1
|
Omer AAM, Kumar S, Söderquist B, Melik W, Bengtsson T, Khalaf H. PLNC8 αβ Potently Inhibits the Flavivirus Kunjin and Modulates Inflammatory and Intracellular Signaling Responses of Alveolar Epithelial Cells. Viruses 2024; 16:1770. [PMID: 39599884 PMCID: PMC11599086 DOI: 10.3390/v16111770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
PLNC8 αβ is a cationic antimicrobial peptide that previously has been reported to express both antibacterial and antiviral properties. This study aimed to further elucidate the antiviral effects of PLNC8 αβ and its impact on virus-induced cytotoxicity and inflammatory signaling in human alveolar epithelial cells (A549) infected with the flavivirus Kunjin. Complementary in silico analyses using molecular dynamics (MD) simulation were conducted to investigate the mechanism of action of PLNC8 αβ by studying the interaction of PLNC8 α and β with models of a flavivirus membrane and a eukaryotic plasma membrane, respectively. Our findings demonstrated that PLNC8 αβ significantly reduces both extracellular and intracellular viral loads, as confirmed by plaque reduction assays and RT-PCR. The peptide also mitigated virus-induced cytotoxicity and inflammation. Notably, PLNC8 αβ modulated the virus-induced dysregulation of key signaling and inflammatory genes, such as TLR9, TLR3, NOD2, FOS, JUN, IL6, and CXCL8. MD simulation revealed that PLNC8 αβ exhibits higher binding affinity for a flavivirus membrane model compared to a model of the plasma membrane, likely due to stronger electrostatic interactions with anionic phospholipids. This selective interaction possibly accounts for a potent antiviral activity of PLNC8 αβ combined with a minimal cytotoxicity toward human cells. Overall, PLNC8 αβ shows significant promise as an antiviral agent against flavivirus infections and warrants further exploration for peptide-based antiviral therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden; (A.A.M.O.); (S.K.); (B.S.); (W.M.); (T.B.)
| |
Collapse
|
2
|
Alghamdi SA, Alissa M, Alghamdi A, Alshehri MA, Albelasi A, Alzahrani KJ, Safhi AY. Interplays Between Matrix Metalloproteinases and Neurotropic Viruses: An Overview. Rev Med Virol 2024; 34:e2585. [PMID: 39349731 DOI: 10.1002/rmv.2585] [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: 08/01/2024] [Revised: 08/28/2024] [Accepted: 09/06/2024] [Indexed: 11/08/2024]
Abstract
Matrix metalloproteinases (MMPs) are a diverse group of proteases involved in various physiological and pathological processes through modulation of extracellular matrix (ECM) components, cytokines, and growth factors. In the central nervous system (CNS), MMPs play a major role in CNS development, plasticity, repair, and reorganisation contributing to learning, memory, and neuroimmune response to injury. MMPs are also linked to various neurological disorders such as Alzheimer's disease, Parkinson's disease, cerebral aneurysm, stroke, epilepsy, multiple sclerosis, and brain cancer suggesting these proteases as key regulatory factors in the nervous system. Moreover, MMPs have been involved in the pathogenesis of neurotropic viral infections via dysregulation of various cellular processes, which may highlight these factors as potential targets for the treatment and control of neurological complications associated with viral pathogens. This review provides an overview of the roles of MMPs in various physiological processes of the CNS and their interactions with neurotropic viral pathogens.
Collapse
Affiliation(s)
- Suad A Alghamdi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Alissa
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdullah Alghamdi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed A Alshehri
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdullah Albelasi
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia
| | - Khalid J Alzahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
3
|
Liang H, Tan W, Wang J, Li M, Pang H, Wang X, Yang L, Jing X. Novel prediction model combining PET/CT metabolic parameters, inflammation markers, and TNM stage: prospects for personalizing prognosis in nasopharyngeal carcinoma. Ann Nucl Med 2024; 38:802-813. [PMID: 38874876 DOI: 10.1007/s12149-024-01949-x] [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: 04/05/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE This study aims to develop a novel prediction model and risk stratification system that could accurately predict progression-free survival (PFS) in patients with nasopharyngeal carcinoma (NPC). METHODS Herein, we included 106 individuals diagnosed with NPC, who underwent 18F-FDG PET/CT scanning before treatment. They were divided into training (n = 76) and validation (n = 30) sets. The prediction model was constructed based on multivariate Cox regression analysis results and its predictive performance was evaluated. Risk factor stratification was performed based on the nomogram scores of each case, and Kaplan-Meier curves were used to evaluate the model's discriminative ability for high- and low-risk groups. RESULTS Multivariate Cox regression analysis showed that N stage, M stage, SUVmax, MTV, HI, and SIRI were independent factors affecting the prognosis of patients with NPC. In the training set, the model considerably outperformed the TNM stage in predicting PFS (AUCs of 0.931 vs. 0.841, 0.892 vs. 0.785, and 0.892 vs. 0.804 at 1-3 years, respectively). The calibration plots showed good agreement between actual observations and model predictions. The DCA curves further justified the effectiveness of the model in clinical practice. Between high- and low-risk group, 3-year PFS rates were significantly different (high- vs. low-risk group: 62.8% vs. 9.8%, p < 0.001). Adjuvant chemotherapy was also effective for prolonging survival in high-risk patients (p = 0.009). CONCLUSION Herein, a novel prediction model was successfully developed and validated to improve the accuracy of prognostic prediction for patients with NPC, with the aim of facilitating personalized treatment.
