1
|
Hang Q, Li W, Guo J, Zuo S, Yang Y, Wu C, Yong W, Li C, Gu J, Hou S. Inhibitory effects of β-galactoside α2,6-sialyltransferase 1 on the Hippo pathway in breast cancer cells. J Biol Chem 2025:110266. [PMID: 40409546 DOI: 10.1016/j.jbc.2025.110266] [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: 11/12/2024] [Revised: 05/01/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025] Open
Abstract
The Hippo signaling pathway is crucial in pathological functions such as tumors. Yes-associated protein (YAP), a well-known downstream effector of the Hippo pathway, has been intensively studied; emerging evidence suggests that multiple cell membrane receptors can regulate the Hippo pathway. However, the mechanistic roles of these upstream pathways remain largely unknown. Here, we identified the β-galactoside α2,6-sialyltransferase 1 (ST6GAL1) catalyzed α2,6-sialylation as a pivotal upstream modulator of Hippo pathway by a glycosyltransferases (GTs) overexpression sub-library screening. Depletion of ST6GAL1 results in increased phosphorylation of LATS1 and YAP, which induces YAP's nuclear localization, transcriptional activity, and multiple biological functions in breast cancer cells, including cell adhesion, spreading, growth, migration, and metastasis. These phenotypes were majorly due to the altered signal transduction of cell surface receptors, as deletion of ST6GAL1 exhibited attenuated GPCR, EGFR, and Integrins response and suppression of dephosphorylation of YAP. Mechanistically, these representative membrane receptors are α2,6-sialylated proteins, and their α2,6-sialylation could be inhibited by β-galactoside α2,3-sialyltransferase 4 (ST3GAL4) via substrate competition. In addition, the α2,6-sialylation is essential for Integrin β1-EGFR/LPAR4 complex formation. Altogether, our findings demonstrate ST6GAL1 is an upstream negative regulator of the Hippo pathway in breast cancer cells, providing a new insight into the regulation between N-glycosylation and Hippo signaling.
Collapse
Affiliation(s)
- Qinglei Hang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China; Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining, Jiangsu 221200, China; Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan.
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Jingya Guo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Shiying Zuo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Yawen Yang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Can Wu
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Wen Yong
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Caimin Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981-8558, Japan.
| | - Sicong Hou
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu 225000, China.
| |
Collapse
|
2
|
Obanya DI, Wootton LM, Morgan EL. Advances in understanding the mechanisms of the human papillomavirus oncoproteins. Biochem Soc Trans 2025:BST20253041. [PMID: 40380881 DOI: 10.1042/bst20253041] [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: 02/13/2025] [Accepted: 05/02/2025] [Indexed: 05/19/2025]
Abstract
High-risk human papillomaviruses (HPVs) are responsible for almost all cervical cancer cases and a growing number of oropharyngeal and anogenital cancers. The primary HPV oncoproteins, E6 and E7, act together to manipulate multiple cellular pathways that can ultimately result in malignant transformation. This includes the deregulation of several signalling pathways that regulate cell proliferation, cell cycle progression and cell survival. Although multiple functions of HPV E6 and E7 in driving oncogenesis are well known, recent studies have uncovered novel oncogenic functions of the HPV oncoproteins, including the manipulation of emerging mechanisms of cancer development, such as epigenetic modifications, cellular plasticity and genomic instability. This review explores current advances in understanding how the HPV oncoproteins interact with these cellular processes, highlighting potential therapeutic targets in HPV-associated cancers.
Collapse
Affiliation(s)
| | | | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, U.K
| |
Collapse
|
3
|
Hou Y, Zhang L, Wang W, Cheng K, Wang H, Ji Y, Su X, Hao M. Down-regulation of miR-125b by HPV16 E6 might promote cervical cancer progression through TAZ/TEAD. Front Oncol 2025; 15:1444874. [PMID: 40303991 PMCID: PMC12037404 DOI: 10.3389/fonc.2025.1444874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Background Abnormal gene expression due to the dysregulation of microRNAs (miRNAs) often occurred in the initiation or progression of cancers. In this attempt, we investigate whether miR-125b regulates biological behaviors of cervical cancer caused by HPV16 through TAZ. Methods Through the application of bioinformatics analysis techniques, differentially expressed miRNAs relevant to cervical cancer were identified. Cervical tissue specimens were collected from 15 patients with HPV16-positive cervical squamous cell carcinoma (stages IA-IIA), 15 patients with high-grade squamous intraepithelial lesion (HSIL), and 10 patients experiencing chronic cervical inflammation at the Second Hospital of Shanxi Medical University between May 2022 and May 2023. The quantitative assessment of miR-125b expression was conducted via real-time quantitative reverse transcription PCR(RT-qPCR). The potential regulatory relationship between miR-125b and TAZ was assessed using a dual-luciferase reporter assay. Within cervical squamous cell carcinoma SiHa cells, models were established using miR-125b mimic and inhibitor constructs to scrutinize cellular physiological processes and assess the expression profiles of miR-125b, TAZ, TEAD, and E6. Additionally, an HPV16 E6 overexpression cellular model was generated, and the expression patterns of miR-125b and its downstream targets were analyzed by RT-qPCR. Results Tissue validation corroborated these findings, demonstrating a significant reduction in miR-125b expression levels in HSIL and cervical squamous cell carcinoma compared to normal cervical tissue (P < 0.05). Functional assays using a miR-125b mimic revealed inhibited proliferation, migration, and invasion of SiHa cells along with enhanced apoptosis, concomitant with decreased expression of HPV16 E6, TAZ, and TEAD mRNA. Conversely, these effects were reversed upon introduction of a miR-125b inhibitor. Notably, overexpression of HPV16 E6 was associated with suppressed miR-125b expression (P < 0.05) and enhanced TAZ and TEAD expression (P < 0.05), as corroborated by western blot analysis. Conclusion HPV E6 promotes the malignant progression of cervical cancer cells by downregulating miR-125b, which targets TAZ, thus regulating the Hippo pathway. Consequently, miR-125b emerges as a promising therapeutic target for HPV-induced cervical cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Min Hao
- Department of Gynecology and Obstetrics, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
4
|
Lv X, Liu J, Islam K, Ruan J, He C, Chen P, Huang C, Wang H, Dhar A, Moness M, Shi D, Murphy S, Zhao X, Yang S, Montoute I, Polakkattil A, Chung A, Ruiz E, Carbajal B, Padavala A, Chen L, Hua G, Chen X, Davis JS, Wang C. Hyperactivated YAP1 is essential for sustainable progression of renal clear cell carcinoma. Oncogene 2025:10.1038/s41388-025-03354-8. [PMID: 40210757 DOI: 10.1038/s41388-025-03354-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025]
Abstract
The most notable progress in renal clear cell carcinoma (ccRCC) in the past decades is the introduction of drugs targeting the VHL-HIF signaling pathway-associated angiogenesis. However, mechanisms underlying the development of VHL mutation-independent ccRCC are unclear. Here we provide evidence that the disrupted Hippo-YAP signaling contributes to the development of ccRCC independent of VHL alteration. We found that YAP1 and its primary target genes are frequently upregulated in ccRCC and the upregulation of these genes is associated with unfavorable patient outcomes. Research results derived from our in vitro and in vivo experimental models demonstrated that, under normoxic conditions, hyperactivated YAP1 drives the expression of FGFs to stimulate the proliferation of tumor and tumor-associated endothelial cells in an autocrine/paracrine manner. When rapidly growing cancer cells create a hypoxic environment, hyperactivated YAP1 in cancer cells induces the production of VEGF, which promotes the angiogenesis of tumor-associated endothelial cells, leading to improved tumor microenvironment and continuous tumor growth. Our study indicates that hyperactivated YAP1 is essential for maintaining ccRCC progression, and targeting the dual role of hyperactivated YAP1 represents a novel strategy to improve renal carcinoma therapy.
Collapse
Affiliation(s)
- Xiangmin Lv
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiyuan Liu
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kazi Islam
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinpeng Ruan
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunbo He
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Peichao Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cong Huang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anjali Dhar
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Dartmouth College, Hanover, NH, USA
| | - Madelyn Moness
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Davie Shi
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Savannah Murphy
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingeng Zhao
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Siyi Yang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Montoute
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Aneeta Polakkattil
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andie Chung
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Ruiz
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Brianna Carbajal
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Stem cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Alekhya Padavala
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Chen
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guohua Hua
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - John S Davis
- Department of Obstetrics and Gynecology, Olson Center for Women's Health, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Cheng Wang
- Department of Obstetrics and Gynecology, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Lao Z, Chen X, Pan B, Fang B, Yang W, Qian Y. Pharmacological regulators of Hippo pathway: Advances and challenges of drug development. FASEB J 2025; 39:e70438. [PMID: 40100056 DOI: 10.1096/fj.202401895rr] [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: 09/06/2024] [Revised: 02/14/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025]
Abstract
The Hippo signaling pathway is crucial in regulating organ size, tumor progression, tissue regeneration, and bone homeostasis. Inactivation of the Hippo pathway results in the nuclear translocation and activation of YAP/TAZ. This activation not only promotes tumor progression but also enhances tissue regeneration, wound healing, and maintenance of bone stability Although its discovery occurred over two decades ago, developing effective inhibitors or activators for the Hippo pathway remains challenging. Recently, however, the pace of advancements in developing Hippo signaling-related agonists and antagonists has accelerated, with some drugs that target TEAD advancing to clinical trials and showing promise for treating related diseases. This review summarizes the progress in research on Hippo signaling-related agonists and inhibitors, offering an in-depth analysis of their regulatory mechanisms, pharmacological properties, and potential in vivo applications.
Collapse
Affiliation(s)
- Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Bin Fang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Lv X, Liu J, Ruan J, Chen P, He C, Zhao X, Huang C, Chen L, Wang H, Hua G, Shi D, Yang S, Moness ML, Montoute I, Dhar A, Chen X, Kumar R, Lu H, Sadreyev R, Yeku O, Wu X, Davis JS, Wang C. Targeting the disrupted Hippo signaling to prevent neoplastic renal epithelial cell immune evasion. Nat Commun 2025; 16:2858. [PMID: 40128178 PMCID: PMC11933345 DOI: 10.1038/s41467-025-57697-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 02/26/2025] [Indexed: 03/26/2025] Open
Abstract
Large-scale cancer genetic/genomic studies demonstrated that papillary renal cell carcinoma (pRCC) is featured with a frequent shallow deletion of the upstream tumor suppressors of the Hippo/YAP signaling pathway, suggesting that this signaling pathway may play a role in pRCC development. Here we develop a transgenic mouse model with a renal epithelial cell-specific hyperactivation of YAP1 and find that hyperactivation of YAP1 can induce dedifferentiation and transformation of renal tubular epithelial cells leading to the development of pRCC. We analyze at the single-cell resolution the cellular landscape alterations during cancer initiation and progression. Our data indicate that the hyperactivated YAP1, via manipulating multiple signaling pathways, induces epithelial cell transformation, MDSC (Myeloid-derived suppressor cells) accumulation, and pRCC development. Interestingly, we find that depletion of MDSC blocks YAP1-induced kidney overgrowth and tumorigenesis. Inhibiting YAP1 activity with MGH-CP1, a recently developed TEAD inhibitor, impedes MDSC accumulation and suppresses tumor development. Our results identify the disrupted Hippo/YAP signaling as a major contributor to pRCC and suggest that targeting the disrupted Hippo pathway represents a plausible strategy to prevent and treat pRCC.
Collapse
Affiliation(s)
- Xiangmin Lv
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jiyuan Liu
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jinpeng Ruan
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peichao Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Chunbo He
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingeng Zhao
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cong Huang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Chen
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hongbo Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Guohua Hua
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Davie Shi
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Siyi Yang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Madelyn L Moness
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Isabelle Montoute
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anjali Dhar
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Cell Biology, Dartmouth College, Hanover, NH, USA
| | - Xingcheng Chen
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Raj Kumar
- Division of Hematology and oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hu Lu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Oladapo Yeku
- Division of Hematology and oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Gynecological Cancer Program, Dana-Farber/Harvard Cancer Center, Boston, MA, USA.
| |
Collapse
|
7
|
Padash Barmchi M, Hassan RN, Afkhami M, Masly JP, Brown H, Collins QP, Grunsted MJ. Drosophila model of HPV18-Induced pathogenesis reveals a role for E6 oncogene in regulation of NF-κB and Wnt to inhibit apoptosis. Tumour Virus Res 2025; 19:200316. [PMID: 40074036 PMCID: PMC12008589 DOI: 10.1016/j.tvr.2025.200316] [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: 01/02/2025] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025] Open
Abstract
Cancers caused by high-risk human papillomavirus (HPV) remain a significant health threat resulting in more than 300,000 deaths, annually. Persistent expression of two HPV oncogenes, E6 and E7, are necessary for cancer development and progression. E6 has several functions contributing to tumorigenesis one of which is blocking programmed cell death, apoptosis. The detailed mechanism of anti-apoptosis function of E6 is not fully understood. Here, using a Drosophila model of HPV18E6 and the human UBE3A-induced pathogenesis, we show that anti-apoptotic function of E6 is conserved in Drosophila. We demonstrate that the Drosophila homologs of human NF-κB transcription factors, Dorsal and Dif are proapoptotic. They induce the expression of Wingless (Wg, the Drosophila homolog of human Wnt), leading to apoptosis. Our results indicate that E6 oncogene inhibits apoptosis by downregulating the expression of Wg, Dorsal, and Dif. Additionally, we find that Dorsal and Dif, not only promote apoptosis but also regulate autophagy and necrosis. Dorsal promotes autophagy while Dif counteracts it, inducing the formation of acidic vacuoles and necrosis. Interestingly, although E6 blocks the proapoptotic function of Dorsal and Dif, it lacks the ability to interfere with their role in apoptosis-independent cell death. Given the high conservation of NF-κB transcription factors our results provide new insight into potential mechanisms mediated by NF-κB to intervene with cell immortalization action of E6 oncoprotein in HPV-infected cells.
