1
|
Gupta A, Devi A, Kamboj M, Narwal A, Kumar A, Singh S. Immunohistochemical Expression of PARK2 and YAP in Oral Epithelial Dysplasia and Oral Squamous Cell Carcinoma. Head Neck Pathol 2025; 19:50. [PMID: 40279065 PMCID: PMC12031683 DOI: 10.1007/s12105-025-01790-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025]
Abstract
INTRODUCTION Oral squamous cell carcinoma is the most prevalent of all the oral cancers. There is no definitive marker available for its early diagnosis and its effective prognosis. YAP serves as a transcriptional regulator in hippo tumor suppressor pathway thereby activating the transcription of genes taking part in cellular proliferation, alteration, migration, and invasion. On the contrary, PARK2 acts as a tumor suppressor and has been widely explored in various malignancies. However, its role in OSCC carcinogenesis is untrodden. AIM To evaluate the Immunohistochemical expression of YAP and PARK2 in oral epithelial dysplasia and Oral Squamous Cell Carcinoma and establish them as prognostic markers. MATERIAL AND METHOD The study sample consisted of 70 formalin fixed paraffin embedded tissue sections of normal oral mucosa (10), oral epithelial dysplasia (30) and oral squamous cell carcinoma (30). Immunohistochemical analysis of YAP and PARK2 was done and final scores were calculated. Further, the markers were graded as low and high expression groups. Statistical analysis was done using chi-square test, cox regression analysis and Spearman's correlation. Kaplan Meier plot for survival analysis was also plotted. RESULT Immunohistochemical expression of YAP depicted a gradual incline from normal oral mucosa to oral squamous cell carcinoma while PARK2 showed a reverse trend. Significant difference of YAP and PARK2 expression between three groups was noted. Inverse moderate degree of correlation was observed between both the markers in OSCC group. CONCLUSION Concomitant immunoexpression of YAP and PARK2 with a moderate degree of inverse correlation from normal oral mucosa to oral squamous cell carcinoma could probably serve as diagnostic and prognostic markers as they might act through a common mechanism, probably hippo/YAP signaling, which could be further confirmed by larger sample size, including longer follow up in future studies.
Collapse
Affiliation(s)
- Akanksha Gupta
- Department of Oral Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pt.BD. Sharma University, Rohtak, Haryana, 124001, India
| | - Anju Devi
- Department of Oral Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pt.BD. Sharma University, Rohtak, Haryana, 124001, India.
| | - Mala Kamboj
- Department of Oral Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pt.BD. Sharma University, Rohtak, Haryana, 124001, India
| | - Anjali Narwal
- Department of Oral Pathology and Microbiology, Post Graduate Institute of Dental Sciences, Pt.BD. Sharma University, Rohtak, Haryana, 124001, India
| | - Adarsh Kumar
- Department of Public Health Dentistry, Post Graduate Institute of Dental Sciences, Pt.BD. Sharma University, Rohtak, Haryana, 124001, India
| | - Sunita Singh
- Department of Pathology, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak City, Haryana, 124001, India
| |
Collapse
|
2
|
Chavan PR, Pandey R, Patil BM, Murti K, Kumar N. Unravelling key signaling pathways for the therapeutic targeting of non-small cell lung cancer. Eur J Pharmacol 2025; 998:177494. [PMID: 40090536 DOI: 10.1016/j.ejphar.2025.177494] [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: 12/11/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
Lung cancer (LC) remains the foremost cause of cancer-related mortality across the globe. Non-small cell lung cancer (NSCLC) is a type of LC that exhibits significant heterogeneity at histological and molecular levels. Genetic alterations in upstream signaling molecules activate cascades affecting apoptosis, proliferation, and differentiation. Disruption of these signaling pathways leads to the proliferation of cancer-promoting cells, progression of cancer, and resistance to its treatment. Recent insights into the function of signaling pathways and their fundamental mechanisms in the onset of various diseases could pave the way for new therapeutic approaches. Recently, numerous drug molecules have been created that target these cell signaling pathways and could be used alongside other standard therapies to achieve synergistic effects in mitigating the pathophysiology of NSCLC. Additionally, many researchers have identified several predictive biomarkers, and alterations in transcription factors and related pathways are employed to create new therapeutic strategies for NSCLC. Findings suggest using specific inhibitors to target cellular signaling pathways in tumor progression to treat NSCLC. This review investigates the role of signaling pathways in NSCLC development and explores novel therapeutic strategies to enhance clinical treatment options for NSCLC.
Collapse
Affiliation(s)
- Pavan Ramrao Chavan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Ruchi Pandey
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Baswant Malesh Patil
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Krishna Murti
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India
| | - Nitesh Kumar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education & Research, Hajipur, Bihar, India.
| |
Collapse
|
3
|
Choi S, Yu E, Park S, Oh SW, Kwon K, Kim G, Ha H, Shin HS, Min S, Song M, Cho JY, Lee J. Protective effect of melatonin against blue light-induced cell damage via the TRPV1-YAP pathway in cultured human epidermal keratinocytes. Biofactors 2025; 51:e70015. [PMID: 40183558 PMCID: PMC11970215 DOI: 10.1002/biof.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Although blue light has been known to negatively affect skin cells, its detailed signaling mechanisms and anti-blue light agents have not been clearly elucidated. We investigated the involvement of Yes-associated protein (YAP)-mediated Hippo signaling in blue light-induced apoptosis, depending on the degree of blue light exposure. Additionally, we elucidated the effects of melatonin on blue light-irradiated keratinocytes and examined their action mechanisms. After blue light irradiation, its effects and antagonizing effects of melatonin on cell proliferation, apoptosis, DNA damage, and transient receptor potential vanilloid 1 (TRPV1)/YAP-mediated signaling were examined in HaCaT cells using western blots, image analysis, flow cytometric analysis, co-immunoprecipitation, and immunocytochemistry. We found that melatonin treatment attenuated the reduced cell viability and increased production of reactive oxygen species (ROS) in response to blue light irradiation. In the experiments to investigate the mechanism of action of blue light and melatonin, we found that YAP changed its binding protein, either p73 or TEAD, depending on the degree of blue light exposure. Melatonin treatment reduced blue light-induced phosphorylation of TRPV1 and MST1/2. Upon treatment with capsazepine, an antagonist of TRPV1, MST1/2 activation also reduced. Furthermore, we found that prolonged blue light irradiation induced DNA damage, which in turn induced YAP-p73 nuclear translocation. These effects were also notably attenuated by melatonin. These findings indicate that depending on the duration of blue light irradiation, two different YAP-mediated Hippo signaling pathways are activated. Additionally, these findings suggest that melatonin could be a potential therapeutic agent for blue light-induced skin damage.
Collapse
Affiliation(s)
- Seoyoung Choi
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Eunbi Yu
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - See‐Hyoung Park
- Department of Bio and Chemical EngineeringHongik UniversitySejong CityKorea
| | - Sae Woong Oh
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Kitae Kwon
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Gyeonghyeon Kim
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Heejun Ha
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Hee Seon Shin
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Seokhyeon Min
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Minkyung Song
- Integrative Research of T cells Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
- Department of Biopharmaceutical ConvergenceSungkyunkwan UniversitySuwon CityKorea
| | - Jae Youl Cho
- Molecular Immunology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| | - Jongsung Lee
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and BioengineeringSungkyunkwan UniversitySuwon CityKorea
| |
Collapse
|
4
|
Zhu J, Zhang Z, Chu Y, Xie Z, Zeng D, Jin L, Li L. Application of Nanomaterial-Mediated Ferroptosis Regulation in Kidney Disease. Int J Nanomedicine 2025; 20:1637-1659. [PMID: 39931533 PMCID: PMC11808220 DOI: 10.2147/ijn.s496644] [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: 10/24/2024] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Kidney diseases are a significant global cause of death and disability, resulting from the destruction of kidney structure and function due to an imbalance between the death of renal parenchymal cells and the proliferation or recruitment of maladaptive cells, caused by various pathogenic factors. Currently, therapies and their efficacy for kidney diseases are limited. Ferroptosis is a newly discovered iron-dependent regulated cell death. The imbalance of iron homeostasis and lipid metabolism affects the occurrence and progression of kidney diseases by triggering ferroptosis, which is considered an important target for the development of kidney disease drugs. However, in clinical practice, targeted ferroptosis therapy for kidney diseases faces obstacles such as poor drug solubility, low drug resistance, and imprecise targeting. With the rapid development of nanomaterials in the medical field, new opportunities have emerged for the precise regulation of ferroptosis in the treatment of kidney diseases. This article provides a detailed introduction to the regulatory mechanisms of ferroptosis, the properties of nanomaterials, and their application in the treatment of kidney diseases, with a focus on discussing the mechanisms of action and therapeutic potential of nanomaterials based on ferroptosis regulation in kidney diseases. The aim of this article is to provide new ideas and directions for future kidney disease treatments.
Collapse
Affiliation(s)
- Jiamin Zhu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Zhen Zhang
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
- School of First Clinical Medical College, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Yanhui Chu
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Zhongyin Xie
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Dongmei Zeng
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Lijiao Jin
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| | - Luxin Li
- Heilongjiang Key Laboratory of Anti-Fibrosis Biotherapy, Mudanjiang Medical University, Mudanjiang, People’s Republic of China
| |
Collapse
|
5
|
Huang L, Wen Y, Guo Q, Zhang C, Yang X, Li M, Liu Y, Li X, Tang J, Zhou X, Qi Q, Zhang H, Liu T. CK2α-mediated phosphorylation of DUB3 promotes YAP1 stability and oncogenic functions. Cell Death Dis 2025; 16:27. [PMID: 39827153 PMCID: PMC11743126 DOI: 10.1038/s41419-024-07323-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: 08/06/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
The aberrant upregulation of Yes-associated protein 1 (YAP1) in a variety of solid cancers contributes to tumor progression and poor clinical outcomes, rendering it an appealing therapeutic target. However, effective therapies to directly target YAP1 remain challenging. In this study, we perform a high-throughput screening and identify Casein kinase II (CK2) as an uncharacterized upstream regulator of YAP1 turnover in cancer cells of ovarian cancer and several other cancer types. Pharmacological inhibition of Casein kinase II by Silmitasertib or genetic depletion of the catalytic subunit of Casein kinase II (CK2α) markedly destabilizes YAP1 and consequently suppresses its oncogenic functions in vitro and in vivo. Moreover, we reveal that DUB3 as a bona fide deubiquitinase of YAP1, which functionally links CK2 and YAP1 stability in a variety of human cancers. Mechanistically, CK2α directly phosphorylates DUB3 at Thr495, thereby facilitating DUB3-mediated deubiquitination process of YAP1. On the contrary, the loss of Thr495 phosphorylation by the phosphorylation-defective mutant DUB3 T495A, the cancer-related mutant DUB3 D496H and CK2 inhibition failed to deubiquitinate and stabilize YAP1 effectively. Notably, upregulated expressions of CK2α and DUB3 in ovarian cancer positively correlate with YAP1 overexpression. Collectively, our findings demonstrate the functional significance of the CK2α-DUB3 axis in YAP1 stabilization and YAP1-driven tumor progression, highlighting that strategies to target this axis might be of benefit in the clinical management of ovarian cancer and several other lethal cancers with aberrantly upregulated YAP1.
Collapse
Affiliation(s)
- Lei Huang
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China
| | - Yalei Wen
- Research Institute for Maternal and Child Health, The Affiliated Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Traditional Chinese Medicine, School of Pharmacy, Jinan University, Guangzhou, China
| | - Qin Guo
- Department of Pathology, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Caishi Zhang
- Jianli Traditional Chinese Medicine Hospital, Jingzhou, China
| | - Xiao Yang
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China
| | - Mei Li
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China
| | - YiXia Liu
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China
| | - Xinying Li
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China
| | - Jiaxin Tang
- School of Pharmacology, Lanzhou University, Lanzhou, China
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiaofeng Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Qi Qi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China.
| | - Haoxing Zhang
- Guangdong Provincial Key Laboratory of Genome Stability and Disease Prevention, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China.
| | - Tongzheng Liu
- Department of General Surgery, Guangzhou Red Cross Hospital/State Key Laboratory of Bioactive Molecules and Druggability Assessment/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China/College of Pharmacy, Jinan University, Guangzhou, China.