Collapse
Affiliation(s)
- Huan Liang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Weilin Tan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Mengdan Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Hua Pang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Xiaohui Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Lu Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China
| | - Xingguo Jing
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, No. 1 Youyi Road, YuanjiagangChongqing, 400016, China.
| |
Collapse
|
4
|
Huang Y, Bergant V, Grass V, Emslander Q, Hamad MS, Hubel P, Mergner J, Piras A, Krey K, Henrici A, Öllinger R, Tesfamariam YM, Dalla Rosa I, Bunse T, Sutter G, Ebert G, Schmidt FI, Way M, Rad R, Bowie AG, Protzer U, Pichlmair A. Multi-omics characterization of the monkeypox virus infection. Nat Commun 2024; 15:6778. [PMID: 39117661 PMCID: PMC11310467 DOI: 10.1038/s41467-024-51074-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Multiple omics analyzes of Vaccinia virus (VACV) infection have defined molecular characteristics of poxvirus biology. However, little is known about the monkeypox (mpox) virus (MPXV) in humans, which has a different disease manifestation despite its high sequence similarity to VACV. Here, we perform an in-depth multi-omics analysis of the transcriptome, proteome, and phosphoproteome signatures of MPXV-infected primary human fibroblasts to gain insights into the virus-host interplay. In addition to expected perturbations of immune-related pathways, we uncover regulation of the HIPPO and TGF-β pathways. We identify dynamic phosphorylation of both host and viral proteins, which suggests that MAPKs are key regulators of differential phosphorylation in MPXV-infected cells. Among the viral proteins, we find dynamic phosphorylation of H5 that influenced the binding of H5 to dsDNA. Our extensive dataset highlights signaling events and hotspots perturbed by MPXV, extending the current knowledge on poxviruses. We use integrated pathway analysis and drug-target prediction approaches to identify potential drug targets that affect virus growth. Functionally, we exemplify the utility of this approach by identifying inhibitors of MTOR, CHUK/IKBKB, and splicing factor kinases with potent antiviral efficacy against MPXV and VACV.
Collapse
Affiliation(s)
- Yiqi Huang
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Valter Bergant
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Vincent Grass
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Quirin Emslander
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - M Sabri Hamad
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Philipp Hubel
- Innate Immunity Laboratory, Max-Planck Institute of Biochemistry, Munich, Germany
- Core Facility Hohenheim, Universität Hohenheim, Stuttgart, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at University Hospital rechts der Isar (BayBioMS@MRI), Technical University of Munich, Munich, Germany
| | - Antonio Piras
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Karsten Krey
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Alexander Henrici
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Yonas M Tesfamariam
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Ilaria Dalla Rosa
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Till Bunse
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Department of Veterinary Sciences, LMU Munich, Munich, Germany
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany
| | - Gregor Ebert
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Florian I Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Michael Way
- Cellular signalling and cytoskeletal function laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Infectious Disease, Imperial College, London, UK
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics and Department of Medicine II, School of Medicine, Technical University of Munich, Munich, Germany
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ulrike Protzer
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany
- Institute of Virology, Technical University of Munich, School of Medicine/Helmholtz Munich, Munich, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany.
