1
|
Ramakrishna NB, Mohamad Sahari UB, Johmura Y, Ali NA, Alghamdi M, Bauer P, Khan S, Ordoñez N, Ferreira M, Pinto Basto J, Alkuraya FS, Faqeih EA, Mori M, Almontashiri NAM, Al Shamsi A, ElGhazali G, Abu Subieh H, Al Ojaimi M, El-Hattab AW, Said Al-Kindi SA, Alhashmi N, Alhabshan F, Al Saman A, Tfayli H, Arabi M, Khalifeh S, Taylor A, Alfadhel M, Jain R, Sinha S, Shenbagam S, Ramachandran R, Altunoğlu U, Jacob A, Thalange N, El Bejjani M, Perrin A, Shin JW, Al-Maawali A, Al-Shidhani A, Al-Futaisi A, Rabea F, Chekroun I, Almarri MA, Ohta T, Nakanishi M, Alsheikh-Ali A, Ali FR, Bertoli-Avella AM, Reversade B, Abou Tayoun A. FBXO22 deficiency defines a pleiotropic syndrome of growth restriction and multi-system anomalies associated with a unique epigenetic signature. Am J Hum Genet 2025; 112:1233-1246. [PMID: 40215970 PMCID: PMC12120182 DOI: 10.1016/j.ajhg.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 05/04/2025] Open
Abstract
FBXO22 encodes an F-box protein, which acts as a substrate-recognition component of the SKP1-CUL1-F-box (SCF) E3 ubiquitin ligase complex. Despite its known roles in the post-translational ubiquitination and degradation of specific substrates, including histone demethylases, the impact of FBXO22 on human development remains unknown. Here, we characterize a pleiotropic syndrome with prominent prenatal onset growth restriction and notable neurodevelopmental delay across 16 cases from 14 families. Through exome and genome sequencing, we identify four distinct homozygous FBXO22 variants with loss-of-function effects segregating with the disease: three predicted to lead to premature translation termination due to frameshift effects and a single-amino-acid-deletion variant, which, we show, impacts protein stability in vitro. We confirm that affected primary fibroblasts with a frameshift mutation are bereft of endogenous FBXO22 and show increased levels of the known substrate histone H3K9 demethylase KDM4B. Accordingly, we delineate a unique epigenetic signature for this disease in peripheral blood via long-read sequencing. Altogether, we identify and demonstrate that FBXO22 deficiency leads to a pleiotropic syndrome in humans, encompassing growth restriction and neurodevelopmental delay, the pathogenesis of which may be explained by broad chromatin alterations.
Collapse
Affiliation(s)
- Navin B Ramakrishna
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Umar Bin Mohamad Sahari
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, Singapore 119260, Singapore
| | - Yoshikazu Johmura
- Division of Cancer and Senescence Biology, Cancer Research Institute, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Nur Ain Ali
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Malak Alghamdi
- Unit of Medical Genetics, Department of Pediatrics, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia
| | | | | | | | | | | | - Fowzan S Alkuraya
- Department of Translational Genomics, Center for Genomic Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Eissa Ali Faqeih
- Section of Medical Genetics, King Fahad Medical City, Children's Specialist Hospital, Riyadh, Saudi Arabia
| | - Mari Mori
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Genetic and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH, USA
| | - Naif A M Almontashiri
- Center for Genetics and Inherited Diseases, Taibah University, 7534 Abdul Muhsin Ibn Abdul Aziz, Al Ihn, Al-Madinah al-Munawwarah 42318, Saudi Arabia; Faculty of Applied Medical Sciences, Taibah University, Janadah Bin Umayyah Road, Tayba, Al-Madinah al-Munawwarah 42353, Saudi Arabia
| | - Aisha Al Shamsi
- Paediatrics Department, Tawam Hospital, Al-Ain, United Arab Emirates
| | - Gehad ElGhazali
- HQ Medical Operations Division, Union 71, Abu Dhabi, United Arab Emirates
| | - Hala Abu Subieh
- Maternal Fetal Medicine Department, Kanad Hospital, Al Ain, United Arab Emirates
| | - Mode Al Ojaimi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ayman W El-Hattab
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | | | - Fahad Alhabshan
- Department of Cardiac Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Abdulaziz Al Saman
- Pediatric Neurology Department, National Neuroscience Institute, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Hala Tfayli
- Pediatric Endocrinology and Diabetes, American University of Beirut Medical Center (AUBMC), Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, Pediatric Cardiology Division, Children's Heart Center, American University of Beirut Medical Center, Beirut, Lebanon
| | - Simone Khalifeh
- Pediatric Neurology Division, American University of Beirut Medical Center, Beirut, Lebanon
| | - Alan Taylor
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Majid Alfadhel
- Genetics and Precision Medicine Department (GPM), King Abdullah Specialized Children's Hospital (KASCH), King Abdulaziz Medical City, Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia; Medical Genomic Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Ministry of National Guard Health Affairs (MNG-HA), Riyadh, Saudi Arabia
| | - Ruchi Jain
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Shruti Sinha
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Shruti Shenbagam
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | - Umut Altunoğlu
- Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul 34010, Turkey
| | - Anju Jacob
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Nandu Thalange
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Mireille El Bejjani
- Al Jalila Children's Specialty Hospital, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Arnaud Perrin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Department of Biochemistry, National University of Singapore, Singapore 119260, Singapore
| | - Almundher Al-Maawali
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Azza Al-Shidhani
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Amna Al-Futaisi
- Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Fatma Rabea
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Ikram Chekroun
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates
| | - Mohamed A Almarri
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Genome Center, Dubai Police GHQ, Dubai, United Arab Emirates
| | - Tomohiko Ohta
- Department of Translational Oncology, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Alawi Alsheikh-Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Dubai Health, Dubai, United Arab Emirates
| | - Fahad R Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates
| | | | - Bruno Reversade
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Medical Genetics Department, Koç University School of Medicine (KUSOM), Istanbul 34010, Turkey; NUS Cardiovascular-Metabolic Disease Translational Research Programme (CVMD-TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Laboratory of Human Genetics & Therapeutics, Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia.
