippo pathway downstream transcriptional co-activator Yki. With the assistance of Yki, Scalloped activated the promoter of Cactus, which is a cytoplasmic inhibitor of the NF-κB pathway, leading to the inhibition of the nuclear translocation of the NF-κB family member Dorsal in shrimp. By inhibiting the Dorsal pathway, Scalloped reduced the expression of immune functional proteins and negatively regulated the immune response against bacterial infection in shrimp. RNAi-mediated silencing of Scalloped significantly enhanced the survival rate of V. parahaemolyticus-infected shrimp and reduced the bacterial load in tissues. These findings demonstrate the potential of Scalloped as a therapeutic target for vibriosis in crustaceans and contribute to our understanding of the shrimp's antibacterial defense and the functional roles of Hippo signaling in animal immunity.
Collapse
Affiliation(s)
- Linwei Yang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Deyu Han
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Zi-Ang Wang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Nuo Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Hongliang Zuo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Zhixun Guo
- South China Sea Fisheries Research Institute (CAFS), Guangzhou, 510300, PR China
| | - Menghuang Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China
| | - Shaoping Weng
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China
| | - Jianguo He
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China
| | - Xiaopeng Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, PR China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou, PR China; Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|