305 results on '"cellular tropism"'
Search Results
2. Tropism and immune response of chikungunya and zika viruses: An overview
- Author
-
Ravindran, Shilpa and Lahon, Anismrita
- Published
- 2023
- Full Text
- View/download PDF
3. AIP56, an AB toxin secreted by Photobacterium damselae subsp. piscicida, has tropism for myeloid cells
- Author
-
Inês Lua Freitas, Maria Fátima Macedo, Liliana Oliveira, Pedro Oliveira, Ana do Vale, and Nuno M.S. dos Santos
- Subjects
photobacteriosis ,virulence factor ,toxins ,cellular tropism ,leukocytes ,macrophages ,Immunologic diseases. Allergy ,RC581-607 - Abstract
IntroductionThe AB-type toxin AIP56 is a key virulence factor of Photobacterium damselae subsp. piscicida (Phdp), inducing apoptosis in fish immune cells. The discovery of AIP56-like and AIP56-related toxins in diverse organisms, including human-associated Vibrio strains, highlights the evolutionary conservation of this toxin family, suggesting that AIP56 and its homologs may share conserved receptors across species. These toxins have potential for biotechnological applications, such as therapeutic protein delivery and immune modulation.MethodsHerein, the cell specificity of AIP56 for immune cells was characterized. The tropism of AIP56 for cells of the sea bass, mouse and human immune system was analyzed by following toxin internalization by flow cytometry and arrival of the toxin in the cytosol by evaluating the cleavage of NF-kB p65 by western blotting.ResultsOnly a small population of sea bass neutrophils internalized AIP56, indicating that most of the neutrophilic destruction during Phdp infection and/or AIP56 intoxication does not result from the direct action of the toxin. Moreover, the cellular tropism of AIP56 for myeloid cells was observed in the three species, including its preference for macrophages. Further, mouse and human M0 and M2-like macrophages internalized more toxin than M1-like macrophages. Despite the limited interaction of lymphoid cells with AIP56, mouse B1-cells were able to internalize the toxin, possibly due to its myeloid features.ConclusionAIP56 has tropism for sea bass, mouse and human myeloid cells, with greater affinity for macrophages. This points to an evolutionary conservation of its receptor(s) and mechanism of action across species, raising the possibility that AIP56-like and -related toxins may also play a role in pathogenesis. These findings are relevant for both pathogenicity and biomedical contexts.
- Published
- 2025
- Full Text
- View/download PDF
4. Transcriptional profiling of zebrafish intestines identifies macrophages as host cells for human norovirus infection
- Author
-
Emma Roux, Reegan J. Willms, Jana Van Dycke, Álvaro Cortes Calabuig, Lore Van Espen, Geert Schoofs, Jelle Matthijnssens, Johan Neyts, Peter de Witte, Edan Foley, and Joana Rocha-Pereira
- Subjects
Human norovirus ,cellular tropism ,macrophages ,intestinal epithelium ,host cell identification ,host-virus interaction ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
Human noroviruses (HuNoVs) are a major cause of diarrheal disease, yet critical aspects of their biology, including cellular tropism, remain unclear. Although research has traditionally focused on the intestinal epithelium, the hypothesis that HuNoV infects macrophages has been recurrently discussed and is investigated here using a zebrafish larval model. Through single-cell RNA sequencing of dissected zebrafish intestines, we unbiasedly identified macrophages as host cells for HuNoV replication, with all three open reading frames mapped to individual macrophages. Notably, HuNoV preferentially infects actively phagocytosing inflammatory macrophages. HuNoV capsid proteins and double-stranded RNA colocalized within intestinal macrophages of infected zebrafish larvae, and the negative-strand RNA intermediate was detected within FACS-sorted macrophages. Flow cytometry confirmed viral replication within these macrophages, constituting approximately 23% of HuNoV’s host cells. Identifying macrophages as host cells prompts a reevaluation of their role in HuNoV pathogenesis, offering new directions for understanding and controlling this infection.
- Published
- 2024
- Full Text
- View/download PDF
5. A novel mouse model for investigating α-synuclein aggregates in oligodendrocytes: implications for the glial cytoplasmic inclusions in multiple system atrophy.
- Author
-
Ishimoto, Tomoyuki, Oono, Miki, Kaji, Seiji, Ayaki, Takashi, Nishida, Katsuya, Funakawa, Itaru, Maki, Takakuni, Matsuzawa, Shu-ichi, Takahashi, Ryosuke, and Yamakado, Hodaka
- Subjects
- *
MULTIPLE system atrophy , *OLIGODENDROGLIA , *ALPHA-synuclein , *LABORATORY mice , *ANIMAL disease models , *FLUORESCENT proteins , *CELL aggregation - Abstract
The aggregated alpha-synuclein (αsyn) in oligodendrocytes (OLGs) is one of the pathological hallmarks in multiple system atrophy (MSA). We have previously reported that αsyn accumulates not only in neurons but also in OLGs long after the administration of αsyn preformed fibrils (PFFs) in mice. However, detailed spatial and temporal analysis of oligodendroglial αsyn aggregates was technically difficult due to the background neuronal αsyn aggregates. The aim of this study is to create a novel mouse that easily enables sensitive and specific detection of αsyn aggregates in OLGs and the comparable analysis of the cellular tropism of αsyn aggregates in MSA brains. To this end, we generated transgenic (Tg) mice expressing human αsyn-green fluorescent protein (GFP) fusion proteins in OLGs under the control of the 2', 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter (CNP-SNCAGFP Tg mice). Injection of αsyn PFFs in these mice induced distinct GFP-positive aggregates in the processes of OLGs as early as one month post-inoculation (mpi), and their number and size increased in a centripetal manner. Moreover, MSA-brain homogenates (BH) induced significantly more oligodendroglial αsyn aggregates than neuronal αsyn aggregates compared to DLB-BH in CNP-SNCAGFP Tg mice, suggestive of their potential tropism of αsyn seeds for OLGs. In conclusion, CNP-SNCAGFP Tg mice are useful for studying the development and tropism of αsyn aggregates in OLGs and could contribute to the development of therapeutics targeting αsyn aggregates in OLGs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Single-cell profiling of African swine fever virus disease in the pig spleen reveals viral and host dynamics.