| | - Ahmad Abou Tayoun
- Dubai Health Genomic Medicine Center, Dubai Health, Dubai, United Arab Emirates; Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai, United Arab Emirates.
| |
Collapse
|
2
|
Sun Z, Zhang Z, Zhang J, Yang Z, Pan S, Li X, Wang X, Zhu X. F-box only protein 25-mediated α-actinin 1 upregulation drives ovarian cancer progression via ERK1/2 signaling in tumor cells and macrophage M2 polarization. Int Immunopharmacol 2025; 153:114479. [PMID: 40117808 DOI: 10.1016/j.intimp.2025.114479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND ACTN1 belongs to the α-actinin family and is considered a tumor-promoting gene in various tumor types; however, the biological function and fundamental molecular mechanisms of ACTN1 in ovarian cancer remain unclear. METHODS The HPA and The Cancer Genome Atlas (TCGA) databases were used to compare the expression of ACTN1 in normal ovarian and OC tissues. The Kaplan-Meier Plotter database was used to analyze the relationship between the expression of ACTN1 and the prognosis of ovarian cancer. The TIMER2.0 database was used to analyze the correlation between the expression of ACTN1 and macrophages. CCK-8, colony formation, and Transwell assays as well as flow cytometry were used to determine the biological properties of the cells. Protein expression was assessed by immunohistochemistry, immunofluorescence, and western blot analysis. A co-culture experiment was used to analyze the effect of ovarian cancer cells on the polarization of macrophages. Co-immunoprecipitation was performed to validate the interaction between FBXO25 and ACTN1. RESULTS ACTN1 was highly expressed in OC tissues and cell lines. Downregulation of ACTN1 attenuated the proliferation, migration, and invasion of OC cells, promoted apoptosis and reduced the aggregation of M2 macrophages and the expression of CD163. The opposite effect was observed following the upregulation of ACTN1. Mechanistically, ACTN1 knockdown reduced ERK1/2 phosphorylation and inhibited epithelial-mesenchymal transition (EMT), whereas its overexpression resulted in the opposite effect. The ERK1/2 inhibitor LY3214996 partially reversed cell proliferation, migration, and M2 polarization of macrophages promoted by ACTN1 overexpression. Moreover, FBXO25, which is upstream of ACTN1 and interacts with it. FBXO25 upregulation partially reversed cell proliferation and migration inhibited by ACTN1 knockdown. CONCLUSION Upregulation of ACTN1 by FBXO25 promotes the progression of ovarian cancer by activating the ERK1/2 signaling pathway and M2 polarization of macrophages. The FBXO25/ACTN1/ERK1/2 axis and M2 macrophages may represent promising targets for developing ovarian cancer treatments.
Collapse
Affiliation(s)
- Zhengwei Sun
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zihan Zhang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jiamin Zhang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ziyi Yang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaosheng Li
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xujing Wang
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Gynecological Diseases, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
3
|
Lu XF, Zhang HW, Chang X, Guo YZ. F-box protein 22: A prognostic biomarker for colon cancer associated with immune infiltration and chemotherapy resistance. World J Gastrointest Oncol 2025; 17:102913. [PMID: 40235877 PMCID: PMC11995338 DOI: 10.4251/wjgo.v17.i4.102913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/10/2025] [Accepted: 02/21/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Colon cancer represents a significant malignant neoplasm within the digestive system, characterized by a high incidence rate and substantial disease burden. The F-box protein 22 (FBXO22) plays a role in forming a specific type of ubiquitin ligase subunit, which is expressed abnormally in various malignant neoplasms and shows a notable relationship with prognosis in patients with cancer. Nevertheless, the function of FBXO22 in the context of colon cancer remains inadequately elucidated. AIM To explore the role of FBXO22 in colon cancer by examining FBXO22 expression patterns and analyzing how the protein affects the prognosis in patients who have undergone surgery. METHODS Samples of cancerous and nearby normal tissues from patients with colon cancer were gathered, along with pertinent clinical data. Expression levels of the FBXO22 gene in both cancerous and paracancerous tissues were assessed through immunohistochemistry. The median H score served as a criterion for categorizing FBXO22 gene expression into high and low levels in cancerous tissues, and the relationship between these expression levels and various pathologic characteristics of patients, such as age, sex, and clinical stage, was analyzed. Colon cancer cell lines HCT116 and DLD-1 were used and divided into three groups: A blank control group, a negative control group, and a si-FBXO22 group. FBXO22 gene mRNA and protein expression were measured 24 hours post-transfection using real-time fluorescence quantitative polymerase chain reaction and western blotting. The proliferation capabilities of the cells in each group were assessed using the Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine assay, while cellular migration and invasion abilities were evaluated using scratch healing and Transwell assays. Various online platforms, including the Timer Immune Estimation Resource, were used to analyze pan-cancer expression, promoter methylation levels, and mutation frequencies of the FBXO22 gene in colon cancer patients. Additionally, the correlation between FBXO22 gene expression, patient prognosis, immune cell infiltration, and the expression of immune molecules in the colon cancer microenvironment was investigated. The relationship between FBXO22 gene expression and chemotherapy resistance, along with the potential mechanisms of action of the FBXO22 gene, were analyzed using The Cancer Genome Atlas dataset and the Genomics of Drug Sensitivity in Cancer drug training set via R software. RESULTS Compared with normal colonic tissues, the FBXO22 gene was highly expressed in colon cancer tissues. Post-operative patients with colon cancer elevated FBXO22 reduced survival and exhibited resistance to various chemotherapeutic agents. FBXO22 expression suppresses the infiltration of anti-tumor immune cells. In vitro, FBXO22 knockdown inhibited the proliferation and migration of colon cancer cells. CONCLUSION The FBXO22 gene is a biomarker of poor prognosis in patients with colon cancer and has potential as a target for immunotherapy and overcoming chemotherapy resistance.