Collapse
Affiliation(s)
| | - Rami N Hassan
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| | - Mehrnaz Afkhami
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - John P Masly
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA
| | - Harrison Brown
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Children's Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Quincy P Collins
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, BC, Canada
| | - Michael J Grunsted
- School of Biological Sciences, University of Oklahoma, Norman, OK, USA; College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
8
|
Zhang Y, Liu Y, Deng Q, Liu L, Zhu W. USP43 drives cervical carcinoma progression through regulation of the Hippo/TAZ pathway. Int Immunopharmacol 2025; 149:114217. [PMID: 39922114 DOI: 10.1016/j.intimp.2025.114217] [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: 11/11/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Cervical carcinoma (CC) poses significant health challenges, with its pathogenesis not fully understood. While ubiquitin specific peptidase 43 (USP43) is implicated in various cancers, its role in CC and regulation of the Hippo/Transcriptional Co-Activator With PDZ-Binding Motif (TAZ) pathway remain unexplored. This study examines USP43's impact on CC progression and its interaction with TAZ. METHODS USP43 expression levels in CC tissues and cell lines were assessed using reverse transcription real-time polymerase chain reaction (RT-qPCR) and Western blot. The effects of USP43 silencing on cell proliferation, migration, and invasion were evaluated through cell counting Kit-8 (CCK-8), 5-ethynyl-2'deoxyuridine (EdU), colony formation, and transwell assays. Immunofluorescence staining and co-immunoprecipitation (Co-IP) assays were used to explore the interaction between USP43 and TAZ. Polyubiquitination assays were performed to evaluate ubiquitination and stability of TAZ, and cycloheximide (CHX) chase experiments determined the half-life of TAZ. In vivo studies using BALB/c nude mice examined the impact of USP43 knockdown on tumor growth and metastasis. RESULTS USP43 was overexpressed in CC tissues and cell lines. Silencing of USP43 reduced cell proliferation, migration, invasion, and the epithelial-mesenchymal transition (EMT) process. Co-IP and ubiquitination assays revealed that USP43 interacted with and stabilized TAZ by inhibiting TAZ ubiquitination. CHX chase experiments confirmed that USP43 prolonged TAZ protein stability. In vivo, USP43 knockdown led to reduced tumor growth and lung metastasis. Overexpression of TAZ reversed the inhibitory effects of USP43 silencing on CC cell proliferation, migration, invasion and EMT CONCLUSION: USP43 promotes CC cell proliferation, migration, invasion, and epithelial-to-mesenchymal transition (EMT) by activating the Hippo/TAZ pathway. These findings enhance our understanding of USP43's role in CC progression and highlight potential therapeutic targets for the treatment of CC.
Collapse
Affiliation(s)
- Yanhua Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China; Department of Obstetrics and Gynecology, Binhai County People's Hospital, Yancheng 224000, Jiangsu, China
| | - Yujing Liu
- Suzhou Research Center of Medical School, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou 215000, Jiangsu, China
| | - Qicheng Deng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Lifen Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China
| | - Weipei Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou 215000, Jiangsu, China.
| |
Collapse
|
9
|
Wootton LM, Morgan EL. Ubiquitin and ubiquitin-like proteins in HPV-driven carcinogenesis. Oncogene 2025; 44:713-723. [PMID: 40011575 PMCID: PMC11888991 DOI: 10.1038/s41388-025-03310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/20/2025] [Accepted: 02/12/2025] [Indexed: 02/28/2025]
Abstract
Persistent infection with high-risk (HR) human papillomaviruses (HPVs) is responsible for approximately 5% of cancer cases worldwide, including a growing number of oropharyngeal and anogenital cancers. The major HPV oncoproteins, E6 and E7, act together to manipulate cellular pathways involved in the regulation of proliferation, the cell cycle and cell survival, ultimately driving malignant transformation. Protein ubiquitination and the ubiquitin proteasome system (UPS) is often deregulated upon viral infection and in oncogenesis. HPV E6 and E7 interact with and disrupt multiple components of the ubiquitination machinery to promote viral persistence, which can also result in cellular transformation and the formation of tumours. This review highlights the ways in which HPV manipulates protein ubiquitination and the ubiquitin-like protein pathways and how this contributes to tumour development. Furthermore, we discuss how understanding the interactions between HPV and the protein ubiquitination could lead to novel therapeutic targets that are of urgent need in HPV+ carcinomas.
Collapse
Affiliation(s)
| | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
10
|
Gnanagurusamy J, Krishnamoorthy S, Muruganatham B, Selvamurugan N, Muthusami S. Analysing the relevance of TGF-β and its regulators in cervical cancer to identify therapeutic and diagnostic markers. Gene 2025; 938:149166. [PMID: 39701195 DOI: 10.1016/j.gene.2024.149166] [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: 09/11/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
The role of transforming growth factor-beta (TGF-β) is dual, such that, it inhibits tumor development in initial stage and promotes metastasis in later stage. The present study is aimed to analyse the relevance of different types of TGF-β and their receptors on the overall survival (OS) and TGF-β driven gene expression in individuals with cervical cancer (CC) using ONCODB and GEPIA databases. The in-silico gene expression analysis showed, TGF-β1 and TGFβR2 are upregulated in cells infected with human papilloma virus (HPV)16, whereas, TGF-β2, TGFβR1 and TGFβR3 expression were downregulated. In HPV 18 infected cells, TGF-β1, TGF-β2 and TGFβR1 were downregulated, meanwhile, TGF-β3, TGFβR2 and TGFβR3 were upregulated. OS analysis of CC patients with different TGF-β expression revealed that, TGF-β1, TGF-β2, TGF-β3 and TGFβR2 were associated with reduced survival rate. Further, we identified four microRNAs (miRNAs) (hsa-miR-21-5p, hsa-miR-29b-3p, hsa-miR-101-3p and hsa-miR-130a-3p) interacted favorably with TGF-β in HPV 16 and 18 positive samples using MIENTURNET. This present review further emphasizes that, targeting TGF-β could be a novel and futuristic approach for CC management and therapeutics.
Collapse
Affiliation(s)
- Jayapradha Gnanagurusamy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Bharathi Muruganatham
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India; Centre for Bioinformatics, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur Chennai 603 203 Tamil Nadu, India
| | - Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India; Centre for Cancer Research, Karpagam Academy of Higher Education, Coimbatore 641021, Tamil Nadu, India.
| |
Collapse
|
11
|
Jia Q, Wang H, Bi B, Han X, Jia Y, Zhang L, Fang L, Thakur A, Cheng JC. Amphiregulin Downregulates E-cadherin Expression by Activating YAP/Egr-1/Slug Signaling in SKOV3 Human Ovarian Cancer Cells. Reprod Sci 2025; 32:404-416. [PMID: 39138796 DOI: 10.1007/s43032-024-01673-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: 03/07/2024] [Accepted: 08/01/2024] [Indexed: 08/15/2024]
Abstract
Amphiregulin (AREG) stimulates human epithelial ovarian cancer (EOC) cell invasion by downregulating E-cadherin expression. YAP is a transcriptional cofactor that has been shown to regulate tumorigenesis. This study aimed to examine whether AREG activates YAP in EOC cells and explore the roles of YAP in AREG-induced downregulation of E-cadherin and cell invasion. Analysis of the Cancer Genome Atlas (TCGA) showed that upregulation of AREG and EGFR were associated with poor survival in human EOC. Treatment of SKOV3 human EOC cells with AREG induced the activation of YAP. In addition, AREG downregulated E-cadherin, upregulated Egr-1 and Slug, and stimulated cell invasion. Using gain- and loss-of-function approaches, we showed that YAP was required for the AREG-upregulated Egr-1 and Slug expression. Furthermore, YAP was also involved in AREG-induced downregulation of E-cadherin and cell invasion. This study provides evidence that AREG stimulates human EOC cell invasion by downregulating E-cadherin expression through the YAP/Egr-1/Slug signaling.
Collapse
Affiliation(s)
- Qiongqiong Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Hailong Wang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Beibei Bi
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Han
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Jia
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lingling Zhang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lanlan Fang
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Avinash Thakur
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Jung-Chien Cheng
- Center for Reproductive Medicine, Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
12
|
Janiszewska J, Kostrzewska-Poczekaj M, Wierzbicka M, Brenner JC, Giefing M. HPV-driven oncogenesis-much more than the E6 and E7 oncoproteins. J Appl Genet 2025; 66:63-71. [PMID: 38907809 PMCID: PMC11761861 DOI: 10.1007/s13353-024-00883-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] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
High-risk human papillomaviruses are well-established drivers of several cancer types including cervical, head and neck, penile as well as anal cancers. While the E6 and E7 viral oncoproteins have proven to be critical for malignant transformation, evidence is also beginning to emerge suggesting that both host pathways and additional viral genes may also be pivotal for malignant transformation. Here, we focus on the role of host APOBEC genes, which have an important role in molecular editing including in the response to the viral DNA and their role in HPV-driven carcinogenesis. Further, we also discuss data developed suggesting the existence of HPV-derived miRNAs in HPV + tumors and their potential role in regulating the host transcriptome. Collectively, while recent advances in these two areas have added complexity to the working model of papillomavirus-induced oncogenesis, these discoveries have also shed a light onto new areas of research that will be required to fully understand the process.
Collapse
Affiliation(s)
- J Janiszewska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Kostrzewska-Poczekaj
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - M Wierzbicka
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
- Research & Development Centre, Regional Specialist Hospital Wroclaw, Wroclaw, Poland
- Faculty of Medicine, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - J C Brenner
- Department of Otolaryngology - Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - M Giefing
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland.
| |
Collapse
|
13
|
Nakagawa R, Beardsley A, Durney S, Hayward MK, Subramanyam V, Meyer NP, Wismer H, Goodarzi H, Weaver VM, Van de Mark D, Goga A. Tumor Cell Spatial Organization Directs EGFR/RAS/RAF Pathway Primary Therapy Resistance through YAP Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.26.615226. [PMID: 39386679 PMCID: PMC11463411 DOI: 10.1101/2024.09.26.615226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Non-small cell lung cancers (NSCLC) harboring common mutations in EGFR and KRAS characteristically respond transiently to targeted therapies against those mutations, but invariably, tumors recur and progress. Resistance often emerges through mutations in the therapeutic target or activation of alternative signaling pathways. Mechanisms of acute tumor cell resistance to initial EGFR (EGFRi) or KRASG12C (G12Ci) pathway inhibition remain poorly understood. Our study reveals that acute response to EGFR/RAS/RAF-pathway inhibition is spatial and culture context specific. In vivo, EGFR mutant tumor xenografts shrink by > 90% following acute EGFRi therapy, and residual tumor cells are associated with dense stroma and have increased nuclear YAP. Interestingly, in vitro EGFRi induced cell cycle arrest in NSCLC cells grown in monolayer, while 3D spheroids preferentially die upon inhibitor treatment. We find differential YAP nuclear localization and activity, driven by the distinct culture conditions, as a common resistance mechanism for selective EGFR/KRAS/BRAF pathway therapies. Forced expression of the YAPS127A mutant partially protects cells from EGFR-mediated cell death in spheroid culture. These studies identify YAP activation in monolayer culture as a non-genetic mechanism of acute EGFR/KRAS/BRAF therapy resistance, highlighting that monolayer vs spheroid cell culture systems can model distinct stages of patient cancer progression.
Collapse
Affiliation(s)
- Rachel Nakagawa
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrew Beardsley
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Sophia Durney
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Mary-Kate Hayward
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
| | - Vishvak Subramanyam
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, USA
| | - Nathaniel P. Meyer
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Harrison Wismer
- Biological Imaging Development CoLab, UCSF, San Francisco, CA, USA
| | - Hani Goodarzi
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Valerie M Weaver
- Department of Surgery and Center for Bioengineering and Tissue Regeneration, University of California San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Daniel Van de Mark
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
| | - Andrei Goga
- Department of Cell & Tissue Biology, University of California, San Francisco, CA, USA
- Department Of Medicine, University of California, San Francisco, San Francisco, CA, USA
- UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
14
|
Wei X, Si A, Zhao S, Fu Y, Li J, Aishanjiang K, Ma Y, Yu C, Yu B, Cui C, Wang H, Kong X, Li S, Kong X, Tong Y, Wu H. CircUCK2(2,3) promotes cancer progression and enhances synergistic cytotoxicity of lenvatinib with EGFR inhibitors via activating CNIH4-TGFα-EGFR signaling. Cell Mol Biol Lett 2025; 30:15. [PMID: 39885395 PMCID: PMC11781035 DOI: 10.1186/s11658-025-00690-1] [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: 08/20/2024] [Accepted: 01/08/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Circular (circ)RNAs have emerged as crucial contributors to cancer progression. Nonetheless, the expression regulation, biological functions, and underlying mechanisms of circRNAs in mediating hepatocellular carcinoma (HCC) progression remain insufficiently elucidated. METHODS We identified circUCK2(2,3) through circRNA sequencing, RT-PCR, and Sanger sequencing. CircUCK2(2,3) levels were measured in two independent HCC cohorts using quantitative real-time PCR (qRT-PCR). We explored the functions of circUCK2(2,3) using gain- and loss-of-function assays. Techniques such as RNA-sequencing, RNA immunoprecipitation (RIP), polysome fractionation, RNA pulldown, dual luciferase reporter assay, inhibitors of EGFR downstream signaling, CRISPR-Cas9, and medium transfer assays were employed to investigate the regulatory mechanisms and the protumoral activities of circUCK2(2,3). Additionally, in vitro cytotoxic assays and patient-derived xenograft (PDX) models assessed the effects of circUCK2(2,3) on the cytotoxic synergy of lenvatinib and EGFR inhibitors. RESULTS CircUCK2(2,3) is upregulated in HCC tissues and serves as an independent risk factor for poor recurrence-free survival. The expression of circUCK2(2,3) is independent on its host gene, UCK2, but is regulated by its upstream promoter and flanking inverted complementary sequences. Functionally, circUCK2(2,3) enhances HCC proliferation, migration, and invasion, both in vitro and in vivo. Mechanistically, by sponging miR-149-5p, circUCK2(2,3) increases CNIH4 levels, which in turn amplifies TGFα secretion, resulting in the activation of EGFR and downstream pAKT and pERK signaling pathways. Moreover, circUCK2(2,3) overexpression sensitizes HCC cells to EGFR inhibitors, and increases the synergistic cytotoxicity of combined lenvatinib and EGFR inhibitor treatment. CONCLUSIONS CircUCK2(2,3) regulates a novel oncogenic pathway, miR-149-5p-CNIH4-TGFα-EGFR, in HCC, presenting a viable therapeutic target and biomarker for the precision treatment of HCC.
