1
|
Lai M, Lai R, He B, Wang X, Chen L, Mo Q. Robust antiviral innate immune response and miRNA regulatory network were identified in ZIKV-infected cells: implications in the pathogenesis of ZIKV infection. Virus Genes 2025; 61:249-264. [PMID: 39955676 DOI: 10.1007/s11262-025-02136-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
Zika virus (ZIKV) infection has emerged as a significant public health concern due to its association with fetal microcephaly and Guillain-Barre syndrome (GBS). Unfortunately, its detailed pathogenesis remains unclear. To better understand how ZIKV evades host antiviral immunity, we analyzed the microarray dataset (GSE98889) of ZIKV-infected primary human brain microvascular endothelial cells (hBMECs) retrieved from the gene expression omnibus (GEO). 160, 1423, 969, 829, and 600 differentially expressed genes (DEGs) were identified at 12, 24, 48, 72, and 216 hours post-ZIKV infection in hBMECs, respectively. Subsequently, 31 common DEGs across all time-points were selected for further analysis. Gene ontology (GO) functional analysis showed these 31 DEGs were mainly involved in the host antiviral innate immune responses. Protein-protein interaction (PPI) network analysis identified 10 hub genes (MX1, OAS1, OAS2, IFI44, IFI44L, IFIT1, IFIT2, IFIT3, IFIH1, and XAF1), which were all interferon-stimulated genes (ISGs) and upregulated. qRT-PCR was used to validate the expression patterns of these 10 hub genes in different ZIKV-infected cell lines. Finally, miRNA-mRNA regulatory network analysis revealed that hsa-miR-129-2-3p, hsa-miR-138-5p, hsa-miR-21-3p, hsa-miR-27a-5p, hsa-miR-449a, and hsa-miR449b-5p were key miRNAs regulating these hub genes. Our study showed that ZIKV infection activated the host innate immune response to restrict ZIKV infection. The common pathways, hub genes, and their regulatory miRNA network offer new insights into virus-host interactions, enhancing our understanding of ZIKV pathogenesis.
Collapse
Affiliation(s)
- Mingshuang Lai
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Rongji Lai
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Baoren He
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Xinwei Wang
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
| | - Limin Chen
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, China
| | - Qiuhong Mo
- The Joint Laboratory On Transfusion-Transmitted Diseases (TTDs) Between Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Nanning Blood Center, Nanning Blood Center, Nanning, China.
| |
Collapse
|
2
|
Cao J, Zhang H, Zhang J, Wang J, Li C, Ma J, Ye Z, Zheng Y, Liu H, Xiao G, Dai W, Zhang L. Screening of botanical drugs reveals the potential anti-human adenovirus activity of berberine. Antiviral Res 2025; 237:106105. [PMID: 39956272 DOI: 10.1016/j.antiviral.2025.106105] [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: 10/11/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Human adenovirus (HAdV) is a significant viral pathogen that causes severe acute respiratory infections (SARIs) in children and immunocompromised patients. Currently, no specific treatment options are available for HAdV infections. This study used a green fluorescence protein-based, high-throughput screening (HTS) assay on a botanical drug library containing 3697 botanical compounds to identify agents that could inhibit HAdV. Four compounds with anti-HAdV-C5 activity in the low-micromolar range were identified and inhibited other wild-type HAdVs known to cause SARIs. Among these compounds, 13-methylberberine chloride presented the highest select index values. Berberine is a commercially available natural product-derived isoquinoline alkaloid with multiple pharmacological effects and is widely used in Asian countries. We systematically evaluated the anti-HAdV activity of six berberine-derived compounds in vitro and performed a time-of-drug-addition assay to explore their antiviral modes of action. Mechanistic studies revealed that berberine and its analogs inhibit HAdV replication by downregulating the MAPK signaling pathway, particularly ERK activation, which is crucial for viral replication and progeny production. Our findings suggest that berberine is a promising candidate for the development of anti-HAdV therapies.
Collapse
Affiliation(s)
- Junyuan Cao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China
| | - Hao Zhang
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Jixiang Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Jinlin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chen Li
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Jin Ma
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Zhengyu Ye
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunting Zheng
- Hubei Jiangxia Laboratory, Wuhan, 430200, China; HuaWu Biotech Wuhan, 430200, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wenhao Dai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China; University of the Chinese Academy of Sciences, Beijing, 100049, China; Hubei Jiangxia Laboratory, Wuhan, 430200, China.
| |
Collapse
|
3
|
Pawan, Devi S. Designing of new trans-stilbene derivative: An entry barrier of Zika virus in host cell. J Mol Graph Model 2025; 135:108935. [PMID: 39731815 DOI: 10.1016/j.jmgm.2024.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
A large population in the world lives in tropical and subtropical regions, showing a high risk of Zika viral infection which leads to a situation of global health emergency and demands extensive research to create effective antiviral medicines. Herein, we introduce the design of a new derivatized trans-stilbene molecule to investigate the inhibition of Zika virus entry into the host cell by molecular docking approach. The synthesized compound has been characterized by different analytical techniques such as FTIR, 1H NMR,13C NMR and UV-visible spectroscopy as well as Mass spectrometry (MS). Moreover, the complete structure elucidation was achieved via X-ray crystallography and DFT analysis. The article describes the life cycle and genome of the Zika virus along with its mechanism of entry inhibition by illustrating the structure and function of the ZIKV envelop (E) protein. The docking studies disclosed that the newly synthesized stilbene compound confers an excellent inhibitory response towards the entry of Zika virus in host cells as supported by calculated docking score and its binding conformation with Zika virus E-protein. Further, the normal mode analysis (NMA) simulation technique is used to predict the conformational states of the target E-protein, which explains the potency of the compound to bind with the Zika virus E-protein. We hope that the present study will help and encourage researchers in the field of medicinal chemistry to develop potential drugs against the Zika virus.
Collapse
Affiliation(s)
- Pawan
- Department of Chemistry, Goswami Ganesh Dutta Sanatan Dharma (GGDSD) College, Chandigarh, 160030, India.
| | - Sonia Devi
- Post Graduate Department of Chemistry, Mehr Chand Mahajan DAV College for Women, Chandigarh, 160036, India
| |
Collapse
|
4
|
Zhang J, Yang Y, Wang Y, Shao X, Wang S, Bao B, Chen Y, Wang F, Han D. Identification of Cells Eliminating and Harboring Mumps Viruses in the Male Reproductive System. J Med Virol 2025; 97:e70250. [PMID: 40008432 DOI: 10.1002/jmv.70250] [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: 07/23/2024] [Revised: 01/16/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
A large spectrum of viruses can infect the male reproductive system (MRS). Although the MRS adopts the local antiviral defense, it has also been considered as a potential viral reservoir. We hypothesize that tissue-specific cells may eliminate invading viruses and harbor viral replication in the MRS. We herein aimed to identify viral eliminators and potential reservoirs in the MRS using mumps virus (MuV) and mouse models. Primary cells, mainly consisting of epithelial cells, stromal cells, and macrophages, were isolated from the epididymis, seminal vesicle, and prostate of mice. MuV infection and replication were analyzed by determining MuV nucleoprotein (MuV-NP) in the primary cells. We demonstrated that a subset of resident macrophages efficiently took up and eliminated MuV, which should be involved in the antiviral defense in the MRS. However, a small subset of epithelial and stromal cells in these organs harbored MuV replication, and these cells could be viral reservoirs. Furthermore, interferon-β (IFN-β) inhibited MuV replication in MuV-replicating cells, suggesting that IFN-β administration may limit viral reservoirs. The results of the present study provide novel insights into the antiviral defense and viral reservoirs in the MRS, which can aid in the development of preventive and therapeutic approaches for viral infection of the MRS.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yixuan Yang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yu Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xinyi Shao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Siqi Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Binghao Bao
- Department of Andrology, China-Japan Friendship Hospital, Beijing, China
| | - Yongmei Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Fei Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Daishu Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Siemer JL, Le TT, Paul A, Boykin DW, Brinton MA, Wilson WD, Germann MW. Conserved Sequences from Dengue Virus Genomes Form Stable G-Quadruplexes. ACS Infect Dis 2025; 11:88-94. [PMID: 39666861 PMCID: PMC11731295 DOI: 10.1021/acsinfecdis.4c00615] [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: 07/29/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Arthropod-borne members of the genus Orthoflavivirus cause significant human disease. Four serotypes of dengue virus are endemic globally, and approximately 50 percent of the world's population lives in a dengue-affected area. Complications from immunoenhancement occurring after a secondary infection with a different dengue serotype make vaccine development challenging. Antiviral therapies that target features conserved in all four serotypes would, therefore, be beneficial. Computational studies identified multiple potential G-quadruplex sites that are conserved in the RNA genome sequences of members of the genus Orthoflavivirus. Biophysical studies confirmed that the NS5-B quadruplex sequences obtained from viruses of each dengue serotype can form quadruplexes in vitro, and binding data showed that known quadruplex binders stabilized NS5-B quadruplexes for all four dengue serotypes.
Collapse
Affiliation(s)
- Jessica L. Siemer
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
| | - Thao T. Le
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
| | - Ananya Paul
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
| | - David. W. Boykin
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
| | - Margo A. Brinton
- Department
of Biology, Georgia State University; Atlanta, Georgia 30303, United States
| | - W. David Wilson
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
| | - Markus W. Germann
- Department
of Chemistry, Georgia State University; Atlanta, Georgia 30303, United States
- Department
of Biology, Georgia State University; Atlanta, Georgia 30303, United States
| |
Collapse
|
6
|
Chung NPY, Cheng CY. Testis Is a Sanctuary Site for HIV-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1469:433-440. [PMID: 40301268 DOI: 10.1007/978-3-031-82990-1_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
In this review, we summarize some recent findings regarding the likely mechanisms by which HIV-1 uses the testis as a sanctuary site for evading the effects of antiviral drugs from reaching the testis, due to the presence of the blood-testis barrier. This discussion also sheds insights into the possibility of eradicating the small viral pool in the testis behind the blood-testis barrier in future functional studies. These findings also bridge the knowledge gap of eradicating viral particles hiding behind the blood-brain barrier in the brain since it is anticipated that the blood-tissue barriers, namely the blood-testis and the blood-brain barriers, are utilizing similar mechanisms to regulate the dynamic nature of their tight junctions.
Collapse
Affiliation(s)
- Nancy P Y Chung
- Department of Biological Sciences, New York City College of Technology, City University of New York, New York, NY, USA
| | - C Yan Cheng
- Department of Biology, St Francis College, New York, NY, USA
| |
Collapse
|
7
|
Du J, Luo H, Ye S, Zhang H, Zheng Z, Liu K. Unraveling IFI44L's biofunction in human disease. Front Oncol 2024; 14:1436576. [PMID: 39737399 PMCID: PMC11682996 DOI: 10.3389/fonc.2024.1436576] [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: 05/22/2024] [Accepted: 11/26/2024] [Indexed: 01/01/2025] Open
Abstract
Interferon-induced protein 44-like (IFI44L) is regarded as an immune-related gene and is a member of interferon-stimulated genes (ISGs). They participate in network transduction, and its own epigenetic modifications, apoptosis, cell-matrix formation, and many other pathways in tumors, autoimmune diseases, and viral infections. The current review provides a comprehensive overview of the onset and biological mechanisms of IFI44L and its potential clinical applications in malignant tumors and non-neoplastic diseases.