Collapse
Affiliation(s)
- Xiao-Fei Lu
- Department of Clinical Medicine, Hebei University of Engineering, Handan 056002, Hebei Province, China
| | - Hong-Wei Zhang
- Department of Gastroenterology, Affiliated Hospital of Hebei Engineering University, Handan 056002, Hebei Province, China
| | - Xiao Chang
- Department of Gastroenterology, Affiliated Hospital of Hebei Engineering University, Handan 056002, Hebei Province, China
| | - Yong-Ze Guo
- Department of Gastroenterology, Affiliated Hospital of Hebei Engineering University, Handan 056002, Hebei Province, China
| |
Collapse
|
4
|
Lei Z, Luo Y, Fu Q, Lu J, Wang C, Zhang L, Zhang Z. Ribosomal protein L6 suppresses hepatocellular carcinoma by modulating FBXO22-mediated p53 degradation. Cell Signal 2025; 127:111612. [PMID: 39842528 DOI: 10.1016/j.cellsig.2025.111612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/30/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
The ribosomal protein L6 (RPL6) is significant in the progression of different cancer types. However, its precise role in hepatocellular carcinoma (HCC) remains unclear. This research demonstrated that the expression levels of RPL6 are notably decreased in HCC tissues. The decreased expression of RPL6 is strongly linked to tumor size, the presence of vascular invasion, and a worse prognosis. Functional experiments revealed that the expression of RPL6 impedes the proliferation of HCC cells and the advancement of xenograft tumors. Mechanistically, we found that RPL6 binds to and is degraded by the E3 ubiquitin ligase FBXO22, thereby inhibiting the polyubiquitination and subsequent degradation of p53 by FBXO22. The enhanced activity of p53 further contributes to cell growth inhibition. In contrast, the levels of p53 decreased significantly following RPL6 depletion, indicating that RPL6 is essential for the stabilization of p53. In summary, RPL6 inhibits the proliferation of HCC cells via the FBXO22/p53 signaling pathway, suggesting its potential as a biomarker and a therapeutic target for HCC.
Collapse
Affiliation(s)
- Zhen Lei
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China; Department of Hepatobiliary Surgery, The First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang 832008, PR China
| | - Yiming Luo
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China
| | - Qinggang Fu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China
| | - Junli Lu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China
| | - Chao Wang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China
| | - Long Zhang
- Department of Hepatopancreatobiliary Surgery, Ganzhou People's Hospital of Jiangxi Province (Ganzhou Hospital Afffliated to Nanchang University), Ganzhou 341000, PR China.
| | - Zhiwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, PR China.
| |
Collapse
|
5
|
Redondo-Calvo F, Rabanal-Ruiz Y, Verdugo-Moreno G, Bejarano-Ramírez N, Bodoque-Villar R, Durán-Prado M, Illescas S, Chicano-Galvez E, Gómez-Romero FJ, Martinez-Alarcón J, Arias-Pardilla J, Lopez-Juarez P, Padin JF, Peinado JR, Serrano-Oviedo L. Longitudinal Assessment of Nasopharyngeal Biomarkers Post-COVID-19: Unveiling Persistent Markers and Severity Correlations. J Proteome Res 2024; 23:5064-5084. [PMID: 39392878 PMCID: PMC11536464 DOI: 10.1021/acs.jproteome.4c00536] [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: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
SARS-CoV-19 infection provokes a variety of symptoms; most patients present mild/moderate symptoms, whereas a small proportion of patients progress to severe illness with multiorgan failure accompanied by metabolic disturbances requiring ICU-level care. Given the importance of the disease, researchers focused on identifying severity-associated biomarkers in infected patients as well as markers associated with patients suffering long-COVID. However, little is known about the presence of biomarkers that remain a few years after SARS-CoV-2 infection once the patients fully recover of the symptoms. In this study, we evaluated the presence of persistent biomarkers in the nasopharyngeal tract two years after SARS-Cov-2 infection in fully asymptomatic patients, taking into account the severity of their infection (mild/moderate and severe infections). In addition to the direct identification of several components of the Coronavirus Infection Pathway in those individuals that suffered severe infections, we describe herein 371 proteins and their associated canonical pathways that define the different adverse effects of SARS-CoV-2 infections. The persistence of these biomarkers for up to two years after infection, along with their ability to distinguish the severity of the infection endured, highlights the surprising presence of persistent nasopharyngeal exudate changes in fully recovered patients.