- German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany.
| |
Collapse
|
5
|
Balhara N, Yadav R, Ranga S, Ahuja P, Tanwar M. Understanding the HPV associated cancers: A comprehensive review. Mol Biol Rep 2024; 51:743. [PMID: 38874682 DOI: 10.1007/s11033-024-09680-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Human papillomavirus (HPV), a common cause of sexually transmitted diseases, may cause warts and lead to various types of cancers, which makes it important to understand the risk factors associated with it. HPV is the leading risk factor and plays a crucial role in the progression of cervical cancer. Viral oncoproteins E6 and E7 play a pivotal role in this process. Beyond cervical cancer, HPV-associated cancers of the mouth and throat are also increasing. HPV can also contribute to other malignancies like penile, vulvar, and vaginal cancers. Emerging evidence links HPV to these cancers. Research on the oncogenic effect of HPV is still ongoing and explorations of screening techniques, vaccination, immunotherapy and targeted therapeutics are all in progress. The present review offers valuable insight into the current understanding of the role of HPV in cancer and its potential implications for treatment and prevention in the future.
Collapse
Affiliation(s)
- Nikita Balhara
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Ritu Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India.
| | - Shalu Ranga
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Parul Ahuja
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Mukesh Tanwar
- Department of Genetics, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| |
Collapse
|
6
|
Calistri NL, Liby TA, Hu Z, Zhang H, Dane M, Gross SM, Heiser LM. TNBC response to paclitaxel phenocopies interferon response which reveals cell cycle-associated resistance mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.596911. [PMID: 38895265 PMCID: PMC11185620 DOI: 10.1101/2024.06.04.596911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Paclitaxel is a standard of care neoadjuvant therapy for patients with triple negative breast cancer (TNBC); however, it shows limited benefit for locally advanced or metastatic disease. Here we used a coordinated experimental-computational approach to explore the influence of paclitaxel on the cellular and molecular responses of TNBC cells. We found that escalating doses of paclitaxel resulted in multinucleation, promotion of senescence, and initiation of DNA damage induced apoptosis. Single-cell RNA sequencing (scRNA-seq) of TNBC cells after paclitaxel treatment revealed upregulation of innate immune programs canonically associated with interferon response and downregulation of cell cycle progression programs. Systematic exploration of transcriptional responses to paclitaxel and cancer-associated microenvironmental factors revealed common gene programs induced by paclitaxel, IFNB, and IFNG. Transcription factor (TF) enrichment analysis identified 13 TFs that were both enriched based on activity of downstream targets and also significantly upregulated after paclitaxel treatment. Functional assessment with siRNA knockdown confirmed that the TFs FOSL1, NFE2L2 and ELF3 mediate cellular proliferation and also regulate nuclear structure. We further explored the influence of these TFs on paclitaxel-induced cell cycle behavior via live cell imaging, which revealed altered progression rates through G1, S/G2 and M phases. We found that ELF3 knockdown synergized with paclitaxel treatment to lock cells in a G1 state and prevent cell cycle progression. Analysis of publicly available breast cancer patient data showed that high ELF3 expression was associated with poor prognosis and enrichment programs associated with cell cycle progression. Together these analyses disentangle the diverse aspects of paclitaxel response and identify ELF3 upregulation as a putative biomarker of paclitaxel resistance in TNBC.
Collapse
Affiliation(s)
- Nicholas L Calistri
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Tiera A. Liby
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Zhi Hu
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Hongmei Zhang
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Mark Dane
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Sean M. Gross
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
| | - Laura M. Heiser
- Biomedical Engineering Department, Oregon Health & Science University, Portland Oregon
- Knight Cancer Institute, Oregon Health & Science University, Portland Oregon
| |
Collapse
|
7
|
Akbari E, Milani A, Seyedinkhorasani M, Bolhassani A. HPV co-infections with other pathogens in cancer development: A comprehensive review. J Med Virol 2023; 95:e29236. [PMID: 37997472 DOI: 10.1002/jmv.29236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/13/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
High-risk human papillomaviruses (HR-HPVs) cause various malignancies in the anogenital and oropharyngeal regions. About 70% of cervical and oropharyngeal cancers are caused by HPV types 16 and 18. Notably, some viruses including herpes simplex virus, Epstein-Barr virus, and human immunodeficiency virus along with various bacteria often interact with HPV, potentially impacting its replication, persistence, and cancer progression. Thus, HPV infection can be significantly influenced by co-infecting agents that influence infection dynamics and disease progression. Bacterial co-infections (e.g., Chlamydia trachomatis) along with bacterial vaginosis-related species also interact with HPV in genital tract leading to viral persistence and disease outcomes. Co-infections involving HPV and diverse infectious agents have significant implications for disease transmission and clinical progression. This review explores multiple facets of HPV infection encompassing the co-infection dynamics with other pathogens, interaction with the human microbiome, and its role in disease development.