Collapse
Affiliation(s)
- Xindong Wei
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, 201203, China
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Anfeng Si
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210015, China
| | - Shuai Zhao
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yi Fu
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jilei Li
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Kedeerya Aishanjiang
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, 831399, China
| | - Yujie Ma
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Chang Yu
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, 201203, China
| | - Bo Yu
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Chunhong Cui
- Basic Medical College, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Hui Wang
- Basic Medical College, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Xianming Kong
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Shibo Li
- Department of Infectious Disease, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, 316100, China.
| | - Xiaoni Kong
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, 201203, China.
| | - Ying Tong
- Department of Liver Surgery, School of Medicine, Renji Hospital, Shanghai JiaoTong University, Shanghai, 200003, China.
| | - Hailong Wu
- Clinical Research Center, Jiading District Central Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 201800, China.
- Collaborative Research Center for Biomedicines, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
- School of Pharmacy, Joint Innovation Laboratory for Cell Therapy Technology, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
15
|
Ajongbolo AO, Langhans SA. YAP/TAZ-associated cell signaling - at the crossroads of cancer and neurodevelopmental disorders. Front Cell Dev Biol 2025; 13:1522705. [PMID: 39936032 PMCID: PMC11810912 DOI: 10.3389/fcell.2025.1522705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
YAP/TAZ (Yes-associated protein/paralog transcriptional co-activator with PDZ-binding domain) are transcriptional cofactors that are the key and major downstream effectors of the Hippo signaling pathway. Both are known to play a crucial role in defining cellular outcomes, including cell differentiation, cell proliferation, and apoptosis. Aside from the canonical Hippo signaling cascade with the key components MST1/2 (mammalian STE20-like kinase 1/2), SAV1 (Salvador homologue 1), MOB1A/B (Mps one binder kinase activator 1A/B) and LATS1/2 (large tumor suppressor kinase 1/2) upstream of YAP/TAZ, YAP/TAZ activation is also influenced by numerous other signaling pathways. Such non-canonical regulation of YAP/TAZ includes well-known growth factor signaling pathways such as the epidermal growth factor receptor (EGFR)/ErbB family, Notch, and Wnt signaling as well as cell-cell adhesion, cell-matrix interactions and mechanical cues from a cell's microenvironment. This puts YAP/TAZ at the center of a complex signaling network capable of regulating developmental processes and tissue regeneration. On the other hand, dysregulation of YAP/TAZ signaling has been implicated in numerous diseases including various cancers and neurodevelopmental disorders. Indeed, in recent years, parallels between cancer development and neurodevelopmental disorders have become apparent with YAP/TAZ signaling being one of these pathways. This review discusses the role of YAP/TAZ in brain development, cancer and neurodevelopmental disorders with a special focus on the interconnection in the role of YAP/TAZ in these different conditions.
Collapse
Affiliation(s)
- Aderonke O. Ajongbolo
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
- Biological Sciences Graduate Program, University of Delaware, Newark, DE, United States
| | - Sigrid A. Langhans
- Division of Neurology and Nemours Biomedical Research, Nemours Children’s Health, Wilmington, DE, United States
| |
Collapse
|
16
|
Pu M, Xiao X, Lv S, Ran D, Huang Q, Zhou M, Lei Q, Kong L, Zhang Q. METTL3-dependent DLG2 inhibits the malignant progression of cervical cancer by inactivating the Hippo/YAP signaling. Hereditas 2025; 162:9. [PMID: 39856747 PMCID: PMC11762078 DOI: 10.1186/s41065-025-00365-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Discs large homolog 2 (DLG2) has been implicated in cancer development, yet its role in cervical cancer remains unclear. This study aims to explore the regulatory mechanism of DLG2 in cervical cancer and its clinical implications. METHODS Quantitative reverse transcription polymerase chain reaction and western blotting assays were employed to detect RNA and protein expression, respectively. Colony formation assay, 5-Ethynyl-2'-deoxyuridine assay, flow cytometry, and transwell assays were conducted for cell functional analysis. A xenograft mouse model assay was performed to analyze tumor tumorigenesis in vivo. m6A RNA immunoprecipitation assay was used to analyze the association of METTL3 and DLG2. RESULTS DLG2 was underexpressed in cervical cancer tissues and cells. Elevating DLG2 levels significantly suppressed cervical cancer cell proliferation, migration, and invasion, while promoting apoptosis. Additionally, DLG2 overexpression led to the deactivation of the Hippo/YAP signaling pathway. In vivo, DLG2 overexpression was shown to reduce tumor formation. We also discovered that METTL3 destabilized DLG2 mRNA through an m6A-dependent mechanism. Moreover, lowering DLG2 expression mitigated the effects of METTL3 silencing on cervical cancer cell malignancy. CONCLUSION DLG2 acted as a tumor suppressor in cervical cancer by inhibiting the Hippo/YAP signaling pathway. The METTL3-dependent regulation of DLG2 mRNA stability could be a critical factor in cervical cancer progression.
Collapse
Affiliation(s)
- Mei Pu
- Department of Obstetrics and Gynecology, Dazhou Vocational and Technical College, Dazhou, 635001, China
| | - Xia Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Shasha Lv
- Department of Obstetrics and Gynecology, Dazhou Vocational and Technical College, Dazhou, 635001, China
| | - Daqing Ran
- Department of Obstetrics and Gynecology, Dazhou Vocational and Technical College, Dazhou, 635001, China
| | - Qian Huang
- Department of Obstetrics and Gynecology, Dazhou Vocational and Technical College, Dazhou, 635001, China
| | - Mingming Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China
| | - Qirong Lei
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, China
| | - Lingshuang Kong
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, No. 181 Hanyu Road, Shapingba District, Chongqing, 400030, China.
| | - Qing Zhang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, China.
- Department of Nursing, The Affiliated Hospital of Southwest Medical University, No. 25 Taiping Street, Jiangyang District, Luzhou, 646000, China.
| |
Collapse
|
17
|
Suvanasuthi R, Therasakvichya S, Kanchanapiboon P, Promptmas C, Chimnaronk S. Analysis of precancerous lesion-related microRNAs for early diagnosis of cervical cancer in the Thai population. Sci Rep 2025; 15:142. [PMID: 39747449 PMCID: PMC11696073 DOI: 10.1038/s41598-024-84080-1] [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: 09/26/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
The incidence rate of cervical cancer (CC) is three times greater in Southeast Asia (SEA), where screening tests are less common than in Northern America, underlining a need for convenient self-diagnostic methods. The expression pattern of microRNAs (miRNAs) has been considered a molecular tool for non-invasive cancer diagnosis and prognosis. This study aimed at the development of the first miRNA biomarker panel for early detection of CC in Thai women. Genome-wide miRNA expression profiling was performed on cervical tissue and discharge samples from high-grade squamous intraepithelial lesion (HSIL) and adenocarcinoma in situ (AIS) subjects. Machine learning was used for handling imbalanced data and feature selection before differential expression analysis to identify significantly dysregulated miRNA panels. Pathway analysis was conducted to provide the cellular functions involved in CC progression. The study identified a shared 18-miRNA panel for both tissue and discharge, with which the prediction model distinguished HSIL and AIS from normal samples with an accuracy of 90.9%. Three dysregulated miRNAs comprised of miR-125b-1-3p, miR-487b-3p, and miR-1180-3p in CC were first described. Most of the miRNAs in the panel were down-regulated, whereas merely miR-142-3p was significantly up-regulated in HSIL and AIS, suggesting a convenient biomarker for detecting precancerous conditions. Moreover, our miRNA panel highlighted important roles played by the cell-cell interaction pathways in CC. Together, our miRNA panel hold promise as a biomarker for the early detection of cervical cancer with cervical discharge, offering the possibility for developing non-invasive diagnostic tools.
Collapse
Affiliation(s)
- Rooge Suvanasuthi
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, 73170, Thailand
- Faculty of Science, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Suwanit Therasakvichya
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Potjanee Kanchanapiboon
- Division of Nuclear Medicine, Department of Radiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Chamras Promptmas
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Sarin Chimnaronk
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.
- Siriraj Center of Research Excellence for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
18
|
Hakim RU, Amin T, Ul Islam SMB. Advances and Challenges in Cervical Cancer: From Molecular Mechanisms and Global Epidemiology to Innovative Therapies and Prevention Strategies. Cancer Control 2025; 32:10732748251336415. [PMID: 40267919 PMCID: PMC12034968 DOI: 10.1177/10732748251336415] [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: 01/30/2025] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
BackgroundIn the global scenario of public health, cervical cancer poses a major threat with high mortality rates, especially in women. New incidence cases and prevalence vary across different regions, as recently shown by GLOBOCAN data. The development of cervical cancer is primarily due to persistent infection by high-risk genotypes of human papillomavirus (HPV), which is a multifaceted process that is influenced by genetic, environmental, and lifestyle factors.PurposeThe goal of this study is to thoroughly investigate cervical cancer, including its etiology, molecular mechanisms, progression, diagnosis strategies, and current therapies. This review further highlights the transformative power of HPV vaccination and screening programs in curbing the disease's burden and potentially promising novel approaches like immunotherapy and targeted therapy.Research DesignThis is a narrative review article that summarizes previous literatures regarding cervical cancer in terms of molecular mechanism, etiology, clinical developments, and prevention.Study SampleThe review encompassed studies from diverse sources, including experimental, observational, and clinical research published between 1992 and 2025.Data Collection and/or AnalysisData were collected through comprehensive literature searches using databases such as PubMed, Scopus, and the Cochrane Library with defined inclusion and exclusion criteria.ResultsNonetheless, there are gaps in research and controversies regarding vaccine coverage, screening practices, and treatment accessibility for poor populations. Precision medicine trends are emerging along with new biomarkers for early detection and personalized treatment, which also form part of this discussion. Key findings include the critical role of prevention measures in controlling the global impact of cervical cancer.ConclusionsThe paper synthesizes the existing knowledge and identifies gaps that require further research, which is significant in augmenting prevention, diagnosis, and treatment of cervical cancer towards addressing its public health implications worldwide.
Collapse
Affiliation(s)
- Raiyan Ul Hakim
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - Tasbir Amin
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| | - S M Bakhtiar Ul Islam
- Department of Biochemistry & Microbiology, North South University, Dhaka, Bangladesh
| |
Collapse
|
19
|
Yin M, Weng Y, Qi T. PLOD2 exacerbates cervical squamous cell carcinoma by suppressing p53 by binding to YAP1. Mol Med Rep 2025; 31:23. [PMID: 39513600 DOI: 10.3892/mmr.2024.13388] [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: 08/15/2024] [Accepted: 10/02/2024] [Indexed: 11/15/2024] Open
Abstract
Procollagen‑lysine, 2‑oxoglutarate 5‑dioxygenase 2 (PLOD2) has been identified as an oncogene involved in the progression of several human cancers. However, its role in cervical squamous cell carcinoma (CESC) and its underlying mechanisms are not well understood. In the present study, several public databases, RT‑qPCR and western blotting were employed to detect the expression of PLOD2 and the prognosis in CESC. Cell counting kit‑8 assay, wound healing assay, Transwell assay, western blotting and flow cytometry were utilized to assess the proliferation, migration and cell apoptosis of CESC cells. Cellular senescence was examined by RT‑qPCR and β‑galactosidase staining. Prediction of PLOD2 binding to Yes‑associated protein 1 (YAP1) was assessed using BioGrid, HDock and co‑immunoprecipitation, and p53 and p21 signaling were assessed using immunofluorescence staining. The findings indicated that the expression of PLOD2 was elevated in CESC tissues and cell lines, and PLOD2 silencing caused the inhibition of CESC cell proliferation, migration and the promotion of apoptosis and senescence of CESC cells. PLOD2 was predicted to be bound to YAP1 and YAP1 overexpression reversed the effects of PLOD2 silencing on CESC cell proliferation, cell migration, apoptosis and senescence. In addition, PLOD2 facilitated CESC progression by regulating the P53 pathway through YAP1. PLOD2 exerted pro‑oncogenic effects on CESC through the p53 pathway by binding to YAP1. These findings provide new perspectives for the future study of PLOD2‑targeted therapy for CESC.