Collapse
|
8
|
Yang W, Zhang C, Liu LB, Bian ZZ, Chang JT, Fan DY, Gao N, Wang PG, An J. Immunocompetent mouse models revealed that S100A4 + monocytes/macrophages facilitate long-term Zika virus infection in the testes. Emerg Microbes Infect 2024; 13:2300466. [PMID: 38164719 PMCID: PMC10773650 DOI: 10.1080/22221751.2023.2300466] [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: 05/14/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
During its global epidemic, Zika virus (ZIKV) attracted widespread attention due to its link with various severe neurological symptoms and potential harm to male fertility. However, the understanding of how ZIKV invades and persists in the male reproductive system is limited due to the lack of immunocompetent small animal models. In this study, immunocompetent murine models were generated by using anti-IFNAR antibody blocked C57BL/6 male mice and human STAT2 (hSTAT2) knock in (KI) male mice. After infection, viral RNA could persist in the testes even after the disappearance of viremia. We also found a population of ZIKV-susceptible S100A4+ monocytes/macrophages that were recruited into testes from peripheral blood and played a crucial role for ZIKV infection in the testis. By using single-cell RNA sequencing, we also proved that S100A4+ monocytes/macrophages had a great impact on the microenvironment of ZIKV-infected testes, thus promoting ZIKV-induced testicular lesions. In conclusion, this study proposed a novel mechanism of long-term ZIKV infection in the male reproductive system.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Chen Zhang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Zhan-Zhan Bian
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Na Gao
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
9
|
Li SY, Kumar S, Gu X, DeFalco T. Testicular immunity. Mol Aspects Med 2024; 100:101323. [PMID: 39591799 PMCID: PMC11624985 DOI: 10.1016/j.mam.2024.101323] [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: 04/02/2024] [Revised: 11/13/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024]
Abstract
The testis is a unique environment where immune responses are suppressed to allow the development of sperm that possess autoimmunogenic antigens. There are several contributors responsible for testicular immune privilege, including the blood-testis barrier, testicular immune cells, immunomodulation by Sertoli cells, and high levels of steroid hormones. Despite multiple mechanisms in place to regulate the testicular immune environment, pathogens that disrupt testicular immunity can lead to long-term effects such as infertility. If testicular immunity is disturbed, autoimmune reactions can also occur, leading to aberrant immune cell infiltration and subsequent attack of autoimmunogenic germ cells. Here we discuss cellular and molecular factors underlying testicular immunity and how testicular infection or autoimmunity compromise immune privilege. We also describe infections and autoimmune diseases that impact the testis. Further research into testicular immunity will reveal how male fertility is maintained and will help update therapeutic strategies for infertility and other testicular disorders.
Collapse
Affiliation(s)
- Shu-Yun Li
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sudeep Kumar
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Xiaowei Gu
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Tony DeFalco
- Reproductive Sciences Center, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| |
Collapse
|
10
|
Obłoza M, Milewska A, Botwina P, Szczepański A, Medaj A, Bonarek P, Szczubiałka K, Pyrć K, Nowakowska M. Curcumin-Poly(sodium 4-styrenesulfonate) Conjugates as Potent Zika Virus Entry Inhibitors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5426-5437. [PMID: 38277775 PMCID: PMC10859898 DOI: 10.1021/acsami.3c13893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
Curcumin, a natural product with recognized antiviral properties, is limited in its application largely due to its poor solubility. This study presents the synthesis of water-soluble curcumin-poly(sodium 4-styrenesulfonate) (Cur-PSSNan) covalent conjugates. The antiflaviviral activity of conjugates was validated in vitro by using the Zika virus as a model. In the development of these water-soluble curcumin-containing derivatives, we used the macromolecules reported by us to also hamper viral infections. Mechanistic investigations indicated that the conjugates exhibited excellent stability and bioavailability. The curcumin and macromolecules in concerted action interact directly with virus particles and block their attachment to host cells, hampering the infection process.
Collapse
Affiliation(s)
- Magdalena Obłoza
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Aleksandra Milewska
- Virogenetics
Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
| | - Paweł Botwina
- Virogenetics
Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
- Department
of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Artur Szczepański
- Virogenetics
Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
| | - Aneta Medaj
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
- Doctoral
School of Exact and Natural Sciences, Jagiellonian
University, Lojasiewicza 11, 30-348 Cracow, Poland
| | - Piotr Bonarek
- Department
of Physical Biochemistry, Faculty of Biochemistry, Biophysics and
Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
| | - Krzysztof Szczubiałka
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Krzysztof Pyrć
- Virogenetics
Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland
| | - Maria Nowakowska
- Faculty
of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| |
Collapse
|
11
|
Yang J, Zhang Z, Liu H, Wang J, Xie S, Li P, Wen J, Wei S, Li R, Ma X, Zhao Y. Network Pharmacology and Experimental Validation of Qingwen Baidu Decoction Therapeutic Potential in COVID-19-related Lung Injury. Comb Chem High Throughput Screen 2024; 27:1286-1302. [PMID: 37957903 DOI: 10.2174/0113862073236899230919062725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 07/15/2023] [Accepted: 08/04/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND AND PURPOSE Coronavirus disease 2019 (COVID-19) is a lifethreatening disease worldwide due to its high infection and serious outcomes resulting from acute lung injury. Qingwen Baidu decoction (QBD), a well-known herbal prescription, has shown significant efficacy in patients with Coronavirus disease 2019. Hence, this study aims to uncover the molecular mechanism of QBD in treating COVID-19-related lung injury. METHODS Traditional Chinese Medicine Systems Pharmacology database (TCMSP), DrugBanks database, and Chinese Knowledge Infrastructure Project (CNKI) were used to retrieve the active ingredients of QBD. Drug and disease targets were collected using UniProt and Online Mendelian Inheritance in Man databases (OMIM). The core targets of QBD for pneumonia were analyzed by the Protein-Protein Interaction Network (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) to reveal the underlying molecular mechanisms. The analysis of key targets using molecular docking and animal experiments was also validated. RESULTS A compound-direct-acting target network mainly containing 171 compounds and 110 corresponding direct targets was constructed. The key targets included STAT3, c-JUN, TNF-α, MAPK3, MAPK1, FOS, PPARG, MAPK8, IFNG, NFκB1, etc. Moreover, 117 signaling pathways mainly involved in cytokine storm, inflammatory response, immune stress, oxidative stress and glucose metabolism were found by KEGG. The molecular docking results showed that the quercetin, alanine, and kaempferol in QBD demonstrated the strongest affinity to STAT3, c- JUN, and TNF-α. Experimental results displayed that QBD could effectively reduce the pathological damage to lung tissue by LPS and significantly alleviate the expression levels of the three key targets, thus playing a potential therapeutic role in COVID-19. CONCLUSION QBD might be a promising therapeutic agent for COVID-19 via ameliorating STAT3-related signals.
Collapse
Affiliation(s)
- Ju Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Zhao Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Honghong Liu
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Jiawei Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Shuying Xie
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Pengyan Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Jianxia Wen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Shizhang Wei
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Ruisheng Li
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| | - Xiao Ma
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yanling Zhao
- Department of Pharmacy, 302 Military Hospital of China, Beijing, 100039, China
| |
Collapse
|
12
|
Hau RK, Wright SH, Cherrington NJ. Addressing the Clinical Importance of Equilibrative Nucleoside Transporters in Drug Discovery and Development. Clin Pharmacol Ther 2023; 114:780-794. [PMID: 37404197 PMCID: PMC11347013 DOI: 10.1002/cpt.2984] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023]
Abstract
The US Food and Drug Administration (FDA), European Medicines Agency (EMA), and Pharmaceuticals and Medical Devices Agency (PMDA) guidances on small-molecule drug-drug interactions (DDIs), with input from the International Transporter Consortium (ITC), recommend the evaluation of nine drug transporters. Although other clinically relevant drug uptake and efflux transporters have been discussed in ITC white papers, they have been excluded from further recommendation by the ITC and are not included in current regulatory guidances. These include the ubiquitously expressed equilibrative nucleoside transporters (ENT) 1 and ENT2, which have been recognized by the ITC for their potential role in clinically relevant nucleoside analog drug interactions for patients with cancer. Although there is comparatively limited clinical evidence supporting their role in DDI risk or other adverse drug reactions (ADRs) compared with the nine highlighted transporters, several in vitro and in vivo studies have identified ENT interactions with non-nucleoside/non-nucleotide drugs, in addition to nucleoside/nucleotide analogs. Some noteworthy examples of compounds that interact with ENTs include cannabidiol and selected protein kinase inhibitors, as well as the nucleoside analogs remdesivir, EIDD-1931, gemcitabine, and fialuridine. Consequently, DDIs involving the ENTs may be responsible for therapeutic inefficacy or off-target toxicity. Evidence suggests that ENT1 and ENT2 should be considered as transporters potentially involved in clinically relevant DDIs and ADRs, thereby warranting further investigation and regulatory consideration.
Collapse
Affiliation(s)
- Raymond K Hau
- Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| | - Stephen H Wright
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Nathan J Cherrington
- Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
13
|
Hau RK, Wright SH, Cherrington NJ. In Vitro and In Vivo Models for Drug Transport Across the Blood-Testis Barrier. Drug Metab Dispos 2023; 51:1157-1168. [PMID: 37258305 PMCID: PMC10449102 DOI: 10.1124/dmd.123.001288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/10/2023] [Accepted: 05/25/2023] [Indexed: 06/02/2023] Open
Abstract
The blood-testis barrier (BTB) is a selectively permeable membrane barrier formed by adjacent Sertoli cells (SCs) in the seminiferous tubules of the testes that develops intercellular junctional complexes to protect developing germ cells from external pressures. However, due to this inherent defense mechanism, the seminiferous tubule lumen can act as a pharmacological sanctuary site for latent viruses (e.g., Ebola, Zika) and cancers (e.g., leukemia). Therefore, it is critical to identify and evaluate BTB carrier-mediated drug delivery pathways to successfully treat these viruses and cancers. Many drugs are unable to effectively cross cell membranes without assistance from carrier proteins like transporters because they are large, polar, and often carry a charge at physiologic pH. SCs express transporters that selectively permit endogenous compounds, such as carnitine or nucleosides, across the BTB to support normal physiologic activity, although reproductive toxicants can also use these pathways, thereby circumventing the BTB. Certain xenobiotics, including select cancer therapeutics, antivirals, contraceptives, and environmental toxicants, are known to accumulate within the male genital tract and cause testicular toxicity; however, the transport pathways by which these compounds circumvent the BTB are largely unknown. Consequently, there is a need to identify the clinically relevant BTB transport pathways in in vitro and in vivo BTB models that recapitulate human pharmacokinetics and pharmacodynamics for these xenobiotics. This review summarizes the various in vitro and in vivo models of the BTB reported in the literature and highlights the strengths and weaknesses of certain models for drug disposition studies. SIGNIFICANCE STATEMENT: Drug disposition to the testes is influenced by the physical, physiological, and immunological components of the blood-testis barrier (BTB). But many compounds are known to cross the BTB by transporters, resulting in pharmacological and/or toxicological effects in the testes. Therefore, models that assess drug transport across the human BTB must adequately account for these confounding factors. This review identifies and discusses the benefits and limitations of various in vitro and in vivo BTB models for preclinical drug disposition studies.