Collapse
Affiliation(s)
- Elahe Akbari
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Milani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
8
|
von Stromberg K, Seddar L, Ip WH, Günther T, Gornott B, Weinert SC, Hüppner M, Bertzbach LD, Dobner T. The human adenovirus E1B-55K oncoprotein coordinates cell transformation through regulation of DNA-bound host transcription factors. Proc Natl Acad Sci U S A 2023; 120:e2310770120. [PMID: 37883435 PMCID: PMC10622919 DOI: 10.1073/pnas.2310770120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/13/2023] [Indexed: 10/28/2023] Open
Abstract
The multifunctional adenovirus E1B-55K oncoprotein can induce cell transformation in conjunction with adenovirus E1A gene products. Previous data from transient expression studies and in vitro experiments suggest that these growth-promoting activities correlate with E1B-55K-mediated transcriptional repression of p53-targeted genes. Here, we analyzed genome-wide occupancies and transcriptional consequences of species C5 and A12 E1B-55Ks in transformed mammalian cells by combinatory ChIP and RNA-seq analyses. E1B-55K-mediated repression correlates with tethering of the viral oncoprotein to p53-dependent promoters via DNA-bound p53. Moreover, we found that E1B-55K also interacts with and represses transcription of numerous p53-independent genes through interactions with transcription factors that play central roles in cancer and stress signaling. Our results demonstrate that E1B-55K oncoproteins function as promiscuous transcriptional repressors of both p53-dependent and -independent genes and further support the model that manipulation of cellular transcription is central to adenovirus-induced cell transformation and oncogenesis.
Collapse
Affiliation(s)
| | - Laura Seddar
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Wing-Hang Ip
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Thomas Günther
- Virus Genomics, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Britta Gornott
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Sophie-Celine Weinert
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Max Hüppner
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Luca D. Bertzbach
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| | - Thomas Dobner
- Department of Viral Transformation, Leibniz Institute of Virology, Hamburg20251, Germany
| |
Collapse
|
9
|
Lyu J, Wang W, Chen B, Wang Y, Sun H, Yuan Y, Ren D. Key Genes and Endoscopic Radical Surgery in Primary Middle Ear Oncocytic Papillomas. Otol Neurotol 2023; 44:e676-e680. [PMID: 37604509 DOI: 10.1097/mao.0000000000003993] [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: 08/23/2023]
Abstract
OBJECTIVE Papillomas originating from the Schneiderian epithelium within the middle ear are extremely rare and may be associated with a high rate of recurrence and malignant transformation. Oncocytic papillomas represent the rarest pathological subtype of such tumors. The current investigation aimed to determine whether there exists a distinct mechanism underlying the incidence of oncocytic papillomas arising primarily within the middle ear, and to explore potential treatment strategies to ensure complete removal and prevent recurrence. STUDY DESIGN Search of the English literature for cases of middle ear papilloma and RNA sequencing analysis of three samples from one new case presenting at the Eye and ENT Hospital, Fudan University (Shanghai, China), with recurrent middle ear oncocytic papilloma, along with two normal mucosal samples. SETTING Academic, tertiary referral hospital. PATIENT AND INTERVENTIONS The patient underwent open mastoidectomy and endoscopic tympanoplasty twice in 6 years. Histopathology confirmed oncocytic papilloma in middle ear. The patient has been free of the disease at 18 months of follow-up without radiation, whereas the RNA-seq analysis of the samples in endoscopic operations remained nonmalignant. RESULTS Only four cases of primary middle ear oncocytic papillomas have been reported. Recurrent masses usually originate from around the eustachian tube, which may explain the pathogenesis of this lesion. RNA-seq analysis was used to identify 1,317 (UP, 239; DOWN, 1078) differentially expressed genes between papillomas and normal mucosa. The involvement of some hub proteins (e.g., FN1, CXCL8, L10, JUN, and FOS) in the pathogenesis of primary middle ear papillomas was found to align with the observed clinical features. CONCLUSION The middle ear oncocytic papillomas were extremely rare and remained incompletely understood. The findings of this first RNA-seq analysis of this rare tumor may serve to enhance comprehension of and aid in the management of middle ear papillomas.