Collapse
Affiliation(s)
- Francisco
Javier Redondo-Calvo
- Department
of Anesthesiology and Critical Care Medicine, University General Hospital, SESCAM, Ciudad Real 13004, Spain
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
- Faculty
of Medicine, University of Castilla-La Mancha, Castilla La Mancha, Ciudad Real 13071, Spain
| | - Yoana Rabanal-Ruiz
- Oxidative
Stress and Neurodegeneration Group, Medical Sciences Department, Medical
School, UCLM, Regional Centre for Biomedical
Research, Research Institute of Castilla-La
Mancha (IDISCAM), University of Castilla-La
Mancha, Ciudad Real 13071, Spain
- Department
of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Gema Verdugo-Moreno
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| | - Natalia Bejarano-Ramírez
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
- Faculty
of Medicine, University of Castilla-La Mancha, Castilla La Mancha, Ciudad Real 13071, Spain
- Department
of Pediatrics, University General Hospital, Ciudad Real 13004, Spain
| | - Raquel Bodoque-Villar
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| | - Mario Durán-Prado
- Oxidative
Stress and Neurodegeneration Group, Medical Sciences Department, Medical
School, UCLM, Regional Centre for Biomedical
Research, Research Institute of Castilla-La
Mancha (IDISCAM), University of Castilla-La
Mancha, Ciudad Real 13071, Spain
- Department
of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Soledad Illescas
- Department
of Microbiology, University General Hospital, Ciudad Real 13004, Spain
| | - Eduardo Chicano-Galvez
- IMIBIC
Mass Spectrometry and Molecular Imaging Unit (IMSMI). Maimonides Biomedical
Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital, University of Cordoba (UCO), Córdoba 14004, Spain
| | - Francisco Javier Gómez-Romero
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| | | | - Javier Arias-Pardilla
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| | - Pilar Lopez-Juarez
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| | - Juan Fernando Padin
- Oxidative
Stress and Neurodegeneration Group, Medical Sciences Department, Medical
School, UCLM, Regional Centre for Biomedical
Research, Research Institute of Castilla-La
Mancha (IDISCAM), University of Castilla-La
Mancha, Ciudad Real 13071, Spain
- Department
of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Juan Ramón Peinado
- Oxidative
Stress and Neurodegeneration Group, Medical Sciences Department, Medical
School, UCLM, Regional Centre for Biomedical
Research, Research Institute of Castilla-La
Mancha (IDISCAM), University of Castilla-La
Mancha, Ciudad Real 13071, Spain
- Department
of Medical Sciences, School of Medicine at Ciudad Real, University of Castilla-La Mancha, Ciudad Real 13071, Spain
| | - Leticia Serrano-Oviedo
- Traslational
Investigation Unit, University General Hospital, SESCAM. Research Institute of Castilla-La Mancha (IDISCAM), Ciudad Real 13004, Spain
| |
Collapse
|
6
|
Yang LL, Zhang XK, Cao Y, Shi LY, Xie SY, Yang YJ, Wu SJ, Sun HZ, Tang XJ, Yuan DL, Zhang D, Xu XF, Li Q, Ying XY. PARP1 acetylation at K119 is essential in regulating the progression and proliferation of cervical cancer cells. Med Oncol 2024; 41:273. [PMID: 39400626 DOI: 10.1007/s12032-024-02315-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/27/2024] [Indexed: 10/15/2024]
Abstract
Cervical cancer, CC, is one of the malignant cancers in women worldwide. Many studies about the genesis and progression of CC have been done at genomic, transcriptional, translational, and epigenetic levels. However, much less is done at post-translational modification (PTM) level. We first used pan-PTM antibodies to compare the pan PTM levels between clinical normal cervical tissues and CC tissues; we then sent the selected samples for label-free identification of acetylation sites. Next, we employed WT or K119A mutant PARP1-EGFP-STREPII plasmid transfection in Hela cells and examined various indexes including colony formation, wound healing, ROS generation, early apoptosis, and immunofluorescence and quantification of proliferation markers (Ki67, PCNA, and p-P53). Last, we examined the levels of multiple important kinases regulating cervical cancer progression. We found that pan-acetylation was the most downregulated in clinical CC samples, whereas the acetylation of PARP1, Poly(ADP-ribose) polymerase-1, was upregulated at K119. Next, we showed that PARP1-WT overexpression significantly suppressed the proliferation and progression in CC cell line Hela, while K119A overexpression didn't show any impact. Finally, PARP1-WT overexpression significantly decreased p-ERK1/2 while didn't affect the phosphorylation levels of other important kinases such as AKT, MTOR, and RPS6. This study discovered a new type of PTM of PARP1 in CC, and showed that PARP1 acetylation at K119 is essential in regulating the proliferation and progression of CC through ERK1/2. Further studies are required to investigate how PARP1 acetylation impact its function.