- The State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China.
| |
Collapse
|
6
|
Wang Z, Zhao M, Huang X, Wang Y, Li W, Qiao J, Yang X. Therapeutic types and advantages of functionalized nanoparticles in inducing ferroptosis in cancer therapy. Ann Med 2024; 56:2396568. [PMID: 39276361 PMCID: PMC11404394 DOI: 10.1080/07853890.2024.2396568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/24/2024] [Accepted: 07/10/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND The clinical efficacy of cancer treatment protocols remains unsatisfactory; however, the emergence of ferroptosis-driven therapy strategies has renewed hope for tumor treatment, owing to their remarkable tumor suppression effects. Biologically based small-molecule inducers are used in conventional method to induce ferroptosis. Nevertheless, some molecular drugs have limited solubility, poor ability to target cells, and fast metabolism, which hinder their ability to induce ferroptosis over a prolonged period. Fortunately, further investigations of ferroptosis and the development of nanotechnology have demonstrated that nanoparticles (NPs) are more efficient in inducing ferroptosis than drugs alone, which opens up new perspectives for cancer therapy. OBJECTIVE In order to organize a profile of recent advance in NPs for inducing ferroptosis in cancer therapy, and NPs were comprehensively classified in a new light.Materials and methods: We comprehensively searched the databases such as PubMed and Embase. The time limit for searching was from the establishment of the database to 2023.11. All literatures were related to "ferroptosis", "nanoparticles", "nanodelivery systems", "tumors", "cancer". RESULTS We summarized and classified the available NPs from a new perspective. The NPs were classified into six categories based on their properties: (1) iron oxide NPs (2) iron - based conversion NPs (3) core-shell structure (4) organic framework (5) silica NPs (6) lipoprotein NPs. According to the therapeutic types of NPs, they can be divided into categories: (1) NPs induced ferroptosis-related immunotherapy (2) NPs loaded with drugs (3) targeted therapy of NPs (4) multidrug resistance therapy (5) gene therapy with NPs (6) energy conversion therapy. CONCLUSIONS The insights gained from this review can provide ideas for the development of original NPs and nanodelivery systems, pave the way for related nanomaterials application in clinical cancer therapy, and advance the application and development of nanotechnology in the medical field.
Collapse
Affiliation(s)
- Ziying Wang
- School of Nursing, Shandong Second Medical University, Weifang, Shandong, China
| | - Miaomiao Zhao
- Department of Pathology, Shandong Second Medical University, Weifang, Shandong, China
| | - Xiaotong Huang
- School of Nursing, Shandong Second Medical University, Weifang, Shandong, China
| | - Yuxin Wang
- School of Pharmacy, Binzhou Medical College, Yantai, Shandong, China
| | - Wentong Li
- Department of Pathology, Shandong Second Medical University, Weifang, Shandong, China
| | - Jianhong Qiao
- Department of Outpatient, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Xiao Yang
- School of Nursing, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
7
|
Shi C, Chen L, Huang K, Yang G, Shi T, Li J, Zheng H. m6A methylation regulators and ncRNAs in osteosarcoma: Potential therapeutic strategies. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:34-42. [PMID: 39461672 DOI: 10.1016/j.pbiomolbio.2024.10.003] [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: 09/05/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 10/29/2024]
Abstract
Osteosarcoma (OS) represents the primary form of bone cancer observed in paediatric and adolescent populations. Nearly 10%-15% of patients have metastases at diagnosis, and the 5-year survival rate was less than 20%. Although numerous investigators have offered significant efforts, the survival rates for patients with OS have remained almost unchanged over the past three decades. The most pervasive and abundant modification of internal transcripts in eukaryotic messenger RNAs (mRNAs) is N6-methyladenosine (m6A), and it is regulated by m6A methylation regulators. A number of recent studies have demonstrated that m6A modifications can regulate the biological activities of tumour cells and are intimately linked with cancer development, prognosis, drug resistance, and therapy. N6-methyladenosine modification of Non-coding RNA (ncRNA) has likewise shown a broad potential in gene regulation and tumor biology. Epigenetic changes induced by mRNAs and ncRNAs methylation are important for a better understanding of OS development and targeted drug development. Therefore, this paper summarises the biological functions of m6A-modified regulators in osteosarcoma and the role of mutual regulation between m6A and ncRNAs in osteosarcoma. Furthermore, the potential clinical applications of m6A modifications in OS are presented for consideration. It provides new directions for the future research and clinical treatment strategies of osteosarcoma.
Collapse
Affiliation(s)
- Ce Shi
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Lei Chen
- Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Kui Huang
- Department of Orthopedics, Feng Xian People's Hospital, Xuzhou, 221700, China
| | - Guanghui Yang
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Tingting Shi
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China
| | - Jinshuang Li
- Department of Cardiology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China.
| | - Hongbing Zheng
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, 223800, China.
| |
Collapse
|
8
|
Zang Y, Lu Y, Yu J, Dong Q, Shi Y, Ying G, Liang Z. FOXP3 inhibits proliferation and migration by competitively inhibiting YAP1 in nasopharyngeal carcinoma. Oral Oncol 2024; 159:107066. [PMID: 39413576 DOI: 10.1016/j.oraloncology.2024.107066] [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: 06/30/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/18/2024]
Abstract
Hippo signalling is involved in the coordination of extracellular signals that control tissue homeostasis and organ size. Yes-associated protein 1 (YAP1) is regulated primarily by Hippo signalling through coactivation of transcription factors with GATA domains called TEADs. However, small-molecule orthosteric inhibitors of YAP1 are difficult to develop due to its tight binding to TEAD4 via a flat interface. Previous studies have shown that chlorpromazine (CPZ) can inhibit YAP1 expression. MTT, colony formation, wound healing, Transwell migration and Western blot assays were performed to explore how CPZ affects nasopharyngeal carcinoma (NPC) cells through FOXP3. In addition, immunofluorescence and live-cell imaging were used to detect YAP1 intracellular localization after CPZ administration. Through the HDOCK website, we predicted protein binding regions between FOXP3 and TEAD4. Western blot and co-IP experiments were used to verify the relationship between FOXP3 and YAP1. The UCSC Xena database, LinkedOmics database and KM plotter website were used to assess the prognostic value of FOXP3 in head and neck squamous cell carcinoma (HNSCC). Age, sex, pathological tumour-node-metastasis (pTMN) stage, grade, smoking status and FOXP3 expression were included in an overall survival nomogram model. Our findings revealed that FOXP3 has the ability to competitively interacts competitively with TEAD4 to inhibit YAP1 expression. By increasing FOXP3 expression, CPZ induces YAP1 nuclear export and phosphorylation, consequently suppressing NPC cell proliferation and migration. Collectively, our findings indicate that FOXP3 competitively binds TEAD4 to regulate YAP1 localization in the nucleus and cytoplasm to suppress NPC progression. Consequently, FOXP3 may be a prognostic indicator for HNSCC.
Collapse
Affiliation(s)
- Yiqing Zang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Yi Lu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jiaxi Yu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Qiuping Dong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China
| | - Yue Shi
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China
| | - Guoguang Ying
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, PR China.
| | - Zheng Liang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
9
|
Zhao Y, Qin C, Lin C, Li Z, Zhao B, Li T, Zhang X, Wang W. Pancreatic ductal adenocarcinoma cells reshape the immune microenvironment: Molecular mechanisms and therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189183. [PMID: 39303859 DOI: 10.1016/j.bbcan.2024.189183] [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: 06/09/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a digestive system malignancy characterized by challenging early detection, limited treatment alternatives, and generally poor prognosis. Although there have been significant advancements in immunotherapy for hematological malignancies and various solid tumors in recent decades, with impressive outcomes in recent preclinical and clinical trials, the effectiveness of these therapies in treating PDAC continues to be modest. The unique immunological microenvironment of PDAC, especially the abnormal distribution, complex composition, and variable activation states of tumor-infiltrating immune cells, greatly restricts the effectiveness of immunotherapy. Undoubtedly, integrating data from both preclinical models and human studies helps accelerate the identification of reliable molecules and pathways responsive to targeted biological therapies and immunotherapies, thereby continuously optimizing therapeutic combinations. In this review, we delve deeply into how PDAC cells regulate the immune microenvironment through complex signaling networks, affecting the quantity and functional status of immune cells to promote immune escape and tumor progression. Furthermore, we explore the multi-modal immunotherapeutic strategies currently under development, emphasizing the transformation of the immunosuppressive environment into an anti-tumor milieu by targeting specific molecular and cellular pathways, providing insights for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Cheng Qin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Chen Lin
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Zeru Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Bangbo Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Tianyu Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Xiangyu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China
| | - Weibin Wang
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100023, PR China; Key Laboratory of Research in Pancreatic Tumor, Chinese Academy of Medical Sciences, Beijing 100023, PR China; National Science and Technology Key Infrastructure on Translational Medicine in Peking Union Medical College Hospital, Beijing 100023, PR China.
| |
Collapse
|
10
|
Diskul-Na-Ayudthaya P, Bae SJ, Bae YU, Van NT, Kim W, Ryu S. ANKRD1 Promotes Breast Cancer Metastasis by Activating NF- κB-MAGE-A6 Pathway. Cancers (Basel) 2024; 16:3306. [PMID: 39409926 PMCID: PMC11476229 DOI: 10.3390/cancers16193306] [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: 06/14/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Early detection and surgical excision of tumors have helped improve the survival rate of patients with breast cancer. However, patients with metastatic cancer typically have a poor prognosis. In this study, we propose that ANKRD1 promotes metastasis of breast cancer. ANKRD1 was found to be highly expressed in the MDA-MB-231 and MDA-LM-2 highly metastatic breast cancer cell lines compared to the non-metastatic breast cancer cell lines (MCF-7, ZR-75-30, T47D) and normal breast cancer cells (MCF-10A). Furthermore, high-grade tumors showed increased levels of ANKRD1 compared to low-grade tumors. Both in vitro and in vivo functional studies demonstrated the essential role of ANKRD1 in cancer cell migration and invasion. The previous studies have suggested a significant role of NF-κB and MAGE-A6 in breast cancer metastasis, but the upstream regulators of this axis are not well characterized. Our study suggests that ANKRD1 promotes metastasis of breast cancer by activating NF-κB as well as MAGE-A6 signaling. Our findings show that ANKRD1 is a potential therapeutic target and a diagnostic marker for breast cancer metastasis.
Collapse
Affiliation(s)
- Penchatr Diskul-Na-Ayudthaya
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
- Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Seon Joo Bae
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Yun-Ui Bae
- Precision Medicine Lung Cancer Center, Konkuk University Medical Center, Konkuk University, Seoul 05030, Republic of Korea;
| | - Ngu Trinh Van
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Wootae Kim
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
| | - Seongho Ryu
- Soonchunhyang Institute of Medi-bio Science (SIMS), Department of Integrated Biomedical Sciences, Soonchunhyang University, Asan-si 31151, Republic of Korea; (P.D.-N.-A.); (S.J.B.); (N.T.V.)
- Department of Pathology, College of Medicine, Soonchunhyang University, Asan-si 311151, Republic of Korea
| |
Collapse
|
11
|
Wang T, Ran B, Luo Y, Ma J, Li J, Li P, Li M, Li D. Functional study of the ST6GAL2 gene regulating skeletal muscle growth and development. Heliyon 2024; 10:e37311. [PMID: 39296044 PMCID: PMC11407927 DOI: 10.1016/j.heliyon.2024.e37311] [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: 01/22/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 09/21/2024] Open
Abstract
ST6GAL2, a member of the sialoglycosyltransferase family, primarily localizes within the cellular Golgi apparatus. However, the role of the ST6GAL2 gene in skeletal muscle growth and development remains elusive. In this study, the impact of the ST6GAL2 gene on the proliferation, differentiation, and apoptosis of primary chicken myoblasts at the cellular level was investigated. Quantitative fluorescent PCR was used to measure the expression levels of genes. Subsequently, using gene knockout mice, we assessed its effects on skeletal muscle growth and development in vivo. Our findings reveal that the ST6GAL2 gene promotes the expression of cell cycle and proliferation-related genes, including CCNB2 and PCNA, and apoptosis-related genes, such as Fas and Caspase-9. At the individual level, double knockout of ST6GAL2 inhibited the formation of both fast and slow muscle fibers in the quadriceps, extensor digitorum longus, and tibial anterior muscle, while promoting their formation in the gastrocnemius and soleus. These results collectively demonstrate that the ST6GAL2 gene facilitates the proliferation, apoptosis, and fusion processes of primary chicken myoblasts. Additionally, it promotes the enlargement of cross-sectional muscle fiber areas and regulates the formation of fast and slow muscle fibers at the individual level, albeit inhibiting muscle fusion. This study provides valuable insights into the role of the ST6GAL2 gene in promoting proliferation of skeletal muscle.