Collapse
Affiliation(s)
| | - Wei Wang
- Department of Otolaryngology, The People's Hospital of Danyang; Affiliated Danyang Hospital of Nantong University, Danyang, Jiangsu Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
10
|
Thiyagarajah K, Basic M, Hildt E. Cellular Factors Involved in the Hepatitis D Virus Life Cycle. Viruses 2023; 15:1687. [PMID: 37632029 PMCID: PMC10459925 DOI: 10.3390/v15081687] [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: 06/30/2023] [Revised: 07/30/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Hepatitis D virus (HDV) is a defective RNA virus with a negative-strand RNA genome encompassing less than 1700 nucleotides. The HDV genome encodes only for one protein, the hepatitis delta antigen (HDAg), which exists in two forms acting as nucleoproteins. HDV depends on the envelope proteins of the hepatitis B virus as a helper virus for packaging its ribonucleoprotein complex (RNP). HDV is considered the causative agent for the most severe form of viral hepatitis leading to liver fibrosis/cirrhosis and hepatocellular carcinoma. Many steps of the life cycle of HDV are still enigmatic. This review gives an overview of the complete life cycle of HDV and identifies gaps in knowledge. The focus is on the description of cellular factors being involved in the life cycle of HDV and the deregulation of cellular pathways by HDV with respect to their relevance for viral replication, morphogenesis and HDV-associated pathogenesis. Moreover, recent progress in antiviral strategies targeting cellular structures is summarized in this article.
Collapse
Affiliation(s)
| | | | - Eberhard Hildt
- Paul-Ehrlich-Institute, Department of Virology, D-63225 Langen, Germany; (K.T.); (M.B.)
| |
Collapse
|
11
|
Fazlalipour M, Ghoreshi ZAS, Molaei HR, Arefinia N. The Role of DNA Viruses in Human Cancer. Cancer Inform 2023; 22:11769351231154186. [PMID: 37363356 PMCID: PMC10286548 DOI: 10.1177/11769351231154186] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/03/2023] [Indexed: 06/28/2023] Open
Abstract
This review discusses the possible involvement of infections-associated cancers in humans, with virus infections contributing 15% to 20% of total cancer cases in humans. DNA virus encoded proteins interact with host cellular signaling pathways and control proliferation, cell death and genomic integrity viral oncoproteins are known to bind cellular Deubiquitinates (DUBs) such as cyclindromatosis tumor suppressor, ubiquitin-specific proteases 7, 11, 15 and 20, and A-20 to improve their intracellular stability and cellular signaling pathways and finally transformation. Human papillomaviruses (cervical carcinoma, oral cancer and laryngeal cancer); human polyomaviruses (mesotheliomas, brain tumors); Epstein-Barr virus (B-cell lymphoproliferative diseases and nasopharyngeal carcinoma); Kaposi's Sarcoma Herpesvirus (Kaposi's Sarcoma and primary effusion lymphomas); hepatitis B (hepatocellular carcinoma (HCC)) cause up to 20% of malignancies around the world.
Collapse
Affiliation(s)
- Mehdi Fazlalipour
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran (IPI), Tehran, Iran
- Research Center for Emerging and Reemerging Infectious diseases, Pasteur Institute of Iran (IPI), Tehran, Iran
| | | | - Hamid Reza Molaei
- Department of Medical Bacteriology and Virology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Nasir Arefinia
- Student Research Committee, Jiroft University of Medical Sciences, Jiroft, Iran
| |
Collapse
|
12
|
Desrochers GF, Filip R, Bastianelli M, Stern T, Pezacki JP. microRNA-27b regulates hepatic lipase enzyme LIPC and reduces triglyceride degradation during hepatitis C virus infection. J Biol Chem 2022; 298:101983. [PMID: 35483451 PMCID: PMC9163519 DOI: 10.1016/j.jbc.2022.101983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 11/26/2022] Open
Abstract
miRNAs are short, noncoding RNAs that negatively and specifically regulate protein expression, the cumulative effects of which can result in broad changes to cell systems and architecture. The miRNA miR-27b is known to regulate lipid regulatory pathways in the human liver and is also induced by the hepatitis C virus (HCV). However, the functional targets of miR-27b are not well established. Herein, an activity-based protein profiling method using a serine hydrolase probe, coupled with stable isotope labeling and mass spectrometry identified direct and indirect targets of miR-27b. The hepatic lipase C (LIPC) stood out as both highly dependent on miR-27b and as a major modulator of lipid pathway misregulation. Modulation of miR-27b using both exogenous miRNA mimics and inhibitors demonstrated that transcription factors Jun, PPARα, and HNF4α, all of which also influence LIPC levels and activity, are regulated by miR-27b. LIPC was furthermore shown to affect the progress of the life cycle of HCV and to decrease levels of intracellular triglycerides, upon which HCV is known to depend. In summary, this work has demonstrated that miR-27b mediates HCV infection by downregulating LIPC, thereby reducing triglyceride degradation, which in turn increases cellular lipid levels.