- Author
-
Zixiang Zhu, Ruoqing Mao, Baohong Liu, Huanan Liu, Zhengwang Shi, Kunpeng Zhang, Huisheng Liu, Danyang Zhang, Jia Liu, Zhenxiang Zhao, Kangli Li, Fan Yang, Weijun Cao, Xiangle Zhang, Chaochao Shen, Dehui Sun, Liyuan Wang, Hong Tian, Yi Ru, and Tao Feng
- Subjects
- *
AFRICAN swine fever virus , *VIRUS diseases , *AFRICAN swine fever , *CLASSICAL swine fever , *SPLEEN , *SWINE farms - Abstract
African swine fever, one of the major viral diseases of swine, poses an imminent threat to the global pig industry. The high-efficient replication of the causative agent .African swine fever virus (ASFV) in various organs in pigs greatly contributes to the disease. However, how ASFV manipulates the cell population to drive high-efficient replication of the virus in vivo remains unclear. Here, we found that the spleen reveals the most severe pathological manifestation with the highest viral loads among various organs in pigs during ASFV infection. By using single-cell-RNA-sequencing technology and multiple methods, we determined that macrophages and monocytes are the major cell types infected by ASFV in the spleen, showing high viral-load heterogeneity. A rare subpopulation of immature monocytes represents the major population infected at late infection stage. ASFV causes massive death of macrophages, but shifts its infection into these monocytes which significantly arise after the infection. The apoptosis, interferon response, and antigen-presentation capacity are inhibited in these monocytes which benefits prolonged infection of ASFV in vivo. Until now, the role of immature monocytes as an important target by ASFV has been overlooked due to that they do not express classical monocyte marker CD14. The present study indicates that the shift of viral infection from macrophages to the immature monocytes is critical for maintaining prolonged ASFV infection in vivo. This study sheds light on ASFV tropism, replica-tion, and infection dynamics, and elicited immune response, which may instruct future research on antiviral strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Pathogenicity and virulence of Marburg virus
- Author
-
Mehedy Hasan Abir, Tanjilur Rahman, Ayan Das, Silvia Naznin Etu, Iqbal Hossain Nafiz, Ahmed Rakib, Saikat Mitra, Talha Bin Emran, Kuldeep Dhama, Ariful Islam, Abolghasem Siyadatpanah, Shafi Mahmud, Bonlgee Kim, and Mohammad Mahmudul Hassan
- Subjects
Marburg virus ,epidemiology ,pathogenicity ,transmission dynamics ,cellular tropism ,virulence ,Infectious and parasitic diseases ,RC109-216 - Abstract
Marburg virus (MARV) has been a major concern since 1967, with two major outbreaks occurring in 1998 and 2004. Infection from MARV results in severe hemorrhagic fever, causing organ dysfunction and death. Exposure to fruit bats in caves and mines, and human-to-human transmission had major roles in the amplification of MARV outbreaks in African countries. The high fatality rate of up to 90% demands the broad study of MARV diseases (MVD) that correspond with MARV infection. Since large outbreaks are rare for MARV, clinical investigations are often inadequate for providing the substantial data necessary to determine the treatment of MARV disease. Therefore, an overall review may contribute to minimizing the limitations associated with future medical research and improve the clinical management of MVD. In this review, we sought to analyze and amalgamate significant information regarding MARV disease epidemics, pathophysiology, and management approaches to provide a better understanding of this deadly virus and the associated infection.
- Published
- 2022
- Full Text
- View/download PDF
8. SARS‐CoV‐2 cellular tropism and direct multiorgan failure in COVID‐19 patients: Bioinformatic predictions, experimental observations, and open questions.
- Author
-
Valyaeva, Anna A., Zharikova, Anastasia A., and Sheval, Eugene V.