Collapse
Affiliation(s)
- Tao Wang
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| | - Bo Ran
- Sichuan Animal Science Academy, Chengdu, 610066, China
| | - Yingyu Luo
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jideng Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Penghao Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu Xi Nan Gynecological Hospital Co., Ltd., 66 Bisheng Road, Chengdu, 610000, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Diyan Li
- School of Pharmacy, Chengdu University, Chengdu, 610106, China
| |
Collapse
|
12
|
Su W, Guo Y, Wang Q, Ma L, Zhang Q, Zhang Y, Geng Y, Jin T, Guo J, Yang R, Niu Z, Ren L, Wang Y, Ning Z, Li W, He W, Sun J, Li T, Li Z, Shan H, Liang H. YAP1 inhibits the senescence of alveolar epithelial cells by targeting Prdx3 to alleviate pulmonary fibrosis. Exp Mol Med 2024; 56:1643-1654. [PMID: 38945958 PMCID: PMC11297023 DOI: 10.1038/s12276-024-01277-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 07/02/2024] Open
Abstract
The senescence of alveolar type II (AT2) cells impedes self-repair of the lung epithelium and contributes to lung injury in the setting of idiopathic pulmonary fibrosis (IPF). Yes-associated protein 1 (YAP1) is essential for cell growth and organ development; however, the role of YAP1 in AT2 cells during pulmonary fibrosis is still unclear. YAP1 expression was found to be downregulated in the AT2 cells of PF patients. Deletion of YAP1 in AT2 cells resulted in lung injury, exacerbated extracellular matrix (ECM) deposition, and worsened lung function. In contrast, overexpression of YAP1 in AT2 cells promoted alveolar regeneration, mitigated pulmonary fibrosis, and improved lung function. In addition, overexpression of YAP1 alleviated bleomycin (BLM) -induced senescence of alveolar epithelial cells both in vivo and in vitro. Moreover, YAP1 promoted the expression of peroxiredoxin 3 (Prdx3) by directly interacting with TEAD1. Forced expression of Prdx3 inhibited senescence and improved mitochondrial dysfunction in BLM-treated MLE-12 cells, whereas depletion of Prdx3 partially abrogated the protective effect of YAP1. Furthermore, overexpression of Prdx3 facilitated self-repair of the injured lung and reduced ECM deposition, while silencing Prdx3 attenuated the antifibrotic effect of YAP1. In conclusion, this study demonstrated that YAP1 alleviates lung injury and pulmonary fibrosis by regulating Prdx3 expression to improve mitochondrial dysfunction and block senescence in AT2 cells, revealing a potential novel therapeutic strategy for pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Su
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yingying Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Qianqian Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Lu Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Qing Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yuhan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yiding Geng
- Department of Systems Biology, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Tongzhu Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Jiayu Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Ruoxuan Yang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhihui Niu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Lingxue Ren
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Yan Wang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhiwei Ning
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Wenyue Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Wenxin He
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China
| | - Jian Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, 519000, Guangdong, China
| | - Tianyu Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China
| | - Zhixin Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai, 200433, China.
| | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China.
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
- Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, 150081, China.
- Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, 150081, China.
| |
Collapse
|
13
|
Lv L, Li S, Kang J, Li Y, Zhao N, Ye D, Qin F, Sun J, Yu T, Wu H. Inhibition of ABI2 ubiquitination-dependent degradation suppresses TNBC cell growth via down-regulating PI3K/Akt signaling pathway. Cancer Cell Int 2024; 24:222. [PMID: 38937761 PMCID: PMC11212232 DOI: 10.1186/s12935-024-03407-0] [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: 02/16/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Triple negative breast cancer (TNBC) is a type of cancer that lacks receptor expression and has complex molecular mechanisms. Recent evidence shows that the ubiquitin-protease system is closely related to TNBC. In this study, we obtain a key ubiquitination regulatory substrate-ABI2 protein by bioinformatics methods, which is also closely related to the survival and prognosis of TNBC. Further, through a series of experiments, we demonstrated that ABI2 expressed at a low level in TNBC tumors, and it has the ability to control cell cycle and inhibit TNBC cell migration, invasion and proliferation. Molecular mechanism studies proved E3 ligase CBLC could increase the ubiquitination degradation of ABI2 protein. Meanwhile, RNA-seq and IP experiments indicated that ABI2, acting as a crucial factor of tumor suppression, can significantly inhibit PI3K/Akt signaling pathway via the interaction with Rho GTPase RAC1. Finally, based on TNBC drug target ABI2, we screened and found that FDA-approved drug Colistimethate sodium(CS) has significant potential in suppressing the proliferation of TNBC cells and inducing cell apoptosis, making it a promising candidate for impeding the progression of TNBC.
Collapse
Affiliation(s)
- Linlin Lv
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, 116024, China
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shujing Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, 116024, China
| | - Jie Kang
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, 116024, China
| | - Yulin Li
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, 116024, China
| | - Nannan Zhao
- Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China
| | - Dongman Ye
- Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China
| | - Fengying Qin
- Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China
| | - Jing Sun
- Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Tao Yu
- Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Huijian Wu
- School of Bioengineering & Key Laboratory of Protein Modification and Disease, Dalian University of Technology, Dalian, Liaoning Province, 116024, China.
| |
Collapse
|
14
|
Lin M, Zheng X, Yan J, Huang F, Chen Y, Ding R, Wan J, Zhang L, Wang C, Pan J, Cao X, Fu K, Lou Y, Feng XH, Ji J, Zhao B, Lan F, Shen L, He X, Qiu Y, Jin J. The RNF214-TEAD-YAP signaling axis promotes hepatocellular carcinoma progression via TEAD ubiquitylation. Nat Commun 2024; 15:4995. [PMID: 38862474 PMCID: PMC11167002 DOI: 10.1038/s41467-024-49045-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: 04/22/2023] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
RNF214 is an understudied ubiquitin ligase with little knowledge of its biological functions or protein substrates. Here we show that the TEAD transcription factors in the Hippo pathway are substrates of RNF214. RNF214 induces non-proteolytic ubiquitylation at a conserved lysine residue of TEADs, enhances interactions between TEADs and YAP, and promotes transactivation of the downstream genes of the Hippo signaling. Moreover, YAP and TAZ could bind polyubiquitin chains, implying the underlying mechanisms by which RNF214 regulates the Hippo pathway. Furthermore, RNF214 is overexpressed in hepatocellular carcinoma (HCC) and inversely correlates with differentiation status and patient survival. Consistently, RNF214 promotes tumor cell proliferation, migration, and invasion, and HCC tumorigenesis in mice. Collectively, our data reveal RNF214 as a critical component in the Hippo pathway by forming a signaling axis of RNF214-TEAD-YAP and suggest that RNF214 is an oncogene of HCC and could be a potential drug target of HCC therapy.
Collapse
Affiliation(s)
- Mengjia Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, and National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaoyun Zheng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
| | - Fei Huang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yilin Chen
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Ran Ding
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jinkai Wan
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenliang Wang
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Jinchang Pan
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Xiaolei Cao
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Kaiyi Fu
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China
| | - Xin-Hua Feng
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Junfang Ji
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Bin Zhao
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China
| | - Fei Lan
- International Co-laboratory of Medical Epigenetics and Metabolism of Ministry of Science and Technology, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, and Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Li Shen
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China
- Department of Orthopedics Surgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Xianglei He
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, 3100014, Zhejiang, China
| | - Yunqing Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Disease, and National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
| | - Jianping Jin
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, Zhejiang, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, 321000, China.
| |
Collapse
|
15
|
Jiang H, Zhang RB, Peng J, Ren L, Wang HD. Disruption of the Hippo pathway promotes the proliferation of childhood acute lymphoblastic leukemia cells, inhibits apoptosis and chemosensitivity. Expert Rev Hematol 2024; 17:269-274. [PMID: 38753450 DOI: 10.1080/17474086.2024.2356255] [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: 01/05/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Despite advancements in chemotherapy and stem cell transplantation, the recurrence and chemoresistance of childhood acute lymphoblastic leukemia (cALL) remain a significant challenge, thus indicating the need for novel therapeutic targets. RESEARCH DESIGN AND METHODS The protein levels of YAP1, p-YAP1, TAZ, and Cyr61 of cALL patients and healthy volunteers were measured by western blot analysis. Then the leukemic cell line SUP-B15 was transfected with sh-YAP1 and pcDNA3.1-YAP1 to knockdown or overexpress YAP1. The viability, chemosensitivity, apoptosis, migration, and invasion of SUP-B15 cells were determined by MTT, flow cytometry, and Transwell assay. RESULTS The cALL patients had higher YAP1, TAZ, and Cyr61 protein expression and lower p-YAP1 protein expression in bone marrow tissues compared with healthy volunteers (p < 0.01). In SUP-B15 cells, YAP1 knockdown upregulated p-YAP1 protein expression (p < 0.01) and downregulated TAZ and Cyr61 protein expression (p < 0.01). In addition, knocking down YAP1 significantly inhibited cell viability, migration, and invasion, and induced apoptosis (p < 0.01). YAP1 knockdown also reduced the IC50 value following treatment with vincristine, daunorubicin, cyclophosphamide, and dexamethasone (p < 0.05). CONCLUSIONS Disruption of the Hippo pathway attenuates the development of cALL by promoting cell proliferation while suppressing apoptosis and drug sensitivity.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Rui-Bo Zhang
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Juan Peng
- Department of Blood Transfusion, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Lan Ren
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| | - Heng-Dong Wang
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, People's Republic of China
| |
Collapse
|
16
|
Balakrishnan K, Chen Y, Dong J. Amplification of Hippo Signaling Pathway Genes Is Governed and Implicated in the Serous Subtype-Specific Ovarian Carcino-Genesis. Cancers (Basel) 2024; 16:1781. [PMID: 38730733 PMCID: PMC11082992 DOI: 10.3390/cancers16091781] [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: 04/13/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Among women, ovarian cancer ranks as the fifth most common cause of cancer-related deaths. This study examined the impact of Hippo signaling pathway on ovarian carcinogenesis. Therefore, the signatures related to Hippo signaling pathway were derived from the molecular signatures database (MSigDB) and were used for further analysis. The Z score-based pathway activation scoring method was employed to investigate the expression patterns of these signatures in the mRNA expression profiles of ovarian cancer cohorts. Compared to other subtype tumors, the results of this study show that the Hippo signaling pathway signatures are dysregulated prominently in serous subtype-specific ovarian carcinogenesis. A receiver operating characteristic (ROC) curve-based results of the Hippo gene set, yes-associated protein 1 (YAP1), and mammalian sterile 20-like kinases 1 (MST1) genes can predict the serous subtype tumors by higher specificity and sensitivity with significant areas under the curve values also further reconfirmed these signaling dysregulations. Moreover, these gene sets were studied further for mutation analysis in the profile of high-grade serous ovarian adenocarcinoma in the cBioPortal database. The OncoPrint results reveal that these Hippo signaling pathway genes are amplified highly during the grade three and stage third or fourth of serous type ovarian tumors. In addition, the results of the Dependency Map (DepMap) plot also clearly show that these genes are amplified significantly across the ovarian cancer cell lines. Finally, overall survival (OS) curve plot investigations also revealed that these gene expressions show poor survival patterns linked to highly expressed conditions in serous subtypes of ovarian cancer patients with significant p-values (p < 0.05). Thus, the current finding would help to develop the targeted therapies treatment for serous subtype ovarian carcinogenesis.
Collapse
Affiliation(s)
| | | | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.B.); (Y.C.)
| |
Collapse
|
17
|
Guo J, Zhao L, Duan M, Yang Z, Zhao H, Liu B, Wang Y, Deng L, Wang C, Jiang X, Jiang X. Demethylases in tumors and the tumor microenvironment: Key modifiers of N 6-methyladenosine methylation. Biomed Pharmacother 2024; 174:116479. [PMID: 38537580 DOI: 10.1016/j.biopha.2024.116479] [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: 12/21/2023] [Revised: 03/09/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
RNA methylation modifications are widespread in eukaryotes and prokaryotes, with N6-methyladenosine (m6A) the most common among them. Demethylases, including Fat mass and obesity associated gene (FTO) and AlkB homolog 5 (ALKBH5), are important in maintaining the balance between RNA methylation and demethylation. Recent studies have clearly shown that demethylases affect the biological functions of tumors by regulating their m6A levels. However, their effects are complicated, and even opposite results have appeared in different articles. Here, we summarize the complex regulatory networks of demethylases, including the most important and common pathways, to clarify the role of demethylases in tumors. In addition, we describe the relationships between demethylases and the tumor microenvironment, and introduce their regulatory mechanisms. Finally, we discuss evaluation of demethylases for tumor diagnosis and prognosis, as well as the clinical application of demethylase inhibitors, providing a strong basis for their large-scale clinical application in the future.