Collapse
Affiliation(s)
| | - Roxana Filip
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
| | - Micheal Bastianelli
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Tiffany Stern
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
13
|
Exploring New Functional Aspects of HTLV-1 RNA-Binding Protein Rex: How Does Rex Control Viral Replication? Viruses 2022; 14:v14020407. [PMID: 35216000 PMCID: PMC8877913 DOI: 10.3390/v14020407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/01/2022] [Accepted: 02/08/2022] [Indexed: 01/27/2023] Open
Abstract
After integration to the human genome as a provirus, human T-cell leukemia virus type 1 (HTLV-1) utilizes host T cell gene expression machinery for viral replication. The viral RNA-binding protein, Rex, is known to transport unspliced/incompletely spliced viral mRNAs encoding viral structural proteins out of the nucleus to enhance virus particle formation. However, the detailed mechanism of how Rex avoids extra splicing of unspliced/incompletely spliced viral mRNAs and stabilizes them for effective translation is still unclear. To elucidate the underlying molecular mechanism of Rex function, we comprehensively analyzed the changes in gene expression and splicing patterns in Rex-overexpressing T cells. In addition, we identified 81 human proteins interacting with Rex, involved in transcription, splicing, translation, and mRNA quality control. In particular, Rex interacts with NONO and SFPQ, which play important roles in the regulation of transcription and splicing. Accordingly, expression profiles and splicing patterns of a wide variety of genes are significantly changed in Rex-expressing T cells. Especially, the level of vPD-L1 mRNA that lacks the part of exon 4, thus encodes soluble PD-L1 was significantly increased in Rex-expressing cells. Overall, by integrated analysis of these three datasets, we showed for the first time that Rex intervenes the host gene expression machinery throughout the pathway, probably to escort viral unstable mRNAs from transcription (start) to translation (end). Upon exerting its function, Rex may alter the expression level and splicing patterns of various genes, thus influencing the phenotype of the host cell.
Collapse
|
14
|
The Involvement of Natural Polyphenols in the Chemoprevention of Cervical Cancer. Int J Mol Sci 2021; 22:ijms22168812. [PMID: 34445518 PMCID: PMC8396230 DOI: 10.3390/ijms22168812] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/25/2022] Open
Abstract
From all types of cancer, cervical cancer manages to be in top four most frequent types, with a 6.5% rate of occurrence. The infectious vector that induces the disease, the high-risk Human papillomavirus (HPV), which is a sexually transmitted virus, is capable of transforming the host cell by modulating some of the principal signaling pathways responsible for cell cycle arrest, proliferation, and survival. Fortunately, like other cancer types, cervical cancer can be treated by chirurgical interventions or chemoradiotherapy, but these methods are not exactly the lucky clover of modern medicine because of the adverse effects they have. That is the reason why in the last years the emphasis has been on alternative medicine, more specifically on phytochemicals, as a substantial number of studies showed that diet contributes to cancer prevention and treatment. All these studies are trying to find new chemopreventive agents with less toxicity but high effectiveness both in vitro and in vivo. The aim of this review is to evaluate the literature in order to underline the advantages and disadvantages of polyphenols, a class of dietary compounds, as chemopreventive and chemotherapeutic agents. This review also aims to present polyphenols from different perspectives, starting with mechanisms of action and ending with their toxicity. The bigger picture illustrates that polyphenols have great potential in cervical cancer prevention, with strong effects on gene modulation.
Collapse
|
15
|
Wang Z, Qiao Y, Chen Z, Liang Y, Cui L, Zhang Y, Li X, Xu L, Wei P, Liu S, Li H. Fos Facilitates Gallid Alpha-Herpesvirus 1 Infection by Transcriptional Control of Host Metabolic Genes and Viral Immediate Early Gene. Viruses 2021; 13:v13061110. [PMID: 34207926 PMCID: PMC8229045 DOI: 10.3390/v13061110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022] Open
Abstract
Gallid alpha-herpesvirus 1, also known as avian infectious laryngotracheitis virus (ILTV), continues to cause huge economic losses to the poultry industry worldwide. Similar to that of other herpesvirus-encoded proteins, the expression of viral genes encoded by ILTV is regulated by a cascade, and the underlying regulatory mechanism remains largely unclear. The viral immediate-early (IE) gene ICP4 plays a prominent role in the initiation of the transcription of early and late genes during ILTV replication. In this study, we identified AP-1 as the key regulator of the transcription of ILTV genes by bioinformatics analysis of genome-wide transcriptome data. Subsequent functional studies of the key members of the AP-1 family revealed that Fos, but not Jun, regulates ILTV infection through AP-1 since knockdown of Fos, but not Jun, by gene silencing significantly reduced ICP4 transcription and subsequent viral genome replication and virion production. Using several approaches, we identified ICP4 as a bona fide target gene of Fos that regulated Fos and has Fos response elements within its promoter. Neither the physical binding of Jun to the promoter of ICP4 nor the transcriptional activity of Jun was observed. In addition, knockdown of Fos reduced the transcription of MDH1 and ATP5A1, genes encoding two host rate-limiting enzymes essential for the production of the TCA intermediates OAA and ATP. The biological significance of the transcriptional regulation of MDH1 and ATP5A1 by Fos in ILTV infection was supported by the fact that anaplerosis of OAA and ATP rescued both ICP4 transcription and virion production in infected cells under when Fos was silenced. Our study identified the transcription factor Fos as a key regulator of ILTV infection through its transcription factor function on both the virus and host sides, improving the current understanding of both avian herpesvirus–host interactions and the roles of AP-1 in viral infection.