- Subjects
- *
SARS-CoV-2 , *COVID-19 , *MULTIPLE organ failure , *COVID-19 pandemic - Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2), the virus that causes coronavirus disease 2019 (COVID‐19), has led to an unprecedented public health emergency worldwide. While common cold symptoms are observed in mild cases, COVID‐19 is accompanied by multiorgan failure in severe patients. Organ damage in COVID‐19 patients is partially associated with the indirect effects of SARS‐CoV‐2 infection (e.g., systemic inflammation, hypoxic‐ischemic damage, coagulopathy), but early processes in COVID‐19 patients that trigger a chain of indirect effects are connected with the direct infection of cells by the virus. To understand the virus transmission routes and the reasons for the wide‐spectrum of complications and severe outcomes of COVID‐19, it is important to identify the cells targeted by SARS‐CoV‐2. This review summarizes the major steps of investigation and the most recent findings regarding SARS‐CoV‐2 cellular tropism and the possible connection between the early stages of infection and multiorgan failure in COVID‐19. The SARS‐CoV‐2 pandemic is the first epidemic in which data extracted from single‐cell RNA‐seq (scRNA‐seq) gene expression data sets have been widely used to predict cellular tropism. The analysis presented here indicates that the SARS‐CoV‐2 cellular tropism predictions are accurate enough for estimating the potential susceptibility of different cells to SARS‐CoV‐2 infection; however, it appears that not all susceptible cells may be infected in patients with COVID‐19. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Characterisation of enterovirus A71 tropism, host cell proteomic impact and sensitivity to novel inhibitors of virus infection
- Author
-
Martin, Gael and Martin, Gael
- Abstract
The persistent surge in viral infections worldwide poses a significant and growing challenge. Effectively managing the impact of viral outbreaks on public health requires a thorough understanding of the underlying pathogens. One of the current demographics most at risk is children under the age of five. In the Asia-Pacific region, a major cause for concern is hand, foot, and mouth disease (HFMD), and is mostly the result of enterovirus A71 (EV-A71) infection. Sadly, there are currently no treatment options against EV-A71 infection available on the market. In this thesis, I will investigate the poorly understood EV-A71 cellular tropism and the influence of EV-A71 on the host-cell metabolic processes with the end purpose of facilitating the design of potent antiviral compounds against EV-A71 infection, using multidisciplinary approaches. [...], Thesis (PhD Doctorate), Doctor of Philosophy, Institute for Glycomics, Griffith Sciences, Full Text
- Published
- 2024
10. Longitudinal analysis of subtype C envelope tropism for memory CD4+ T cell subsets over the first 3 years of untreated HIV-1 infection
- Author
-
Matthew J. Gartner, Paul R. Gorry, Carolin Tumpach, Jingling Zhou, Ashanti Dantanarayana, J. Judy Chang, Thomas A. Angelovich, Paula Ellenberg, Annemarie E. Laumaea, Molati Nonyane, Penny L. Moore, Sharon R. Lewin, Melissa J. Churchill, Jacqueline K. Flynn, and Michael Roche
- Subjects
Cellular tropism ,CD4+ T cells ,Subtype C HIV-1 ,Coreceptor usage ,Envelope ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Background HIV-1 infects a wide range of CD4+ T cells with different phenotypic properties and differing expression levels of entry coreceptors. We sought to determine the viral tropism of subtype C (C-HIV) Envelope (Env) clones for different CD4+ T cell subsets and whether tropism changes during acute to chronic disease progression. HIV-1 envs were amplified from the plasma of five C-HIV infected women from three untreated time points; less than 2 months, 1-year and 3-years post-infection. Pseudoviruses were generated from Env clones, phenotyped for coreceptor usage and CD4+ T cell subset tropism was measured by flow cytometry. Results A total of 50 C-HIV envs were cloned and screened for functionality in pseudovirus infection assays. Phylogenetic and variable region characteristic analysis demonstrated evolution in envs between time points. We found 45 pseudoviruses were functional and all used CCR5 to mediate entry into NP2/CD4/CCR5 cells. In vitro infection assays showed transitional memory (TM) and effector memory (EM) CD4+ T cells were more frequently infected (median: 46% and 25% of total infected CD4+ T cells respectively) than naïve, stem cell memory, central memory and terminally differentiated cells. This was not due to these subsets contributing a higher proportion of the CD4+ T cell pool, rather these subsets were more susceptible to infection (median: 5.38% EM and 2.15% TM cells infected), consistent with heightened CCR5 expression on EM and TM cells. No inter- or intra-participant changes in CD4+ T cell subset tropism were observed across the three-time points. Conclusions CD4+ T cell subsets that express more CCR5 were more susceptible to infection with C-HIV Envs, suggesting that these may be the major cellular targets during the first 3 years of infection. Moreover, we found that viral tropism for different CD4+ T cell subsets in vitro did not change between Envs cloned from acute to chronic disease stages. Finally, central memory, naïve and stem cell memory CD4+ T cell subsets were susceptible to infection, albeit inefficiently by Envs from all time-points, suggesting that direct infection of these cells may help establish the latent reservoir early in infection.
- Published
- 2020
- Full Text
- View/download PDF
11. Molecular Mechanism of Porcine Epidemic Diarrhea Virus Cell Tropism
- Author
-
Zhiwei Li, Zhiqian Ma, Linfang Dong, Ting Yang, Yang Li, Dian Jiao, Weiguo Han, Haixue Zheng, and Shuqi Xiao
- Subjects
spike ,cellular tropism ,coronavirus ,porcine epidemic diarrhea virus ,reverse genetic analysis ,Microbiology ,QR1-502 - Abstract
ABSTRACT In the 21st century, several human and swine coronaviruses (CoVs) have emerged suddenly and caused great damage to people's lives and property. The porcine epidemic diarrhea virus (PEDV), leading to enormous economic losses to the pork industry and remains a large challenge. PEDV showed extensive cell tropism, and we cannot ignore the potential risk of cross-species transmission. However, the mechanism of adaptation and cell tropism of PEDV remains largely unknown and in vitro isolation of PEDV remains a huge challenge, which seriously impedes the development of vaccines. In this study, we confirmed that the spike (S) protein determines the adaptability of PEDV to monkey Vero cells and LLC-PK1 porcine cells, and isolated exchange of S1 and S2 subunits of adaptive strains did not make PEDV adapt to cells. Further, we found that the cellular adaptability of rCH/SX/2016-SHNXP depends on S1 and the first half of S2 (S3), and the 803L and 976H of the S2 subunit are critical for rCH/SX/2016-S1HNXP+S3HNXP adaptation to Vero cells. These findings highlight the decisive role of PEDV S protein in cell tropism and the potential role of coronaviruses S protein in cross-species transmissibility. Besides, our work also provides some different insight into finding PEDV receptors and developing PEDV and other coronaviruses vaccines. IMPORTANCE CoVs can spill from an animal reservoir into a naive host to cause diseases in humans or domestic animals. PEDV results in high mortality in piglets, which has caused immense economic losses in the pork industry. Virus isolation is the first step in studying viral pathogenesis and developing effective vaccines. However, the molecular mechanism of PEDV cell tropism is largely unknown, and isolation of endemic PEDV strains remains a major challenge. This study confirmed that the S gene is the decisive gene of PEDV adaptability to monkey Vero cells and porcine LLC-PK1 cells by the PEDV reverse genetics system. Isolated exchange of S1 and S2 of adaptive strains did not make PEDV adapt to cells, and the 803L and 976H of S2 subunit are critical for rCH/SX/2016-S1HNXP+S3HNXP adaptation to Vero cells. These results illustrate the decisive role of PEDV S protein in cell tropism and highlight the potential role of coronaviruses S protein in cross-species transmissibility. Besides, our finding also provides some unique insight into identifying PEDV functional receptors and has guiding significance for developing PEDV and other coronavirus vaccines.