Collapse
Affiliation(s)
- Junchen Guo
- Departmentof Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Liang Zhao
- Department of Anorectal Surgery, Shenyang Anorectal Hospital, Shenyang, Liaoning 110002, China
| | - Meiqi Duan
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Zhi Yang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - He Zhao
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Baiming Liu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Yihan Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Liping Deng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Chen Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Xiaodi Jiang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110002, China.
| | - Xiaofeng Jiang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China.
| |
Collapse
|
18
|
Kunachowicz D, Król-Kulikowska M, Raczycka W, Sleziak J, Błażejewska M, Kulbacka J. Heat Shock Proteins, a Double-Edged Sword: Significance in Cancer Progression, Chemotherapy Resistance and Novel Therapeutic Perspectives. Cancers (Basel) 2024; 16:1500. [PMID: 38672583 PMCID: PMC11048091 DOI: 10.3390/cancers16081500] [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: 03/19/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Heat shock proteins (Hsps) are involved in one of the adaptive mechanisms protecting cells against environmental and metabolic stress. Moreover, the large role of these proteins in the carcinogenesis process, as well as in chemoresistance, was noticed. This review aims to draw attention to the possibilities of using Hsps in developing new cancer therapy methods, as well as to indicate directions for future research on this topic. In order to discuss this matter, a thorough review of the latest scientific literature was carried out, taking into account the importance of selected proteins from the Hsp family, including Hsp27, Hsp40, Hsp60, Hsp70, Hsp90 and Hsp110. One of the more characteristic features of all Hsps is that they play a multifaceted role in cancer progression, which makes them an obvious target for modern anticancer therapy. Some researchers emphasize the importance of directly inhibiting the action of these proteins. In turn, others point to their possible use in the design of cancer vaccines, which would work by inducing an immune response in various types of cancer. Due to these possibilities, it is believed that the use of Hsps may contribute to the progress of oncoimmunology, and thus help in the development of modern anticancer therapies, which would be characterized by higher effectiveness and lower toxicity to the patients.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (D.K.); (M.K.-K.)
| | - Magdalena Król-Kulikowska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (D.K.); (M.K.-K.)
| | - Wiktoria Raczycka
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Jakub Sleziak
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Marta Błażejewska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (W.R.); (J.S.); (M.B.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine Santariškių g. 5, LT-08406 Vilnius, Lithuania
- DIVE IN AI, 53-307 Wroclaw, Poland
| |
Collapse
|
19
|
Radu P, Zurzu M, Tigora A, Paic V, Bratucu M, Garofil D, Surlin V, Munteanu AC, Coman IS, Popa F, Strambu V, Ramboiu S. The Impact of Cancer Stem Cells in Colorectal Cancer. Int J Mol Sci 2024; 25:4140. [PMID: 38673727 PMCID: PMC11050141 DOI: 10.3390/ijms25084140] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Despite incessant research, colorectal cancer (CRC) is still one of the most common causes of fatality in both men and women worldwide. Over time, advancements in medical treatments have notably enhanced the survival rates of patients with colorectal cancer. Managing metastatic CRC involves a complex tradeoff between the potential benefits and adverse effects of treatment, considering factors like disease progression, treatment toxicity, drug resistance, and the overall impact on the patient's quality of life. An increasing body of evidence highlights the significance of the cancer stem cell (CSC) concept, proposing that CSCs occupy a central role in triggering cancer. CSCs have been a focal point of extensive research in a variety of cancer types, including CRC. Colorectal cancer stem cells (CCSCs) play a crucial role in tumor initiation, metastasis, and therapy resistance, making them potential treatment targets. Various methods exist for isolating CCSCs, and understanding the mechanisms of drug resistance associated with them is crucial. This paper offers an overview of the current body of research pertaining to the comprehension of CSCs in colorectal cancer.
Collapse
Affiliation(s)
- Petru Radu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mihai Zurzu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Anca Tigora
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Vlad Paic
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Mircea Bratucu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Dragos Garofil
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Valeriu Surlin
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Alexandru Claudiu Munteanu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| | - Ionut Simion Coman
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
- General Surgery Department, “Bagdasar-Arseni” Clinical Emergency Hospital, 12 Berceni Road, 041915 Bucharest, Romania
| | - Florian Popa
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Victor Strambu
- Tenth Department of Surgery, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania; (P.R.); (A.T.); (V.P.); (M.B.); (D.G.); (I.S.C.); (F.P.); (V.S.)
| | - Sandu Ramboiu
- Sixth Department of Surgery, University of Medicine and Pharmacy of Craiova, Craiova Emergency Clinical 7 Hospital, 200642 Craiova, Romania; (V.S.); (A.C.M.); (S.R.)
| |
Collapse
|
20
|
Schiavoni G, Messina B, Scalera S, Memeo L, Colarossi C, Mare M, Blandino G, Ciliberto G, Bon G, Maugeri-Saccà M. Role of Hippo pathway dysregulation from gastrointestinal premalignant lesions to cancer. J Transl Med 2024; 22:213. [PMID: 38424512 PMCID: PMC10903154 DOI: 10.1186/s12967-024-05027-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/25/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND First identified in Drosophila melanogaster, the Hippo pathway is considered a major regulatory cascade controlling tissue homeostasis and organ development. Hippo signaling components include kinases whose activity regulates YAP and TAZ final effectors. In response to upstream stimuli, YAP and TAZ control transcriptional programs involved in cell proliferation, cytoskeletal reorganization and stemness. MAIN TEXT While fine tuning of Hippo cascade components is essential for maintaining the balance between proliferative and non-proliferative signals, pathway signaling is frequently dysregulated in gastrointestinal cancers. Also, YAP/TAZ aberrant activation has been described in conditions characterized by chronic inflammation that precede cancer development, suggesting a role of Hippo effectors in triggering carcinogenesis. In this review, we summarize the architecture of the Hippo pathway and discuss the involvement of signaling cascade unbalances in premalignant lesions of the gastrointestinal tract, providing a focus on the underlying molecular mechanisms. CONCLUSIONS The biology of premalignant Hippo signaling dysregulation needs further investigation in order to elucidate the evolutionary trajectories triggering cancer inititation and develop effective early therapeutic strategies targeting the Hippo/YAP pathway.
Collapse
Affiliation(s)
- Giulia Schiavoni
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Beatrice Messina
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Scalera
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
| | | | - Marzia Mare
- Medical Oncology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Bon
- Cellular Network and Molecular Therapeutic Target Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144, Rome, Italy.
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
21
|
Payne S, Neal A, De Val S. Transcription factors regulating vasculogenesis and angiogenesis. Dev Dyn 2024; 253:28-58. [PMID: 36795082 PMCID: PMC10952167 DOI: 10.1002/dvdy.575] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Transcription factors (TFs) play a crucial role in regulating the dynamic and precise patterns of gene expression required for the initial specification of endothelial cells (ECs), and during endothelial growth and differentiation. While sharing many core features, ECs can be highly heterogeneous. Differential gene expression between ECs is essential to pattern the hierarchical vascular network into arteries, veins and capillaries, to drive angiogenic growth of new vessels, and to direct specialization in response to local signals. Unlike many other cell types, ECs have no single master regulator, instead relying on differing combinations of a necessarily limited repertoire of TFs to achieve tight spatial and temporal activation and repression of gene expression. Here, we will discuss the cohort of TFs known to be involved in directing gene expression during different stages of mammalian vasculogenesis and angiogenesis, with a primary focus on development.
Collapse
Affiliation(s)
- Sophie Payne
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Alice Neal
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| | - Sarah De Val
- Department of Physiology, Anatomy and GeneticsInstitute of Developmental and Regenerative Medicine, University of OxfordOxfordUK
| |
Collapse
|
22
|
Huang X, Li S, Ding R, Li Y, Li C, Gu R. Antitumor effects of polysaccharides from medicinal lower plants: A review. Int J Biol Macromol 2023; 252:126313. [PMID: 37579902 DOI: 10.1016/j.ijbiomac.2023.126313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 08/16/2023]
Abstract
Cancer is one of the leading causes of death worldwide, yet the drugs currently approved for cancer treatment are associated with significant side effects, making it urgent to develop alternative drugs with low side effects. Polysaccharides are natural polymers with ketone or aldehyde groups, which are widely found in plants and have various biological activities such as immunomodulation, antitumor and hypolipidemic. The lower plants have attracted much attention for their outstanding anticancer effects, and many studies have shown that medicinal lower plant polysaccharides (MLPPs) have antitumor activity against various cancers and are promising alternatives with potential development in the food and pharmaceutical fields. Therefore, this review describes the structure and mechanism of action of MLPPs with antitumor activity. In addition, the application of MLPPs in cancer treatment is discussed, and the future development of MLPPs is explored.
Collapse
Affiliation(s)
- Xi Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ding
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Canlin Li
- School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Gu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; School of Ethnic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
23
|
Fnaiche A, Mélin L, Suárez NG, Paquin A, Vu V, Li F, Allali-Hassani A, Bolotokova A, Allemand F, Gelin M, Cotelle P, Woo S, LaPlante SR, Barsyte-Lovejoy D, Santhakumar V, Vedadi M, Guichou JF, Annabi B, Gagnon A. Development of LM-41 and AF-2112, two flufenamic acid-derived TEAD inhibitors obtained through the replacement of the trifluoromethyl group by aryl rings. Bioorg Med Chem Lett 2023; 95:129488. [PMID: 37770003 DOI: 10.1016/j.bmcl.2023.129488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
The Hippo pathway regulates organ size and tissue homeostasis by controlling cell proliferation and apoptosis. The YAP-TEAD transcription factor, the downstream effector of the Hippo pathway, regulates the expression of genes such as CTGF, Cyr61, Axl and NF2. Aberrant Hippo activity has been identified in multiple types of cancers. Flufenamic acid (FA) was reported to bind in a liphophilic TEAD palmitic acid (PA) pocket, leading to reduction of the expression of Axl and NF2. Here, we show that the replacement of the trifluoromethyl moiety in FA by aromatic groups, directly connected to the scaffold or separated by a linker, leads to compounds with better affinity to TEAD. Co-crystallization studies show that these compounds bind similarly to FA, but deeper within the PA pocket. Our studies identified LM-41 and AF-2112 as two TEAD binders that strongly reduce the expression of CTGF, Cyr61, Axl and NF2. LM-41 gave the strongest reduction of migration of human MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Ahmed Fnaiche
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Léa Mélin
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Narjara González Suárez
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Alexis Paquin
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada
| | - Victoria Vu
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Fengling Li
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Albina Bolotokova
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | - Frédéric Allemand
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Muriel Gelin
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Philippe Cotelle
- Université de Lille, CHU Lille, INSERM-UMR-S-1172-JPArc-Centre de Recherche Jean-Pierre Aubert, Neurosciences et Cancer, F-59000 Lille, France
| | - Simon Woo
- INRS-Centre Armand Frappier Santé Biotechnologie, Université du Québec, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | - Steven R LaPlante
- INRS-Centre Armand Frappier Santé Biotechnologie, Université du Québec, 531 Boulevard des Prairies, Laval, Québec H7V 1B7, Canada
| | - Dalia Barsyte-Lovejoy
- Structural Genomics Consortium, 101 College Street, Toronto, Ontario M5G 1L7, Canada
| | | | - Masoud Vedadi
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France.
| | - Borhane Annabi
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada.
| | - Alexandre Gagnon
- Département de chimie, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montréal, Québec H3C 3P8, Canada.
| |
Collapse
|
24
|
Zhou L, Guo H, Liao Q, Zou J, Le Y, Fang Z, Xiong J, Huang S, Deng J, Xiang X. miR-3133 inhibits gastrointestinal cancer progression through activation of Hippo and p53 signalling pathways via multi-targets. J Cell Mol Med 2023; 27:3090-3106. [PMID: 37555915 PMCID: PMC10568676 DOI: 10.1111/jcmm.17880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 07/09/2023] [Accepted: 07/19/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Malignant cell growth and chemoresistance, the main obstacles in treating gastrointestinal cancer (GIC), rely on the Hippo and p53 signalling pathways. However, the upstream regulatory mechanisms of these pathways remain complex and poorly understood. METHODS Immunohistochemistry (IHC), western blot and RT-qPCR were used to analyse the expression of RNF146, miR-3133 and key components of Hippo and p53 pathway. CCK-8, colony formation, drug sensitivity assays and murine xenograft models were used to investigate the effect of RNF146 and miR-3133 in GIC. Further exploration of the upstream regulatory mechanism was performed using bioinformatics analysis, dual-luciferase reporter gene, immunoprecipitation assays and bisulfite sequencing PCR (BSP). RESULTS Clinical samples, in vitro and in vivo experiments demonstrated that RNF146 exerts oncogenic effects in GIC by regulating the Hippo pathway. Bioinformatics analysis identified a novel miRNA, miR-3133, as an upstream regulatory factor of RNF146. fluorescence in situ hybridization and RT-qPCR assays revealed that miR-3133 was less expressed in gastrointestinal tumour tissues and was associated with adverse pathological features. Functional assays and animal models showed that miR-3133 promoted the proliferation and chemotherapy sensitivity of GIC cells. miR-3133 affected YAP1 protein expression by targeting RNF146, AGK and CUL4A, thus activating the Hippo pathway. miR-3133 inhibited p53 protein degradation and extended p53's half-life by targeting USP15, SPIN1. BSP experiments confirmed that miR-3133 promoter methylation is an important reason for its low expression. CONCLUSION miR-3133 inhibits GIC progression by activating the Hippo and p53 signalling pathways via multi-targets, including RNF146, thereby providing prognostic factors and valuable potential therapeutic targets for GIC.