Collapse
Affiliation(s)
- Zhitao Wang
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yangyang Qiao
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Zhijie Chen
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yumeng Liang
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Lu Cui
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Yanhui Zhang
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Xuefeng Li
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Li Xu
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
| | - Ping Wei
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Correspondence: (P.W.); (S.L.); (H.L.); Tel.: +86-451-51051700 (H.L.)
| | - Shengwang Liu
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
- Correspondence: (P.W.); (S.L.); (H.L.); Tel.: +86-451-51051700 (H.L.)
| | - Hai Li
- State Key Laboratory of Veterinary Biotechnology, National Poultry Laboratory Animal Resource Center, Harbin Veterinary Research Institute, the Chinese Academy of Agricultural Sciences, Harbin 150069, China; (Z.W.); (Y.Q.); (Z.C.); (Y.L.); (L.C.); (Y.Z.); (X.L.); (L.X.)
- Correspondence: (P.W.); (S.L.); (H.L.); Tel.: +86-451-51051700 (H.L.)
| |
Collapse
|
16
|
Yang K, Dong W. SIRT1-Related Signaling Pathways and Their Association With Bronchopulmonary Dysplasia. Front Med (Lausanne) 2021; 8:595634. [PMID: 33693011 PMCID: PMC7937618 DOI: 10.3389/fmed.2021.595634] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic and debilitating disease that can exert serious and overwhelming effects on the physical and mental health of premature infants, predominantly due to intractable short- and long-term complications. Oxidative stress is one of the most predominant causes of BPD. Hyperoxia activates a cascade of hazardous events, including mitochondrial dysfunction, uncontrolled inflammation, reduced autophagy, increased apoptosis, and the induction of fibrosis. These events may involve, to varying degrees, alterations in SIRT1 and its associated targets. In the present review, we describe SIRT1-related signaling pathways and their association with BPD. Our intention is to provide new insights into the molecular mechanisms that regulate BPD and identify potential therapeutic targets for this debilitating condition.
Collapse
Affiliation(s)
- Kun Yang
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenbin Dong
- Department of Newborn Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
17
|
Rasi Bonab F, Baghbanzadeh A, Ghaseminia M, Bolandi N, Mokhtarzadeh A, Amini M, Dadashzadeh K, Hajiasgharzadeh K, Baradaran B, Bannazadeh Baghi H. Molecular pathways in the development of HPV-induced cervical cancer. EXCLI JOURNAL 2021; 20:320-337. [PMID: 33746665 PMCID: PMC7975633 DOI: 10.17179/excli2021-3365] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Recently, human papillomavirus (HPV) has gained considerable attention in cervical cancer research studies. It is one of the most important sexually transmitted diseases that can affect 160 to 289 out of 10000 persons every year. Due to the infectious nature of this virus, HPV can be considered a serious threat. The knowledge of viral structure, especially for viral oncoproteins like E6, E7, and their role in causing cancer is very important. This virus has different paths (PI3K/Akt, Wnt/β-catenin, ERK/MAPK, and JAK/STAT) that are involved in the transmission of signaling paths through active molecules like MEK (pMEK), ERK (pERK), and Akt (pAkt). It's eventually through these paths that cancer is developed. Precise knowledge of these paths and their signals give us the prognosis to adopt appropriate goals for prevention and control of these series of cancer.