- Published
- 2022
- Full Text
- View/download PDF
12. Futuristic Methods for Determining HIV Co-receptor Use
- Author
-
Flynn, Jacqueline K., Gartner, Matthew, Laumaea, Annamarie, Gorry, Paul R., Shapshak, Paul, editor, Balaji, Seetharaman, editor, Kangueane, Pandjassarame, editor, Chiappelli, Francesco, editor, Somboonwit, Charurut, editor, Menezes, Lynette J., editor, and Sinnott, John T., editor
- Published
- 2019
- Full Text
- View/download PDF
13. Tilapia lake virus immunoglobulin G (TiLV IgG) antibody: Immunohistochemistry application reveals cellular tropism of TiLV infection.
- Author
-
Piewbang, Chutchai, Tattiyapong, Puntanat, Techangamsuwan, Somporn, and Surachetpong, Win
- Subjects
- *
IMMUNOGLOBULIN G , *TILAPIA , *MOZAMBIQUE tilapia , *VIRAL tropism , *IMMUNOGLOBULINS , *TROPISMS , *IMMUNOHISTOCHEMISTRY - Abstract
Tilapia lake virus (TiLV) is a notable contagious agent that causes massive economic losses in the tilapia industry globally. Evaluations of the histological changes associated with TiLV infection are not only crucial for diagnosis, but also to gain an understanding of the disease. We therefore synthesized a rabbit polyclonal immunoglobulin G antibody against TiLV and developed an immunohistochemical (IHC) procedure to detect TiLV localization in the tissues of infected fish for comparison with in situ hybridization (ISH) testing. A total of four different sample cohorts derived from TiLV-infected fish was used to validate the IHC procedure. The TiLV IHC application was successfully developed and facilitated nuclear and cytoplasmic immunolabelling in the intestines, gills, brain, liver, pancreas, spleen, and kidneys that corresponded with the ISH results. Apart from the ISH results, TiLV-IHC signals were clearly evident in the endothelial cells of various organs, the circulating leukocytes in the blood vessels, and the areas of tissue inflammation. Among the tested sample cohorts, the intestines, gills, and brain had IHC-positive signals, highlighting the possibility of these organs as common TiLV targets. Immunological staining pattern and distribution corresponded with the TiLV viral load but not the inoculation route. The TiLV IHC was also capable of detecting TiLV infection in the experimentally challenged ornamental cichlids, Mozambique tilapia, giant gourami, and naturally infected tilapia, indicating the dynamic range of IHC for TiLV detection. Overall, our study delivers the first IHC platform to detect TiLV infection and provides novel evidence of cellular tropism during TiLV infection. Our findings also reveal the TiLV distribution pattern of infected fish and propose the endotheliotropism and lymphotropism of this virus, which requires further elaboration. Importantly, this new IHC procedure could be applied to study the pathogenesis and interaction of TiLV in future research. • A polyclonal rabbit immunoglobulin G (IgG) against tilapia lake virus (TiLV) is synthesized. • Immunohistochemistry (IHC) to detect the TiLV infection is developed and optimized with variations in the antigen retrieval methods and primary antibody concentration. • The optimized IHC is compared with in situ hybridization (ISH) technique and validated by performing in different sample cohorts. • The developed IHC gains high affinity for TiLV infection detection and the IHC results indicates novel cellular tropism of the TiLV. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
14. 日本脑炎病毒感染仔猪扁桃体的细胞嗜性研究.
- Author
-
张秋婷, 关如婷, 潘俊慧, 杨兴淼, 谢盛达, 余都, 王学飞, and 曹瑞兵
- Subjects
- *
ANTIGEN presenting cells , *JAPANESE encephalitis viruses , *LYMPHOID tissue , *VIRAL tropism , *PIGLETS , *DENDRITIC cells , *TONSILS - Abstract
[Objectives]The experiment aimed to identify the cellular tropism of Japanese encephalitis virus(JEV)in the pig tonsils. [Methods] In this study, three healthy commercial piglets with negative JEV antigen and antibody were inoculated with 2× 107 TCID50. mL-1 JEV NJ2008 strain per piglet by injection intravenously and intradermally in the neck region, and one control piglet was injected with 2 mL saline. The piglets were kept in isolation for one week, during which the samples of nose swab, blood and tonsil tissue were collected. We detected the virus in the samples by qPCR, immunohistochemistry and immunofluorescence. [Results]The temperatures of all three piglets after inoculated poison were normal. No obvious clinical symptoms were observed in all experimental piglets during 7 days. Three piglets inoculated with JEV NJ2008 showed viremia for 2 days from the day of inoculation, and JEV could be detected from nasal swabs collected for 7 days. JEV mainly infected the soft palatal tonsils of pigs. No JEV positive cells were found in the pharyngeal tonsils, tube tonsils and parapharyngeal tonsils of infected piglets. In the soft palatal tonsil of pigs, JEV infected cells were mainly distributed around the lymphoid follicles and in the diffuse lymphoid tissues. Most of JEV antigen positive cells were irregular in shape, large in nucleus and light in color, with the characteristics of macrophages or dendritic cells. Then through the colocalization of CD11b, CD163 and MHC, cell surface markers and virus antigens, JEV positive cells were identified as mainly antigen presenting cell, such as macrophages and dendritic cells. [Conclusions]In this experiment, we found that in the early stage of JEV infection within one week, the tonsils of piglets showed a persistent infection. The cellular tropism of JEV in pig tonsil were mainly macrophages and dendritic cells. These results pave the way for studying the mechanism of persistent infection of JEV in tonsil. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
15. Longitudinal analysis of subtype C envelope tropism for memory CD4+ T cell subsets over the first 3 years of untreated HIV-1 infection.