Collapse
Affiliation(s)
- Ling Zhou
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Hui Guo
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Quan Liao
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jianping Zou
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Yi Le
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Ziling Fang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jianping Xiong
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Shanshan Huang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Jun Deng
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| | - Xiaojun Xiang
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangChina
- Jiangxi Key Laboratory for Individualized Cancer TherapyNanchangChina
| |
Collapse
|
25
|
Yang S, Guo LJ, Liang Y, He ZM, Luo J, Mu YD. ADCY6 is a potential prognostic biomarker and suppresses OTSCC progression via Hippo signaling pathway. Kaohsiung J Med Sci 2023; 39:978-988. [PMID: 37574908 DOI: 10.1002/kjm2.12725] [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: 04/08/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 08/15/2023] Open
Abstract
Oral tongue squamous cell carcinoma (OTSCC) is a malignant tumor. Recently, studies have found that adenylate cyclase 6 (ADCY6) plays a pivotal role in many lethal tumors formation processes. The role of ADCY6 in OTSCC remains unknown. The expression of ADCY6 in OTSCC tissue samples was detected. The clinical significance of ADCY6 in OTSCC was analyzed by statistical methods. OTSCC cell lines were selected to analyze the biological function of ADCY6. Meanwhile, the effect of ADCY6 on the growth of OTSCC in vivo was explored using subcutaneous tumorigenesis assay. WB assay was used to detect the underlying signaling pathway. Cell function recovery test used to investigate the mechanism of ADCY6-promoting OTSCC malignant biological behavior via Hippo signaling pathway. We report that ADCY6 was obviously downregulated in OTSCC tissue samples and cell lines. Importantly, lower expression of ADCY6 indicates a poorer prognosis in patients with OTSCC, and its expression is significantly correlated with TNM stage and tumor size. Functionally, forced expression of ADCY6 can significantly inhibit the proliferation, migration, invasion, and promote apoptosis of OTSCC cells. Mechanistically, we demonstrated that ADCY6 upregulation impaired Hippo signaling pathway to reduce the malignant biological behavior of OTSCC. Generally, our findings suggest that ADCY6 suppressed Hippo signaling pathway to regulate malignant biological behavior in OTSCC, which provide new cues for further exploring the mechanism of occurrence and development of OTSCC.
Collapse
Affiliation(s)
- Sen Yang
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Oral and Maxillofacial Surgery, Suining Central Hospital, Sichuan, China
| | - Li-Juan Guo
- Department of Medical Cosmetology, Suining Central Hospital, Sichuan, China
| | - Yong Liang
- Institute of Electronic and Information Engineering of UESTC in Guangdong, University of Electronic Science and Technology of China, Dongguan, China
| | - Zhi-Ming He
- Institute of Electronic and Information Engineering of UESTC in Guangdong, University of Electronic Science and Technology of China, Dongguan, China
| | - Jia Luo
- Department of Stomatology Center, Suining Central Hospital, Sichuan, China
| | - Yan-Dong Mu
- Department of Stomatology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
26
|
Devos M, Dias Nunes J, Donfack Jiatsa N, Demeestere I. Regulation of follicular activation signaling pathways by in vitro inhibition of YAP/TAZ activity in mouse ovaries. Sci Rep 2023; 13:15346. [PMID: 37714905 PMCID: PMC10504383 DOI: 10.1038/s41598-023-41954-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/04/2023] [Indexed: 09/17/2023] Open
Abstract
The Hippo pathway plays a crucial role in the regulation of follicular activation, which constitutes the first step of the folliculogenesis process. Disruption of this pathway occurs in several non-physiological contexts, after fragmentation for ovarian tissue cryopreservation procedures or chemotherapy exposure, leading to massive follicular growth and depletion. This study aimed to investigate the effect of controlling the Hippo pathway using verteporfin (VERT) during in vitro ovarian culture and to evaluate its potential preventive effects on chemotherapy-induced follicle activation using a mouse model. After exposure of cut ovaries to different concentrations of VERT for 3 h, a dose-dependent effect of VERT was observed that reached significant inhibition of YAP activity at 3 µmol/L. To assess the potential effect of controlling chemotherapy-induced Hippo pathway disruption, whole mouse ovaries were exposed to VERT alone or as a co-treatment with 4-hydroperoxycylophosphamide (4HC). VERT co-treatment prevented chemotherapy-induced YAP activation but had a limited impact on downstream effector gene, Ccn2. Surprisingly, VERT co-treatment also prevented mTOR and survival signaling pathway alterations following chemotherapy exposure. These results suggest an interaction between the two main signaling pathways regulating follicle activation and a protective effect of VERT on 4HC-induced DNA damage.
Collapse
Affiliation(s)
- Melody Devos
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Campus Erasme CP636, Route de Lennik 808, 1070, Brussels, Belgium
| | - Joana Dias Nunes
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Campus Erasme CP636, Route de Lennik 808, 1070, Brussels, Belgium
| | - Nathalie Donfack Jiatsa
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Campus Erasme CP636, Route de Lennik 808, 1070, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Campus Erasme CP636, Route de Lennik 808, 1070, Brussels, Belgium.
- Fertility Clinic, HUB-Erasme Hospital, Université Libre de Bruxelles (ULB), Route de Lennik 808, 1070, Brussels, Belgium.
| |
Collapse
|
27
|
Xie H, Guo L, Ma Q, Zhang W, Yang Z, Wang Z, Peng S, Wang K, Wen S, Shang Z, Niu Y. YAP is required for prostate development, regeneration, and prostate stem cell function. Cell Death Discov 2023; 9:339. [PMID: 37689711 PMCID: PMC10492789 DOI: 10.1038/s41420-023-01637-1] [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: 06/06/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/11/2023] Open
Abstract
Prostate development and regeneration depend on prostate stem cell function, the delicate balance of stem cell self-renewal and differentiation. However, mechanisms modulating prostate stem cell function remain poorly identified. Here, we explored the roles of Yes-associated protein 1 (YAP) in prostate stem cells, prostate development and regeneration. Using YAPfl/fl, CD133-CreER mice, we found that stem cell-specific YAP-deficient mice had compromised branching morphogenesis and epithelial differentiation, resulting in damaged prostate development. YAP inhibition also significantly affected the regeneration process of mice prostate, leading to impaired regenerated prostate. Furthermore, YAP ablation in prostate stem cells significantly reduced its self-renewal activity in vitro, and attenuated prostate regeneration of prostate grafts in vivo. Further analysis revealed a decrease in Notch and Hedgehog pathways expression in YAP inhibition cells, and treatment with exogenous Shh partially restored the self-renewal ability of prostate sphere cells. Taken together, our results revealed the roles of YAP in prostate stem cell function and prostate development and regeneration through regulation of the Notch and Hedgehog signaling pathways.
Collapse
Affiliation(s)
- Hui Xie
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Linpei Guo
- Gene and Immunotherapy Center, The Second Hospital of Shandong University, 250033, Jinan, Shandong, China
| | - Qianwang Ma
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Wenyi Zhang
- Department of Radiology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhao Yang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhun Wang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Shuanghe Peng
- Department of Pathology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Keruo Wang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Simeng Wen
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China
| | - Zhiqun Shang
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China.
| | - Yuanjie Niu
- Department of Urology, Tianjin Institute of Urology, The second hospital of Tianjin Medical University, 300211, Tianjin, China.
| |
Collapse
|
28
|
Guo H, Zeng H, Hu Y, Jiang L, Lei L, Hung J, Fu C, Li H, Long Y, Chen J, Zeng Q. UVB promotes melanogenesis by regulating METTL3. J Cell Physiol 2023; 238:2161-2171. [PMID: 37417881 DOI: 10.1002/jcp.31077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/08/2023]
Abstract
Ultraviolet (UV) radiation is the primary exogenous inducer of skin pigmentation, although the mechanism has not been fully elucidated. N6-methyladenosine (m6 A) modification is one of the key epigenetic form of gene regulation that affects multiple biological processes. The aim of this study was to explore the role and underlying mechanisms of m6 A modification in UVB-induced melanogenesis. Low-dose UVB increased global m6 A modification in melanocytes (MCs) and MNT1 melanoma cell line. The GEPIA database predicted that methyltransferase METTL3 is positively correlated with the melanogenic transcription factor MITF in the sun-exposed skin tissues. After METTL3 respectively overexpressed and knocked down in the MNT1, the melanin content and melanogenesis-related genes were significantly upregulated after overexpression of METTL3, especially with UVB irradiation, and downregulated after METTL3 knockdown. METTL3 levels were also higher in melanocytic nevi with high melanin content. METTL3 overexpression and knockdown also altered the protein level of YAP1. SRAMP analysis predicted four high-potential m6 A modification sites on YAP1 mRNA, of which three were confirmed by methylated RNA immunoprecipitation. Inhibition of YAP1 expression can partially reverse melanogenesis induced by overexpression of METTL3. In conclusion, UVB irradiation promotes global m6 A modification in MCs and upregulates METTL3, which increases the expression level of YAP1 through m6 A modification, thereby activating the co-transcription factor TEAD1 and promoting melanogenesis.
Collapse
Affiliation(s)
- Haoran Guo
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hongliang Zeng
- Center of Medical Laboratory Animal, Hunan Academy of Chinese Medicine, Changsha, China
| | - Yibo Hu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Li Lei
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jinhua Hung
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Chuhan Fu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Long
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Qinghai Zeng
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Julius P, Siyumbwa SN, Maate F, Moonga P, Kang G, Kaile T, West JT, Wood C, Angeletti PC. Yes-associated protein-1 overexpression in ocular surface squamous neoplasia; a potential diagnostic marker and therapeutic target. Front Oncol 2023; 13:1213426. [PMID: 37476371 PMCID: PMC10354641 DOI: 10.3389/fonc.2023.1213426] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Yes-associated protein-1 (YAP-1) is a Hippo system transcription factor, which serves as an oncogene in squamous cell carcinoma, and several solid tumors when the Hippo pathway is dysregulated. Yet, the activity of YAP-1 in ocular surface squamous neoplasia (OSSN) has not been determined. Here, we investigate the relationship between YAP-1 overexpression and OSSN. Using a cross-sectional study design, we recruited 227 OSSN patients from the University Teaching Hospitals in Lusaka, Zambia. Immunohistochemistry was used to assess YAP-1 protein overexpression in tumor tissue relative to surrounding benign squamous epithelium. OSSN patient samples (preinvasive, n = 62, 27% and invasive, n = 165, 73%) were studied. One hundred forty-nine invasive tumors contained adjacent preinvasive tissue, bringing the total number of preinvasive lesions examined to 211 (62 + 149). There was adjacent benign squamous epithelium in 50.2% (114/227) of OSSN samples. Nuclear YAP- 1 was significantly overexpressed in preinvasive (Fisher's (F): p <.0001, Monte Carlo (MC): p <.0001) and invasive (F: p <.0001, MC: p <.0001) OSSN in comparison to adjacent benign squamous epithelium when analyzed for basal keratinocyte positive count, staining intensity, expression pattern, and Immunostaining intensity-distribution index. YAP-1 expression did not differ between preinvasive and invasive OSSN (p >.05), keratinizing and non- keratinizing cancer (p >.05), or between T1/T2 and T3/T4 stages in invasive tumors (p >.05). However, grade 2 and 3 tumors had significantly stronger nucleus YAP-1 overexpression intensity than grade 1 tumors (F: p = .0078, MC: p = .0489). By immunohistochemistry, we identified significant overexpression (upregulation of YAP-1 protein expression) in preinvasive and invasive OSSN lesions compared to neighboring benign squamous epithelium. YAP-1 expression was significantly higher in poorly and moderately differentiated invasive squamous cancer than in well-differentiated carcinomas. Overexpression of YAP-1 within the margin of preinvasive and invasive OSSN, but not in the neighboring normal epithelium, indicates that it plays a role in the development and progression of OSSN.