Collapse
Affiliation(s)
- Meilin Yin
- Clinical Laboratory Department, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100021, P.R. China
| | - Yanhua Weng
- Clinical Laboratory Department, Beijing Da Wang Lu Emergency Hospital, Beijing 100021, P.R. China
| | - Tianshu Qi
- Clinical Laboratory Department, Beijing Chaoyang District Maternal and Child Health Care Hospital, Beijing 100021, P.R. China
| |
Collapse
|
20
|
Li J, Ma Y, Wu Q, Ping P, Li J, Xu X. The potential role of HPV oncoproteins in the PD-L1/PD-1 pathway in cervical cancer: new perspectives on cervical cancer immunotherapy. Front Oncol 2024; 14:1488730. [PMID: 39735605 PMCID: PMC11671370 DOI: 10.3389/fonc.2024.1488730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Cervical cancer (CC) is a common malignant tumour of the female reproductive system that is highly harmful to women's health. The efficacy of traditional surgery, radiotherapy and chemotherapy is limited, especially for recurrent and metastatic CC. With continuous progress in diagnostic and treatment technology, immunotherapy has become a new approach for treating CC and has become a new therapy for recurrent and metastatic CC. However, immunotherapy is not effective for all patients with CC. Therefore, factors related to immunotherapy efficacy in CC patients have become the focus of researchers. High-risk human papillomavirus (HPV) infection is an important factor that drives CC development and affects its progression and prognosis. Increasing attention has been given to the mechanism of the E5, E6 and E7 proteins, which are encoded by the HPV gene, in the occurrence and development of CC and their interaction with programmed cell death ligand-1/programmed cell death-1 (PD-L1/PD-1). Although some preliminary studies have been conducted on these topics, a comprehensive and systematic review of these topics is not available. This review comprehensively summarizes related articles from journals with impact factors greater than 3 and published in the past 5 years; it also reviews studies on the mechanism of HPV and CC, the mechanism of PD-L1/PD-1 axis regulation in CC, and the mechanism by which the interaction between HPV-related oncoproteins and the PD-L1/PD-1 pathway affects the development and prognosis of CC. This study provides theoretical support for the use of immunotherapies for CC, provides a basis for the selection of specific medications that target different HPV-related proteins, and provides a new perspective for the discovery of new immunotherapy targets for CC.
Collapse
Affiliation(s)
| | | | | | | | - Juan Li
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| | - Xiaoying Xu
- Department of Radiotherapy Oncology, The Second Affiliated Hospital of Dalian Medical
University, Dalian, China
| |
Collapse
|
21
|
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
|
22
|
Liu Y, Zhou J, Liu W, Le Y, Zhang L, Zhang Z, Zhou L, Li L. The E3 ubiquitin ligase RNF6 facilitates the progression of cervical cancer by inhibiting the Hippo/Yap pathway. Cell Div 2024; 19:32. [PMID: 39551725 PMCID: PMC11571774 DOI: 10.1186/s13008-024-00136-8] [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: 04/25/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
PURPOSE Cervical cancer (CC), a significant global health threat, necessitates comprehensive understanding for improved therapeutic interventions. Many research indicates that dysregulation of the Hippo-YAP1 pathway leads to uncontrolled proliferation and invasion of tumor cells, promoting the progression of various cancers. This article aims to elucidate the role of RNF6 in CC and its regulation of the Hippo-YAP1 signaling pathway. METHODS The public tumor dataset analyses, immunohistochemistry, and western blotting were used to explore the expression of RNF6 in CC. Gain- and loss-of-function assays were conducted to elucidate the role of RNF6 in the proliferation and invasion of CC cells. Transcriptome sequencing was used to explore RNF6's role in cervical cancer, with validation of its regulation of the Hippo-YAP1 pathway through western blotting and RT-qPCR. Co-transfection of YAP overexpression plasmids into RNF6-silenced CC cells were preformed to confirm YAP1's pivotal role in RNF6-mediated CC progression. Animal experiments were preformed to further validate RNF6 interference's inhibitory effect on CC proliferation in vivo. RESULTS Clinical samples and bioinformatics analysis revealed high expression of RNF6 in CC, and closely associated with advanced FIGO (International Federation of Gynecology and Obstetrics) stage, larger tumor size, and poor prognosis. Cellular functional experiments demonstrate that RNF6 promotes the proliferation, invasion, and migration of CC cells, while knockdown of RNF6 yields the opposite effect. Transcriptome sequencing further reveals that RNF6 may promote CC progression through the Hippo-YAP signaling pathway. Western blotting and RT-qPCR further unveil that RNF6 enhances the upregulation of YAP1 protein levels, thereby activating downstream oncogenes CTGF and CYR61 transcription. Additionally, exogenous overexpression of YAP1 reverses the inhibitory effect of RNF6 silencing on CC proliferation and invasion. Furthermore, RNF6 interference significantly attenuates tumor growth in vivo experiments. CONCLUSION Our research reveals that RNF6 is highly expressed in CC, driving malignant progression by upregulating YAP1 protein expression and enhancing the transcription of downstream target genes CTGF and CYR61, offering potential therapeutic targets for CC treatment.
Collapse
Affiliation(s)
- Yawen Liu
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, 318 Bayi Road, 330006, Nanchang, Jiangxi Province, P.R. China
| | - Juanjuan Zhou
- Department of Oncology, the First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, 330006, Nanchang, Jiangxi Province, P.R. China
- Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, 330006, Nanchang, Jiangxi, China
| | - Weiqi Liu
- Department of Oncology, the First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, 330006, Nanchang, Jiangxi Province, P.R. China
- Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, 330006, Nanchang, Jiangxi, China
| | - Yi Le
- Department of Oncology, the First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, 330006, Nanchang, Jiangxi Province, P.R. China
- Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, 330006, Nanchang, Jiangxi, China
| | - Lingling Zhang
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, 318 Bayi Road, 330006, Nanchang, Jiangxi Province, P.R. China
| | - Ziyu Zhang
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, 318 Bayi Road, 330006, Nanchang, Jiangxi Province, P.R. China
| | - Ling Zhou
- Department of Oncology, the First Affiliated Hospital of Nanchang University, 1519 Dongyue Avenue, 330006, Nanchang, Jiangxi Province, P.R. China.
- Department of Jiangxi Key Laboratory for Individualized Cancer Therapy, 17 Yongwai Street, 330006, Nanchang, Jiangxi, China.
| | - Ling Li
- Department of Oncology, Jiangxi Maternal and Child Health Hospital, 318 Bayi Road, 330006, Nanchang, Jiangxi Province, P.R. China.
| |
Collapse
|
23
|
Sharma R, Sharma S, Shriwas P, Mehta L, Vu AH, Mouw JK, Koo J, Huang C, Matsuk VY, Tucker-Burden C, Joseph G, Behera M, Sun SY, Roy MA, Gilbert-Ross M, Leal T, Marcus AI, Shanmugam M. Intra-tumoral YAP and TAZ heterogeneity drives collective NSCLC invasion that is targeted by SUMOylation inhibitor TAK-981. iScience 2024; 27:111133. [PMID: 39524367 PMCID: PMC11544388 DOI: 10.1016/j.isci.2024.111133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/15/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) collective invasion is supported by cooperativity of proliferative (follower) and invasive (leader) cells. H1299-isolated follower cells exhibit higher Yes-associated protein (YAP) expression, while leader cells were found to express elevated transcriptional coactivator with PDZ-binding motif (TAZ/WWTR1) expression. Suppressing TAZ (not YAP) in leader cells reduced invasion. TAZ-regulated leader cell invasion is associated with activation of the EGFR-PI3K-AKT axis. NSCLC patient samples also demonstrated heterogeneity in YAP and TAZ expression. YAP and TAZ regulate proliferation of follower and leader cells. Our results highlight the need to inhibit both YAP and TAZ to effectively target their regulation of collective invasion. We identify that the SUMOylation inhibitor TAK-981 reduces YAP and TAZ expression, decreasing tumor burden and metastasis in a murine NSCLC model. Our study reveals an intra-tumoral division of labor, driven by differential YAP and TAZ expression, which can be effectively targeted with TAK-981 for NSCLC therapy.
Collapse
Affiliation(s)
- Richa Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shagun Sharma
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Pratik Shriwas
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Labdhi Mehta
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - An H. Vu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Janna K. Mouw
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Junghui Koo
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Chunzi Huang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Veronika Y. Matsuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Carol Tucker-Burden
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Gregory Joseph
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Madhusmita Behera
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Melissa A. Roy
- Division of Pathology, Emory National Primate Research Center, Atlanta, GA, USA
| | - Melissa Gilbert-Ross
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Ticiana Leal
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Adam I. Marcus
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| | - Mala Shanmugam
- Department of Hematology and Medical Oncology, Winship Cancer Institute, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
24
|
Arega S, Dey S, Pani S, Dash SR, Budhwar R, Kundu CN, Ganguly N. Determining the effect of long non-coding RNA maternally expressed gene 3 (lncRNA MEG3) on the transcriptome profile in cervical cancer cell lines. Genomics 2024; 116:110957. [PMID: 39510199 DOI: 10.1016/j.ygeno.2024.110957] [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: 06/14/2024] [Revised: 10/16/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
This study investigates the role of the long non-coding RNA Maternally Expressed Gene3 (lncRNA MEG3) gene in cervical cancer, as evidenced by its downregulation in cancerous cell lines. The study demonstrates the effects of the overexpression of lncRNA MEG3 in cervical cancer cell lines, particularly in C33A and CaSki. Through comprehensive analyses, including Next-Generation Sequencing (NGS), alterations in global mRNA expression were analyzed. In C33A cells, 67 genes were upregulated, while 303 genes were downregulated. Similarly, in CaSki cells, 221 genes showed upregulation and 248 genes displayed downregulation. Gene ontology and KEGG pathway analyses were conducted to gain insight into potential mechanisms. Furthermore, the study delves into gene regulatory networks, uncovering intricate interactions among genes. The RNA sequencing data were confirmed for eight genes: PAX3, EGR2, ROR1, NRP1, OAS2, STRA6, CA9, and EDN2 by Real-time PCR. The findings illuminate the complex landscape of gene expression alterations and pathways impacted by the overexpression of lncRNA MEG3. The impact of MEG3 on the overall cervical cancer cells' mRNA profile is reported for the first time. New biomarkers for the prognosis of cervical cancer are also reported in this study. Moreover, identifying specific genes within the regulatory networks provides valuable insights into potential therapeutic targets for managing cervical cancer.
Collapse
Affiliation(s)
- Solomon Arega
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India.
| | - Suchanda Dey
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Sunil Pani
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Somya Ranjan Dash
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Roli Budhwar
- Bionivid Technology Pvt. Ltd, Bangalore 560043, India
| | - Chanakya Nath Kundu
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| | - Niladri Ganguly
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT), Deemed to be University, Bhubaneswar 751024, India
| |
Collapse
|
25
|
Tan N, Li Y, Ying J, Chen W. Histological transformation in lung adenocarcinoma: Insights of mechanisms and therapeutic windows. J Transl Int Med 2024; 12:452-465. [PMID: 39513032 PMCID: PMC11538883 DOI: 10.1515/jtim-2024-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Histological transformation from lung adenocarcinoma (ADC) to small cell lung carcinoma (SCLC), large cell neuroendocrine carcinoma (LCNEC), squamous cell carcinoma (SCC), and sarcomatoid carcinoma (PSC) after targeted therapies is recognized as a mechanism of resistance in ADC treatments. Patients with transformed lung cancer typically experience a poor prognosis and short survival time. However, effective treatment options for these patients are currently lacking. Therefore, understanding the mechanisms underlying histological transformation is crucial for the development of effective therapies. Hypotheses including intratumoral heterogeneity, cancer stem cells, and alteration of suppressor genes have been proposed to explain the mechanism of histological transformation. In this review, we provide a comprehensive overview of the known molecular features and signaling pathways of transformed tumors, and summarized potential therapies based on previous findings.
Collapse
Affiliation(s)
- Nuopei Tan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
He C, Lv X, Liu J, Ruan J, Chen P, Huang C, Angeletti PC, Hua G, Moness ML, Shi D, Dhar A, Yang S, Murphy S, Montoute I, Chen X, Islam KN, George S, Ince TA, Drapkin R, Guda C, Davis JS, Wang C. HPV-YAP1 oncogenic alliance drives malignant transformation of fallopian tube epithelial cells. EMBO Rep 2024; 25:4542-4569. [PMID: 39271776 PMCID: PMC11467260 DOI: 10.1038/s44319-024-00233-3] [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: 12/06/2023] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
High grade serous ovarian carcinoma (HGSOC) is the most common and aggressive ovarian malignancy. Accumulating evidence indicates that HGSOC may originate from human fallopian tube epithelial cells (FTECs), although the exact pathogen(s) and/or molecular mechanism underlying the malignant transformation of FTECs is unclear. Here we show that human papillomavirus (HPV), which could reach FTECs via retrograde menstruation or sperm-carrying, interacts with the yes-associated protein 1 (YAP1) to drive the malignant transformation of FTECs. HPV prevents FTECs from natural replicative and YAP1-induced senescence, thereby promoting YAP1-induced malignant transformation of FTECs. HPV also stimulates proliferation and drives metastasis of YAP1-transformed FTECs. YAP1, in turn, stimulates the expression of the putative HPV receptors and suppresses the innate immune system to facilitate HPV acquisition. These findings provide critical clues for developing new strategies to prevent and treat HGSOC.