- Author
-
Gartner, Matthew J., Gorry, Paul R., Tumpach, Carolin, Zhou, Jingling, Dantanarayana, Ashanti, Chang, J. Judy, Angelovich, Thomas A., Ellenberg, Paula, Laumaea, Annemarie E., Nonyane, Molati, Moore, Penny L., Lewin, Sharon R., Churchill, Melissa J., Flynn, Jacqueline K., and Roche, Michael
- Subjects
VIRAL tropism ,T cells ,HIV ,TROPISMS ,STEM cells ,ACUTE diseases ,INFECTION - Abstract
Background: HIV-1 infects a wide range of CD4
+ T cells with different phenotypic properties and differing expression levels of entry coreceptors. We sought to determine the viral tropism of subtype C (C-HIV) Envelope (Env) clones for different CD4+ T cell subsets and whether tropism changes during acute to chronic disease progression. HIV-1 envs were amplified from the plasma of five C-HIV infected women from three untreated time points; less than 2 months, 1-year and 3-years post-infection. Pseudoviruses were generated from Env clones, phenotyped for coreceptor usage and CD4+ T cell subset tropism was measured by flow cytometry. Results: A total of 50 C-HIV envs were cloned and screened for functionality in pseudovirus infection assays. Phylogenetic and variable region characteristic analysis demonstrated evolution in envs between time points. We found 45 pseudoviruses were functional and all used CCR5 to mediate entry into NP2/CD4/CCR5 cells. In vitro infection assays showed transitional memory (TM) and effector memory (EM) CD4+ T cells were more frequently infected (median: 46% and 25% of total infected CD4+ T cells respectively) than naïve, stem cell memory, central memory and terminally differentiated cells. This was not due to these subsets contributing a higher proportion of the CD4+ T cell pool, rather these subsets were more susceptible to infection (median: 5.38% EM and 2.15% TM cells infected), consistent with heightened CCR5 expression on EM and TM cells. No inter- or intra-participant changes in CD4+ T cell subset tropism were observed across the three-time points. Conclusions: CD4+ T cell subsets that express more CCR5 were more susceptible to infection with C-HIV Envs, suggesting that these may be the major cellular targets during the first 3 years of infection. Moreover, we found that viral tropism for different CD4+ T cell subsets in vitro did not change between Envs cloned from acute to chronic disease stages. Finally, central memory, naïve and stem cell memory CD4+ T cell subsets were susceptible to infection, albeit inefficiently by Envs from all time-points, suggesting that direct infection of these cells may help establish the latent reservoir early in infection. [ABSTRACT FROM AUTHOR]- Published
- 2020
- Full Text
- View/download PDF
16. 2014 年~2019 年PRRSV 主要流行毒株在我国的变化.
- Author
-
张洪亮, 张文立, 许浒, 宋帅杰, 赵静, 相丽润, 冷超粮, 李真, 刘春晓, 汤艳东, 陈家锃, 彭金美, 王倩, 安同庆, 童光志, 蔡雪辉, and 田志军
- Abstract
Copyright of Chinese Journal of Preventive Veterinary Medicine / Zhongguo Yufang Shouyi Xuebao is the property of Chinese Journal of Preventive Veterinary Medicine Editorial Office and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2020
- Full Text
- View/download PDF
17. Distinct Expression Patterns of AAV8 Vectors with Broadly Active Promoters from Subretinal Injections of Neonatal Mouse Eyes at Two Different Ages
- Author
-
Xiong, Wenjun, Cepko, Constance, Bowes Rickman, Catherine, editor, LaVail, Matthew M., editor, Anderson, Robert E., editor, Grimm, Christian, editor, Hollyfield, Joe, editor, and Ash, John, editor
- Published
- 2016
- Full Text
- View/download PDF
18. The nsp2 Hypervariable Region of Porcine Reproductive and Respiratory Syndrome Virus Strain JXwn06 Is Associated with Viral Cellular Tropism to Primary Porcine Alveolar Macrophages.