Collapse
Affiliation(s)
- Peter Julius
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Stepfanie N. Siyumbwa
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Fred Maate
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - Phyllis Moonga
- University Teaching Hospital, Eye Hospital, Lusaka, Zambia
| | - Guobin Kang
- Department of Interdisciplinary Oncology, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Trevor Kaile
- Department of Pathology and Microbiology, School of Medicine, University of Zambia, Lusaka, Zambia
| | - John T. West
- Department of Interdisciplinary Oncology, Louisiana State University Health Science Center, New Orleans, LA, United States
| | - Charles Wood
- Department of Interdisciplinary Oncology, Louisiana State University Health Science Center, New Orleans, LA, United States
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, NE, United States
| | - Peter C. Angeletti
- Nebraska Center for Virology and the School of Biological Sciences, University of Nebraska-Lincoln, NE, United States
| |
Collapse
|
30
|
Rai U, Senapati D, Arora MK. Insights on the role of anti-inflammatory and immunosuppressive agents in the amelioration of diabetes. Diabetol Int 2023; 14:134-144. [PMID: 37090130 PMCID: PMC10113422 DOI: 10.1007/s13340-022-00607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/30/2022] [Indexed: 11/18/2022]
Abstract
Diabetes is a major health problem worldwide. It is a chronic metabolic disorder that produces overt hyperglycemic condition that occurs either when the pancreas does not produce enough insulin due to excessive destruction of pancreatic β-cells (type 1 diabetes) or due to development of insulin resistance (type 2 diabetes). An autoimmune condition known as type 1 diabetes (T1D) results in the targeted immune death of β-cells that produce insulin. The only available treatment for T1D at the moment is the lifelong use of insulin. Multiple islet autoantibody positivity is used to diagnose T1D. There are four standard autoantibodies observed whose presence shows the development of T1D: antibodies against insulin, glutamic acid decarboxylase (GAD65), zinc T8 transporter (ZnT8), and tyrosine phosphatase-like protein (ICA512). In type 2 diabetes (T2D), an inflammatory response precipitates as a consequence of the immune response to high blood glucose level along with the presence of inflammation mediators produced by macrophages and adipocytes in fat tissue. The slow and chronic inflammatory condition of adipose tissue produces insulin resistance leading to increased stress on pancreatic β-cells to produce more insulin to compensate for the insulin resistance. Thus, this stress condition exacerbates the apoptosis of β-cells leading to insufficient production of insulin, resulting in hyperglycemia which signifies late stage T2D. Therefore, the therapeutic utilization of immunosuppressive agents may be a better alternative over the use of insulin and oral hypoglycemic agents for the treatment of T1D and T2D, respectively. This review enlightens the immune intervention for the prevention and amelioration of T1D and T2D in humans with main focus on the antigen-specific immune suppressive therapy.
Collapse
Affiliation(s)
- Uddipak Rai
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| | - Dhirodatta Senapati
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| | - Mandeep Kumar Arora
- School of Pharmaceutical and Population Health Informatics, DIT University, 248009, Dehradun, Uttarakhand India
| |
Collapse
|
31
|
Li T, Li Y. Quercetin acts as a novel anti-cancer drug to suppress cancer aggressiveness and cisplatin-resistance in nasopharyngeal carcinoma (NPC) through regulating the yes-associated protein/Hippo signaling pathway. Immunobiology 2023; 228:152324. [PMID: 36608594 DOI: 10.1016/j.imbio.2022.152324] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 12/06/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Quercetin has been proven to be effective for cancer treatment, including nasopharyngeal carcinoma (NPC). Also, Quercetin sensitizes cancer cells to current chemical drugs to improve their therapeutic efficacy. However, up until now, the molecular mechanisms that quercetin exerted its therapeutic effects on NPC have not been fully delineated. METHODS Cell proliferation abilities were examined by CCK-8 assay and colony formation assay. Real-Time qPCR and Western Blot analysis were used to detect gene expressions at RNA and protein levels. Cell mobility was determined by wound scratch assay and transwell assay. Cell death was detected using flow cytometry (FCM). Tumorigenesis of the NPC cells was determined by in vivo tumor-bearing mice models. Hematoxylin and eosin (H&E) and TUNEL staining were used to detect the tumor metastasis to lung tissues and dead cells, respectively. RESULTS Here, we validated that quercetin exerted its anti-tumor effects and increased cisplatin-sensitivity in NPC in vitro and in vivo. Specifically, quercetin inhibited NPC cell proliferation, viability, mobility, epithelial-mesenchymal transition (EMT), and tumorigenesis, and induced cell death, resulting in the inhibition of NPC progression. In addition, the NPC cells were subjected to a continuously increasing doses of cisplatin to generate cisplatin-resistant NPC (NPC/CDDP) cells. Interestingly, quercetin significantly enhanced the cytotoxic effects of high-dose cisplatin on NPC/CDDP cells. Furthermore, the potential underlying mechanisms were uncovered, and the results evidenced that quercetin inhibited Yes-associated protein (YAP) expression and its translocation to the nucleus, leading to the recovery of the Hippo pathway, inhibition of cancer progression, and increase in cisplatin-resistance. Mechanistically, upregulation of YAP by its gene manipulating vectors abrogated the inhibiting effects of quercetin on NPC malignant phenotypes, which also made NCP/CDDP cells irresponsive to high-dose cisplatin-quercetin co-treatments. CONCLUSION Collectively, our data evidenced that quercetin inhibited YAP to recover the Hippo pathway, which further inhibited NPC pathogenesis and increased susceptibility of NCP/CDDP cells to cisplatin treatment.
Collapse
Affiliation(s)
- Tao Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou City, Henan Province 450052, China
| | - Yujie Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou City, Henan Province 450052, China.
| |
Collapse
|
32
|
Liu Z, Hayashi H, Matsumura K, Ogata Y, Sato H, Shiraishi Y, Uemura N, Miyata T, Higashi T, Nakagawa S, Mima K, Imai K, Baba H. Hyperglycaemia induces metabolic reprogramming into a glycolytic phenotype and promotes epithelial-mesenchymal transitions via YAP/TAZ-Hedgehog signalling axis in pancreatic cancer. Br J Cancer 2023; 128:844-856. [PMID: 36536047 PMCID: PMC9977781 DOI: 10.1038/s41416-022-02106-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Hyperglycaemia is a well-known initial symptom in patients with pancreatic ductal adenocarcinoma (PDAC). Metabolic reprogramming in cancer, described as the Warburg effect, can induce epithelial-mesenchymal transition (EMT). METHODS The biological impact of hyperglycaemia on malignant behaviour in PDAC was examined by in vitro and in vivo experiments. RESULTS Hyperglycaemia promoted EMT by inducing metabolic reprogramming into a glycolytic phenotype via yes-associated protein (YAP)/PDZ-binding motif (TAZ) overexpression, accompanied by GLUT1 overexpression and enhanced phosphorylation Akt in PDAC. In addition, hyperglycaemia enhanced chemoresistance by upregulating ABCB1 expression and triggered PDAC switch into pure basal-like subtype with activated Hedgehog pathway (GLI1 high, GATA6 low expression) through YAP/TAZ overexpression. PDAC is characterised by abundant stroma that harbours tumour-promoting properties and chemoresistance. Hyperglycaemia promotes the production of collagen fibre-related proteins (fibronectin, fibroblast activation protein, COL1A1 and COL11A1) by stimulating YAP/TAZ expression in cancer-associated fibroblasts (CAFs). Knockdown of YAP and/or TAZ or treatment with YAP/TAZ inhibitor (K975) abolished EMT, chemoresistance and a favourable tumour microenvironment even under hyperglycemic conditions in vitro and in vivo. CONCLUSION Hyperglycaemia induces metabolic reprogramming into glycolytic phenotype and promotes EMT via YAP/TAZ-Hedgehog signalling axis, and YAP/TAZ could be a novel therapeutic target in PDAC.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yoko Ogata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Tatsunori Miyata
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Takaaki Higashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Shigeki Nakagawa
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Kosuke Mima
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Katsunori Imai
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto, 860-8556, Japan.
| |
Collapse
|
33
|
Yang M, Zhang H, Gao S, Huang W. DEPDC1 and KIF4A synergistically inhibit the malignant biological behavior of osteosarcoma cells through Hippo signaling pathway. J Orthop Surg Res 2023; 18:145. [PMID: 36849972 PMCID: PMC9972622 DOI: 10.1186/s13018-023-03572-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 02/01/2023] [Indexed: 03/01/2023] Open
Abstract
The treatment of osteosarcoma (OS) is still mainly surgery combined with systematic chemotherapy, and gene therapy is expected to improve the survival rate of patients. This study aimed to explore the effect of DEP domain 1 protein (DEPDC1) and kinesin super-family protein 4A (KIF4A) in OS and understand its mechanism. Th expression of DEPDC1 and KIF4A in OS cells was detected by RT-PCR and western blot. The viability, proliferation, invasion and migration of OS cells and tube formation of human umbilical vein endothelial cells (HUVECs) after indicated treatment were in turn detected by CCK-8 assay, EdU staining, wound healing assay, transwell assay and tube formation assay. The interaction between DEPDC1 and KIF4A was predicted by STRING and confirmed by co-immunoprecipitation. The expression of epithelial-mesenchymal transition (EMT)-related proteins, tube formation-related proteins and Hippo signaling pathway proteins was detected by western blot. As a result, the expression of DEPDC1 and KIF4A was all increased in U2OS cells. Down-regulation of DEPDC1 suppressed the viability, proliferation, invasion and migration of U2OS cells and tube formation of HUVECs, accompanied by the increased expression of E-cadherin and decreased expression of N-cadherin, Vimentin and VEGF. DEPDC1 was confirmed to be interacted with KIF4A. Upregulation of KIF4A partially reversed the effect of DEPDC1 interference on the above biological behaviors of U2OS cells. Down-regulation of DEPDC1 promoted the expression of p-LATS1 and p-YAP in Hippo signaling pathway, which was reversed by upregulation of KIF4A. In conclusion, down-regulation of DEPDC1 inhibited the malignant biological behavior of OS cells through the activation of Hippo signaling pathway, which could be reversed by upregulation of KIF4A.
Collapse
Affiliation(s)
- Mingming Yang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016, People's Republic of China.
| | - Hang Zhang
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| | - Shichang Gao
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| | - Wei Huang
- grid.452206.70000 0004 1758 417XDepartment of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, Yuan Jia Gang, Yu Zhong District, Chongqing, 400016 People’s Republic of China
| |
Collapse
|
34
|
Messina B, Lo Sardo F, Scalera S, Memeo L, Colarossi C, Mare M, Blandino G, Ciliberto G, Maugeri-Saccà M, Bon G. Hippo pathway dysregulation in gastric cancer: from Helicobacter pylori infection to tumor promotion and progression. Cell Death Dis 2023; 14:21. [PMID: 36635265 PMCID: PMC9837097 DOI: 10.1038/s41419-023-05568-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/27/2022] [Accepted: 01/06/2023] [Indexed: 01/13/2023]
Abstract
The Hippo pathway plays a critical role for balancing proliferation and differentiation, thus regulating tissue homeostasis. The pathway acts through a kinase cascade whose final effectors are the Yes-associated protein (YAP) and its paralog transcriptional co‑activator with PDZ‑binding motif (TAZ). In response to a variety of upstream signals, YAP and TAZ activate a transcriptional program that modulates cellular proliferation, tissue repair after injury, stem cell fate decision, and cytoskeletal reorganization. Hippo pathway signaling is often dysregulated in gastric cancer and in Helicobacter pylori-induced infection, suggesting a putative role of its deregulation since the early stages of the disease. In this review, we summarize the architecture and regulation of the Hippo pathway and discuss how its dysregulation fuels the onset and progression of gastric cancer. In this setting, we also focus on the crosstalk between Hippo and other established oncogenic signaling pathways. Lastly, we provide insights into the therapeutic approaches targeting aberrant YAP/TAZ activation and discuss the related clinical perspectives and challenges.
Collapse
Affiliation(s)
- Beatrice Messina
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Federica Lo Sardo
- Oncogenomic and Epigenetic Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Stefano Scalera
- SAFU Laboratory, Department of Research, Advanced Diagnostic, and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo Memeo
- Pathology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
| | | | - Marzia Mare
- Medical Oncology Unit, Mediterranean Institute of Oncology, Viagrande, Italy
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marcello Maugeri-Saccà
- Clinical Trial Center, Biostatistics and Bioinformatics Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Division of Medical Oncology 2, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giulia Bon
- Cellular Network and Molecular Therapeutic Target Unit, Department of Research, Diagnosis and Innovative Technologies, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| |
Collapse
|
35
|
Ma FY, Zhou XH, Liang Q. Advances in understanding of role and mechanism of Hippo signaling pathway in colorectal cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:14-19. [DOI: 10.11569/wcjd.v31.i1.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors, and most patients have a poor prognosis. Many studies have shown that the Hippo signaling pathway plays a key role in the occurrence and development of CRC by regulating CRC cell proliferation and apoptosis, tumor invasion and metastasis, autophagy, metabolic reprogramming, drug resistance, and other processes. This article reviews the latest progress in research of the expression of key molecules of the Hippo signaling pathway in CRC as well as the understanding of the mechanism by which this pathway regulates the occurrence and development of CRC.