Collapse
Affiliation(s)
- Raymond K Hau
- College of Pharmacy, Department of Pharmacology & Toxicology, (R.K.H., N.J.C.) and College of Medicine, Department of Physiology, The University of Arizona, Tucson, Arizona (S.H.W.)
| | - Stephen H Wright
- College of Pharmacy, Department of Pharmacology & Toxicology, (R.K.H., N.J.C.) and College of Medicine, Department of Physiology, The University of Arizona, Tucson, Arizona (S.H.W.)
| | - Nathan J Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology, (R.K.H., N.J.C.) and College of Medicine, Department of Physiology, The University of Arizona, Tucson, Arizona (S.H.W.)
| |
Collapse
|
14
|
Hau RK, Wright SH, Cherrington NJ. Drug Transporters at the Human Blood-Testis Barrier. Drug Metab Dispos 2023; 51:560-571. [PMID: 36732077 PMCID: PMC10158500 DOI: 10.1124/dmd.122.001186] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/06/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Transporters are involved in the movement of many physiologically important molecules across cell membranes and have a substantial impact on the pharmacological and toxicological effect of xenobiotics. Many transporters have been studied in the context of disposition to, or toxicity in, organs such as the kidney and liver; however, transporters in the testes are increasingly gaining recognition for their role in drug transport across the blood-testis barrier (BTB). The BTB is an epithelial membrane barrier formed by adjacent Sertoli cells (SCs) in the seminiferous tubules that form intercellular junctional complexes to protect developing germ cells from the external environment. Consequently, many charged or large polar molecules cannot cross this barrier without assistance from a transporter. SCs express a variety of drug uptake and efflux transporters to control the flux of endogenous and exogenous molecules across the BTB. Recent studies have identified several transport pathways in SCs that allow certain drugs to circumvent the human BTB. These pathways may exist in other species, such as rodents and nonhuman primates; however, there is (1) a lack of information on their expression and/or localization in these species, and (2) conflicting reports on localization of some transporters that have been evaluated in rodents compared with humans. This review outlines the current knowledge on the expression and localization of pharmacologically relevant drug transporters in human testes and calls attention to the insufficient and contradictory understanding of testicular transporters in other species that are commonly used in drug disposition and toxicity studies. SIGNIFICANCE STATEMENT: While the expression, localization, and function of many xenobiotic transporters have been studied in organs such as the kidney and liver, the characterization of transporters in the testes is scarce. This review summarizes the expression and localization of common pharmacologically-relevant transporters in human testes that have significant implications for the development of drugs that can cross the blood-testis barrier. Potential expression differences between humans and rodents highlighted here suggest rodents may be inappropriate for some testicular disposition and toxicity studies.
Collapse
Affiliation(s)
- Raymond K Hau
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| | - Stephen H Wright
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| | - Nathan J Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology (R.K.H., N.J.C.) and College of Medicine, Department of Physiology (S.H.W.), The University of Arizona, Tucson, Arizona
| |
Collapse
|
15
|
Yang W, Liu LB, Liu FL, Wu YH, Zhen ZD, Fan DY, Sheng ZY, Song ZR, Chang JT, Zheng YT, An J, Wang PG. Single-cell RNA sequencing reveals the fragility of male spermatogenic cells to Zika virus-induced complement activation. Nat Commun 2023; 14:2476. [PMID: 37120617 PMCID: PMC10148584 DOI: 10.1038/s41467-023-38223-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/21/2023] [Indexed: 05/01/2023] Open
Abstract
Zika virus (ZIKV) is a potential threat to male reproductive health but the mechanisms underlying its influence on testes during ZIKV infection remain obscure. To address this question, we perform single-cell RNA sequencing using testes from ZIKV-infected mice. The results reveal the fragility of spermatogenic cells, especially spermatogonia, to ZIKV infection and show that the genes of the complement system are significantly upregulated mainly in infiltrated S100A4 + monocytes/macrophages. Complement activation and its contribution to testicular damage are validated by ELISA, RT‒qPCR and IFA and further verify in ZIKV-infected northern pigtailed macaques by RNA genome sequencing and IFA, suggesting that this might be the common response to ZIKV infection in primates. On this basis, we test the complement inhibitor C1INH and S100A4 inhibitors sulindac and niclosamide for their effects on testis protection. C1INH alleviates the pathological change in the testis but deteriorates ZIKV infection in general. In contrast, niclosamide effectively reduces S100A4 + monocyte/macrophage infiltration, inhibits complement activation, alleviates testicular damage, and rescues the fertility of male mice from ZIKV infection. This discovery therefore encourages male reproductive health protection during the next ZIKV epidemic.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Feng-Liang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Yang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zheng-Ran Song
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yong-Tang Zheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, 650107, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
16
|
Benchimol GDC, Santos JB, Lopes ASDC, Oliveira KG, Okada EST, de Alcantara BN, Pereira WLA, Leão DL, Martins ACC, Carneiro LA, Imbeloni AA, Makiama ST, de Castro LPPA, Coutinho LN, Casseb LMN, Vasconcelos PFDC, Domingues SFS, Medeiros DBDA, Scalercio SRRDA. Zika Virus Infection Damages the Testes in Pubertal Common Squirrel Monkeys ( Saimiri collinsi). Viruses 2023; 15:615. [PMID: 36992324 PMCID: PMC10051343 DOI: 10.3390/v15030615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 02/26/2023] Open
Abstract
During the Zika virus (ZIKV) outbreak and after evidence of its sexual transmission was obtained, concerns arose about the impact of the adverse effects of ZIKV infection on human fertility. In this study, we evaluated the clinical-laboratory aspects and testicular histopathological patterns of pubertal squirrel monkeys (Saimiri collinsi) infected with ZIKV, analyzing the effects at different stages of infection. The susceptibility of S. collinsi to ZIKV infection was confirmed by laboratory tests, which detected viremia (mean 1.63 × 106 RNA copies/µL) and IgM antibody induction. Reduced fecal testosterone levels, severe testicular atrophy and prolonged orchitis were observed throughout the experiment by ultrasound. At 21 dpi, testicular damage associated with ZIKV was confirmed by histopathological and immunohistochemical (IHC) analyses. Tubular retraction, the degeneration and necrosis of somatic and germ cells in the seminiferous tubules, the proliferation of interstitial cells and an inflammatory infiltrate were observed. ZIKV antigen was identified in the same cells where tissue injuries were observed. In conclusion, squirrel monkeys were found to be susceptible to the Asian variant of ZIKV, and this model enabled the identification of multifocal lesions in the seminiferous tubules of the infected group evaluated. These findings may suggest an impact of ZIKV infection on male fertility.
Collapse
Affiliation(s)
- Gabriela da Costa Benchimol
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | - Josye Bianca Santos
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Postgraduate Program in Animal Reproduction in the Amazon (ReproAmazon), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
| | | | | | | | | | - Washington Luiz Assunção Pereira
- Laboratory of Animal Pathology (LABOPAT), Institute of Health and Animal Production, Federal Rural University of the Amazon, Belém 66077-830, Pará, Brazil
| | - Danuza Leite Leão
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Mamirauá Institute for Sustainable Development, Tefé 69553-225, Amazonas, Brazil
| | | | | | | | | | | | - Leandro Nassar Coutinho
- Laboratory of Animal Pathology (LABOPAT), Institute of Health and Animal Production, Federal Rural University of the Amazon, Belém 66077-830, Pará, Brazil
| | - Lívia Medeiros Neves Casseb
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | - Pedro Fernando da Costa Vasconcelos
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Department of Pathology, Center of Biologic and Health Sciences, State University of Pará, Belém 66050-540, Pará, Brazil
| | - Sheyla Farhayldes Souza Domingues
- Laboratory of Amazon Animal Biotechnology and Medicine (BIOMEDAM), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- Postgraduate Program in Animal Reproduction in the Amazon (ReproAmazon), Federal University of Pará, Castanhal 68740-970, Pará, Brazil
- School of Veterinary Medicine, Federal University of Pará, Castanhal 68740-970, Pará, Brazil
| | - Daniele Barbosa de Almeida Medeiros
- Department of Arbovirology and Hemorrhagic Fevers, Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
- Postgraduate Program in Virology (PPGV), Evandro Chagas Institute, Ananindeua 67030-000, Pará, Brazil
| | | |
Collapse
|
17
|
Zeng Y, Chen HQ, Zhang Z, Fan J, Li JZ, Zhou SM, Wang N, Yan SP, Cao J, Liu JY, Zhou ZY, Liu WB. IFI44L as a novel epigenetic silencing tumor suppressor promotes apoptosis through JAK/STAT1 pathway during lung carcinogenesis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 319:120943. [PMID: 36584854 DOI: 10.1016/j.envpol.2022.120943] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Numerous evidence showed that the occurrence and development of lung cancer is closely related to environmental pollution. Therefore, new environmental response predictive markers are urgently needed for early diagnosis and screening of lung cancer. Interferon-induced protein 44-like (IFI44L) has been shown to be related in a variety of tumors, but its function and mechanism during lung carcinogenesis still have remained largely unknown. In this study, gene expression and methylation status were analyzed through online tools and malignant transformation models. Differentially expressed cell models and xenograft tumor models were established and used to clarify the gene function. RT-qPCR, western blotting, immunohistochemistry, and co-immunoprecipitation (Co-IP) were used to explore the mechanism. Results showed that IFI44L was dramatically downexpressed during lung carcinogenesis, and its low expression may be attributed to DNA methylation. Overexpression of IFI44L obviously inhibited cell growth and promoted apoptosis. After knockdown of IFI44L expression, the proliferation ability was remarkably increased and the apoptosis was significantly reduced. Functional enrichment showed that IFI44L was involved in apoptosis and JAK/STAT1 signaling pathway, and was highly correlated with downstream molecules. After overexpression of IFI44L, the expression of P-STAT1 and downstream molecules XAF1, OAS1, OAS2 and OAS3 were significantly increased. After knockdown of STAT1 expression, the pro-apoptotic effect of IFI44L was reduced. Co-IP results showed that IFI44L had protein interaction with STAT1. Results proved that IFI44L promoted STAT1 phosphorylation and activated the JAK/STAT1 signaling pathway by directly binding to STAT1 protein, thereby leading to cell apoptosis. Our study revealed that IFI44L promotes cell apoptosis and exerts tumor suppressors by activating the JAK/STAT1 signaling pathway. It further suggests that IFI44L has clinical therapeutic potential and may be a promising biomarker during lung carcinogenesis.
Collapse
Affiliation(s)
- Yong Zeng
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Hong-Qiang Chen
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zhe Zhang
- Department of Breast and Thyroid Surgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, PR China
| | - Jun Fan
- Department of Breast and Thyroid Surgery, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, PR China
| | - Jing-Zhi Li
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China; School of Public Health, Xinxiang Medical University, Xinxiang, Henan, 453003, PR China
| | - Shi-Meng Zhou
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China; School of Public Health, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Na Wang
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China; Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, Guizhou, 550025, PR China
| | - Su-Peng Yan
- Department of Sanitary Equipment and Metrology, School of Biomedical Engineering and Medical Imaging, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Jin-Yi Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Zi-Yuan Zhou
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China
| | - Wen-Bin Liu
- Department of Environmental Health, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China; Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, PR China.
| |
Collapse
|
18
|
Yang W, Zhang C, Wu YH, Liu LB, Zhen ZD, Fan DY, Song ZR, Chang JT, Wang PG, An J. Mice 3D testicular organoid system as a novel tool to study Zika virus pathogenesis. Virol Sin 2023; 38:66-74. [PMID: 36241087 PMCID: PMC10006202 DOI: 10.1016/j.virs.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022] Open
Abstract
Zika virus (ZIKV) poses a serious threat to global public health due to its close relationship with neurological and male reproductive damage. However, deficiency of human testicular samples hinders the in-depth research on ZIKV-induced male reproductive system injury. Organoids are relatively simple in vitro models, which could mimic the pathological changes of corresponding organs. In this study, we constructed a 3D testicular organoid model using primary testicular cells from adult BALB/c mice. Similar to the testis, this organoid system has a blood-testis barrier (BTB)-like structure and could synthesize testosterone. ZIKV tropism of testicular cells and ZIKV-induced pathological changes in testicular organoid was also similar to that in mammalian testis. Therefore, our results provide a simple and reproducible in vitro testicular model for the investigations of ZIKV-induced testicular injury.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Department of Neurosurgery, Capital Medical University Sanbo Brain Hospital, Beijing, 100093, China
| | - Chen Zhang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Li-Bo Liu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zheng-Ran Song
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jia-Tong Chang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100093, China.
| |
Collapse
|
19
|
Abstract
Zika virus (ZIKV) is an emerging virus from the Flaviviridae family that is transmitted to humans by mosquito vectors and represents an important health problem. Infections in pregnant women are of major concern because of potential devastating consequences during pregnancy and have been associated with microcephaly in newborns. ZIKV has a unique ability to use the host machinery to promote viral replication in a tissue-specific manner, resulting in characteristic pathological disorders. Recent studies have proposed that the host ubiquitin system acts as a major determinant of ZIKV tropism by providing the virus with an enhanced ability to enter new cells. In addition, ZIKV has developed mechanisms to evade the host immune response, thereby allowing the establishment of viral persistence and enhancing viral pathogenesis. We discuss recent reports on the mechanisms used by ZIKV to replicate efficiently, and we highlight potential new areas of research for the development of therapeutic approaches.