- Author
-
Jiangwei Song, Peng Gao, Can Kong, Lei Zhou, Xinna Ge, Xin Guo, Jun Han, and Hanchun Yang
- Subjects
- *
HYPERVARIABLE regions , *VIRAL tropism , *ALVEOLAR macrophages , *PORCINE reproductive & respiratory syndrome , *RNA synthesis - Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) poses a major threat to global pork production and has been notorious for its rapid genetic evolution in the field. The nonstructural protein 2 (nsp2) replicase protein represents the fastest evolving region of PRRSV, but the underlying biological significance has remained poorly understood. By deletion mutagenesis, we discovered that the nsp2 hypervariable region plays an important role in controlling the balance of genomic mRNA and a subset of subgenomic mRNAs. More significantly, we revealed an unexpected link of the nsp2 hypervariable region to viral tropism. Specifically, a mutant of the Chinese highly pathogenic PRRSV strain JXwn06 carrying a deletion spanning nsp2 amino acids 323 to 521 (nsp2Δ323--521) in its hypervariable region was found to lose infectivity in primary porcine alveolar macrophages (PAMs), although it could replicate relatively efficiently in the supporting cell line MARC-145. Consequently, this mutant failed to establish an infection in piglets. Further dissection of the viral life cycle revealed that the mutant had a defect (or defects) lying in the steps between virus penetration and negative-stranded RNA synthesis. Taken together, our results reveal novel functions of nsp2 in the PRRSV life cycle and provide important insights into the mechanisms of PRRSV RNA synthesis and cellular tropism. IMPORTANCE The PRRSV nsp2 replicase protein undergoes rapid and broad genetic variations in its middle region in the field, but the underlying significance has remained enigmatic. Here, we demonstrate that the nsp2 hypervariable region not only plays an important regulatory role in maintaining the balance of different viral mRNA species but also regulates PRRSV tropism to primary PAMs. Our results reveal novel functions for PRRSV nsp2 and have important implications for understanding the mechanisms of PRRSV RNA synthesis and cellular tropism. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
19. Experimental Models of SARS-CoV-2 Infection: Possible Platforms to Study COVID-19 Pathogenesis and Potential Treatments
- Author
-
Alireza Sahebi Shahem-abadi, Jaleel A. Miyan, Silke Haerteis, Sareh Pandamooz, Carl-Philipp Meinung, Afshin Borhani-Haghighi, Benjamin Jurek, Mehdi Dianatpour, James A. Downing, Zahra Baharvand, and Mohammad Saied Salehi
- Subjects
Pharmacology ,RM ,Protective immunity ,Coronavirus disease 2019 (COVID-19) ,SARS-CoV-2 ,business.industry ,Transmission (medicine) ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,COVID-19 ,virus diseases ,Models, Theoretical ,Toxicology ,medicine.disease_cause ,Bioinformatics ,Antiviral Agents ,Pathogenesis ,Global health ,Humans ,Medicine ,business ,Pandemics ,Cellular Tropism ,Coronavirus - Abstract
In December 2019, a novel coronavirus crossed species barriers to infect humans and was effectively transmitted from person to person, leading including vaccines and antiviral drugs that could prevent or limit the burden or transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global health priority. It is thus of utmost importance to assess possible therapeutic strategies against SARS-CoV-2 using experimental models that recapitulate aspects of the human disease. Here, we review available models currently being developed and used to study SARS-CoV-2 infection and highlight their application to screen potential therapeutic approaches, including repurposed antiviral drugs and vaccines. Each identified model provides a valuable insight into SARS-CoV-2 cellular tropism, replication kinetics, and cell damage that could ultimately enhance understanding of SARS-CoV-2 pathogenesis and protective immunity. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 62 is January 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
- Published
- 2022
- Full Text
- View/download PDF
20. Henipavirus Receptor Usage and Tropism
- Author
-
Pernet, Olivier, Wang, Yao E, Lee, Benhur, Lee, Benhur, editor, and Rota, Paul A., editor
- Published
- 2012
- Full Text
- View/download PDF
21. Biology and Cellular Tropism of a Unique Astrovirus Strain: Murine Astrovirus 2
- Author
-
Sean P Kelly, Neil S. Lipman, Chuanwu Wang, Adam O. Michel, Rodolfo J Ricart Arbona, and Kenneth S Henderson
- Subjects
General Veterinary ,biology ,viruses ,CD3 ,virus diseases ,biology.organism_classification ,Tropism ,Virology ,General Biochemistry, Genetics and Molecular Biology ,Astrovirus ,Mice, Inbred C57BL ,Mice ,Cytokeratin ,Immune system ,Mice, Inbred NOD ,biology.protein ,Tissue tropism ,Animals ,Astroviridae ,Viral shedding ,Biology ,Viral load ,Cellular Tropism ,Original Research - Abstract
Murine astrovirus 2 (MuAstV2) is a novel murine astrovirus recently identified in laboratory and wild mice. MuAstV2 readily transmits between immunocompetent mice yet fails to transmit to highly immunocompromised mouse strains—a unique characteristic when contrasted with other murine viruses including other astroviruses. We characterized the viral shedding kinetics and tissue tropism of MuAstV2 in immunocompetent C57BL/6NCrl mice and evaluated the apparent resistance of highly immunocompromised NOD- Prkdcem26Cd52Il2rgem26Cd22 /NjuCrl mice to MuAstV2 after oral inoculation. Temporal patterns of viral shedding were determined by serially measuring fecal viral RNA. Tissue tropism and viral load were characterized and quantified by using in-situ hybridization (ISH) targeting viral RNA. Cellular tropism was characterized by evaluating fluorescent colocalization of viral ISH with various immunohistochemical markers. We found a rapid increase of fecal viral RNA in B6 mice, which peaked at 5 d after inoculation (dpi) followed by cessation of shedding by 168 dpi. The small intestine had the highest percentage of hybridization (3.09% of tissue area) of all tissues in which hybridization occurred at 5 dpi. The thymus displayed the next highest degree of hybridization (2.3%) at 7 dpi, indicating extraintestinal viral spread. MuAstV2 RNA hybridization was found to colocalize with only 3 of the markers evaluated: CD3 (T cells), Iba1 (macrophages), and cytokeratin (enterocytes). A higher percentage of CD3 cells and Iba1 cells hybridized with MuAstV2 as compared with cytokeratin at 2 dpi (CD3, 59%; Iba1, 46%; cytokeratin, 6%) and 35 dpi (CD3, 14%; Iba1, 55%; cytokeratin, 3%). Neither fecal viral RNA nor viral hybridization was noted in NCG mice at the time points examined. In addition, mice of mixed genetic background were inoculated, and only those with a functioning Il2rg gene shed MuAstV2. Results from this study suggest that infection of, or interaction with, the immune system is required for infection by or replication of MuAstV2.
- Published
- 2021
- Full Text
- View/download PDF
22. hCD46 receptor is not required for measles vaccine Schwarz strain replication in vivo: Type-I IFN is the species barrier in mice.