Collapse
Affiliation(s)
- Fu-Yan Ma
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Xi-Han Zhou
- Department of Gastroenterology, Affiliated Hospital of Youjiang Medical College Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| | - Qiao Liang
- Graduate School of Youjiang Medical College for Nationalities, Baise 533000, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
36
|
Wang T, Li K, Liu H, Luo E. Focusing on Hippo Pathway in Stem Cells of Oral Origin, Enamel Formation and Periodontium Regeneration. Organogenesis 2022; 18:2082236. [PMID: 35786361 PMCID: PMC9897286 DOI: 10.1080/15476278.2022.2082236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hippo pathway is a cellular regulatory pathway composed of core molecules such as MST1/2, LATS1/2, SAV1, MOB1A/B and downstream YAP/TAZ. Fully involved in regulating cell proliferation, differentiation, migration and apoptosis, the Hippo pathway is critical in regulating stem cells of oral origin, for instance, DPSCs and PDLSCs, enamel formation and periodontium regeneration. Here, we summarized the Hippo pathway involved in these progresses and concluded crosstalks of the Hippo pathway with BCL-2, ERK1/2, ROCK, TGF-β/BMP and Wnt/β-catenin pathways, hoping to provide foundation for further clinical therapy.
Collapse
Affiliation(s)
- Tianyi Wang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kehan Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - En Luo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China,CONTACT En Luo State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu, Sichuan610041, China
| |
Collapse
|
37
|
Wang L, Xu W, Mei Y, Wang X, Liu W, Zhu Z, Ni Z. CHRDL2 promotes cell proliferation by activating the YAP/TAZ signaling pathway in gastric cancer. Free Radic Biol Med 2022; 193:158-170. [PMID: 36206931 DOI: 10.1016/j.freeradbiomed.2022.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 12/24/2022]
Abstract
The encoding product of Chordin-like 2 (CHRDL2) is a member of the chordin family of proteins, which has been shown to be aberrantly expressed in several types of solid tumors. The regulatory underlying mechanisms of CHRDL2, however, remain poorly understood in gastric cancer (GC). In the present study, we determined that CHRDL2 was abnormally upregulated in human gastric cancer tissues compared with adjacent normal tissues. We also showed that CHRDL2 was positively associated with T stage, the pathological stage, distant metastasis, and poor patient prognosis. Furthermore, the serum level of CHRDL2 was obviously higher in GC patients than normal people, and is positively correlated with later TNM stage, deeper T stage, later N stage and poorer differentiation. Moreover, we verified that overexpressing CHRDL2 promoted the proliferation and cell cycle transition of GC cells both in vitro and in vivo, whereas the opposite results were observed in CHRDL2-depleted cells. In addition, the phosphorylation levels of Yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ) and the total levels MST2 were decreased in CHRDL2 overexpressing cells. Consistent with previous findings, we observed the converse results in CHRDL2-silenced GC cells. Additionally, knockdown of YAP and overexpression of STK3 (MST2) could reverse the effects of CHRDL2 overexpression-induced proliferation of GC cells in vitro. Taken together, CHRDL2 plays a key role by activating the YAP/TAZ pathway in gastric cancer. Therefore, CHRDL2 could serve as a potential therapeutic tool for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Lingquan Wang
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Xu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Mei
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xufeng Wang
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wentao Liu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhenggang Zhu
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Zhentian Ni
- Department of General Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China; Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
38
|
Targeted Disruption of Lats1 and Lats2 in Mice Impairs Testis Development and Alters Somatic Cell Fate. Int J Mol Sci 2022; 23:ijms232113585. [DOI: 10.3390/ijms232113585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Hippo signaling plays an essential role in the development of numerous tissues. Although it was previously shown that the transcriptional effectors of Hippo signaling Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) can fine-tune the regulation of sex differentiation genes in the testes, the role of Hippo signaling in testis development remains largely unknown. To further explore the role of Hippo signaling in the testes, we conditionally deleted the key Hippo kinases large tumor suppressor homolog kinases 1 and -2 (Lats1 and Lats2, two kinases that antagonize YAP and TAZ transcriptional co-regulatory activity) in the somatic cells of the testes using an Nr5a1-cre strain (Lats1flox/flox;Lats2flox/flox;Nr5a1-cre). We report here that early stages of testis somatic cell differentiation were not affected in this model but progressive testis cord dysgenesis was observed starting at gestational day e14.5. Testis cord dysgenesis was further associated with the loss of polarity of the Sertoli cells and the loss of SOX9 expression but not WT1. In parallel with testis cord dysgenesis, a loss of steroidogenic gene expression associated with the appearance of myofibroblast-like cells in the interstitial space was also observed in mutant animals. Furthermore, the loss of YAP phosphorylation, the accumulation of nuclear TAZ (and YAP) in both the Sertoli and interstitial cell populations, and an increase in their transcriptional co-regulatory activity in the testes suggest that the observed phenotype could be attributed at least in part to YAP and TAZ. Taken together, our results suggest that Hippo signaling is required to maintain proper differentiation of testis somatic cells.
Collapse
|
39
|
Research progress on the role of cholesterol in hepatocellular carcinoma. Eur J Pharmacol 2022; 938:175410. [DOI: 10.1016/j.ejphar.2022.175410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
|
40
|
Qin X, Luo H, Deng Y, Yao X, Zhang J, He B. Resveratrol inhibits proliferation and induces apoptosis via the Hippo/YAP pathway in human colon cancer cells. Biochem Biophys Res Commun 2022; 636:197-204. [DOI: 10.1016/j.bbrc.2022.10.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/30/2022]
|
41
|
miR-187/PDLIM1 Gets Involved in Gastric Cancer Progression and Cisplatin Sensitivity of Cisplatin by Mediating the Hippo-YAP Signaling Pathway. JOURNAL OF ONCOLOGY 2022; 2022:5456016. [PMID: 36164345 PMCID: PMC9509220 DOI: 10.1155/2022/5456016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/30/2022] [Accepted: 08/04/2022] [Indexed: 11/22/2022]
Abstract
Gastric cancer (GC) is one of the most prevalent malignancies in the digestive system across the world. The function and mechanism of PDLIM1, a cancer-suppressing gene, in gastric cancer progression remain unclear. This study is aimed at investigating the expression features and function of PDLIM1 in GC. RT-qPCR and western blot were used to compare the profiles of PDLIM1 and miR-187 between GC and normal tissues. The cell models of PDLIM1 overexpression and low expression were established in gastric cancer cell lines MKN45 and AGS. CCK8 and BrdU assays measured cell proliferation. Flow cytometry monitored cell apoptosis. Transwell analyzed cell invasion and migration. The influence of miR-187 overexpression on gastric cancer development was assessed. We predicted the targeted correlation between miR-187 and PDLIM1 through bioinformatics, which was corroborated via dual luciferase activity assay and RIP. Meanwhile, the cell model of PDLIM1 overexpression was built in AGS cells transfected with miR-187 mimics. A rescue experiment was conducted to assess the impact of PDLIM1 overexpression on the procancer function of miR-187. As a result, in contrast with normal paracancer tissues, PDLIM1 was substantially downregulated in GC tissues. Moreover, PDLIM1 overexpression considerably dampened proliferation, invasion, and migration in GC cells, boosted the cell apoptosis, and bolstered their sensitivity to cisplatin. PDLIM1 knockdown or miR-187 overexpression dramatically fostered GC cell proliferation, invasion, and migration and repressed cell apoptosis. Mechanism studies demonstrated that PDLIM1 vigorously restrained the profiles of the Hippo-YAP signaling pathway and the downstream target genes. miR-187 targeted PDLIM1, while miR-187 overexpression cramped PDLIM1 expression. The rescue experiment suggested that PDLIM1 overexpression weakened the procancer function of miR-187 in GC cells. In conclusion, our study demonstrated that PDLIM1 presented a low expression in GC tissues, while miR-187/PDLIM1 participated in GC development and cisplatin sensitivity by mediating the Hippo-YAP signaling pathway.
Collapse
|
42
|
Li L, Luo J, Fang JY, Zhang R, Ma JB, Zhu ZP. Expression characteristics of the yes-associated protein in breast cancer: A meta-analysis. Medicine (Baltimore) 2022; 101:e30176. [PMID: 36042678 PMCID: PMC9410612 DOI: 10.1097/md.0000000000030176] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The yes-associated protein (YAP) gene plays an important role in many malignant tumors, but its clinical significance in breast cancer remains unclear. This study aimed to explore the significance of YAP expression in breast cancer using meta-analysis. METHODS Seven databases will be searched to collect the case-control studies published on the association between YAP expression and clinical pathogenic features in breast cancer until December 2021: PubMed, EMBASE, Web of Science, China National Knowledge Infrastructure, Chinese Scientific Journal Database, Wan Fang Database, and the Chinese Biomedical Literature Database. To perform meta-analysis, STATA 14.0 and RevMan5 software were used with odds ratio (OR) and 95% confidence interval (95% CI) as the effect index, and publication bias and sensitivity analysis were subsequently tested. RESULTS Form a total of 10 articles used in this study, 8 studies consisted of nontriple negative breast cancer (non-TNBC) and the other 2 of TNBC. Meta-analysis indicated a positive expression rate of YAP in non-TNBC tissues that was lower than in normal breast tissue (OR = 0.15, 95% CI = 0.10-0.21, P < .001). In contrast, the positive rate of YAP expression in TNBC was significantly higher than that in normal breast tissue (OR = 18.23, 95% CI = 8.20-40.52, P < .001). Furthermore, the positive expression rate was higher in the patients with lymph node metastasis, higher tumor node metastasis stage and histologic grade, and larger diameter in TNBC. However, there was no statistical difference in the positive expression rate of YAP between non-TNBC patients and lymph node metastasis, tumor node metastasis stage, histologic grade, and tumor size. CONCLUSIONS YAP may participate in the occurrence and development of non-TNBC as a tumor suppressor gene; however, it may also be a carcinogenic factor in TNBC and may be a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Lan Li
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Jin Luo
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Jing-Yi Fang
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Rui Zhang
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Jian-Bo Ma
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Zheng-Peng Zhu
- Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
- *Correspondence: Zheng-Peng Zhu, Department of Pathology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Daling Road, Zhangwan District, Shiyan 16, Hubei Province, China (e-mail: )
| |
Collapse
|
43
|
Liu T, Guo W, Luo K, Li L, Dong J, Liu M, Shi X, Wang Z, Zhang J, Yin J, Qiu N, Lu M, Chen D, Jia X, Liu H, Gu Y, Xiong Y, Zheng G, Xu G, He Z, Zhang Z. Smoke-induced SAV1 Gene Promoter Hypermethylation Disrupts YAP Negative Feedback and Promotes Malignant Progression of Non-small Cell Lung Cancer. Int J Biol Sci 2022; 18:4497-4512. [PMID: 35864957 PMCID: PMC9295071 DOI: 10.7150/ijbs.73428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/02/2022] [Indexed: 11/26/2022] Open
Abstract
YAP (gene symbol YAP1) as a potential oncoprotein, is positively correlated with the malignancy of various tumors. However, overexpression of YAP alone in multiple normal tissue cells has failed to induce tumor formation and the underlying mechanism is poorly understood. Herein, we show that YAP activation directly induces transcription of its negative regulator, SAV1, to constitute a negative feedback loop, which plays a vital role in maintaining lung epithelial cell homeostasis and was dysregulated in non-small cell lung cancer (NSCLC). Notably, smoking promotes the hypermethylation of the SAV1 promoter region, which disrupts YAP negative feedback by inactivating the Hippo pathway. Besides, exogenous overexpression of SAV1 can act as a traffic protein, activating the Hippo signaling and concurrently inhibiting the WNT pathway to decrease cancer cell growth. Furthermore, using the lung cancer organoids, we found that lentivirus-mediated SAV1 gene transfer combined with methylation inhibitor and YAP-TEAD inhibitor is a potential feasible clinical medication regimen for the lung cancer patient, especially among the smoking population. Thus, this SAV1 mediated feedback loop provides an efficient mechanism to establish the robustness and homeostasis of YAP regulation and as a potential target of gene therapy for the smoking NSCLC population.