Collapse
Affiliation(s)
- Chunbo He
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiangmin Lv
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jiyuan Liu
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Jinpeng Ruan
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Peichao Chen
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Cong Huang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Peter C Angeletti
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, 68583, USA
| | - Guohua Hua
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Madelyn Leigh Moness
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Davie Shi
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Anjali Dhar
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Siyi Yang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Savannah Murphy
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Isabelle Montoute
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Xingcheng Chen
- Fred & Pamela Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kazi Nazrul Islam
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Sophia George
- Department of Obstetrics & Gynecology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, 33136, USA
| | - Tan A Ince
- New York Presbyterian Brooklyn Methodist Hospital and Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Ronny Drapkin
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Chittibabu Guda
- Department of Cellular and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Western Iowa and Nebraska Veteran's Affairs Medical Center, Omaha, NE, 68105, USA
| | - Cheng Wang
- Vincent Center for Reproductive Biology, Vincent Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Olson Center for Women's Health, Department of Obstetrics & Gynecology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
27
|
Wu J, Wang X, Li X, Zhu Z, Cui Z, Zhang T, Zou W, Han G. A dual-labeling molecule for efficient drug discovery of mitochondrial-lysosomal interactions. Mol Cell Probes 2024; 76:101968. [PMID: 38960210 DOI: 10.1016/j.mcp.2024.101968] [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: 04/18/2024] [Revised: 06/20/2024] [Accepted: 07/01/2024] [Indexed: 07/05/2024]
Abstract
The close association between organelle interactions, such as mitochondrial-lysosomal interactions, and various diseases, including tumors, remains a challenge for drug discovering and identification. Conventional evaluation methods are often complex and multistep labeling procedures often generate false positives, such as cell damage. To overcome these limitations, we employed a single dual-color reporting molecule called Coupa, which labels mitochondria and lysosomes as blue and red, respectively. This facilitates the evaluation and discovering of drugs targeting mitochondria-lysosome contact (MLC). Using Coupa, we validated the effectiveness of various known antitumor drugs in intervening MLC by assessing their effect on key aspects, such as status, localization, and quantity. This provides evidence for the accuracy and applicability of our dual-color reporting molecule. Notably, we observed that several structural isomers of drugs, including Urolithin (A/B/C), exhibited distinct effects on MLC. In addition, Verteporfin and TEAD were found to induce anti-tumor effects by controlling MLC at the organelle level, suggesting a potential new mechanism of action. Collectively, Coupa offers a novel scientific tool for discovering drugs that target mitochondrial-lysosomal interactions. It not only distinguished the differential effects of structurally similar drugs on the same target, but also reveals new mechanisms underlying the reported antitumor properties of existing drugs. Ultimately, our findings contribute to the advancement of drug discovery and provide valuable insights into the complex interactions between organelles in a disease context.
Collapse
Affiliation(s)
- Jinfang Wu
- College of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Xiaolei Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China
| | - Xiang Li
- College of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Zixuan Zhu
- College of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Zhongcheng Cui
- College of Pharmacy, Jinzhou Medical University, Jinzhou, China
| | - Tao Zhang
- Department of General Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China.
| | - Weiwei Zou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China; Medicine-Engineering Integration & Medical Equipment Innovation Institute of Anhui Medical University, Hefei, Anhui, China.
| | - Guanying Han
- Medical College of Jinzhou Medical University, Jinzhou, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China.
| |
Collapse
|
28
|
Zwick A, Braun FL, Weber LJ, Linder M, Linxweiler M, Lohse S. Engineering Dimeric EGFR-directed IgA Antibodies Reveals a Central Role of CD147 during Neutrophil-mediated Tumor Cell Killing of Head and Neck Squamous Cancer Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:148-160. [PMID: 38787053 DOI: 10.4049/jimmunol.2300544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 04/18/2024] [Indexed: 05/25/2024]
Abstract
Human IgA Abs engage neutrophils for cancer immunotherapy more effectively than IgG Abs. Previous studies demonstrated that engineering approaches improved biochemical and functional properties. In this study, we report a novel, to our knowledge, IgA2 Ab against the epidermal growth factor receptor generated by protein engineering and polymerization. The resulting molecule demonstrated a covalent linkage of L and H chains and an effective polymerization by the joining chain. The engineered dimer outperformed its monomeric variant in functional experiments on Fab-mediated modes of action and binding to the Fc receptor. The capacity to engage neutrophils for Ab-dependent cell-mediated cytotoxicity (ADCC) of adherent growing target cancer cells was cell line dependent. Although the engineered dimer displayed a long-term efficacy against the vulva carcinoma cell line A431, there was a notable in-efficacy against human papillomavirus (HPV)- head and neck squamous cell carcinoma (HNSCC) cell lines. However, the highly engineered IgA Abs triggered a neutrophil-mediated cytotoxicity against HPV+ HNSCC cell lines. Short-term ADCC efficacy correlated with the target cells' epidermal growth factor receptor expression and the ability of cancer cell-conditioned media to enhance the CD147 surface level on neutrophils. Notably, the HPV+ HNSCC cell lines demonstrated a significant increment in releasing soluble CD147 and a reduced induction of membranous CD147 on neutrophils compared with HPV- cells. Although membranous CD147 on neutrophils may impair proper IgA-Fc receptor binding, soluble CD147 enhanced the IgA-neutrophil-mediated ADCC in a dose-dependent manner. Thus, engineering IgA Abs and impedance-based ADCC assays provided valuable information regarding the target-effector cell interaction and identified CD147 as a putative critical parameter for neutrophil-mediated cytotoxicity.
Collapse
Affiliation(s)
- Anabel Zwick
- Institute of Virology, Saarland University Medical Center, Homburg, Germany
| | - Felix Leon Braun
- Institute of Virology, Saarland University Medical Center, Homburg, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg/Saar, Germany
| | | | - Manuel Linder
- Institute of Virology, Saarland University Medical Center, Homburg, Germany
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery; Saarland University Medical Center, Homburg/Saar, Germany
| | - Stefan Lohse
- Institute of Virology, Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
29
|
Patterson MR, Cogan JA, Cassidy R, Theobald DA, Wang M, Scarth JA, Anene CA, Whitehouse A, Morgan EL, Macdonald A. The Hippo pathway transcription factors YAP and TAZ play HPV-type dependent roles in cervical cancer. Nat Commun 2024; 15:5809. [PMID: 38987584 PMCID: PMC11237029 DOI: 10.1038/s41467-024-49965-9] [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: 09/11/2023] [Accepted: 06/21/2024] [Indexed: 07/12/2024] Open
Abstract
Human papillomaviruses (HPVs) cause most cervical cancers and an increasing number of anogenital and oral carcinomas, with most cases caused by HPV16 or HPV18. HPV hijacks host signalling pathways to promote carcinogenesis. Understanding these interactions could permit identification of much-needed therapeutics for HPV-driven malignancies. The Hippo signalling pathway is important in HPV+ cancers, with the downstream effector YAP playing a pro-oncogenic role. In contrast, the significance of its paralogue TAZ remains largely uncharacterised in these cancers. We demonstrate that TAZ is dysregulated in a HPV-type dependent manner by a distinct mechanism to that of YAP and controls proliferation via alternative cellular targets. Analysis of cervical cancer cell lines and patient biopsies revealed that TAZ expression was only significantly increased in HPV18+ and HPV18-like cells and TAZ knockdown reduced proliferation, migration and invasion only in HPV18+ cells. RNA-sequencing of HPV18+ cervical cells revealed that YAP and TAZ have distinct targets, suggesting they promote carcinogenesis by different mechanisms. Thus, in HPV18+ cancers, YAP and TAZ play non-redundant roles. This analysis identified TOGARAM2 as a previously uncharacterised TAZ target and demonstrates its role as a key effector of TAZ-mediated proliferation, migration and invasion in HPV18+ cancers.
Collapse
Affiliation(s)
- Molly R Patterson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| | - Joseph A Cogan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Rosa Cassidy
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Daisy A Theobald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Miao Wang
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - James A Scarth
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Chinedu A Anene
- Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Biomedical Science Research, Leeds Beckett University, Leeds, UK
| | - Adrian Whitehouse
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Ethan L Morgan
- School of Life Sciences, University of Sussex, Brighton, UK
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
30
|
Patterson MR, Meijers AS, Ryder EL, Wootton LM, Scarth JA, Evans D, Turner AL, Wasson CW, Darell JE, Theobald DA, Cogan JA, James CD, Wang M, Ladbury JE, Morgan IM, Samson A, Morgan EL, Macdonald A. E7-mediated repression of miR-203 promotes LASP1-dependent proliferation in HPV-positive cervical cancer. Oncogene 2024; 43:2184-2198. [PMID: 38789663 PMCID: PMC11226402 DOI: 10.1038/s41388-024-03067-4] [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: 02/02/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024]
Abstract
Human papillomaviruses (HPV) are a major cause of malignancy, contributing to ~5% of all human cancers worldwide, including most cervical cancer cases and a growing number of anogenital and oral cancers. The major HPV viral oncogenes, E6 and E7, manipulate many host cellular pathways that promote cell proliferation and survival, predisposing infected cells to malignant transformation. Despite the availability of highly effective vaccines, there are still no specific anti-viral therapies targeting HPV or treatments for HPV-associated cancers. As such, a better understanding of viral-host interactions may allow the identification of novel therapeutic targets. Here, we demonstrate that the actin-binding protein LASP1 is upregulated in cervical cancer and significantly correlates with a poorer overall survival. In HPV positive cervical cancer, LASP1 depletion significantly inhibited the oncogenic phenotype in vitro, whilst having minimal effects in HPV negative cervical cancer cells. Furthermore, we demonstrate that the LASP1 SH3 domain is essential for LASP1-mediated oncogenicity in these cells. Mechanistically, we show that HPV E7 regulates LASP1 at the post-transcriptional level by repressing the expression of miR-203, which negatively regulates LASP1 mRNA levels by binding to its 3'UTR. Finally, we demonstrate that LASP1 expression is required for the growth of HPV positive cervical cancer cells in an in vivo tumourigenicity model. Together, these data demonstrate that HPV induces LASP1 expression to promote proliferation and survival in cervical cancer, thus identifying a potential therapeutic target in these cancers.
Collapse
Affiliation(s)
- Molly R Patterson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aniek S Meijers
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Emma L Ryder
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | | | - James A Scarth
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Debra Evans
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, UK
| | - Amy L Turner
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Christopher W Wasson
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, St-James University Teaching Hospital, Leeds, UK
| | - Janne E Darell
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Daisy A Theobald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Joseph A Cogan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Claire D James
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | - Miao Wang
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - John E Ladbury
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK
| | - Iain M Morgan
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
- VCU Massey Cancer Center, VCU, Richmond, VA, USA
| | - Adel Samson
- Leeds Institute of Medical Research, St James's University Hospital, University of Leeds, Leeds, UK
| | - Ethan L Morgan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
- School of Life Sciences, University of Sussex, Brighton, UK.
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK.
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
31
|
Xu C, Wu J, Ye J, Si Y, Zhang J, Wu B, Pan L, Fu J, Ren Q, Xie S, Tang B, Xiao Y, Hong T. Multiomics integration-based immunological characterizations of adamantinomatous craniopharyngioma in relation to keratinization. Cell Death Dis 2024; 15:439. [PMID: 38906852 PMCID: PMC11192745 DOI: 10.1038/s41419-024-06840-1] [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: 11/09/2023] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/23/2024]
Abstract
Although adamantinomatous craniopharyngioma (ACP) is a tumour with low histological malignancy, there are very few therapeutic options other than surgery. ACP has high histological complexity, and the unique features of the immunological microenvironment within ACP remain elusive. Further elucidation of the tumour microenvironment is particularly important to expand our knowledge of potential therapeutic targets. Here, we performed integrative analysis of 58,081 nuclei through single-nucleus RNA sequencing and spatial transcriptomics on ACP specimens to characterize the features and intercellular network within the microenvironment. The ACP environment is highly immunosuppressive with low levels of T-cell infiltration/cytotoxicity. Moreover, tumour-associated macrophages (TAMs), which originate from distinct sources, highly infiltrate the microenvironment. Using spatial transcriptomic data, we observed one kind of non-microglial derived TAM that highly expressed GPNMB close to the terminally differentiated epithelial cell characterized by RHCG, and this colocalization was verified by asmFISH. We also found the positive correlation of infiltration between these two cell types in datasets with larger cohort. According to intercellular communication analysis, we report a regulatory network that could facilitate the keratinization of RHCG+ epithelial cells, eventually causing tumour progression. Our findings provide a comprehensive analysis of the ACP immune microenvironment and reveal a potential therapeutic strategy base on interfering with these two types of cells.
Collapse
Affiliation(s)
- Chunming Xu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jie Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jiye Ye
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yuancheng Si
- Department of Mathematics, University of Manchester, Manchester, UK
- The School of Economics, Fudan University, Shanghai, China
| | - Jinshi Zhang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bowen Wu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Laisheng Pan
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jun Fu
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Quan Ren
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shenhao Xie
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bin Tang
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yingqun Xiao
- Department of Pathology, Affiliated Infectious Disease Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Tao Hong
- Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
32
|
Xu L, Zhang C, Bao J, Han G, Wang C, Cai Y, Xu G, Sun H, Liu M. Alpha-lipoic Acid Prevents Bone Loss in Type 2 Diabetes and Postmenopausal Osteoporosis Coexisting Conditions by Modulating the YAP/Glut4 Pathway. Cell Biochem Biophys 2024; 82:669-685. [PMID: 38261247 DOI: 10.1007/s12013-024-01216-w] [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: 12/14/2022] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
This study aims to characterize the bone-protecting effects of Alpha-lipoic acid (ALA), a potent antioxidant, against the detrimental effects of the coexistence of type 2 diabetes mellitus (T2DM) and postmenopausal osteoporosis (POP) and identify the possible mechanisms with particular reference to its modulation of YAP/Glut4 pathway. The T2DM and POP coexisting model was induced in mice by high fat diet (HFD) + Streptozocin (STZ) + ovariectomy (OVX). The mice in the treatment groups were given ALA for 10 weeks. In the in vitro study, MC3T3-E1 cells were induced with 500 μM methylglyoxal for 24 h with or without pretreatment with ALA for 24 h. The oxidative and antioxidative biomarkers, bone microarchitecture, histo-morphology, and related protein expression of apoptosis, osteogenic differentiation and the YAP/Glut4 pathway were detected. The results showed ALA could improve glucose tolerance, inhibit oxidative stress and apoptosis and alleviate bone loss. Further study by siRNA technology revealed that the YAP/Glut4 pathway was implicated in the pathogenesis of bone loss due to the coexistence of T2DM and POP. Taken together, the present study has demonstrated for the first time that ALA exerts potent protective effects against bone loss in T2DM and POP coexisting conditions by modulating the YAP/Glut4 pathway.