Collapse
Affiliation(s)
- Li-Li Yang
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Xue-Ke Zhang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China
| | - Ying Cao
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
| | - Li-Ya Shi
- Reproductive Medicine Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200082, China
- Obstetrics and Gynecology, Shanghai East Hospital Ji'an Hospital, Ji'an, 343006, China
| | - Shi-Ya Xie
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China
| | - Yan-Jie Yang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China
| | - Shao-Jun Wu
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Hong-Zhan Sun
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China
| | - Xue-Jun Tang
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China
| | - Dong-Lan Yuan
- Taizhou People's Hospital Affiliated to Nanjing Medical University, 366 Taihu Road, Taizhou, 225300, China.
| | - Dong Zhang
- State Key Lab of Reproductive Medicine and Offspring Health, Nanjing Medical University, 101 Longmian Ave., Nanjing, 211166, China.
| | - Xiao-Feng Xu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, 230022, Anhui, China.
| | - Qian Li
- Department of Gynecology, Nanjing Women and Children's Healthcare Hospital, 123 Tianfei Lane, Mochou Road, Nanjing, 210018, China.
| | - Xiao-Yan Ying
- The Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjia Garden, Nanjing, 210029, China.
| |
Collapse
|
7
|
Zhang H, Bai Y, Li J, Chen T, Shang G. FBXO22 promotes osteosarcoma progression via regulation of FOXO1 for ubiquitination and degradation. J Cell Mol Med 2024; 28:e70021. [PMID: 39153212 PMCID: PMC11330286 DOI: 10.1111/jcmm.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yang Bai
- Department of NursingShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiatong Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
8
|
Wu T, Jin X, Huang C, Yu X, Xu B, Gao W, Qiu X, Bao M, Zhao D, Feng G, Zheng B, Huang X. E3 ligase FBXO22 is not significant for spermatogenesis and male fertility in mice. Am J Transl Res 2024; 16:1834-1844. [PMID: 38883371 PMCID: PMC11170574 DOI: 10.62347/stda4237] [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: 04/11/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND F-box-only protein 22 (FBXO22), an important substrate receptor of the SKP1-Cullin-F-box (SCF) ubiquitin ligases, has been reported to be involved in many biological processes, including tumorigenesis, neurological disorders, cellular senescence, and DNA damage. However, the specific role of FBXO22 during spermatogenesis is poorly understood. METHODS We produced Fbxo22 conditional knockout (cKO) and global knockout (KO) mice and assessed their sperm masurements using a computer-assisted sperm analysis (CASA) system. Additionally, we conducted histologic staining and immunostaining to examine the impact of Fbxo22 loss on spermatogenesis. RESULTS Our results revealed that there were no notable differences in semen quality, fertility test results, or histologic findings in Fbxo22-KO and Fbxo22-cKO mice compared to the control group. CONCLUSIONS Our study demonstrated that Fbxo22 is not significant for spermatogenesis or male fertility in mice. These findings will help researchers avoid redundant efforts and serve as a foundational resource for genetic studies on human fertility.
Collapse
Affiliation(s)
- Tiantian Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University Nanjing 211166, Jiangsu, China
| | - Xin Jin
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University Suzhou 215002, Jiangsu, China
| | - Chao Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University Suzhou 215002, Jiangsu, China
| | - Xiangling Yu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University Wuxi 214122, Jiangsu, China
| | - Bingya Xu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University Wuxi 214122, Jiangsu, China
| | - Wenxin Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University Nanjing 211166, Jiangsu, China
| | - Xiya Qiu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University Suzhou 215002, Jiangsu, China
| | - Mingyuan Bao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University Nanjing 211166, Jiangsu, China
| | - Dan Zhao
- Fourth Affiliated Hospital of Jiangsu University Zhenjiang 212008, Jiangsu, China
| | - Guannan Feng
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University Suzhou 215002, Jiangsu, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University Suzhou 215002, Jiangsu, China
| | - Xiaoyan Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University Nanjing 211166, Jiangsu, China
| |
Collapse
|
9
|
Csergeová L, Krbušek D, Janoštiak R. CIP/KIP and INK4 families as hostages of oncogenic signaling. Cell Div 2024; 19:11. [PMID: 38561743 PMCID: PMC10985988 DOI: 10.1186/s13008-024-00115-z] [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: 12/12/2023] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
CIP/KIP and INK4 families of Cyclin-dependent kinase inhibitors (CKIs) are well-established cell cycle regulatory proteins whose canonical function is binding to Cyclin-CDK complexes and altering their function. Initial experiments showed that these proteins negatively regulate cell cycle progression and thus are tumor suppressors in the context of molecular oncology. However, expanded research into the functions of these proteins showed that most of them have non-canonical functions, both cell cycle-dependent and independent, and can even act as tumor enhancers depending on their posttranslational modifications, subcellular localization, and cell state context. This review aims to provide an overview of canonical as well as non-canonical functions of CIP/KIP and INK4 families of CKIs, discuss the potential avenues to promote their tumor suppressor functions instead of tumor enhancing ones, and how they could be utilized to design improved treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Lucia Csergeová
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | - David Krbušek
- BIOCEV-First Faculty of Medicine, Charles University, Prague, Czechia
| | | |
Collapse
|
10
|
Shen Z, Dong T, Yong H, Deng C, Chen C, Chen X, Chen M, Chu S, Zheng J, Li Z, Bai J. FBXO22 promotes glioblastoma malignant progression by mediating VHL ubiquitination and degradation. Cell Death Discov 2024; 10:151. [PMID: 38519492 PMCID: PMC10959977 DOI: 10.1038/s41420-024-01919-2] [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: 08/24/2023] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor. Despite comprehensive treatment with traditional surgery, radiotherapy, and chemotherapy, the median survival rate is <14.6% and the 5-year survival rate is only 5%. FBXO22, a substrate receptor of the SCF ubiquitin ligases, has been reported to play a promoting role in melanoma, liver cancer, cervical cancer, and other cancers. However, the function of FBXO22 in GBM has not been reported. In the present study, we demonstrate that FBXO22 is highly expressed in glioma and is positively correlated with worse pathological features and shorter survival of GBM patients. We revealed that FBXO22 promotes GBM cell proliferation, angiogenesis, migration, and tumorigenesis in vitro and in vivo. In terms of mechanism, we reveal that FBXO22 decreases VHL expression by directly mediating VHL ubiquitination degradation, which ultimately increases HIF-1α and VEGFA expression. In addition, our data confirm that there are positive correlations among FBXO22, HIF-1α, and VEGFA expression, and there is a negative correlation between FBXO22 and VHL protein expression in glioma patients. Our study strongly indicates that FBXO22 is a promising diagnostic marker and therapeutic target for glioma patients.