Collapse
Affiliation(s)
- Farnaz Rasi Bonab
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Laboratory Sciences, Marand Branch, Islamic Azad University, Marand, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moslem Ghaseminia
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kianoosh Dadashzadeh
- Department of Laboratory Sciences, Marand Branch, Islamic Azad University, Marand, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Gammacoronavirus Avian Infectious Bronchitis Virus and Alphacoronavirus Porcine Epidemic Diarrhea Virus Exploit a Cell-Survival Strategy via Upregulation of cFOS to Promote Viral Replication. J Virol 2021; 95:JVI.02107-20. [PMID: 33239458 PMCID: PMC7851560 DOI: 10.1128/jvi.02107-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Coronaviruses have evolved a variety of strategies to optimize cellular microenvironment for efficient replication. In this study, we report the induction of AP-1 transcription factors by coronavirus infection based on genome-wide analyses of differentially expressed genes in cells infected with avian coronavirus infectious bronchitis virus (IBV). Most members of the AP-1 transcription factors were subsequently found to be upregulated during the course of IBV and porcine epidemic diarrhea virus (PEDV) infection of cultured cells as well as in IBV-infected chicken embryos. Further characterization of the induction kinetics and functional roles of cFOS in IBV replication demonstrated that upregulation of cFOS at early to intermediate phases of IBV replication cycles suppresses IBV-induced apoptosis and promotes viral replication. Blockage of nuclear translocation of cFOS by peptide inhibitor NLSP suppressed IBV replication and apoptosis, ruling out the involvement of the cytoplasmic functions of cFOS in the replication of IBV. Furthermore, knockdown of ERK1/2 and inhibition of JNK and p38 kinase activities reduced cFOS upregulation and IBV replication. This study reveals an important function of cFOS in the regulation of coronavirus-induced apoptosis, facilitating viral replication.IMPORTANCE The ongoing pandemic of coronavirus disease 2019 (COVID-19), caused by a newly emerged zoonotic coronavirus (SARS-CoV-2), highlights the importance of coronaviruses as human and animal pathogens and our knowledge gaps in understanding the cellular mechanisms, especially mechanisms shared among human and animal coronaviruses, exploited by coronaviruses for optimal replication and enhanced pathogenicity. This study reveals that upregulation of cFOS, along with other AP-1 transcription factors, as a cell-survival strategy is such a mechanism utilized by coronaviruses during their replication cycles. Through induction and regulation of apoptosis of the infected cells at early to intermediate phases of the replication cycles, subtle but appreciable differences in coronavirus replication efficiency were observed when the expression levels of cFOS were manipulated in the infected cells. As the AP-1 transcription factors are multi-functional, further studies of their regulatory roles in proinflammatory responses may provide new insights into the pathogenesis and virus-host interactions during coronavirus infection.
Collapse
|
19
|
Gao F, Zhou L, Li M, Liu W, Yang S, Li W. Inhibition of ERKs/Akt-Mediated c-Fos Expression Is Required for Piperlongumine-Induced Cyclin D1 Downregulation and Tumor Suppression in Colorectal Cancer Cells. Onco Targets Ther 2020; 13:5591-5603. [PMID: 32606774 PMCID: PMC7304781 DOI: 10.2147/ott.s251295] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Background Deregulation of Cyclin D1 and cell cycle progression plays a critical role in tumorigenesis. The natural compound piperlongumine (PL) exhibits potential anticancer effects in various cancer models, but the underlying mechanism needs further elucidation. Methods The inhibitory effect of PL on colorectal cancer (CRC) cells was determined by anchorage-dependent and -independent assays. The protein level of Cyclin D1 was examined by immunoblot (IB) and immunohistochemical staining (IHC). The mRNA level was determined by qRT-PCR. Phosphorylation of histone H3 was analyzed by immunofluorescence (IF). The cell cycle was examined by flow cytometry. The in vivo antitumor effect was validated by the xenograft mouse model. Results Cyclin D1 was overexpressed in CRC tissues and cells, and was required for maintaining cell growth, colony formation, and in vivo tumorigenesis. PL decreased the protein level of c-Fos, which eventually reduced the transcriptional activity of AP-1 and the mRNA level of Cyclin D1. Mechanism study showed that PL impaired EGF-induced activation of ERK1/2 and Akt signalings, which resulted in a reduction of c-Fos transcription. Furthermore, PL reduced the half-life of c-Fos and caused the ubiquitination-dependent degradation of c-Fos. Finally, the in vivo antitumor effect of PL on CRC cells was examined using a xenograft mouse model. Conclusion Our data indicate that PL is a promising antitumor agent that deserves further study for CRC treatment.
Collapse
Affiliation(s)
- Feng Gao
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China.,Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Li Zhou
- Department of Pathology, Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Ming Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China.,Changsha Stomatological Hospital, Changsha, Hunan 410004, People's Republic of China.,School of Stomatology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China
| | - Wenbin Liu
- Department of Pathology, Hunan Cancer Hospital, Changsha, Hunan 410013, People's Republic of China
| | - Shuting Yang
- Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Wei Li
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China.,Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| |
Collapse
|