Collapse
MESH Headings
- Thioctic Acid/pharmacology
- Thioctic Acid/therapeutic use
- Animals
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/drug therapy
- Mice
- Female
- Osteoporosis, Postmenopausal/metabolism
- Osteoporosis, Postmenopausal/drug therapy
- Glucose Transporter Type 4/metabolism
- YAP-Signaling Proteins/metabolism
- Oxidative Stress/drug effects
- Signal Transduction/drug effects
- Adaptor Proteins, Signal Transducing/metabolism
- Mice, Inbred C57BL
- Apoptosis/drug effects
- Cell Line
- Diet, High-Fat/adverse effects
- Humans
- Ovariectomy/adverse effects
- Antioxidants/pharmacology
- Cell Differentiation/drug effects
- Transcription Factors/metabolism
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/complications
- Osteogenesis/drug effects
Collapse
Affiliation(s)
- Lei Xu
- Office of Ethics Committee, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chunfang Zhang
- Department of Pathology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiawu Bao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Guozhu Han
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yuanqing Cai
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Gang Xu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Mozhen Liu
- Department of Orthopaedics, the First Affiliated Hospital, Dalian Medical University, Dalian, China.
| |
Collapse
|
33
|
Sayeed K, Parameswaran S, Beucler MJ, Edsall LE, VonHandorf A, Crowther A, Donmez O, Hass M, Richards S, Forney C, Wright J, Leong MML, Murray-Nerger LA, Gewurz BE, Kaufman KM, Harley JB, Zhao B, Miller WE, Kottyan LC, Weirauch MT. Human cytomegalovirus infection coopts chromatin organization to diminish TEAD1 transcription factor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.588762. [PMID: 38645179 PMCID: PMC11030363 DOI: 10.1101/2024.04.12.588762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Human cytomegalovirus (HCMV) infects up to 80% of the world's population. Here, we show that HCMV infection leads to widespread changes in human chromatin accessibility and chromatin looping, with hundreds of thousands of genomic regions affected 48 hours after infection. Integrative analyses reveal HCMV-induced perturbation of Hippo signaling through drastic reduction of TEAD1 transcription factor activity. We confirm extensive concordant loss of TEAD1 binding, active H3K27ac histone marks, and chromatin looping interactions upon infection. Our data position TEAD1 at the top of a hierarchy involving multiple altered important developmental pathways. HCMV infection reduces TEAD1 activity through four distinct mechanisms: closing of TEAD1-bound chromatin, reduction of YAP1 and phosphorylated YAP1 levels, reduction of TEAD1 transcript and protein levels, and alteration of TEAD1 exon-6 usage. Altered TEAD1-based mechanisms are highly enriched at genetic risk loci associated with eye and ear development, providing mechanistic insight into HCMV's established roles in these processes.
Collapse
Affiliation(s)
- Khund Sayeed
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Matthew J. Beucler
- Department of Molecular Genetics, Biochemistry & Microbiology, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Lee E. Edsall
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrew VonHandorf
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Audrey Crowther
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Omer Donmez
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Matthew Hass
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Scott Richards
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Carmy Forney
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Jay Wright
- Department of Molecular Genetics, Biochemistry & Microbiology, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Merrin Man Long Leong
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Laura A. Murray-Nerger
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
- Department of Microbiology, Harvard Program in Virology, Harvard Medical School, Boston, MA, 02115, USA
- Center for Integrated Solutions to Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Ben E. Gewurz
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Kenneth M. Kaufman
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Research Service, Cincinnati VA Medical Center, Cincinnati, OH 45229, USA
| | - John B. Harley
- Research Service, Cincinnati VA Medical Center, Cincinnati, OH 45229, USA
| | - Bo Zhao
- Department of Medicine, Division of Infectious Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - William E. Miller
- Department of Molecular Genetics, Biochemistry & Microbiology, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Leah C. Kottyan
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Matthew T. Weirauch
- Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
- Division of Allergy and Immunology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Developmental Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, USA
| |
Collapse
|
34
|
Zhang X, Li J, Yang L, Zhu Y, Gao R, Zhang T, Chen X, Fu J, He G, Shi H, Peng S, Wu X. Targeted proteomics-determined multi-biomarker profiles developed classifier for prognosis and immunotherapy responses of advanced cervical cancer. Front Immunol 2024; 15:1391524. [PMID: 38835778 PMCID: PMC11148239 DOI: 10.3389/fimmu.2024.1391524] [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: 02/26/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Background Cervical cancer (CC) poses a global health challenge, with a particularly poor prognosis in cases of recurrence, metastasis, or advanced stages. A single biomarker is inadequate to predict CC prognosis or identify CC patients likely to benefit from immunotherapy, presumably owing to tumor complexity and heterogeneity. Methods Using advanced Olink proteomics, we analyzed 92 oncology-related proteins in plasma from CC patients receiving immunotherapy, based upon the comparison of protein expression levels of pre-therapy with those of therapy-Cycle 6 in the partial response (PR) group and progressive disease (PD) group, respectively. Results 55 proteins were identified to exhibit differential expression trends across pre-therapy and post-therapy in both PR and PD groups. Enriched GO terms and KEGG pathways were associated with vital oncological and immunological processes. A logistic regression model, using 5 proteins (ITGB5, TGF-α, TLR3, WIF-1, and ERBB3) with highest AUC values, demonstrated good predictive performance for prognosis of CC patients undergoing immunotherapy and showed potential across different cancer types. The effectiveness of these proteins in prognosis prediction was further validated using TCGA-CESC datasets. A negative correlation and previously unidentified roles of WIF-1 in CC immunotherapy was also first determined. Conclusion Our findings reveal multi-biomarker profiles effectively predicting CC prognosis and identifying patients benefitting most from immunotherapy, especially for those with limited treatment options and traditionally poor prognosis, paving the way for personalized immunotherapeutic treatments and improved clinical strategies.
Collapse
Affiliation(s)
- Xu Zhang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Jin Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liuke Yang
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Youwei Zhu
- Clinical Center of Bio-Therapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Rongrong Gao
- Clinical Center for Biotherapy at Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tiancheng Zhang
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Xuwen Chen
- Shanghai Kelin Clinical Bioinformatics Institute, Shanghai, China
| | - Jun Fu
- LC-Bio Technology Co., Ltd, Hangzhou, China
| | - Gaoyang He
- LC-Bio Technology Co., Ltd, Hangzhou, China
| | - Huijuan Shi
- NHC Key Laboratory of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Laboratory of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Shenjie Peng
- Shanghai Medical College of Fudan University, Fudan University, Shanghai, China
| | - XiaoHua Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Kumar R, Hong W. Hippo Signaling at the Hallmarks of Cancer and Drug Resistance. Cells 2024; 13:564. [PMID: 38607003 PMCID: PMC11011035 DOI: 10.3390/cells13070564] [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: 01/08/2024] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024] Open
Abstract
Originally identified in Drosophila melanogaster in 1995, the Hippo signaling pathway plays a pivotal role in organ size control and tumor suppression by inhibiting proliferation and promoting apoptosis. Large tumor suppressors 1 and 2 (LATS1/2) directly phosphorylate the Yki orthologs YAP (yes-associated protein) and its paralog TAZ (also known as WW domain-containing transcription regulator 1 [WWTR1]), thereby inhibiting their nuclear localization and pairing with transcriptional coactivators TEAD1-4. Earnest efforts from many research laboratories have established the role of mis-regulated Hippo signaling in tumorigenesis, epithelial mesenchymal transition (EMT), oncogenic stemness, and, more recently, development of drug resistances. Hippo signaling components at the heart of oncogenic adaptations fuel the development of drug resistance in many cancers for targeted therapies including KRAS and EGFR mutants. The first U.S. food and drug administration (US FDA) approval of the imatinib tyrosine kinase inhibitor in 2001 paved the way for nearly 100 small-molecule anti-cancer drugs approved by the US FDA and the national medical products administration (NMPA). However, the low response rate and development of drug resistance have posed a major hurdle to improving the progression-free survival (PFS) and overall survival (OS) of cancer patients. Accumulating evidence has enabled scientists and clinicians to strategize the therapeutic approaches of targeting cancer cells and to navigate the development of drug resistance through the continuous monitoring of tumor evolution and oncogenic adaptations. In this review, we highlight the emerging aspects of Hippo signaling in cross-talk with other oncogenic drivers and how this information can be translated into combination therapy to target a broad range of aggressive tumors and the development of drug resistance.
Collapse
Affiliation(s)
- Ramesh Kumar
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology, and Research), Singapore 138673, Singapore;
| | | |
Collapse
|
36
|
Sun Y, Tang L, Kan X, Tan L, Song C, Qiu X, Liao Y, Nair V, Ding C, Liu X, Sun Y. Oncolytic Newcastle disease virus induced degradation of YAP through E3 ubiquitin ligase PRKN to exacerbate ferroptosis in tumor cells. J Virol 2024; 98:e0189723. [PMID: 38411946 PMCID: PMC10949840 DOI: 10.1128/jvi.01897-23] [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: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Ferroptosis, a form of programmed cell death characterized by iron-dependent lipid peroxidation, has recently gained considerable attention in the field of cancer therapy. There is significant crosstalk between ferroptosis and several classical signaling pathways, such as the Hippo pathway, which suppresses abnormal growth and is frequently aberrant in tumor tissues. Yes-associated protein 1 (YAP), the core effector molecule of the Hippo pathway, is abnormally expressed and activated in a variety of malignant tumor tissues. We previously proved that the oncolytic Newcastle disease virus (NDV) activated ferroptosis to kill tumor cells. NDV has been used in tumor therapy; however, its oncolytic mechanism is not completely understood. In this study, we demonstrated that NDV exacerbated ferroptosis in tumor cells by inducing ubiquitin-mediated degradation of YAP at Lys90 through E3 ubiquitin ligase parkin (PRKN). Blocking YAP degradation suppressed NDV-induced ferroptosis by suppressing the expression of Zrt/Irt-like protein 14 (ZIP14), a metal ion transporter that regulates iron uptake. These findings demonstrate that NDV exacerbated ferroptosis in tumor cells by inducing YAP degradation. Our study provides new insights into the mechanism of NDV-induced ferroptosis and highlights the critical role that oncolytic viruses play in the treatment of drug-resistant cancers.IMPORTANCEThe oncolytic Newcastle disease virus (NDV) is being developed for use in cancer treatment; however, its oncolytic mechanism is still not completely understood. The Hippo pathway, which is a tumor suppressor pathway, is frequently dysregulated in tumor tissues due to aberrant yes-associated protein 1 (YAP) activation. In this study, we have demonstrated that NDV degrades YAP to induce ferroptosis and promote virus replication in tumor cells. Notably, NDV was found to induce ubiquitin-mediated degradation of YAP at Lys90 through E3 ubiquitin ligase parkin (PRKN). Our study reveals a new mechanism by which NDV induces ferroptosis and provides new insights into NDV as an oncolytic agent for cancer treatment.
Collapse
Affiliation(s)
- Yifan Sun
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Lanlan Tang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Xianjin Kan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| | - Venugopal Nair
- Avian Oncogenic viruses group, UK-China Centre of Excellence on Avian Disease Research, The Pirbright Institute, Guildford, United Kingdom
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Science, Shanghai, China
| |
Collapse
|
37
|
Byerly CD, Zhu B, Warwick PA, Patterson LL, Pittner NA, McBride JW. Ehrlichia chaffeensis TRP120 ubiquitinates tumor suppressor APC to modulate Hippo and Wnt signaling. Front Cell Dev Biol 2024; 12:1327418. [PMID: 38562145 PMCID: PMC10982408 DOI: 10.3389/fcell.2024.1327418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Ehrlichia chaffeensis: TRP120 is a multifunctional effector that acts as a ligand mimic to activate evolutionary conserved eukaryotic signaling pathways Notch, Wnt, Hedgehog and Hippo. In addition, TRP120 is also a HECT E3 ubiquitin ligase known to ubiquitinate several host cell regulatory proteins (FBW7, PCGF5 and ENO-1) for degradation. We previously determined that TRP120 ubiquitinates the Notch negative regulator, FBW7, to maintain Notch signaling and promote infection. In this study, we investigated a potential mechanism used by Ehrlichia chaffeensis to maintain Hippo and Wnt signaling by ubiquitinating the tumor suppressor, adenomatous polyposis coli (APC), a negative regulator of Wnt and Hippo signaling. We determined that APC was rapidly degraded during E. chaffeensis infection despite increased APC transcription. Moreover, RNAi knockdown of APC significantly increased E. chaffeensis infection and coincided with increased active Yap and β-catenin in the nucleus. We observed strong nuclear colocalization between TRP120 and APC in E. chaffeensis-infected THP-1 cells and after ectopic expression of TRP120 in HeLa cells. Additionally, TRP120 interacted with both APC full length and truncated isoforms via co-immunoprecipitation. Further, TRP120 ubiquitination of APC was demonstrated in vitro and confirmed by ectopic expression of a TRP120 HECT Ub ligase catalytic site mutant. This study identifies APC as a TRP120 HECT E3 Ub ligase substrate and demonstrates that TRP120 ligase activity promotes ehrlichial infection by degrading tumor suppressor APC to positively regulate Hippo and Wnt signaling.