Collapse
Affiliation(s)
- Zhigang Shen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Tao Dong
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hongmei Yong
- Department of Oncology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huaian, Jiangsu, China
| | - Chuyin Deng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changxiu Chen
- Department of Pediatrics, the Affiliated Huaihai Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xintian Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Miaolei Chen
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sufang Chu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Zhongwei Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Laboratory of Tumor Epigenetics, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, China.
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
11
|
Mei J, Pan L, Huang M, Bao D, Gao H, Wang D. DDOST is associated with tumor immunosuppressive microenvironment in cervical cancer. Discov Oncol 2024; 15:69. [PMID: 38460058 PMCID: PMC10924880 DOI: 10.1007/s12672-024-00927-z] [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: 10/26/2023] [Accepted: 03/05/2024] [Indexed: 03/11/2024] Open
Abstract
Evidence has revealed that DDOST plays an important role in cancer development and progression. However, there are no reports on functions of DDOST in cervical tumorigenesis. Hence, we investigated the relationship of DDOST with prognosis, mutation, promoter methylation, immune cell infiltration, and drug sensitivity using bioinformatics techniques. Our results demonstrated that DDOST was significantly upregulated in a variety of tumor types and correlated with poor prognosis, including cervical cancer. Cox regression analysis dissected that high DDOST expression was associated with poor survival in cervical cancer patients. Immune infiltration analysis defined that DDOST was negatively correlated with CD8 T cells and NK cells. Strikingly, the sensitivity to multiple drugs was negatively correlated with the expression of DDOST. Therefore, our findings uncovered that DDOST could play an essential role in the tumor microenvironment and tumor immune regulation in cervical cancer, which indicated that DDOST could be a useful biomarker for prognosis and a potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Jie Mei
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Liuliu Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Min Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Dandan Bao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hui Gao
- Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| | - Danhan Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
12
|
Sheng B, Pan S, Ye M, Liu H, Zhang J, Zhao B, Ji H, Zhu X. Single-cell RNA sequencing of cervical exfoliated cells reveals potential biomarkers and cellular pathogenesis in cervical carcinogenesis. Cell Death Dis 2024; 15:130. [PMID: 38346944 PMCID: PMC10861450 DOI: 10.1038/s41419-024-06522-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: 04/29/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024]
Abstract
Cervical cancer (CC) is a common gynecological malignancy. Despite the current screening methods have been proved effectively and significantly decreased CC morbidity and mortality, deficiencies still exist. Single-cell RNA sequencing (scRNA-seq) approach can identify the complex and rare cell populations at single-cell resolution. By scRNA-seq, the heterogeneity of tumor microenvironment across cervical carcinogenesis has been mapped and described. Whether these alterations could be detected and applied to CC screening is unclear. Herein, we performed scRNA-seq of 56,173 cervical exfoliated cells from 15 samples, including normal cervix, low-grade squamous intraepithelial lesion (LSIL), high-grade squamous intraepithelial lesion (HSIL), and malignancy. The present study delineated the alteration of immune and epithelial cells derived during the cervical lesion progression. A subset of lipid-associated macrophage was identified as a tumor-promoting element and could serve as a biomarker for predicting the progression of LSIL into HSIL, which was then verified by immunofluorescence. Furthermore, cell-cell communication analysis indicated the SPP1-CD44 axis might exhibit a protumor interaction between epithelial cell and macrophage. In this study, we investigated the cervical multicellular ecosystem in cervical carcinogenesis and identified potential biomarkers for early detection.