Collapse
Affiliation(s)
- Ting Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Wei Guo
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Kai Luo
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Lei Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Jing Dong
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Meijun Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Xingyuan Shi
- Department of Central Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Zhiyuan Wang
- Department of Central Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jianlei Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Jiang Yin
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Ni Qiu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Minying Lu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Danyang Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Xiaoting Jia
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Hao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Yixue Gu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Yan Xiong
- Department of Central Laboratory, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Guopei Zheng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China.,The State Key Laboratory of Respiratory, Guangzhou, Guangdong, P. R. China
| | - Gang Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Zhimin He
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| | - Zhijie Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of "Translational Medicine on Malignant Tumor Treatment", Guangzhou city, Guangdong, P. R. China
| |
Collapse
|
44
|
Dong P, Cai Z, Li B, Zhu Y, Chan AKY, Chiang MWL, Au CH, Sung WK, Cheung TT, Lo CM, Man K, Lee NP. HFE promotes mitotic cell division through recruitment of cytokinetic abscission machinery in hepatocellular carcinoma. Oncogene 2022; 41:4185-4199. [PMID: 35882980 DOI: 10.1038/s41388-022-02419-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
HFE (Hemochromatosis) is a conventional iron level regulator and its loss of function due to gene mutations increases the risk of cancers including hepatocellular carcinoma (HCC). Likewise, studies focusing on HFE overexpression in cancers are all limited to linking up these events as a consequence of iron level deregulation. No study has explored any iron unrelated role of HFE in cancers. Here, we first reported HFE as an oncogene in HCC and its undescribed function on promoting abscission in cytokinesis during mitotic cell division, independent of its iron-regulating ability. Clinical analyses revealed HFE upregulation in tumors linking to large tumor size and poor prognosis. Functionally and mechanistically, HFE promoted cytokinetic abscission via facilitating ESCRT abscission machinery recruitment to the abscission site through signaling a novel HFE/ALK3/Smads/LIF/Hippo/YAP/YY1/KIF13A axis. Pharmacological blockage of HFE signaling axis impeded tumor phenotypes in vitro and in vivo. Our data on HFE-driven HCC unveiled a new mechanism utilized by cancer cells to propel rapid cell division. This study also laid the groundwork for tumor intolerable therapeutics development given the high cytokinetic dependency of cancer cells and their vulnerability to cytokinetic blockage.
Collapse
Affiliation(s)
- Pingping Dong
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong.,Department of Radiation Oncology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziqing Cai
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Bingfeng Li
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Yueqin Zhu
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alice K Y Chan
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong.,Po Leung Kuk Tong Nai Kan Junior Secondary College, Hong Kong, Hong Kong
| | - Michael W L Chiang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Hang Au
- Hong Kong Genome Institute, Hong Kong, Hong Kong
| | - Wing Kin Sung
- Hong Kong Genome Institute, Hong Kong, Hong Kong.,School of Computing, National University of Singapore, Singapore, Singapore.,Computational and Systems Biology, Genome Institute of Singapore, Singapore, Singapore
| | - Tan To Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong.
| | - Nikki P Lee
- Department of Surgery, The University of Hong Kong, Hong Kong, Hong Kong. .,Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Hong Kong, Hong Kong.
| |
Collapse
|
45
|
Shen Y, Jing D, Zhao Z. The effect of AKT in extracellular matrix stiffness induced osteogenic differentiation of hBMSCs. Cell Signal 2022; 99:110404. [PMID: 35835331 DOI: 10.1016/j.cellsig.2022.110404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/02/2022] [Accepted: 07/08/2022] [Indexed: 02/05/2023]
Abstract
Extracellular matrix (ECM) stiffness is an important biophysical factor in human bone marrow mesenchymal stem cells (hBMSCs) differentiation. Although there is evidence that Yes-associated protein (YAP) plays an important role in ECM elasticity induced osteogenesis, but the regulatory mechanism and signaling pathways have not been distinctly uncovered. In this study, hBMSCs were cultured on collagen-coated polydimethylsiloxane hydrogels with stiffness corresponding to Young's moduli of 0.5 kPa and 32 kPa, and gene chip analyses revealed the phosphoinositide 3-kinase (PI3K)-AKT pathway was highly correlated with ECM stiffness. Following western blots indicated that AKT phosphorylation was evidently affected in 5th-7th days after ECM stiffness stimulation, while PI3K showed little difference. The AKT activator SC79 and inhibitor MK2206 were utilized to modulate AKT phosphorylation. SC79 and MK2206 caused alteration in the mRNA expression and protein level of alkaline phosphatase (ALP), collagen type I alpha 1 (COL1A1) and runt related transcription factor 2 (RUNX2). On 32 kPa substrates, YAP enrichment in nucleus were significantly promoted by SC79 and remarkably decreased by MK2206. Besides, the ratio of YAP/p-YAP is upregulated by SC79 on both 32 kPa and 0.5 kPa substrates. In conclusion, these findings suggest that AKT is involved in the modulation of ECM stiffness induced osteogenesis, and AKT phosphorylation also influences the subcellular localization and activation of YAP.
Collapse
Affiliation(s)
- Yu Shen
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, China.
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
46
|
Yu T, Liu Y, Xue J, Sun X, Zhu D, Ma L, Guo Y, Jin T, Cao H, Chen Y, Zhu T, Li X, Liang H, Du Z, Shan H. Gankyrin modulated non-small cell lung cancer progression via glycolysis metabolism in a YAP1-dependent manner. Cell Death Dis 2022; 8:312. [PMID: 35810157 PMCID: PMC9271063 DOI: 10.1038/s41420-022-01104-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/10/2022] [Accepted: 06/27/2022] [Indexed: 11/09/2022]
Abstract
Non-small cell lung cancer (NSCLC) is highly malignant and heterogeneous form of lung cancer and involves various oncogene alterations. Glycolysis, an important step in tumor metabolism, is closely related to cancer progression. In this study, we investigated the biological function and mechanism of action of Gankyrin in glycolysis and its association with NSCLC. Analyzed of data from The Cancer Genome Atlas as well as NSCLC specimens and adjacent tissues demonstrated that Gankyrin expression was upregulated in NSCLC tissues compared to adjacent normal tissues. Gankyrin was found to significantly aggravate cancer-related phenotypes, including cell viability, migration, invasion, and epithelial mesenchymal transition (EMT), whereas Gankyrin silencing alleviated the malignant phenotype of NSCLC cells. Our results reveal that Gankyrin exerted its function by regulating YAP1 expression and increasing its nuclear translocation. Importantly, YAP1 actuates glycolysis, which involves glucose uptake, lactic acid production, and ATP generation and thus might contribute to the tumorigenic effect of Gankyrin. Furthermore, the Gankyrin-accelerated glycolysis in NSCLC cells was reversed by YAP1 deficiency. Gankyrin knockdown reduced A549 cell tumorigenesis and EMT and decreased YAP1 expression in a subcutaneous xenograft nude mouse model. In conclusion, both Gankyrin and YAP1 play important roles in tumor metabolism, and Gankyrin-targeted inhibition may be a potential anti-cancer therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, P. R. China.,Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China
| | - Yanyan Liu
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, P. R. China
| | - Junwen Xue
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Xiang Sun
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Di Zhu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Lu Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Yingying Guo
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Tongzhu Jin
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Huiying Cao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Yingzhun Chen
- Department of Pathology, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Tong Zhu
- Department of General Surgery, the Fourth Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Xuelian Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China
| | - Haihai Liang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China
| | - Zhimin Du
- Zhuhai People's Hospital, Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, P. R. China. .,Institute of Clinical Pharmacy, the Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China.
| | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, P. R. China. .,Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, 150081, P. R. China. .,Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin, Heilongjiang, 150081, P. R. China.
| |
Collapse
|
47
|
Zeng Y, Yin L, Zhou J, Zeng R, Xiao Y, Black AR, Hu T, Singh PK, Yin F, Batra SK, Yu F, Chen Y, Dong J. MARK2 regulates chemotherapeutic responses through class IIa HDAC-YAP axis in pancreatic cancer. Oncogene 2022; 41:3859-3875. [PMID: 35780183 PMCID: PMC9339507 DOI: 10.1038/s41388-022-02399-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 01/04/2023]
Abstract
Despite paclitaxel's wide use in cancer treatment, patient response rate is still low and drug resistance is a major clinical obstacle. Through a Phos-tag-based kinome-wide screen, we identified MARK2 as a critical regulator for paclitaxel chemosensitivity in PDAC. We show that MARK2 is phosphorylated by CDK1 in response to antitubulin chemotherapeutics and in unperturbed mitosis. Phosphorylation is essential for MARK2 in regulating mitotic progression and paclitaxel cytotoxicity in PDAC cells. Mechanistically, our findings also suggest that MARK2 controls paclitaxel chemosensitivity by regulating class IIa HDACs. MARK2 directly phosphorylates HDAC4 specifically during antitubulin treatment. Phosphorylated HDAC4 promotes YAP activation and controls expression of YAP target genes induced by paclitaxel. Importantly, combination of HDAC inhibition and paclitaxel overcomes chemoresistance in organoid culture and preclinical PDAC animal models. The expression levels of MARK2, HDACs, and YAP are upregulated and positively correlated in PDAC patients. Inhibition of MARK2 or class IIa HDACs potentiates paclitaxel cytotoxicity by inducing mitotic abnormalities in PDAC cells. Together, our findings identify the MARK2-HDAC axis as a druggable target for overcoming chemoresistance in PDAC.
Collapse
Affiliation(s)
- Yongji Zeng
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ling Yin
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jiuli Zhou
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Renya Zeng
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tuo Hu
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pankaj K Singh
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Feng Yin
- Department of Pathology and Anatomic Sciences, University of Missouri, Columbia, MO, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Fang Yu
- Department of Biostatistics, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yuanhong Chen
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
48
|
Barberino RS, Silva RLS, Palheta Junior RC, Smitz JEJ, Matos MHT. Protective Effects of Antioxidants on Cyclophosphamide-Induced Ovarian Toxicity. Biopreserv Biobank 2022; 21:121-141. [PMID: 35696235 DOI: 10.1089/bio.2021.0159] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The most common limitation of anticancer chemotherapy is the injury to normal cells. Cyclophosphamide, which is one of the most widely used alkylating agents, can cause premature ovarian insufficiency and infertility since the ovarian follicles are extremely sensitive to their effects. Although little information is available about the pathogenic mechanism of cyclophosphamide-induced ovarian damage, its toxicity is attributed to oxidative stress, inflammation, and apoptosis. The use of compounds with antioxidant and cytoprotective properties to protect ovarian function from deleterious effects during chemotherapy would be a significant advantage. Thus, this article reviews the mechanism by which cyclophosphamide exerts its toxic effects on the different cellular components of the ovary, and describes 24 cytoprotective compounds used to ameliorate cyclophosphamide-induced ovarian injury and their possible mechanisms of action. Understanding these mechanisms is essential for the development of efficient and targeted pharmacological complementary therapies that could protect and prolong female fertility.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Regina Lucia S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta Junior
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, Free University Brussels-VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley-UNIVASF, Petrolina, Brazil
| |
Collapse
|
49
|
Zinc oxide nanoparticles promotes ferroptosis to repress cancer cell survival and inhibits invasion and migration by targeting miR-27a-3p/YAP axis in renal cell carcinoma. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.103753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
50
|
Neinaa YMEH, El-Aziz Mohamed DA, Ali SAEM, Gaballah HH, El-Tatawy RA. YAP1 Expression in Lichen Planus and Squamous Cell Carcinoma: Role in Disease Pathogenesis and Potential Therapeutic Target. Am J Dermatopathol 2022; 44:348-354. [PMID: 35234193 DOI: 10.1097/dad.0000000000002152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Lichen planus (LP), especially oral type, reported a potential risk of malignant transformation to squamous cell carcinoma (SCC). Yes-Associated Protein (YAP1), a key component of the Hippo pathway, acts as a transcription cofactor regulating expression of genes involved in cell proliferation, apoptosis, and migration. Therefore, it has been implicated in carcinogenesis of a wide variety of human cancers. OBJECTIVES To study YAP1 expression in LP and SCC in comparison to normal control (NC) specimens. PATIENTS AND METHODS This study was conducted on 50 NC specimens, 50 LP specimens, and 50 SCC specimens. They were categorized into 2 main groups; cutaneous (25 NC, 25 LP, 25 SCC), and oral (25 NC, 25 LP, 25 SCC). All specimens were examined for YAP1 antibody expression by immunohistochemistry and YAP1 mRNA expression by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS In both cutaneous and oral groups; significant upregulation of YAP1 expressions was observed in SCC specimens followed by LP and then NC specimens in the same sequence. Its expression in SCC was found to be significantly higher in poorly and moderately differentiated types than well differentiated types. CONCLUSION YAP1 may have a potential role in the pathogenesis of LP and oncogenesis and progression of SCC. Moreover, it could be considered as a novel therapeutic target for such cases.
Collapse
Affiliation(s)
| | | | | | | | - Rania Ahmed El-Tatawy
- Dermatology and Venereology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|