Collapse
Affiliation(s)
| | | | | | | | | | - Jere W. McBride
- Departments of Pathology, Microbiology and Immunology, Center for Biodefense and Emerging Infectious Diseases, Sealy Institute for Vaccine Sciences and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
38
|
Rodriguez I, Rossi NM, Keskus AG, Xie Y, Ahmad T, Bryant A, Lou H, Paredes JG, Milano R, Rao N, Tulsyan S, Boland JF, Luo W, Liu J, O'Hanlon T, Bess J, Mukhina V, Gaykalova D, Yuki Y, Malik L, Billingsley KJ, Blauwendraat C, Carrington M, Yeager M, Mirabello L, Kolmogorov M, Dean M. Insights into the mechanisms and structure of breakage-fusion-bridge cycles in cervical cancer using long-read sequencing. Am J Hum Genet 2024; 111:544-561. [PMID: 38307027 PMCID: PMC10940022 DOI: 10.1016/j.ajhg.2024.01.002] [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: 08/18/2023] [Revised: 12/20/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024] Open
Abstract
Cervical cancer is caused by human papillomavirus (HPV) infection, has few approved targeted therapeutics, and is the most common cause of cancer death in low-resource countries. We characterized 19 cervical and four head and neck cancer cell lines using long-read DNA and RNA sequencing and identified the HPV types, HPV integration sites, chromosomal alterations, and cancer driver mutations. Structural variation analysis revealed telomeric deletions associated with DNA inversions resulting from breakage-fusion-bridge (BFB) cycles. BFB is a common mechanism of chromosomal alterations in cancer, and our study applies long-read sequencing to this important chromosomal rearrangement type. Analysis of the inversion sites revealed staggered ends consistent with exonuclease digestion of the DNA after breakage. Some BFB events are complex, involving inter- or intra-chromosomal insertions or rearrangements. None of the BFB breakpoints had telomere sequences added to resolve the dicentric chromosomes, and only one BFB breakpoint showed chromothripsis. Five cell lines have a chromosomal region 11q BFB event, with YAP1-BIRC3-BIRC2 amplification. Indeed, YAP1 amplification is associated with a 10-year-earlier age of diagnosis of cervical cancer and is three times more common in African American women. This suggests that individuals with cervical cancer and YAP1-BIRC3-BIRC2 amplification, especially those of African ancestry, might benefit from targeted therapy. In summary, we uncovered valuable insights into the mechanisms and consequences of BFB cycles in cervical cancer using long-read sequencing.
Collapse
Affiliation(s)
- Isabel Rodriguez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Nicole M Rossi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Ayse G Keskus
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Yi Xie
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Tanveer Ahmad
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Asher Bryant
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Hong Lou
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jesica Godinez Paredes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Rose Milano
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Nina Rao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA; Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Sonam Tulsyan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Joseph F Boland
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Wen Luo
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jia Liu
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Tim O'Hanlon
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Jazmyn Bess
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Vera Mukhina
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Maryland School of Medical Center, Baltimore, MD, USA
| | - Daria Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical System, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Yuko Yuki
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Laksh Malik
- Center for Alzheimer's and Related Dementias, National Institute on Aging, Bethesda, MD, USA
| | | | - Cornelis Blauwendraat
- Center for Alzheimer's and Related Dementias, National Institute on Aging, Bethesda, MD, USA; Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
| | - Mary Carrington
- Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Meredith Yeager
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Lisa Mirabello
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Mikhail Kolmogorov
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA.
| |
Collapse
|
39
|
Melrose J. Hippo cell signaling and HS-proteoglycans regulate tissue form and function, age-dependent maturation, extracellular matrix remodeling, and repair. Am J Physiol Cell Physiol 2024; 326:C810-C828. [PMID: 38223931 DOI: 10.1152/ajpcell.00683.2023] [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: 12/11/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
This review examined how Hippo cell signaling and heparan sulfate (HS)-proteoglycans (HSPGs) regulate tissue form and function. Despite being a nonweight-bearing tissue, the brain is regulated by Hippo mechanoresponsive cell signaling pathways during embryonic development. HS-proteoglycans interact with growth factors, morphogens, and extracellular matrix components to regulate development and pathology. Pikachurin and Eyes shut (Eys) interact with dystroglycan to stabilize the photoreceptor axoneme primary cilium and ribbon synapse facilitating phototransduction and neurotransduction with bipolar retinal neuronal networks in ocular vision, the primary human sense. Another HSPG, Neurexin interacts with structural and adaptor proteins to stabilize synapses and ensure specificity of neural interactions, and aids in synaptic potentiation and plasticity in neurotransduction. HSPGs also stabilize the blood-brain barrier and motor neuron basal structures in the neuromuscular junction. Agrin and perlecan localize acetylcholinesterase and its receptors in the neuromuscular junction essential for neuromuscular control. The primary cilium is a mechanosensory hub on neurons, utilized by YES associated protein (YAP)-transcriptional coactivator with PDZ-binding motif (TAZ) Hippo, Hh, Wnt, transforming growth factor (TGF)-β/bone matrix protein (BMP) receptor tyrosine kinase cell signaling. Members of the glypican HSPG proteoglycan family interact with Smoothened and Patched G-protein coupled receptors on the cilium to regulate Hh and Wnt signaling during neuronal development. Control of glycosyl sulfotransferases and endogenous protease expression by Hippo TAZ YAP represents a mechanism whereby the fine structure of HS-proteoglycans can be potentially modulated spatiotemporally to regulate tissue morphogenesis in a similar manner to how Hippo signaling controls sialyltransferase expression and mediation of cell-cell recognition, dysfunctional sialic acid expression is a feature of many tumors.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, University of Sydney, Northern Sydney Local Health District, Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Sydney Medical School-Northern, University of Sydney at Royal North Shore Hospital, St. Leonards, New South Wales, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
40
|
Dehghani A, Khajepour F, Dehghani M, Razmara E, Zangouey M, Abadi MFS, Nezhad RBA, Dabiri S, Garshasbi M. Hsa-miR-194-5p and hsa-miR-195-5p are down-regulated expressed in high dysplasia HPV-positive Pap smear samples compared to normal cytology HPV-positive Pap smear samples. BMC Infect Dis 2024; 24:182. [PMID: 38342922 PMCID: PMC10860252 DOI: 10.1186/s12879-023-08942-1] [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: 06/04/2023] [Accepted: 12/20/2023] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND The human papillomavirus (HPV) infection may affect the miRNA expression pattern during cervical cancer (CC) development. To demonstrate the association between high-risk HPVs and the development of cervix dysplasia, we examined the expression patterns of hsa-miR-194-5p and hsa-miR-195-5p in Pap smear samples from southeast Iranian women. We compared samples that were HPV-positive but showed no abnormality in the cytological examination to samples that were HPV-positive and had severe dysplasia. METHODS Pap smear samples were obtained from 60 HPV-positive (HPV-16/18) patients with histologically confirmed severe dysplasia (cervical intra-epithelial neoplasia (CIN 3) or carcinoma in situ) and the normal cytology group. The expression of hsa-miR-194-5p and hsa-miR-195-5p was analyzed by real-time quantitative PCR, using specific stem-loop primers and U6 snRNA as the internal reference gene. Clinicopathological features were associated with miRNA expression levels. Furthermore, functional enrichment analysis was conducted using in silico tools. The Kaplan-Meier survival method was also obtained to discriminate survival-significant candidate miRNAs in CC, and receiver operating characteristic (ROC) curves were constructed to assess the diagnostic value. RESULTS Compared to HPV-positive cytologically normal Pap smear samples, hsa-miR-194-5p and hsa-miR-195-5p relative expression decreased significantly in HPV-positive patients with a severe dysplasia Pap smear. Kaplan-Meier analysis indicated a significant association between the miR-194 decrease and poor CC survival. In essence, ROC curve analysis showed that miR-194-5p and miR-195-5p could serve as valuable markers for the development of cervix dysplasia in individuals who are positive for high-risk HPVs. CONCLUSIONS This study revealed that hsa-miR-194-5p and hsa-miR-195-5p may possess tumor suppressor capabilities in the context of cervical dysplasia progression. However, it remains uncertain whether these microRNAs are implicated in the transition of patients with high dysplasia to cervical cancer. We also showed the potential capability of candidate miRNAs as novel diagnostic biomarkers related to cervical dysplasia progression.
Collapse
Affiliation(s)
- Ali Dehghani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fardin Khajepour
- Department of Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Dehghani
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Ehsan Razmara
- Australian Regenerative Medicine Institute, Monash University, Melbourne, Australia
| | - Mohammadreza Zangouey
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Reza Bahram Abadi Nezhad
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
41
|
Zhang C, Yuan L, Zou Q, Shao C, Jia Y, Li J, Liao Y, Zhao X, Wen W, Jing X, Yang G, Wang W, Jiang H, Yao S. CircMAST1 inhibits cervical cancer progression by hindering the N4-acetylcytidine modification of YAP mRNA. Cell Mol Biol Lett 2024; 29:25. [PMID: 38331765 PMCID: PMC10854152 DOI: 10.1186/s11658-024-00540-6] [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: 11/19/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Cervical cancer (CCa) is the fourth most common cancer among females, with high incidence and mortality rates. Circular RNAs (circRNAs) are key regulators of various biological processes in cancer. However, the biological role of circRNAs in cervical cancer (CCa) remains largely unknown. This study aimed to elucidate the role of circMAST1 in CCa. METHODS CircRNAs related to CCa progression were identified via a circRNA microarray. The relationship between circMAST1 levels and clinicopathological features of CCa was evaluated using the clinical specimens and data of 131 patients with CCa. In vivo and in vitro experiments, including xenograft animal models, cell proliferation assay, transwell assay, RNA pull-down assay, whole-transcriptome sequencing, RIP assay, and RNA-FISH, were performed to investigate the effects of circMAST1 on the malignant behavior of CCa. RESULTS CircMAST1 was significantly downregulated in CCa tissues, and low expression of CircMAST1 was correlated with a poor prognosis. Moreover, our results demonstrated that circMAST1 inhibited tumor growth and lymph node metastasis of CCa. Mechanistically, circMAST1 competitively sequestered N-acetyltransferase 10 (NAT10) and hindered Yes-associated protein (YAP) mRNA ac4C modification to promote its degradation and inhibit tumor progression in CCa. CONCLUSIONS CircMAST1 plays a major suppressive role in the tumor growth and metastasis of CCa. In particular, circMAST1 can serve as a potential biomarker and novel target for CCa.
Collapse
Affiliation(s)
- Chunyu Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Li Yuan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Qiaojian Zou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Caixia Shao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Yan Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Jiaying Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Yan Liao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Xueyuan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Weijia Wen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Xu Jing
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Guofen Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China.
| | - Hongye Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China.
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, China.
| |
Collapse
|
42
|
Thilakasiri P, O'Keefe RN, To SQ, Chisanga D, Eissmann MF, Carli ALE, Duscio B, Baloyan D, Dmello RS, Williams D, Mariadason J, Poh AR, Pal B, Kile BT, Vissers JH, Harvey KF, Buchert M, Shi W, Ernst M, Chand AL. Mechanisms of cellular crosstalk in the gastric tumor microenvironment are mediated by YAP1 and STAT3. Life Sci Alliance 2024; 7:e202302411. [PMID: 37957015 PMCID: PMC10643184 DOI: 10.26508/lsa.202302411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Deregulation of the Hippo pathway is a driver for cancer progression and treatment resistance. In the context of gastric cancer, YAP1 is a biomarker for poor patient prognosis. Although genomic tumor profiling provides information of Hippo pathway activation, the present study demonstrates that inhibition of Yap1 activity has anti-tumor effects in gastric tumors driven by oncogenic mutations and inflammatory cytokines. We show that Yap1 is a key regulator of cell metabolism, proliferation, and immune responses in normal and neoplastic gastric epithelium. We propose that the Hippo pathway is targetable across gastric cancer subtypes and its therapeutic benefits are likely to be mediated by both cancer cell-intrinsic and -extrinsic mechanisms.
Collapse
Affiliation(s)
- Pathum Thilakasiri
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ryan N O'Keefe
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Sarah Q To
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Moritz F Eissmann
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Annalisa LE Carli
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Belinda Duscio
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Baloyan
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Rhynelle S Dmello
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - David Williams
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
- Department of Pathology, Austin Health, Heidelberg, Australia
| | - John Mariadason
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashleigh R Poh
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Bhupinder Pal
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Benjamin T Kile
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, Australia
| | | | - Kieran F Harvey
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Anatomy and Developmental Biology, and Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Michael Buchert
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Matthias Ernst
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Ashwini L Chand
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| |
Collapse
|
43
|
Patterson MR, Meijers AS, Ryder EL, Scarth JA, Evans D, Turner AL, Wasson CW, Darell JE, Theobald D, Cogan J, James CD, Wang M, Ladbury JE, Morgan IM, Samson A, Morgan EL, Macdonald A. E7-mediated repression of miR-203 promotes LASP1-dependent proliferation in HPV-positive cervical cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574687. [PMID: 38293147 PMCID: PMC10827106 DOI: 10.1101/2024.01.08.574687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Human papillomaviruses (HPV) are a major cause of malignancy, contributing to ∼5% of all human cancers worldwide, including most cervical cancer cases and a growing number of ano-genital and oral cancers. The major HPV viral oncogenes, E6 and E7, manipulate many host cellular pathways that promote cell proliferation and survival, predisposing infected cells to malignant transformation. Despite the availability of highly effective vaccines, there are still no specific anti-viral therapies targeting HPV or treatments for HPV-associated cancers. As such, a better understanding of viral-host interactions may allow the identification of novel therapeutic targets. Here, we demonstrate that the actin-binding protein LASP1 is upregulated in cervical cancer and significantly correlates with a poorer overall survival. In HPV positive cervical cancer, LASP1 depletion significantly inhibited proliferation in vitro , whilst having minimal effects in HPV negative cervical cancer cells. Furthermore, we show that the LASP1 SH3 domain is essential for LASP1-mediated proliferation in these cells. Mechanistically, we show that HPV E7 regulates LASP1 at the post-transcriptional level by repressing the expression of miR-203, which negatively regulated LASP1 mRNA levels by binding to its 3'UTR. Finally, we demonstrated that LASP1 expression is required for the growth of HPV positive cervical cancer cells in an in vivo tumourigenicity model. Together, these data demonstrate that HPV induces LASP1 expression to promote proliferation and survival role in cervical cancer, thus identifying a potential therapeutic target in these cancers.