Collapse
Affiliation(s)
- Maria I Giraldo
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA; ,
| | - Maria Gonzalez-Orozco
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA; ,
| | - Ricardo Rajsbaum
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA; ,
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
- Current affiliation: Center for Virus-Host-Innate-Immunity; Rutgers Biomedical and Health Sciences, Institute for Infectious and Inflammatory Diseases; and Department of Medicine, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, New Jersey, USA;
| |
Collapse
|
20
|
Wang S, Zhang Y, Lou J, Yong H, Shan S, Liu Z, Song M, Zhang C, Kou R, Liu Z, Yu W, Zhao X, Song F. The therapeutic potential of berberine chloride against SARM1-dependent axon degeneration in acrylamide-induced neuropathy. Phytother Res 2023; 37:77-88. [PMID: 36054436 DOI: 10.1002/ptr.7594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 07/11/2022] [Accepted: 08/06/2022] [Indexed: 01/19/2023]
Abstract
Chronic acrylamide (ACR) intoxication causes typical pathology of axon degeneration. Moreover, sterile-α and toll/interleukin 1 receptor motif-containing protein 1 (SARM1), the central executioner of the programmed axonal destruction process under various insults, is up-regulated in ACR neuropathy. However, it remains unclear whether inhibitors targeting SARM1 are effective or not. Among all the pharmacological antagonists, berberine chloride (BBE), a natural phytochemical and the first identified non-competitive inhibitor of SARM1, attracts tremendous attention. Here, we observed the protection of 100 μM BBE against ACR-induced neurites injury (2 mM ACR, 24 hr) in vitro, and further evaluated the neuroprotective effect of BBE (100 mg/kg p.o. three times a week for 4 weeks) in ACR-intoxicated rats (40 mg/kg i.p. three times a week for 4 weeks). The expression of SARM1 was also detected. BBE intervention significantly inhibited the overexpression of SARM1, ameliorated axonal degeneration, alleviated pathological changes in the sciatic nerve and spinal cord, and improved neurobehavioral symptoms in ACR-poisoned rats. Thus, BBE exhibits a strong neuroprotective effect against the SARM1-dependent axon destruction in ACR neuropathy. Meanwhile, our study underscores the need for appropriate inhibitor selection in diverse situations that would benefit from blocking the SARM1-dependent axonal destruction pathway.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yifan Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jianwei Lou
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Hui Yong
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shulin Shan
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhidan Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Mingxue Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Cuiqin Zhang
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Ruirui Kou
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhaoxiong Liu
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Wenhao Yu
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Xiulan Zhao
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.,School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Fuyong Song
- Department of Toxicology and Nutrition, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
21
|
Abstract
Proliferative diabetic retinopathy (PDR) is a world-wide leading cause of blindness among adults and may be associated with the influence of genetic factors. It is significant to search for genetic biomarkers of PDR. In our study, we collected genomic data about PDR from gene expression omnibus (GEO) database. Differentially expressed gene (DEG) analysis and weighted gene co-expression network analysis (WGCNA) were carried out. The gene module with the highest gene significance (GS) was defined as the key module. Hub genes were identified by Venn diagram. Then we verified the expression of hub genes in validation data sets and built a diagnostic model by least absolute shrinkage and selection operator (LASSO) regression. Enrichment analysis, including gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), gene set enrichment analysis (GSEA) and construction of a protein-protein interaction (PPI) network were conducted. In GSE60436, we identified 466 DEGs. WGCNA established 14 gene modules, and the blue module (GS = 0.64), was the key module. Interferon (IFN)-induced protein 44-like (IFI44L) and complement C1q tumor necrosis factor-related protein 5 (C1QTNF5) were identified as hub genes. The expression of hub genes in GEO datasets was verified and a diagnostic model was constructed by LASSO as follows: index = IFI44L * 0.0432 + C1QTNF5 * 0.11246. IFI44L and C1QTNF5 might affect the disease progression of PDR by regulating metabolism-related and inflammatory pathways. IFI44L and C1QTNF5 may play important roles in the disease process of PDR, and a LASSO regression model suggested that the 2 genes could serve as promising biomarkers of PDR.
Collapse
Affiliation(s)
- Mingxin Shang
- He Eye Specialist Hospital, Shenyang, Liaoning Province, China
| | - Yao Zhang
- He Eye Specialist Hospital, Shenyang, Liaoning Province, China
| | - Tongtong Zhang
- He Eye Specialist Hospital, Shenyang, Liaoning Province, China
- * Correspondence: Tongtong Zhang, He Eye Specialist Hospital, No.128 North Huanghe Street, Shenyang, Liaoning Province 110034, China (e-mail: )
| |
Collapse
|
22
|
Wang F, Zhang J, Wang Y, Chen Y, Han D. Viral tropism for the testis and sexual transmission. Front Immunol 2022; 13:1040172. [PMID: 36439102 PMCID: PMC9682072 DOI: 10.3389/fimmu.2022.1040172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/24/2022] [Indexed: 10/17/2023] Open
Abstract
The mammalian testis adopts an immune privileged environment to protect male germ cells from adverse autoimmune reaction. The testicular immune privileged status can be also hijacked by various microbial pathogens as a sanctuary to escape systemic immune surveillance. In particular, several viruses have a tropism for the testis. To overcome the immune privileged status and mount an effective local defense against invading viruses, testicular cells are well equipped with innate antiviral machinery. However, several viruses may persist an elongated duration in the testis and disrupt the local immune homeostasis, thereby impairing testicular functions and male fertility. Moreover, the viruses in the testis, as well as other organs of the male reproductive system, can shed to the semen, thus allowing sexual transmission to partners. Viral infection in the testis, which can impair male fertility and lead to sexual transmission, is a serious concern in research on known and on new emerging viruses. To provide references for our scientific peers, this article reviews research achievements and suggests future research focuses in the field.
Collapse
Affiliation(s)
| | | | | | - Yongmei Chen
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Daishu Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
23
|
Han Y, Tan L, Zhou T, Yang L, Carrau L, Lacko LA, Saeed M, Zhu J, Zhao Z, Nilsson-Payant BE, Lira Neto FT, Cahir C, Giani AM, Chai JC, Li Y, Dong X, Moroziewicz D, Paull D, Zhang T, Koo S, Tan C, Danziger R, Ba Q, Feng L, Chen Z, Zhong A, Wise GJ, Xiang JZ, Wang H, Schwartz RE, tenOever BR, Noggle SA, Rice CM, Qi Q, Evans T, Chen S. A human iPSC-array-based GWAS identifies a virus susceptibility locus in the NDUFA4 gene and functional variants. Cell Stem Cell 2022; 29:1475-1490.e6. [PMID: 36206731 PMCID: PMC9550219 DOI: 10.1016/j.stem.2022.09.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 06/09/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022]
Abstract
Population-based studies to identify disease-associated risk alleles typically require samples from a large number of individuals. Here, we report a human-induced pluripotent stem cell (hiPSC)-based screening strategy to link human genetics with viral infectivity. A genome-wide association study (GWAS) identified a cluster of single-nucleotide polymorphisms (SNPs) in a cis-regulatory region of the NDUFA4 gene, which was associated with susceptibility to Zika virus (ZIKV) infection. Loss of NDUFA4 led to decreased sensitivity to ZIKV, dengue virus, and SARS-CoV-2 infection. Isogenic hiPSC lines carrying non-risk alleles of SNPs or deletion of the cis-regulatory region lower sensitivity to viral infection. Mechanistic studies indicated that loss/reduction of NDUFA4 causes mitochondrial stress, which leads to the leakage of mtDNA and thereby upregulation of type I interferon signaling. This study provides proof-of-principle for the application of iPSC arrays in GWAS and identifies NDUFA4 as a previously unknown susceptibility locus for viral infection.
Collapse
Affiliation(s)
- Yuling Han
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Lei Tan
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Center for Energy Metabolism and Reproduction, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ting Zhou
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Stem Cell Research Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Liuliu Yang
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Lucia Carrau
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Lauretta A Lacko
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Mohsan Saeed
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA; Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA; National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA 02118, USA
| | - Jiajun Zhu
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Zeping Zhao
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | | | | | - Clare Cahir
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; The Tri-Institutional PhD Program in Chemical Biology, New York, NY, USA
| | - Alice Maria Giani
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Jin Chou Chai
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Yang Li
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Xue Dong
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Dorota Moroziewicz
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | - Daniel Paull
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | - Tuo Zhang
- Genomic Resource Core Facility, Weill Cornell Medical College, New York, NY 10065, USA
| | - Soyeon Koo
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Weill Cornell Neuroscience PhD Program, New York, NY, USA
| | - Christina Tan
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Ron Danziger
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Qian Ba
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingling Feng
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA; Key Laboratory of Pesticide and Chemical Biology (CCNU), Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, Hubei 430079, China
| | - Zhengming Chen
- Department of Population Health Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Aaron Zhong
- Stem Cell Research Facility, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Gilbert J Wise
- Department of Urology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jenny Z Xiang
- Genomic Resource Core Facility, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hui Wang
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Benjamin R tenOever
- Department of Microbiology, New York University, 430 E 29th Street, New York, NY 10016, USA
| | - Scott A Noggle
- The New York Stem Cell Foundation Research Institute, 619 West 54th Street, 3rd Floor, New York, NY 10019, USA
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Qibin Qi
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
24
|
de Albuquerque BHDR, de Oliveira MTFC, Aderaldo JF, de Medeiros Garcia Torres M, Lanza DCF. Human seminal virome: a panel based on recent literature. Basic Clin Androl 2022; 32:16. [PMID: 36064315 PMCID: PMC9444275 DOI: 10.1186/s12610-022-00165-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/16/2022] [Indexed: 02/06/2023] Open
Abstract
Background The seminal virome and its implications for fertility remain poorly understood. To date, there are no defined panels for the detection of viruses of clinical interest in seminal samples. Results In this study, we characterized the human seminal virome based on more than 1,000 studies published over the last five years. Conclusions The number of studies investigating viruses that occur in human semen has increased, and to date, these studies have been mostly prospective or related to specific clinical findings. Through the joint analysis of all these studies, we have listed the viruses related to the worsening of seminal parameters and propose a new panel with the main viruses already described that possibly affect male fertility and health. This panel can assist in evaluating semen quality and serve as a tool for investigation in cases of infertility.
Collapse
|
25
|
Mungin JW, Chen X, Liu B. Interferon Epsilon Signaling Confers Attenuated Zika Replication in Human Vaginal Epithelial Cells. Pathogens 2022; 11:853. [PMID: 36014974 PMCID: PMC9415962 DOI: 10.3390/pathogens11080853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging flavivirus that causes congenital birth defects and neurological compilations in the human host. Although ZIKV is primarily transmitted through infected mosquitos, recent studies reveal sexual contact as a potential transmission route. In vagina-bearing individuals, the vaginal epithelium constitutes the first line of defense against viruses. However, it is unclear how ZIKV interacts with the vaginal epithelium to initiate ZIKV transmission. In this study, we demonstrate that exposing ZIKV to human vaginal epithelial cells (hVECs) resulted in de novo viral RNA replication, increased envelope viral protein production, and a steady, extracellular release of infectious viral particles. Interestingly, our data show that, despite an increase in viral load, the hVECs did not exhibit significant cytopathology in culture as other cell types typically do. Furthermore, our data reveal that the innate antiviral state of hVECs plays a crucial role in preventing viral cytopathology. For the first time, our data show that interferon epsilon inhibits ZIKV replication. Collectively, our results in this study provide a novel perspective on the viral susceptibility and replication dynamics during ZIKV infection in the human vaginal epithelium. These findings will be instrumental towards developing therapeutic agents aimed at eliminating the pathology caused by the virus.