Collapse
Affiliation(s)
- Bo Sheng
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuya Pan
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Miaomiao Ye
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hejing Liu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiamin Zhang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Zhao
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
13
|
Song G, Sun Z, Chu M, Zhang Z, Chen J, Wang Z, Zhu X. FBXO28 promotes cell proliferation, migration and invasion via upregulation of the TGF-beta1/SMAD2/3 signaling pathway in ovarian cancer. BMC Cancer 2024; 24:122. [PMID: 38267923 PMCID: PMC10807113 DOI: 10.1186/s12885-024-11893-8] [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: 08/24/2023] [Accepted: 01/17/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Ovarian cancer is one of the most common gynecological malignancies due to the lack of early symptoms, early diagnosis and limited screening. Therefore, it is necessary to understand the molecular mechanism underlying the occurrence and progression of ovarian cancer and to identify a basic biomarker for the early diagnosis and clinical treatment of ovarian cancer. METHODS The association between FBXO28 and ovarian cancer prognosis was analyzed using Kaplan‒Meier survival analysis. The difference in FBXO28 mRNA expression between normal ovarian tissues and ovarian tumor tissues was obtained from The Cancer Genome Atlas (TCGA), and Genotype-Tissue Expression (GTEx) cohorts. The expression levels of the FBXO28 protein in ovarian cancer tissues and normal ovarian tissues were measured via immunohistochemical staining. Western blotting was used to determine the level of FBXO28 expression in ovarian cancer cells. The CCK-8, the colony formation, Transwell migration and invasion assays were performed to evaluate cell proliferation and motility. RESULTS We found that a higher expression level of FBXO28 was associated with poor prognosis in ovarian cancer patients. Analysis of the TCGA and GTEx cohorts showed that the FBXO28 mRNA level was lower in normal ovarian tissue samples than in ovarian cancer tissue samples. Compared with that in normal ovarian tissues or cell lines, the expression of FBXO28 was greater in ovarian tumor tissues or tumor cells. The upregulation of FBXO28 promoted the viability, proliferation, migration and invasion of ovarian cancer cells. Finally, we demonstrated that FBXO28 activated the TGF-beta1/Smad2/3 signaling pathway in ovarian cancer. CONCLUSIONS In conclusion, FBXO28 enhanced oncogenic function via upregulation of the TGF-beta1/Smad2/3 signaling pathway in ovarian cancer.
Collapse
Affiliation(s)
- Gendi Song
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhengwei Sun
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China
| | - Man Chu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zihan Zhang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiajia Chen
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhiwei Wang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
14
|
Sheng B, Zhao B, Dong Y, Zhang J, Wu S, Ji H, Zhu X. Copine 1 predicts poor clinical outcomes by promoting M2 macrophage activation in ovarian cancer. Carcinogenesis 2023; 44:748-759. [PMID: 37747823 PMCID: PMC10773812 DOI: 10.1093/carcin/bgad067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/05/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVE Copine 1 (CPNE1), a membrane-binding protein, influences the prognosis of various cancers. According to cBioPortal, CPNE1 amplification is a prevalent genetic mutation in ovarian cancer but with unknown oncogenic mechanism. METHODS This study analysed the CPNE1 expression in ovarian cancer using online datasets, as validated by immunohistochemistry (IHC), quantitative polymerase chain reaction (qPCR) and western blotting. Concurrently, the prognostic value of CPNE1 was accessed. Cell Counting Kit-8, colony formation, transwells and xenograft experiments were performed to evaluate the functions of CPNE1 during ovarian cancer carcinogenesis. CPNE1 and its related genes were analysed by g:Profiler and Tumour Immune Estimation Resource. Furthermore, human monocytic THP-1 cells were co-cultured with ES2 cells to investigate the effect of CPNE1 on macrophage polarization. RESULTS The results of bioinformatic analysis, IHC, qPCR and western blotting indicated a higher CPNE1 in ovarian cancer. CPNE1 overexpression demonstrated an association with a poor prognosis of ovarian cancer. Functionally, CPNE1 overexpression increased ES2 and SKOV3 cell proliferation, invasion and migration in vitro and promoted ovarian tumour xenograft growth in vivo, while CPNE1 knockdown led to opposite effects. Additionally, CPNE1 expression demonstrated an association with immune cell infiltration in ovarian cancer, especially macrophage. CPNE1 promoted protumour M2 macrophage polarization by upregulating cluster of differentiation 163 (CD163), CD206 and interleukin-10. CONCLUSIONS Our study revealed that CPNE1 mediated M2 macrophage polarization and provided a therapeutic target for ovarian cancer.
Collapse
Affiliation(s)
- Bo Sheng
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Bo Zhao
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yue Dong
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jiamin Zhang
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Suni Wu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huihui Ji
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
15
|
Zhang L, Shi Z, Zhang F, Chen B, Qiu W, Cai L, Lin X. Ubiquitination-related biomarkers in metastatic melanoma patients and their roles in tumor microenvironment. Front Oncol 2023; 13:1170190. [PMID: 37274231 PMCID: PMC10235493 DOI: 10.3389/fonc.2023.1170190] [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: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 06/06/2023] Open
Abstract
Background Skin cutaneous melanoma (SKCM) is the deadliest type of cutaneous malignancy. Ubiquitination is a process of protein sorting and degradation that exhibits multiple functions in the progression of various tumors. This study aimed to characterize a set of genes for ubiquitination in SKCM. Methods The expression patterns of ubiquitin-associated genes (URGs) and the corresponding clinical information in SKCM tissues were comprehensively analyzed based on The Cancer Genome Atlas (TCGA) database. We performed univariate and multivariate Cox proportional regression models to characterize the risk scores and identify four critical genes related to prognostic ubiquitination (HCLS1, CORO1A, NCF1 and CCRL2), which were used to construct the prognostic signatures. We also studied the effects of HCLS1, CORO1A and CCRL2 on tumor metastasis-related indicators at the cellular level through in vitro experiments. Results SKCM patients in the low-risk group showing a longer survival than those in the high-risk group. Characteristic risk scores correlated with several clinicopathological variables and reflected the infiltration of multiple immune cells. In addition, the knockdown of CLS1, CORO1A and CCRL2 affected cellular malignant biological behavior through the EMT signaling pathway. Conclusion This study provides a novel and prospective strategy to improve the clinical survival of SKCM patients.