- Author
-
Mura, M., Ruffié, C., Billon-Denis, E., Combredet, C., Tournier, J.N., and Tangy, F.
- Subjects
- *
MEASLES virus , *TROPISMS , *IMMUNOGENETICS , *TYPE I interferons , *LABORATORY mice - Abstract
Abstract Measles virus has been successfully attenuated on chicken embryo cells to obtain a highly efficient and safe live attenuated vaccine, administered thus far to billions of children. Measles virus attenuation has long been described to involve a modification of cellular tropism with the use of human CD46 ubiquitous receptor. Nevertheless, the use of this receptor in vivo is not obvious. In this study we use four different mouse models to decipher the respective part of hCD46 receptor and type-I interferon response in measles host restriction. We observed that only type-I interferon restricts viral replication of attenuated MV Schwarz strain in mice, independently of the presence of hCD46 receptor. By comparing measles virus immunogenicity in the different models, we confirmed that there was no impact on the absence of this receptor on the immune response. Therefore, we propose to simplify the mouse model. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
23. The S2 Subunit of Infectious Bronchitis Virus Beaudette Is a Determinant of Cellular Tropism.
- Author
-
Bickerton, Erica, Maier, Helena J., Stevenson-Leggett, Phoebe, Armesto, Maria, and Britton, Paul
- Subjects
- *
BRONCHITIS , *VIRAL tropism , *GLYCOPROTEINS , *RECOMBINANT viruses , *MICROBIAL virulence - Abstract
The spike (S) glycoprotein of the avian gammacoronavirus infectious bronchitis virus (IBV) is comprised of two subunits (S1 and S2), has a role in virulence in vivo, and is responsible for cellular tropism in vitro. We have previously demonstrated that replacement of the S glycoprotein ectodomain from the avirulent Beaudette strain of IBV with the corresponding region from the virulent M41-CK strain resulted in a recombinant virus, BeauR-M41(S), with the in vitro cell tropism of M41-CK. The IBV Beaudette strain is able to replicate in both primary chick kidney cells and Vero cells, whereas the IBV M41-CK strain replicates in primary cells only. In order to investigate the region of the IBV S responsible for growth in Vero cells, we generated a series of recombinant IBVs expressing chimeric S glycoproteins, consisting of regions from the Beaudette and M41-CK S gene sequences, within the genomic background of Beaudette. The S2, but not the S1, subunit of the Beaudette S was found to confer the ability to grow in Vero cells. Various combinations of Beaudette-specific amino acids were introduced into the S2 subunit of M41 to determine the minimum requirement to confer tropism for growth in Vero cells. The ability of IBV to grow and produce infectious progeny virus in Vero cells was subsequently narrowed down to just 3 amino acids surrounding the S2= cleavage site. Conversely, swapping of the 3 Beaudette-associated amino acids with the corresponding ones from M41 was sufficient to abolish Beaudette growth in Vero cells. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
24. Risk Assessment for Highly Pathogenic Avian Influenza A(H5N6/H5N8) Clade 2.3.4.4 Viruses
- Author
-
J. S. Malik Peiris, Daniel K.W. Chu, Michael C. W. Chan, Christine H T Bui, John M. Nicholls, Kenrie P Y Hui, Ka-Chun Ng, Richard J. Webby, Denise I. T. Kuok, and Hin Wo Yeung
- Subjects
Microbiology (medical) ,China ,Chemokine ,Epidemiology ,animal diseases ,viruses ,Infectious and parasitic diseases ,RC109-216 ,medicine.disease_cause ,alveolar epithelial cells ,human airway organoids ,Proinflammatory cytokine ,Birds ,Risk Assessment for Highly Pathogenic Avian Influenza A(H5N6/H5N8) Clade 2.3.4.4 Viruses ,Influenza A Virus, H1N1 Subtype ,Influenza, Human ,medicine ,Animals ,Humans ,Influenza A Virus, H5N8 Subtype ,Clade ,Tropism ,clade 2.3.4.4 ,HPAI H5Nx ,Influenza A Virus, H5N1 Subtype ,biology ,Host (biology) ,Research ,tropism ,innate host responses ,risk assessment ,virus diseases ,Outbreak ,Virology ,Influenza A virus subtype H5N1 ,Infectious Diseases ,Influenza in Birds ,biology.protein ,Hong Kong ,Medicine ,influenza ,Cellular Tropism - Abstract
The numerous global outbreaks and continuous reassortments of highly pathogenic avian influenza (HPAI) A(H5N6/H5N8) clade 2.3.4.4 viruses in birds pose a major risk to the public health. We investigated the tropism and innate host responses of 5 recent HPAI A(H5N6/H5N8) avian isolates of clades 2.3.4.4b, e, and h in human airway organoids and primary human alveolar epithelial cells. The HPAI A(H5N6/H5N8) avian isolates replicated productively but with lower competence than the influenza A(H1N1)pdm09, HPAI A(H5N1), and HPAI A(H5N6) isolates from humans in both or either models. They showed differential cellular tropism in human airway organoids; some infected all 4 major epithelial cell types: ciliated cells, club cells, goblet cells, and basal cells. Our results suggest zoonotic potential but low transmissibility of the HPAI A(H5N6/H5N8) avian isolates among humans. These viruses induced low levels of proinflammatory cytokines/chemokines, which are unlikely to contribute to the pathogenesis of severe disease.