Collapse
Affiliation(s)
| | | | - Bindong Liu
- Centers for AIDS Health Disparity Research, Department of Microbiology, Immunology, and Physiology, Meharry Medical College, Nashville, TN 37208, USA; (J.W.M.J.); (X.C.)
| |
Collapse
|
26
|
Ball EE, Pesavento PA, Van Rompay KKA, Keel MK, Singapuri A, Gomez-Vazquez JP, Dudley DM, O’Connor DH, Breitbach ME, Maness NJ, Schouest B, Panganiban A, Coffey LL. Zika virus persistence in the male macaque reproductive tract. PLoS Negl Trop Dis 2022; 16:e0010566. [PMID: 35788751 PMCID: PMC9299295 DOI: 10.1371/journal.pntd.0010566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/20/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022] Open
Abstract
Zika virus (ZIKV) is unique among mosquito-borne flaviviruses in that it is also vertically and sexually transmitted by humans. The male reproductive tract is thought to be a ZIKV reservoir; however, the reported magnitude and duration of viral persistence in male genital tissues vary widely in humans and non-human primate models. ZIKV tissue and cellular tropism and potential effects on male fertility also remain unclear. The objective of this study was to resolve these questions by analyzing archived genital tissues from 51 ZIKV-inoculated male macaques and correlating data on plasma viral kinetics, tissue tropism, and ZIKV-induced pathological changes in the reproductive tract. We hypothesized that ZIKV would persist in the male macaque genital tract for longer than there was detectable viremia, where it would localize to germ and epithelial cells and associate with lesions. We detected ZIKV RNA and infectious virus in testis, epididymis, seminal vesicle, and prostate gland. In contrast to prepubertal males, sexually mature macaques were significantly more likely to harbor persistent ZIKV RNA or infectious virus somewhere in the genital tract, with detection as late as 60 days post-inoculation. ZIKV RNA localized primarily to testicular stem cells/sperm precursors and epithelial cells, including Sertoli cells, epididymal duct epithelium, and glandular epithelia of the seminal vesicle and prostate gland. ZIKV infection was associated with microscopic evidence of inflammation in the epididymis and prostate gland of sexually mature males, pathologies that were absent in uninfected controls, which could have significant effects on male fertility. The findings from this study increase our understanding of persistent ZIKV infection which can inform risk of sexual transmission during assisted reproductive therapies as well as potential impacts on male fertility. Zika virus (ZIKV) spread since 2015 led to establishment of urban epidemic cycles involving humans and Aedes mosquitoes. ZIKV is also sexually and vertically transmitted and causes congenital Zika syndrome. Together, these features show that ZIKV poses significant global public health risks. By virtue of similar reproductive anatomy and physiology to humans, macaques serve as a useful model for ZIKV infection. However, macaque studies to date have been limited by small sample size, typically 1 to 5 animals. Although mounting evidence identifies the male reproductive tract as a significant ZIKV reservoir, data regarding the duration of ZIKV persistence, potential for sexual transmission, and male genitourinary sequelae remain sparse. Here, we analyzed archived genital tissues from more than 50 ZIKV-inoculated male macaques. Our results show that ZIKV can persist in the male macaque reproductive tract after the resolution of viremia, with virus localization to sperm precursors and epithelial cells, and microscopic evidence of inflammation in the epididymis and prostate gland. Our findings help explain cases of sexual transmission of ZIKV in humans, which also carries a risk for transmission via assisted fertility procedures, even after resolution of detectable viremia.
Collapse
Affiliation(s)
- Erin E. Ball
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
- United States Army, Veterinary Corps
| | - Patricia A. Pesavento
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Koen K. A. Van Rompay
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
- California National Primate Research Center, University of California, Davis, California, United States of America
| | - M. Kevin Keel
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Anil Singapuri
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Jose P. Gomez-Vazquez
- Center for Animal Disease Modeling and Surveillance, University of California, Davis, California, United States of America
| | - Dawn M. Dudley
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - David H. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Meghan E. Breitbach
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Nicholas J. Maness
- Division of Microbiology, Tulane National Primate Research Center, Covington, Los Angeles, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Blake Schouest
- Division of Microbiology, Tulane National Primate Research Center, Covington, Los Angeles, United States of America
| | - Antonito Panganiban
- Division of Microbiology, Tulane National Primate Research Center, Covington, Los Angeles, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Lark L. Coffey
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
27
|
Kuassivi ON, Abiven H, Satie AP, Cartron M, Mahé D, Aubry F, Mathieu R, Rebours V, Le Tortorec A, Dejucq-Rainsford N. Human Testicular Germ Cells, a Reservoir for Zika Virus, Lack Antiviral Response Upon Zika or Poly(I:C) Exposure. Front Immunol 2022; 13:909341. [PMID: 35784373 PMCID: PMC9248283 DOI: 10.3389/fimmu.2022.909341] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Zika virus (ZIKV) is an emerging teratogenic arbovirus that persists in semen and is sexually transmitted. We previously demonstrated that ZIKV infects the human testis and persists in testicular germ cells (TGCs) for several months after patients’ recovery. To decipher the mechanisms underlying prolonged ZIKV replication in TGCs, we compared the innate immune response of human testis explants and isolated TGCs to ZIKV and to Poly(I:C), a viral RNA analog. Our results demonstrate the weak innate responses of human testis to both ZIKV and Poly(I:C) as compared with other tissues or species. TGCs failed to up-regulate antiviral effectors and type I IFN upon ZIKV or Poly(I:C) stimulation, which might be due to a tight control of PRR signaling, as evidenced by the absence of activation of the downstream effector IRF3 and elevated expression of repressors. Importantly, exogenous IFNβ boosted the innate immunity of TGCs and inhibited ZIKV replication in the testis ex vivo, raising hopes for the prevention of ZIKV infection and persistence in this organ.
Collapse
Affiliation(s)
- Ohiniba Nadège Kuassivi
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Hervé Abiven
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Anne-Pascale Satie
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Matéo Cartron
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Dominique Mahé
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Florence Aubry
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Romain Mathieu
- Service d’Urologie, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Valérie Rebours
- Unité de Coordination Hospitalière des Prélèvements d’Organes et de Tissus, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | - Anna Le Tortorec
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- Université de Rennes, INSERM, Ecole des Hautes Etudes en Santé Publique (EHESP), Institut de recherche en santé, environnement et travail (Irset) – UMR_S1085, Rennes, France
- *Correspondence: Nathalie Dejucq-Rainsford,
| |
Collapse
|
28
|
Barnard TR, Wang AB, Sagan SM. A highly sensitive strand-specific multiplex RT-qPCR assay for quantitation of Zika virus replication. J Virol Methods 2022; 307:114556. [PMID: 35654259 DOI: 10.1016/j.jviromet.2022.114556] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 05/28/2022] [Indexed: 10/18/2022]
Abstract
Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) is widely used to quantify viral RNA genomes for diagnostics and research, yet conventional RT-qPCR protocols are unable to accurately distinguish between the different viral RNA species that exist during infection. Here we show that false-priming and self-priming occur during reverse transcription with several published Zika virus (ZIKV) primer sets. We developed a RT-qPCR assay using tagged primers and thermostable reverse transcriptase, which greatly reduced the occurrence of nonspecific cDNA products. Furthermore, we optimized the assay for use in multiplex qPCR which allows for simultaneous quantitative detection of positive-strand and negative-strand ZIKV RNA along with an internal control from both human and mosquito cells. Importantly, this assay is sensitive enough to study early stages of virus infection in vitro. Strikingly, using this assay, we detected ZIKV negative-strand RNA as early as 3 h post-infection in mammalian cell culture, at a time point prior to the onset of positive-strand RNA synthesis. Overall, the strand-specific RT-qPCR assay developed herein is a valuable tool to quantify ZIKV RNA and to study viral replication dynamics during infection. The application of these findings has the potential to increase accuracy of RNA detection methods for a variety of viral pathogens.
Collapse
Affiliation(s)
- Trisha R Barnard
- Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
| | - Alex B Wang
- Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
| | - Selena M Sagan
- Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada; Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
29
|
Miller SR, McGrath ME, Zorn KM, Ekins S, Wright SH, Cherrington NJ. Remdesivir and EIDD-1931 Interact with Human Equilibrative Nucleoside Transporters 1 and 2: Implications for Reaching SARS-CoV-2 Viral Sanctuary Sites. Mol Pharmacol 2021; 100:548-557. [PMID: 34503974 PMCID: PMC8626781 DOI: 10.1124/molpharm.121.000333] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 09/07/2021] [Indexed: 11/22/2022] Open
Abstract
Equilibrative nucleoside transporters (ENTs) are present at the blood-testis barrier (BTB), where they can facilitate antiviral drug disposition to eliminate a sanctuary site for viruses detectable in semen. The purpose of this study was to investigate ENT-drug interactions with three nucleoside analogs, remdesivir, molnupiravir, and molnupiravir's active metabolite, β-d-N4-hydroxycytidine (EIDD-1931), and four non-nucleoside molecules repurposed as antivirals for coronavirus disease 2019 (COVID-19). The study used three-dimensional pharmacophores for ENT1 and ENT2 substrates and inhibitors and Bayesian machine learning models to identify potential interactions with these transporters. In vitro transport experiments demonstrated that remdesivir was the most potent inhibitor of ENT-mediated [3H]uridine uptake (ENT1 IC50: 39 μM; ENT2 IC50: 77 μM), followed by EIDD-1931 (ENT1 IC50: 259 μM; ENT2 IC50: 467 μM), whereas molnupiravir was a modest inhibitor (ENT1 IC50: 701 μM; ENT2 IC50: 851 μM). Other proposed antivirals failed to inhibit ENT-mediated [3H]uridine uptake below 1 mM. Remdesivir accumulation decreased in the presence of 6-S-[(4-nitrophenyl)methyl]-6-thioinosine (NBMPR) by 30% in ENT1 cells (P = 0.0248) and 27% in ENT2 cells (P = 0.0054). EIDD-1931 accumulation decreased in the presence of NBMPR by 77% in ENT1 cells (P = 0.0463) and by 64% in ENT2 cells (P = 0.0132), which supported computational predictions that both are ENT substrates that may be important for efficacy against COVID-19. NBMPR failed to decrease molnupiravir uptake, suggesting that ENT interaction is likely inhibitory. Our combined computational and in vitro data can be used to identify additional ENT-drug interactions to improve our understanding of drugs that can circumvent the BTB. SIGNIFICANCE STATEMENT: This study identified remdesivir and EIDD-1931 as substrates of equilibrative nucleoside transporters 1 and 2. This provides a potential mechanism for uptake of these drugs into cells and may be important for antiviral potential in the testes and other tissues expressing these transporters.
Collapse
Affiliation(s)
- Siennah R Miller
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Meghan E McGrath
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Kimberley M Zorn
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Sean Ekins
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Stephen H Wright
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| | - Nathan J Cherrington
- College of Pharmacy, Department of Pharmacology & Toxicology (S.R.M., M.E.M., N.J.C.) and Department of Physiology (S.H.W.), University of Arizona, Tucson, Arizona; and Collaborations Pharmaceuticals, Inc., Raleigh, North Carolina (K.M.Z., S.E.)
| |
Collapse
|
30
|
Wang Q, Wang F, Chen R, Liu W, Gao N, An J, Chen Y, Wu H, Han D. Differential Effects of Viral Nucleic Acid Sensor Signaling Pathways on Testicular Sertoli and Leydig Cells. Endocrinology 2021; 162:6359040. [PMID: 34453520 DOI: 10.1210/endocr/bqab180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Indexed: 12/17/2022]
Abstract
The human testis can be infected by a large number of RNA and DNA viruses. While various RNA virus infections may induce orchitis and impair testicular functions, DNA virus infection rarely affects the testis. Mechanisms underlying the differential effects of RNA and DNA viral infections on the testis remain unclear. In the current study, we therefore examined the effects of viral RNA and DNA sensor signaling pathways on mouse Sertoli cells (SC) and Leydig cells (LC). The local injection of viral RNA analogue polyinosinic-polycytidylic acid [poly(I:C)] into the testis markedly disrupted spermatogenesis, whereas the injection of the herpes simplex virus (HSV) DNA analogue HSV60 did not affect spermatogenesis. Poly(I:C) dramatically induced the expression of the proinflammatory cytokines tumor necrosis factor α (TNF-α) and interleukin 6 in SC and LC through Toll-like receptor 3 and interferon β promoter stimulator 1 signaling pathways, impairing the integrity of the blood-testis barrier and testosterone synthesis. Poly(I:C)-induced TNF-α production thus plays a critical role in the impairment of cell functions. In contrast, HSV60 predominantly induced the expression of type 1 interferons and antiviral proteins via the DNA sensor signaling pathway, which did not affect testicular cell functions. Accordingly, the Zika virus induced high levels of TNF-α in SC and LC and impaired their respective cellular functions, whereas Herpes simplex virus type 2 principally induced antiviral responses and did not impair such functions. These results provide insights into the mechanisms by which RNA viral infections impair testicular functions.