Collapse
|
16
|
Hou B, Chen T, Zhang H, Li J, Wang P, Shang G. The E3 ubiquitin ligases regulate PD-1/PD-L1 protein levels in tumor microenvironment to improve immunotherapy. Front Immunol 2023; 14:1123244. [PMID: 36733484 PMCID: PMC9887025 DOI: 10.3389/fimmu.2023.1123244] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
The tumor microenvironment (TME) is the tumor surrounding environment, which is critical for tumor development and progression. TME is also involved in clinical intervention and treatment outcomes. Modulation of TME is useful for improving therapy strategies. PD-L1 protein on tumor cells interacts with PD-1 protein on T cells, contributing to T cell dysfunction and exhaustion, blockage of the immune response. Evidence has demonstrated that the expression of PD-1/PD-L1 is associated with clinical response to anti-PD-1/PD-L1 therapy in cancer patients. It is important to discuss the regulatory machinery how PD-1/PD-L1 protein is finely regulated in tumor cells. In recent years, studies have demonstrated that PD-1/PD-L1 expression was governed by various E3 ubiquitin ligases in TME, contributing to resistance of anti-PD-1/PD-L1 therapy in human cancers. In this review, we will discuss the role and molecular mechanisms of E3 ligases-mediated regulation of PD-1 and PD-L1 in TME. Moreover, we will describe how E3 ligases-involved PD-1/PD-L1 regulation alters anti-PD-1/PD-L1 efficacy. Altogether, targeting E3 ubiquitin ligases to control the PD-1/PD-L1 protein levels could be a potential strategy to potentiate immunotherapeutic effects in cancer patients.
Collapse
Affiliation(s)
- Bo Hou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ting Chen
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jiatong Li
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui, China
| | - Guanning Shang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China,*Correspondence: Guanning Shang,
| |
Collapse
|
17
|
Dai J, Pan Y, Chen Y, Yao S. A panel of seven immune-related genes can serve as a good predictive biomarker for cervical squamous cell carcinoma. Front Genet 2022; 13:1024508. [PMID: 36406134 PMCID: PMC9667556 DOI: 10.3389/fgene.2022.1024508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 10/12/2022] [Indexed: 09/15/2023] Open
Abstract
Objective: Cervical cancer is one of the most common gynecological malignancies. The interaction between tumor microenvironment and immune infiltration is closely related to the progression of cervical squamous cell carcinoma (CSCC) and patients' prognosis. Herein, a panel of immune-related genes was established for more accurate prognostic prediction. Methods: The transcriptome information of tumor and normal samples were obtained from TCGA-CSCC and GTEx. Differentially expressed genes (DEGs) were defined from it. Immune-related genes (IRGs) were retrieved from the ImmPort database. After removing the transcriptome data which not mentioned in GSE44001, IR-DEGs were preliminarily identified. Then, TCGA-CSCC samples were divided into training and testing set (3:1) randomly. Univariate Cox analysis, LASSO regression analysis and multivariate Cox analysis were used in turn to construct the signature to predict the overall survival (OS) and disease-free survival (DFS). External validation was performed in GSE44001, and initial clinical validation was performed by qRT-PCR. Function enrichment analysis, immune infiltration analysis and establishment of nomogram were conducted as well. Results: A prognostic prediction signature consisting of seven IR-DEGs was established. High expression of NRP1, IGF2R, SERPINA3, TNF and low expression of ICOS, DES, HCK suggested that CSCC patients had shorter OS (POS<0.001) and DFS (PDFS<0.001). AUC values of 1-, 3-, five- year OS were 0.800, 0.831 and 0.809. Analyses in other validation sets showed good consistency with the results in training set. The signature can serve as an independent prognostic factor for OS (HR = 1.166, p < 0.001). AUC values of 1-, 3-, five- year OS based on the nomogram were 0.769, 0.820 and 0.807. Functional enrichment analysis suggested that these IR-DEGs were associated with receptor interaction and immune cell activity. Immune infiltration analysis indicated that patients in high-risk group had lower immune infiltration, weaker immune function, and were more likely to benefit from immune checkpoint inhibitor therapy. Through qRT-PCR on clinical samples, expression of NRP1, IGF2R, SERPINA3 and TNF were significantly upregulated in tumor tissue, while ICOS and DES were significantly downregulated. Conclusion: To conclude, the immune-related signature can provide strong support for exploration of immune infiltration, prediction of prognosis and response to immunotherapy through stratify CSCC patients into subgroups.
Collapse
Affiliation(s)
| | | | | | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|