Collapse
|
44
|
Kim N, Yeo MK, Sun P, Lee D, Kim DK, Lee SI, Chung C, Kang DH, Lee JE. Cathepsin C regulates tumor progression via the Yes-associated protein signaling pathway in non-small cell lung cancer. Am J Cancer Res 2024; 14:97-113. [PMID: 38323275 PMCID: PMC10839315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024] Open
Abstract
Cathepsin C (CTSC), also known as dipeptidyl peptidase I, is a cathepsin with lysosomal exocysteine protease activity and a central coordinator for the activation of neutrophil-derived serine proteases in the lysosomes of neutrophils. Although the role of CTSC in various cancers, including liver and breast cancers, has recently been reported, its role in non-small cell lung cancer (NSCLC) is largely unknown. This study aimed to investigate the functional role of CTSC in NSCLC and the molecular mechanisms underlying CTSC involvement in disease progression. CTSC overexpression markedly enhanced the growth, motility, and invasiveness of NSCLC cells in vitro and in vivo. CTSC knockdown using shRNA in NSCLC cells reversed the migratory and invasive behavior of NSCLC cells. CTSC also induced epithelial-mesenchymal transition through the Yes-associated protein signaling pathway. In addition, our analyses of clinical samples confirmed that high CTSC expression was associated with lymph node metastasis and recurrence in lung adenocarcinoma. In conclusion, CTSC plays an important role in the progression of NSCLC. Thus, targeting CTSC may be a promising treatment option for patients with NSCLC.
Collapse
Affiliation(s)
- Nayoung Kim
- Cancer Research Institute, Chungnam National UniversityDaejeon 35015, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Pureum Sun
- Research Institute for Medical Sciences, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Dahye Lee
- Infection Control Convergence Research Center, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Duk Ki Kim
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Song-I Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Da Hyun Kang
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| | - Jeong Eun Lee
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National UniversityDaejeon 34134, Republic of Korea
| |
Collapse
|
45
|
Sharma S, Chauhan D, Kumar S, Kumar R. Impact of HPV strains on molecular mechanisms of cervix cancer. Microb Pathog 2024; 186:106465. [PMID: 38036109 DOI: 10.1016/j.micpath.2023.106465] [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: 08/19/2023] [Revised: 11/07/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
PURPOSE Cervical cancer accounts for a high number of deaths worldwide. Risk factors are extensive for cervix cancer but Human papillomavirus (HPV) plays a prime role in its development. Different strains of HPV are prevalent globally, which show different grades of mortality and morbidity among women. This study is planned to evaluate the molecular mechanism of different strains of HPV infection and progression leading to cervix cancer. METHODS This review includes different research articles on cervix cancer progression reported from India and all over the world. RESULTS HPV 16 and 18 are prevalent strains using heparan sulfate-independent and dependent pathways for viral replication inside the cell. It also uses transcription mechanisms through NF-kappa B, FOXA-1, and AP-1 genes while strains like HPV-35, 45, and 52 are also predominant in India, which showed a very slow mechanism of progression due to which mortality rate is low after their infection with these strains. CONCLUSION HPV uses E6 and E7 proteins which activate NF-kappa B and AP-1 pathway which suppresses the tumor suppressor gene and activates cytokine production, causing inflammation and leading to a decrease in apoptosis due to Caspase-3 activation. In contrast, the E7 protein involves HOXA genes and decreases apoptotic factors due to which mortality and incidence rates are low in viruses that use E7 motifs. Some HPV strains employ the cap-dependent pathway, which is also associated with lower mortality and infection rates.
Collapse
Affiliation(s)
- Sunidhi Sharma
- Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh, 176215, India.
| | - Disha Chauhan
- Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh, 176215, India.
| | - Sunil Kumar
- Central University of Himachal Pradesh, Dharamshala, Himachal Pradesh, 176215, India.
| | - Ranjit Kumar
- Nagaland University, Lumami, Nagaland, 798627, India.
| |
Collapse
|
46
|
Peng Q, Wang L, Zuo L, Gao S, Jiang X, Han Y, Lin J, Peng M, Wu N, Tang Y, Tian H, Zhou Y, Liao Q. HPV E6/E7: insights into their regulatory role and mechanism in signaling pathways in HPV-associated tumor. Cancer Gene Ther 2024; 31:9-17. [PMID: 38102462 DOI: 10.1038/s41417-023-00682-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/09/2023] [Accepted: 10/24/2023] [Indexed: 12/17/2023]
Abstract
Human papillomavirus (HPV) is a class of envelope-free double-stranded DNA virus. HPV infection has been strongly associated with the development of many malignancies, such as cervical, anal and oral cancers. The viral oncoproteins E6 and E7 perform central roles on HPV-induced carcinogenic processes. During tumor development, it usually goes along with the activation of abnormal signaling pathways. E6 and E7 induces changes in cell cycle, proliferation, invasion, metastasis and other biological behaviors by affecting downstream tumor-related signaling pathways, thus promoting malignant transformation of cells and ultimately leading to tumorigenesis and progression. Here, we summarized that E6 and E7 proteins promote HPV-associated tumorigenesis and development by regulating the activation of various tumor-related signaling pathways, for example, the Wnt/β-catenin, PI3K/Akt, and NF-kB signaling pathway. We also discussed the importance of HPV-encoded E6 and E7 and their regulated tumor-related signaling pathways for the diagnosis and effective treatment of HPV-associated tumors.
Collapse
Affiliation(s)
- Qiu Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Lujuan Wang
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Liang Zuo
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Shuichao Gao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yaqian Han
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Jinguan Lin
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Mingjing Peng
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Nayiyuan Wu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Yanyan Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
| | - Hao Tian
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
| | - Yujuan Zhou
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- University of South China, Hengyang, 421001, Hunan, China.
- Public Service Platform of Tumor organoids Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Qianjin Liao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China.
- University of South China, Hengyang, 421001, Hunan, China.
- Public Service Platform of Tumor organoids Technology, 283 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
47
|
Huang J, Wei W, Kang F, Tan S, Li Y, Lu X, Wang N. ANXA3, associated with YAP1 regulation, participates in the proliferation and chemoresistance of cervical cancer cells. Genes Genomics 2023; 45:1575-1586. [PMID: 37843781 DOI: 10.1007/s13258-023-01461-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: 04/28/2023] [Accepted: 10/01/2023] [Indexed: 10/17/2023]
Abstract
BACKGROUND Cervical cancer, as one of the most common cancers in women, remains a major health threat worldwide. Annexin A3 (ANXA3), a component of the annexin family, is upregulated in numerous cancers, with no explicit role in cervical cancer. OBJECTIVE This study aims to investigate the function of ANXA3 in cervical cancer. METHODS Differential expression genes between the cervical cancer tissues of patients and the controls were analyzed in The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA) database. Using transfection approaches to either upregulate or downregulate ANXA3, its role in cell proliferation and chemosensitivity of human cervical cancer cell lines (HeLa and C33A) was evaluated. Furthermore, the binding activity between YAP1 and ANXA3 was also explored. RESULTS Genomics analysis indicated that differential genes were mostly associated with cell cycle progression and DNA replication. ANXA3 was highly expressed in the cervical cancer tissues and closely linked to malignancy degree. Knockdown of ANXA3 in cervical cancer cells inhibited cell cycle progression. A similar result was observed in the reduction of cyclin D, CDK4, cyclin E, and CDK2 in cervical cancer cells with ANXA3 silencing. Cervical cancer cells obtained high sensitivity to cisplatin (DDP) when ANXA3 was downregulated. Conversely, these capabilities were the opposite in cervical cancer cells overexpressing ANXA3. Furthermore, the expression levels of ANXA3 and YAP1 were positively correlated. YAP1 upregulation was positively connected with malignant behaviors, which were reversed by ANXA3 downregulation. CONCLUSION In light of our findings, targeting ANXA3 expressed in cervical cancer might contribute to more potential therapeutic strategies.
Collapse
Affiliation(s)
- Jiazhen Huang
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Wei Wei
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Fuli Kang
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Shuang Tan
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Yibing Li
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Xiaohang Lu
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China
| | - Ning Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Dalian Medical University, No. 467, Zhongshan Road, Dalian, People's Republic of China.
| |
Collapse
|
48
|
Nakajima M, Tanaka K, Yoneshima Y, Yamashita S, Shibahara D, Iwama E, Okamoto I. YAP mediates resistance to EGF-induced apoptosis in EGFR-mutated non-small cell lung cancer cells. Biochem Biophys Res Commun 2023; 681:120-126. [PMID: 37774569 DOI: 10.1016/j.bbrc.2023.09.067] [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: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 10/01/2023]
Abstract
Mechanisms underlying the growth and survival of non-small cell lung cancer (NSCLC) cells positive for activating mutations of the epidermal growth factor receptor gene (EGFR) have remained unclear. We here examined the functional relation between such mutant forms of EGFR and Yes-associated protein (YAP), a transcriptional coactivator of the Hippo signaling pathway that regulates cell proliferation and survival. Under the condition of serum deprivation, epidermal growth factor (EGF) induced activation of YAP in NSCLC cell lines positive for mutated EGFR but not in those wild type (WT) for EGFR. Similar EGF-induced activation of YAP was apparent in A549 lung cancer cells forcibly expressing mutant EGFR but not in those overexpressing the WT receptor. Furthermore, EGF induced apoptotic cell death in serum-deprived A549 cells overexpressing the WT form of EGFR but not in those expressing mutant EGFR, and knockdown of YAP rendered the latter cells sensitive to this effect of EGF. Our results thus suggest that activation of YAP mediates resistance of EGFR-mutated NSCLC cells to EGF-induced apoptosis and thereby contributes specifically to the survival of such cells.
Collapse
Affiliation(s)
- Maako Nakajima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kentaro Tanaka
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Yasuto Yoneshima
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Sho Yamashita
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Daisuke Shibahara
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Eiji Iwama
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Isamu Okamoto
- Department of Respiratory Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
49
|
Vemula S, Bonala S, Vadde NK, Natu JZ, Basha R, Vadde R, Ahmad S. Drug resistance and immunotherapy in gynecologic cancers. Life Sci 2023; 332:122104. [PMID: 37730109 DOI: 10.1016/j.lfs.2023.122104] [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: 07/11/2023] [Revised: 09/07/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023]
Abstract
Gynecologic malignancies (GMs) are relatively less focused cancers by oncologists and researchers. The five-year survival rate of patients with GMs remained almost the same during the last decade. The development of drug resistance GMs makes it even more challenging to tackle due to tumor heterogeneity, genomic instability, viral/non-viral antigens, and etiological tumor origin. A precision medicine approach, including gene therapies, is under testing to restore tumor responsiveness to therapeutics and immunotherapy. With more data being uncovered, immunotherapy is emerging as a viable alternative for achieving promising results. This review highlights the drug resistance mechanisms and immunotherapeutic approaches to managing GMs better. The approval of immune therapeutic drugs in recent years shifted this notion. It provided hope for researchers, clinicians, and patients with GMs to experience the anti-cancer benefits of these therapies.
Collapse
Affiliation(s)
| | | | | | - Jay Z Natu
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, AL, USA
| | - Raasil Basha
- Department of Biology-Environmental Health, Missouri Southern State University, Joplin, MO, USA
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa, Andhra Pradesh, India.
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, USA.
| |
Collapse
|
50
|
Gelbard MK, Munger K. Human papillomaviruses: Knowns, mysteries, and unchartered territories. J Med Virol 2023; 95:e29191. [PMID: 37861365 PMCID: PMC10608791 DOI: 10.1002/jmv.29191] [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: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023]
Abstract
There has been an explosion in the number of papillomaviruses that have been identified and fully sequenced. Yet only a minute fraction of these has been studied in any detail. Most of our molecular research efforts have focused on the E6 and E7 proteins of "high-risk," cancer-associated human papillomaviruses (HPVs). Interactions of the high-risk HPV E6 and E7 proteins with their respective cellular targets, the p53 and the retinoblastoma tumor suppressors, have been investigated in minute detail. Some have thus questioned if research on papillomaviruses remains an exciting and worthwhile area of investigation. However, fundamentally new insights on the biological activities and cellular targets of the high-risk HPV E6 and E7 proteins have been discovered and previously unstudied HPVs have been newly associated with human diseases. HPV infections continue to be an important cause of human morbidity and mortality and since there are no antivirals to combat HPV infections, research on HPVs should remain attractive to new investigators and biomedical funding agencies, alike.
Collapse
Affiliation(s)
- Maya K. Gelbard
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences
- Department of Developmental, Molecular and Cellular Biology, Tufts University School of Medicine, Boston, MA 02111
| | - Karl Munger
- Genetics, Molecular and Cellular Biology Program, Graduate School of Biomedical Sciences
- Department of Developmental, Molecular and Cellular Biology, Tufts University School of Medicine, Boston, MA 02111
| |
Collapse
|