Collapse
Affiliation(s)
- Qing Wang
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wang
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Ran Chen
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Weihua Liu
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Na Gao
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yongmei Chen
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Han Wu
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Daishu Han
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Wu S, Frank I, Derby N, Martinelli E, Cheng CY. HIV-1 Establishes a Sanctuary Site in the Testis by Permeating the BTB Through Changes in Cytoskeletal Organization. Endocrinology 2021; 162:6338140. [PMID: 34343260 PMCID: PMC8407494 DOI: 10.1210/endocr/bqab156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Indexed: 11/19/2022]
Abstract
Studies suggest that HIV-1 invades the testis through initial permeation of the blood-testis barrier (BTB). The selectivity of the BTB to antiretroviral drugs makes this site a sanctuary for the virus. Little is known about how HIV-1 crosses the BTB and invades the testis. Herein, we used 2 approaches to examine the underlying mechanism(s) by which HIV-1 permeates the BTB and gains entry into the seminiferous epithelium. First, we examined if recombinant Tat protein was capable of perturbing the BTB and making the barrier leaky, using the primary rat Sertoli cell in vitro model that mimics the BTB in vivo. Second, we used HIV-1-infected Sup-T1 cells to investigate the activity of HIV-1 infection on cocultured Sertoli cells. Using both approaches, we found that the Sertoli cell tight junction permeability barrier was considerably perturbed and that HIV-1 effectively permeates the BTB by inducing actin-, microtubule-, vimentin-, and septin-based cytoskeletal changes in Sertoli cells. These studies suggest that HIV-1 directly perturbs BTB function, potentially through the activity of the Tat protein.
Collapse
Affiliation(s)
- Siwen Wu
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Ines Frank
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | - Nina Derby
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Elena Martinelli
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - C Yan Cheng
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| |
Collapse
|
32
|
Duan Q, Liu T, Huang C, Shao Q, Ma Y, Wang W, Liu T, Sun J, Fang J, Huang G, Chen Z. The Chinese Herbal Prescription JieZe-1 Inhibits Membrane Fusion and the Toll-like Receptor Signaling Pathway in a Genital Herpes Mouse Model. Front Pharmacol 2021; 12:707695. [PMID: 34630083 PMCID: PMC8497740 DOI: 10.3389/fphar.2021.707695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022] Open
Abstract
Chinese herbal prescription JieZe-1 is effective for genital herpes with no visible adverse effects clinically. It showed an excellent anti-HSV-2 effect in vitro. However, its mechanism of anti-HSV-2 effect in vivo remains unclear. This study was designed to evaluate the anti-HSV-2 effect of JieZe-1 and berberine in a genital herpes mouse model and explore the underlying mechanism. The fingerprint of JieZe-1 was determined by high-performance liquid chromatography. First, we optimized a mouse model of genital herpes. Next, the weight, symptom score, morphological changes, viral load, membrane fusion proteins, critical proteins of the Toll-like receptor signaling pathway, cytokines, and immune cells of vaginal tissue in mice at different time points were measured. Finally, we treated the genital herpes mouse model with JieZe-1 gel (2.5, 1.5, and 0.5 g/ml) and tested the above experimental indexes at 12 h and on the 9th day after modeling. JieZe-1 improved the symptoms, weight, and histopathological damage of genital herpes mice, promoted the keratin repair of tissues, and protected organelles to maintain the typical morphology of cells. It downregulated the expression of membrane fusion proteins, critical proteins of the Toll-like receptor signaling pathway, cytokines, and immune cells. The vaginal, vulvar, and spinal cord viral load and vaginal virus shedding were also significantly reduced. In summary, JieZe-1 shows significant anti-HSV-2 efficacy in vivo. The mechanism is related to the inhibition of membrane fusion, the Toll-like receptor signaling pathway, inflammatory cytokines, and cellular immunity. However, berberine, the main component of JieZe-1 monarch medicine, showed no efficacy at a concentration of 891.8 μM (0.3 mg/ml).
Collapse
Affiliation(s)
- Qianni Duan
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Liu
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong Huang
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Shao
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonggui Ma
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenjia Wang
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianli Liu
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Sun
- Department of Biochemistry and Molecular Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianguo Fang
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangying Huang
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Chen
- Department of TCM, Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Ratanakomol T, Roytrakul S, Wikan N, Smith DR. Berberine Inhibits Dengue Virus through Dual Mechanisms. Molecules 2021; 26:5501. [PMID: 34576974 PMCID: PMC8470584 DOI: 10.3390/molecules26185501] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/04/2021] [Accepted: 09/08/2021] [Indexed: 11/17/2022] Open
Abstract
Mosquito transmitted viruses, particularly those of the genus Flavivirus, are a significant healthcare burden worldwide, especially in tropical and sub-tropical areas. However, effective medicines for these viral infections remains lacking. Berberine (BBR) is an alkaloid found in some plants used in traditional medicines in Southeast Asia and elsewhere, and BBR has been shown to possess anti-viral activities. During a screen for potential application to mosquito transmitted viruses, BBR was shown to have virucidal activity against dengue virus (DENV; IC50 42.87 µM) as well as against Zika virus (IC50 11.42 µM) and chikungunya virus (IC50 14.21 µM). BBR was shown to have cellular effects that lead to an increase in cellular DENV E protein without a concomitant effect on DENV nonstructural proteins, suggesting an effect on viral particle formation or egress. While BBR was shown to have an effect of ERK1/2 activation this did not result in defects in viral egress mechanisms. The primary effect of BBR on viral production was likely to be through BBR acting through AMPK activation and disruption of lipid metabolism. Combined these results suggest that BBR has a dual effect on DENV infection, and BBR may have the potential for development as an anti-DENV antiviral.
Collapse
Affiliation(s)
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Rangsit 12120, Thailand;
| | - Nitwara Wikan
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand;
| | - Duncan R. Smith
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand;
| |
Collapse
|
34
|
Identification of potential biomarkers in dengue via integrated bioinformatic analysis. PLoS Negl Trop Dis 2021; 15:e0009633. [PMID: 34347790 PMCID: PMC8336846 DOI: 10.1371/journal.pntd.0009633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 07/07/2021] [Indexed: 11/19/2022] Open
Abstract
Dengue fever virus (DENV) is a global health threat that is becoming increasingly critical. However, the pathogenesis of dengue has not yet been fully elucidated. In this study, we employed bioinformatics analysis to identify potential biomarkers related to dengue fever and clarify their underlying mechanisms. The results showed that there were 668, 1901, and 8283 differentially expressed genes between the dengue-infected samples and normal samples in the GSE28405, GSE38246, and GSE51808 datasets, respectively. Through overlapping, a total of 69 differentially expressed genes (DEGs) were identified, of which 51 were upregulated and 18 were downregulated. We identified twelve hub genes, including MX1, IFI44L, IFI44, IFI27, ISG15, STAT1, IFI35, OAS3, OAS2, OAS1, IFI6, and USP18. Except for IFI44 and STAT1, the others were statistically significant after validation. We predicted the related microRNAs (miRNAs) of these 12 target genes through the database miRTarBase, and finally obtained one important miRNA: has-mir-146a-5p. In addition, gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment were carried out, and a protein–protein interaction (PPI) network was constructed to gain insight into the actions of DEGs. In conclusion, our study displayed the effectiveness of bioinformatics analysis methods in screening potential pathogenic genes in dengue fever and their underlying mechanisms. Further, we successfully predicted IFI44L and IFI6, as potential biomarkers with DENV infection, providing promising targets for the treatment of dengue fever to a certain extent. Dengue fever is a mosquito borne viral disease caused by a single stranded RNA virus with four serotypes. DENV infection can cause various diseases, such as breakbone fever, haemorrhagic fever, and shock syndrome. As one of the most viral diseases leading to incidence rate and mortality in animal arthropods, Dengue fever has become an increasingly serious global health threat. However, the pathogenesis of dengue fever has not been fully elucidated. In this study, we used bioinformatics analysis to identify potential biomarkers associated with dengue fever and elucidate their underlying mechanisms. Finally, we predicted that IFI44L and IFI6 might be potential biomarkers of DENV infection. This finding provides a promising target for the treatment of dengue fever to a certain extent. In addition, the Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment, protein–protein interaction (PPI) network were implemented to analyze the key differentially expressed genes after DENV infection, and the related mechanisms were illuminated by this study.
Collapse
|
35
|
Yao R, Ianevski A, Kainov D. Safe-in-Man Broad Spectrum Antiviral Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:313-337. [PMID: 34258746 DOI: 10.1007/978-981-16-0267-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Emerging and re-emerging viral diseases occur with regularity within the human population. The conventional 'one drug, one virus' paradigm for antivirals does not adequately allow for proper preparedness in the face of unknown future epidemics. In addition, drug developers lack the financial incentives to work on antiviral drug discovery, with most pharmaceutical companies choosing to focus on more profitable disease areas. Safe-in-man broad spectrum antiviral agents (BSAAs) can help meet the need for antiviral development by already having passed phase I clinical trials, requiring less time and money to develop, and having the capacity to work against many viruses, allowing for a speedy response when unforeseen epidemics arise. In this chapter, we discuss the benefits of repurposing existing drugs as BSAAs, describe the major steps in safe-in-man BSAA drug development from discovery through clinical trials, and list several database resources that are useful tools for antiviral drug repositioning.
Collapse
Affiliation(s)
- Rouan Yao
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Institute of Technology, University of Tartu, Tartu, Estonia.
- Institute for Molecule Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
36
|
Pletnev AG, Maximova OA, Liu G, Kenney H, Nagata BM, Zagorodnyaya T, Moore I, Chumakov K, Tsetsarkin KA. Epididymal epithelium propels early sexual transmission of Zika virus in the absence of interferon signaling. Nat Commun 2021; 12:2469. [PMID: 33927207 PMCID: PMC8084954 DOI: 10.1038/s41467-021-22729-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/23/2021] [Indexed: 11/21/2022] Open
Abstract
Recognition of Zika virus (ZIKV) sexual transmission (ST) among humans challenges our understanding of the maintenance of mosquito-borne viruses in nature. Here we dissected the relative contributions of the components of male reproductive system (MRS) during early male-to-female ZIKV transmission by utilizing mice with altered antiviral responses, in which ZIKV is provided an equal opportunity to be seeded in the MRS tissues. Using microRNA-targeted ZIKV clones engineered to abolish viral infectivity to different parts of the MRS or a library of ZIKV genomes with unique molecular identifiers, we pinpoint epithelial cells of the epididymis (rather than cells of the testis, vas deferens, prostate, or seminal vesicles) as a most likely source of the sexually transmitted ZIKV genomes during the early (most productive) phase of ZIKV shedding into the semen. Incorporation of this mechanistic knowledge into the development of a live-attenuated ZIKV vaccine restricts its ST potential.
Collapse
Affiliation(s)
- Alexander G Pletnev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olga A Maximova
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Guangping Liu
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kenney
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bianca M Nagata
- Infectious Disease and Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tatiana Zagorodnyaya
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Ian Moore
- Infectious Disease and Pathogenesis Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Konstantin Chumakov
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Konstantin A Tsetsarkin
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
37
|
Enzyme-Linked Immunosorbent Assay and Quantitative Reverse Transcription PCR as a Technique to Analyze Inflammation. Methods Mol Biol 2021. [PMID: 32367360 DOI: 10.1007/978-1-0716-0581-3_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Inflammation is part of a defense reaction of live tissues that is triggered by pathogens, chemical reagents, trauma, and radiation. Understanding the inflammatory process triggered by Zika virus (ZIKV) is important to better understand the pathogen-host interaction. The evaluation of this process can be done using tools such as enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription PCR (RT-qPCR). Both techniques have been an indispensable tool not just for immunologists but for all interested in understanding the inflammatory process.
Collapse
|
38
|
Zika RNA and Flavivirus-Like Antigens in the Sperm Cells of Symptomatic and Asymptomatic Subjects. Viruses 2021; 13:v13020152. [PMID: 33494175 PMCID: PMC7909808 DOI: 10.3390/v13020152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/26/2020] [Accepted: 01/08/2021] [Indexed: 01/03/2023] Open
Abstract
Zika virus (ZIKV) RNA has been found to remain in human semen for up to one year after infection, but the presence of Flavivirus antigens in the different compartments of semen has been largely unexplored. Following the introduction of ZIKV in Nicaragua (2016), a prospective study of patients with clinical symptoms consistent with ZIKV was conducted in León to investigate virus shedding in different fluids. ZIKV infection was confirmed in 16 male subjects (≥18 years of age) by RT-qPCR in either blood, saliva or urine. Of these, three provided semen samples at 7, 14, 21, 28, 60 and 180 days postsymptom onset (DPSO) for Flavivirus antigens and RNA studies. These cases were compared with 19 asymptomatic controls. Flavivirus antigens were examined by immunofluorescence (IF) using the 4G2 Mabs, and confocal microscopy was used to explore fluorescence patterns. The three (100%) symptomatic subjects and 3 (16%) of the 19 asymptomatic subjects had Flavivirus antigens and viral RNA in the spermatozoa fraction. The percentage of IF Flavivirus-positive spermatozoa cells ranged from 1.9% to 25% in specimens from symptomatic subjects, as compared with 0.8% to 3.8% in specimens from asymptomatic controls. A marked IF-pattern in the cytoplasmic droplets and tail of the spermatozoa was observed. The sperm concentrations (45 × 106/mL vs. 63.5 × 106/mL, p = 0.041) and the total motility percentage (54% vs. 75%, p = 0.009) were significantly lower in specimens from ZIKV-positive than in those of ZIKV-negative. In conclusion, this study demonstrated the presence of Flavivirus antigens and RNA within a time frame of 28 DPSO in sperm cells of symptomatic and asymptomatic subjects during the ZIKV epidemic. These findings have implications for public health, in terms of nonarthropod-born, silent transmission facilitated by sperm cells and potential transmission from asymptomatic males to pregnant women, with consequences to the fetus.
Collapse
|
39
|
Chloroquine and Sulfadoxine Derivatives Inhibit ZIKV Replication in Cervical Cells. Viruses 2020; 13:v13010036. [PMID: 33383619 PMCID: PMC7823661 DOI: 10.3390/v13010036] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/22/2022] Open
Abstract
Despite the severe morbidity caused by Zika fever, its specific treatment is still a challenge for public health. Several research groups have investigated the drug repurposing of chloroquine. However, the highly toxic side effect induced by chloroquine paves the way for the improvement of this drug for use in Zika fever clinics. Our aim is to evaluate the anti-Zika virus (ZIKV) effect of hybrid compounds derived from chloroquine and sulfadoxine antimalarial drugs. The antiviral activity of hybrid compounds (C-Sd1 to C-Sd7) was assessed in an in-vitro model of human cervical and Vero cell lines infected with a Brazilian (BR) ZIKV strain. First, we evaluated the cytotoxic effect on cultures treated with up to 200 µM of C-Sds and observed CC50 values that ranged from 112.0 ± 1.8 to >200 µM in cervical cells and 43.2 ± 0.4 to 143.0 ± 1.3 µM in Vero cells. Then, the cultures were ZIKV-infected and treated with up to 25 µM of C-Sds for 48 h. The treatment of cervical cells with C-Sds at 12 µM induced a reduction of 79.8% ± 4.2% to 90.7% ± 1.5% of ZIKV-envelope glycoprotein expression in infected cells as compared to 36.8% ± 2.9% of infection in vehicle control. The viral load was also investigated and revealed a reduction of 2- to 3-logs of ZIKV genome copies/mL in culture supernatants compared to 6.7 ± 0.7 × 108 copies/mL in vehicle control. The dose-response curve by plaque-forming reduction (PFR) in cervical cells revealed a potent dose-dependent activity of C-Sds in inhibiting ZIKV replication, with PFR above 50% and 90% at 6 and 12 µM, respectively, while 25 µM inhibited 100% of viral progeny. The treatment of Vero cells at 12 µM led to 100% PFR, confirming the C-Sds activity in another cell type. Regarding effective concentration in cervical cells, the EC50 values ranged from 3.2 ± 0.1 to 5.0 ± 0.2 µM, and the EC90 values ranged from 7.2 ± 0.1 to 11.6 ± 0.1 µM, with selectivity index above 40 for most C-Sds, showing a good therapeutic window. Here, our aim is to investigate the anti-ZIKV activity of new hybrid compounds that show highly potent efficacy as inhibitors of ZIKV in-vitro infection. However, further studies will be needed to investigate whether these new chemical structures can lead to the improvement of chloroquine antiviral activity.
Collapse
|
40
|
Yang W, Wu YH, Liu SQ, Sheng ZY, Zhen ZD, Gao RQ, Cui XY, Fan DY, Qin ZH, Zheng AH, Wang PG, An J. S100A4+ macrophages facilitate zika virus invasion and persistence in the seminiferous tubules via interferon-gamma mediation. PLoS Pathog 2020; 16:e1009019. [PMID: 33315931 PMCID: PMC7769614 DOI: 10.1371/journal.ppat.1009019] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/28/2020] [Accepted: 10/01/2020] [Indexed: 12/12/2022] Open
Abstract
Testicular invasion and persistence are features of Zika virus (ZIKV), but their mechanisms are still unknown. Here, we showed that S100A4+ macrophages, a myeloid macrophage subpopulation with susceptibility to ZIKV infection, facilitated ZIKV invasion and persistence in the seminiferous tubules. In ZIKV-infected mice, S100A4+ macrophages were specifically recruited into the interstitial space of testes and differentiated into interferon-γ-expressing M1 macrophages. With interferon-γ mediation, S100A4+ macrophages down-regulated Claudin-1 expression and induced its redistribution from the cytosol to nucleus, thus increasing the permeability of the blood-testis barrier which facilitated S100A4+ macrophages invasion into the seminiferous tubules. Intraluminal S100A4+ macrophages were segregated from CD8+ T cells and consequently helped ZIKV evade cellular immunity. As a result, ZIKV continued to replicate in intraluminal S100A4+ macrophages even when the spermatogenic cells disappeared. Deficiencies in S100A4 or interferon-γ signaling both reduced ZIKV infection in the seminiferous tubules. These results demonstrated crucial roles of S100A4+ macrophages in ZIKV infection in testes.
Collapse
Affiliation(s)
- Wei Yang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan-Hua Wu
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuang-Qing Liu
- Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Zi-Yang Sheng
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zi-Da Zhen
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rui-Qi Gao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Yun Cui
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Science and Technology, Capital Institute of Pediatrics, Beijing, China
| | - Dong-Ying Fan
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhi-Hai Qin
- Institute of Biophysics, Chinese Academy of Science, Beijing, China
| | - Ai-Hua Zheng
- Institute of Zoology, Chinese Academy of Science, Beijing, China
| | - Pei-Gang Wang
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- * E-mail: (PGW); , (JA)
| | - Jing An
- Department of Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
- * E-mail: (PGW); , (JA)
| |
Collapse
|
41
|
Wang R, Gornalusse GG, Kim Y, Pandey U, Hladik F, Vojtech L. Potent Restriction of Sexual Zika Virus Infection by the Lipid Fraction of Extracellular Vesicles in Semen. Front Microbiol 2020; 11:574054. [PMID: 33133043 PMCID: PMC7550675 DOI: 10.3389/fmicb.2020.574054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/07/2020] [Indexed: 12/31/2022] Open
Abstract
Sexual Zika virus (ZIKV) transmission from men to women occurs less frequently than the often-detected high viral loads in semen would suggest, but worries that this transmission route predisposes to fetal damage in pregnant women remain. To better understand sexual ZIKV pathogenesis, we studied the permissiveness of the human female genital tract to infection and the effect of semen on this process. ZIKV replicates in vaginal tissues and primary epithelial cells from the vagina, ectocervix, and endocervix and induces an innate immune response, but also continues to replicate without cytopathic effect. Infection of genital cells and tissues is strongly inhibited by extracellular vesicles (EV) in semen at physiological vesicle-to-virus ratios. Liposomes with the same composition as semen EVs also impair infection, indicating that the EV’s lipid fraction, rather than their protein or RNA cargo, is responsible for this anti-viral effect. Thus, EVs in semen potently restrict ZIKV transmission, but the virus propagates well once infection in the recipient mucosa has been established.
Collapse
Affiliation(s)
- Ruofan Wang
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Germán G Gornalusse
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Yeseul Kim
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Urvashi Pandey
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States.,Division of Allergy and Infectious Diseases, Department of Medicine, University of Washington, Seattle, WA, United States.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| |
Collapse
|
42
|
Blitvich BJ, Magalhaes T, Laredo-Tiscareño SV, Foy BD. Sexual Transmission of Arboviruses: A Systematic Review. Viruses 2020; 12:v12090933. [PMID: 32854298 PMCID: PMC7552039 DOI: 10.3390/v12090933] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are primarily maintained in nature in transmission cycles between hematophagous arthropods and vertebrate hosts, but an increasing number of arboviruses have been isolated from or indirectly detected in the urogenital tract and sexual secretions of their vertebrate hosts, indicating that further investigation on the possibility of sexual transmission of these viruses is warranted. The most widely recognized sexually-transmitted arbovirus is Zika virus but other arboviruses, including Crimean-Congo hemorrhagic fever virus and dengue virus, might also be transmitted, albeit occasionally, by this route. This review summarizes our current understanding on the ability of arboviruses to be sexually transmitted. We discuss the sexual transmission of arboviruses between humans and between vertebrate animals, but not arthropod vectors. Every taxonomic group known to contain arboviruses (Asfarviridae, Bunyavirales, Flaviviridae, Orthomyxoviridae, Reoviridae, Rhabdoviridae and Togaviridae) is covered.
Collapse
Affiliation(s)
- Bradley J. Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
- Correspondence: ; Tel.: +1-515-294-9861; Fax: +1-515-294-8500
| | - Tereza Magalhaes
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| | - S. Viridiana Laredo-Tiscareño
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Brian D. Foy
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| |
Collapse
|
43
|
Botwina P, Obłoza M, Szczepański A, Szczubiałka K, Nowakowska M, Pyrć K. In Vitro Inhibition of Zika Virus Replication with Poly(Sodium 4-Styrenesulfonate). Viruses 2020; 12:E926. [PMID: 32842540 PMCID: PMC7551931 DOI: 10.3390/v12090926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 12/28/2022] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne pathogen associated with microcephaly and other congenital abnormalities in newborns as well as neurologic complications in adults. The explosive transmission of the virus in the last ten years put it in the limelight and improved our understanding of its biology and pathology. Currently, no vaccine or drugs are available to prevent or treat ZIKV infections. Knowing the potential of flaviviruses to broaden their geographic distribution, as observed for the West Nile virus, it is of importance to develop novel antiviral strategies. In this work, we identified poly(sodium 4-styrenesulfonate) (PSSNa) as a new polymeric ZIKV inhibitor. We demonstrated that PSSNa inhibits ZIKV replication in vitro both in animal and human cells, while no cytotoxicity is observed. Our mechanistic studies indicated that PSSNa acts mostly through direct binding to ZIKV particle and blocking its attachment to the host cells.
Collapse
Affiliation(s)
- Paweł Botwina
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (P.B.); (A.S.)
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Magdalena Obłoza
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.O.); (K.S.); (M.N.)
| | - Artur Szczepański
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (P.B.); (A.S.)
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387 Krakow, Poland
| | - Krzysztof Szczubiałka
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.O.); (K.S.); (M.N.)
| | - Maria Nowakowska
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (M.O.); (K.S.); (M.N.)
| | - Krzysztof Pyrć
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, 30-387 Krakow, Poland; (P.B.); (A.S.)
| |
Collapse
|
44
|
Schmidt JK, Mean KD, Puntney RC, Alexander ES, Sullivan R, Simmons HA, Zeng X, Weiler AM, Friedrich TC, Golos TG. Zika virus in rhesus macaque semen and reproductive tract tissues: a pilot study of acute infection†. Biol Reprod 2020; 103:1030-1042. [PMID: 32761051 DOI: 10.1093/biolre/ioaa137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/09/2019] [Accepted: 07/30/2020] [Indexed: 12/19/2022] Open
Abstract
Although sexual transmission of Zika virus (ZIKV) is well-documented, the viral reservoir(s) in the male reproductive tract remains uncertain in humans and immune-intact animal models. We evaluated the presence of ZIKV in a rhesus macaque pilot study to determine persistence in semen, assess the impact of infection on sperm functional characteristics, and define the viral reservoir in the male reproductive tract. Five adult male rhesus monkeys were inoculated with 105 PFU of Asian-lineage ZIKV isolate PRVABC59, and two males were inoculated with the same dose of African-lineage ZIKV DAKAR41524. Viremia and viral RNA (vRNA) shedding in semen were monitored, and a cohort of animals were necropsied for tissue collection to assess tissue vRNA burden and histopathology. All animals exhibited viremia for limited periods (1-11 days); duration of shedding did not differ significantly between viral isolates. There were sporadic low levels of vRNA in the semen from some, but not all animals. Viral RNA levels in reproductive tract tissues were also modest and present in the epididymis in three of five cases, one case in the vas deferens, but not detected in testis, seminal vesicles or prostate. ZIKV infection did not impact semen motility parameters as assessed by computer-assisted sperm analysis. Despite some evidence of prolonged ZIKV RNA shedding in human semen and high tropism of ZIKV for male reproductive tract tissues in mice deficient in Type 1 interferon signaling, in the rhesus macaques assessed in this pilot study, we did not consistently find ZIKV RNA in the male reproductive tract.
Collapse
Affiliation(s)
- Jenna K Schmidt
- Wisconsin National Primate Research Center, Madison, WI, USA
| | | | - Riley C Puntney
- Wisconsin National Primate Research Center, Madison, WI, USA
| | | | - Ruth Sullivan
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Frederick, MD, USA
| | - Andrea M Weiler
- Wisconsin National Primate Research Center, Madison, WI, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, Madison, WI, USA.,Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, Madison, WI, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
45
|
van der Kuyl AC, Berkhout B. Viruses in the reproductive tract: On their way to the germ line? Virus Res 2020; 286:198101. [PMID: 32710926 DOI: 10.1016/j.virusres.2020.198101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 01/13/2023]
Abstract
Studies of vertebrate genomes have indicated that all species contain in their chromosomes stretches of DNA with sequence similarity to viral genomes. How such 'endogenous' viral elements (EVEs) ended up in host genomes is usually explained in general terms such as 'they entered the germ line at some point during evolution'. This seems a correct statement, but is also rather imprecise. The vast number of endogenous viral sequences suggest that common routes to the 'germ line' may exist, as relying on chance alone may not easily explain the abundance of EVEs in modern mammalian genomes. An increasing number of virus types have been detected in human semen and a growing number of studies have reported on viral infections that cause male infertility or subfertility and on viral infections that threaten in vitro fertilisation practices. Thus, it is timely to survey the pathway(s) that viruses can use to gain access to the human germ line. Embryo transfer and semen quality studies in livestock form another source of relevant information because virus infection during reproduction is clearly unwanted, as is the case for the human situation. In this review, studies on viruses in the male and female reproductive tract and in the early embryo will be discussed to propose a plausible viral route to the mammalian germ line.
Collapse
Affiliation(s)
- Antoinette Cornelia van der Kuyl
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands.
| | - Ben Berkhout
- Laboratory of Experimental Virology, Department of Medical Microbiology, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Abstract
Alphaviruses are enveloped positive-sense RNA viruses that can cause serious human illnesses such as polyarthritis and encephalitis. Despite their widespread distribution and medical importance, there are no licensed vaccines or antivirals to combat alphavirus infections. Berberine chloride (BBC) is a pan-alphavirus inhibitor that was previously identified in a replicon-based small-molecule screen. This work showed that BBC inhibits alphavirus replication but also suggested that BBC might have additional effects later in the viral life cycle. Here, we show that BBC has late effects that target the virus nucleocapsid (NC) core. Infected cells treated with BBC late in infection were unable to form stable cytoplasmic NCs or assembly intermediates, as assayed by gradient sedimentation. In vitro studies with recombinant capsid protein (Cp) and purified genomic RNA (gRNA) showed that BBC perturbs core-like particle formation and potentially traps the assembly process in intermediate states. Particles produced from BBC-treated cells were less infectious, despite efficient particle production and only minor decreases in genome packaging. In addition, BBC treatment of free virus particles strongly decreased alphavirus infectivity. In contrast, the infectivity of the negative-sense RNA virus vesicular stomatitis virus was resistant to BBC treatment of infected cells or free virus. Together, our data indicate that BBC alters alphavirus Cp-gRNA interactions and oligomerization and suggest that this may cause defects in NC assembly and in disassembly during subsequent virus entry. Thus, BBC may be considered a novel alphavirus NC assembly inhibitor.IMPORTANCE The alphavirus chikungunya virus (CHIKV) is an example of an emerging human pathogen with increased and rapid global spread. Although an acute CHIKV infection is rarely fatal, many patients suffer from debilitating chronic arthralgia for years. Antivirals against chikungunya and other alphaviruses have been identified in vitro, but to date none have been shown to be efficacious and have been licensed for human use. Here, we investigated a small molecule, berberine chloride (BBC), and showed that it inhibited infectious virus production by several alphaviruses including CHIKV. BBC acted on a late step in the alphavirus exit pathway, namely the formation of the nucleocapsid containing the infectious viral RNA. Better understanding of nucleocapsid formation and its inhibition by BBC will provide important information on the mechanisms of infectious alphavirus production and may enable their future targeting in antiviral strategies.
Collapse
|
47
|
Rashid MU, Zahedi-Amiri A, Glover KKM, Gao A, Nickol ME, Kindrachuk J, Wilkins JA, Coombs KM. Zika virus dysregulates human Sertoli cell proteins involved in spermatogenesis with little effect on tight junctions. PLoS Negl Trop Dis 2020; 14:e0008335. [PMID: 32511241 PMCID: PMC7279580 DOI: 10.1371/journal.pntd.0008335] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV), a neglected tropical disease until its re-emergence in 2007, causes microcephaly in infants and Guillain-Barré syndrome in adults. Its re-emergence and spread to more than 80 countries led the World Health Organization in 2016 to declare a Public Health Emergency. ZIKV is mainly transmitted by mosquitos, but can persist in infected human male semen for prolonged periods and may be sexually transmitted. Testicular Sertoli cells support ZIKV replication and may be a reservoir for persistent ZIKV infection. Electrical impedance analyses indicated ZIKV infection rapidly disrupted Vero cell monolayers but had little effect upon human Sertoli cells (HSerC). We determined ZIKV-induced proteomic changes in HSerC using an aptamer-based multiplexed technique (SOMAscan) targeting >1300 human proteins. ZIKV infection caused differential expression of 299 proteins during three different time points, including 5 days after infection. Dysregulated proteins are involved in different bio-functions, including cell death and survival, cell cycle, maintenance of cellular function, cell signaling, cellular assembly, morphology, movement, molecular transport, and immune response. Many signaling pathways important for maintenance of HSerC function and spermatogenesis were highly dysregulated. These included IL-6, IGF1, EGF, NF-κB, PPAR, ERK/MAPK, and growth hormone signaling. Down-regulation of the PPAR signaling pathway might impact cellular energy supplies. Upstream molecule analysis also indicated microRNAs involved in germ cell development were downregulated by infection. Overall, this study leads to a better understanding of Sertoli cellular mechanisms used by ZIKV during persistent infection and possible ZIKV impacts on spermatogenesis.
Collapse
Affiliation(s)
- Mahamud-ur Rashid
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
| | - Ali Zahedi-Amiri
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
| | - Kathleen K. M. Glover
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
| | - Ang Gao
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
| | - Michaela E. Nickol
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
| | - Jason Kindrachuk
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
| | - John A. Wilkins
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
- University of Manitoba, Department of Internal Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Kevin M. Coombs
- University of Manitoba, Department of Medical Microbiology and Infectious Diseases, Winnipeg, Manitoba, Canada
- Manitoba Centre for Proteomics & Systems Biology, Winnipeg, Manitoba, Canada
- Children’s Hospital Research Institute of Manitoba, John Buhler Research Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
48
|
Immune-profiling of ZIKV-infected patients identifies a distinct function of plasmacytoid dendritic cells for immune cross-regulation. Nat Commun 2020; 11:2421. [PMID: 32415086 PMCID: PMC7229207 DOI: 10.1038/s41467-020-16217-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/22/2020] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne pathogen with increasing public health significance. To characterize immune responses to ZIKV, here we examine transcriptional signatures of CD4 T, CD8 T, B, and NK cells, monocytes, myeloid dendritic cells (mDCs), and plasmacytoid dendritic cells (pDCs) from three individuals with ZIKV infection. While gene expression patterns from most cell subsets display signs of impaired antiviral immune activity, pDCs from infected host have distinct transcriptional response associated with activation of innate immune recognition and type I interferon signaling pathways, but downregulation of key host factors known to support ZIKV replication steps; meanwhile, pDCs exhibit a unique expression pattern of gene modules that are correlated with alternative cell populations, suggesting collaborative interactions between pDCs and other immune cells, particularly B cells. Together, these results point towards a discrete but integrative function of pDCs in the human immune responses to ZIKV infection.
Collapse
|
49
|
Song D, Hao J, Fan D. Biological properties and clinical applications of berberine. Front Med 2020; 14:564-582. [DOI: 10.1007/s11684-019-0724-6] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023]
|
50
|
The Cellular Impact of the ZIKA Virus on Male Reproductive Tract Immunology and Physiology. Cells 2020; 9:cells9041006. [PMID: 32325652 PMCID: PMC7226248 DOI: 10.3390/cells9041006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
Zika virus (ZIKV) has been reported by several groups as an important virus causing pathological damage in the male reproductive tract. ZIKV can infect and persist in testicular somatic and germ cells, as well as spermatozoa, leading to cell death and testicular atrophy. ZIKV has also been detected in semen samples from ZIKV-infected patients. This has huge implications for human reproduction. Global scientific efforts are being applied to understand the mechanisms related to arboviruses persistency, pathogenesis, and host cellular response to suggest a potential target to develop robust antiviral therapeutics and vaccines. Here, we discuss the cellular modulation of the immunologic and physiologic properties of the male reproductive tract environment caused by arboviruses infection, focusing on ZIKV. We also present an overview of the current vaccine effects and therapeutic targets against ZIKV infection that may impact the testis and male fertility.
Collapse
|