- Published
- 2021
- Full Text
- View/download PDF
25. Pathogenicity and virulence of Marburg virus
- Author
-
Abir, MH, Rahman, T, Das, A, Etu, SN, Nafiz, IH, Rakib, A, Mitra, S, Emran, TB, Dhama, K, Islam, Ariful, Siyadatpanah, A, Mahmud, S, Kim, B, Hassan, MM, Abir, MH, Rahman, T, Das, A, Etu, SN, Nafiz, IH, Rakib, A, Mitra, S, Emran, TB, Dhama, K, Islam, Ariful, Siyadatpanah, A, Mahmud, S, Kim, B, and Hassan, MM
- Abstract
Marburg virus (MARV) has been a major concern since 1967, with two major outbreaks occurring in 1998 and 2004. Infection from MARV results in severe hemorrhagic fever, causing organ dysfunction and death. Exposure to fruit bats in caves and mines, and human-to-human transmission had major roles in the amplification of MARV outbreaks in African countries. The high fatality rate of up to 90% demands the broad study of MARV diseases (MVD) that correspond with MARV infection. Since large outbreaks are rare for MARV, clinical investigations are often inadequate for providing the substantial data necessary to determine the treatment of MARV disease. Therefore, an overall review may contribute to minimizing the limitations associated with future medical research and improve the clinical management of MVD. In this review, we sought to analyze and amalgamate significant information regarding MARV disease epidemics, pathophysiology, and management approaches to provide a better understanding of this deadly virus and the associated infection.
- Published
- 2022
26. Murine astrovirus tropism for goblet cells and enterocytes facilitates an IFN-λ response in vivo and in enteroid cultures
- Author
-
Gowri Kalugotla, Harshad Ingle, Belgacem Mihi, Megan T. Baldridge, Misty Good, Michael S. Diamond, Rodney D. Newberry, Keely G. McDonald, Yuhao Li, Pritesh Desai, Sanghyun Lee, Ebrahim Hassan, Jana Gawron, Elizabeth A. Kennedy, and Heyde Makimaa
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Immunology ,Biology ,Article ,Microbiology ,Pathogenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Organ Culture Techniques ,Th2 Cells ,In vivo ,Astroviridae Infections ,medicine ,Immunology and Allergy ,Animals ,Humans ,Tropism ,Cells, Cultured ,Mice, Knockout ,Innate immune system ,Coinfection ,Intestinal epithelium ,Immunity, Innate ,Gastroenteritis ,Mice, Inbred C57BL ,Chronic infection ,Viral Tropism ,030104 developmental biology ,Cytokine ,Enterocytes ,Astroviridae ,Cytokines ,Goblet Cells ,Cellular Tropism ,030215 immunology - Abstract
Although they globally cause viral gastroenteritis in children, astroviruses are understudied due to the lack of well-defined animal models. While murine astroviruses (muAstVs) chronically infect immunodeficient mice, a culture system and understanding of their pathogenesis is lacking. Here, we describe a platform to cultivate muAstV using air–liquid interface (ALI) cultures derived from mouse enteroids, which support apical infection and release. Chronic muAstV infection occurs predominantly in the small intestine and correlates with higher interferon-lambda (IFN-λ) expression. MuAstV stimulates IFN-λ production in ALI, recapitulating our in vivo findings. We demonstrate that goblet cells and enterocytes are targets for chronic muAstV infection in vivo, and that infection is enhanced by parasite co-infection or type 2 cytokine signaling. Depletion of goblet cells from ALI limits muAstV infection in vitro. During chronic infection, muAstV stimulates IFN-λ production in infected cells and induces ISGs throughout the intestinal epithelium in an IFN-λ-receptor-dependent manner. Collectively, our study provides insights into the cellular tropism and innate immune responses to muAstV and establishes an enteroid-based culture system to propagate muAstV in vitro.
- Published
- 2021
27. An approach to cellular tropism of SARS-CoV-2 through protein–protein interaction and enrichment analysis
- Author
-
Selene Zarate, Daniel Ortega-Bernal, Rafael Bojalil, and Maria de los Ángeles Cárdenas
- Subjects
Multidisciplinary ,Host Microbial Interactions ,SARS-CoV-2 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,fungi ,COVID-19 ,Humans ,Angiotensin-Converting Enzyme 2 ,Biology ,Tropism ,Virology ,Cellular Tropism ,Protein–protein interaction - Abstract
COVID-19, caused by SARS-CoV-2, is a primarily pulmonary disease that can affect several organs, directly or indirectly. To date, there are many questions about the different pathological mechanisms. Here, we generate an approach to identify the cellular-level tropism of SARS-CoV-2 using human proteomics, virus-host interactions, and enrichment analysis. Through a network-based approach, the molecular context was visualized and analyzed. This procedure was also performed for SARS-CoV-1. We obtained proteomes and interactomes from 145 different cells corresponding to 57 different tissues. We discarded the cells without the proteins known for interacting with the virus, such as ACE2 or TMPRSS2. Of the remaining cells, a gradient of susceptibility to infection was observed. In addition, we identified proteins associated with the coagulation cascade that can be directly or indirectly affected by viral proteins. As a whole we identified 55 cells that could be potentially controlled by the virus, with different susceptibilities, mainly being pneumocytes, heart, kidney, liver, or small intestine cells. These results help to explain the molecular context and provide elements for possible treatments in the current situation. This strategy may be useful for other viruses, especially those with limited reported PPI, such as a new virus.
- Published
- 2022
- Full Text
- View/download PDF
28. Epstein-Barr Virus Type 2 Infects T Cells in Healthy Kenyan Children.
- Author
-
Coleman, Carrie B., Daud, Ibrahim I., Ogolla, Sidney O., Ritchie, Julie A., Smith, Nicholas A., Sumba, Peter O., Dent, Arlene E., and Rochford, Rosemary
- Subjects
- *
CHILDREN , *EPSTEIN-Barr virus , *T cells , *BREAST milk , *SALIVA , *MOTHERS , *DISEASES , *COLLECTION & preservation of biological specimens , *DNA , *EPSTEIN-Barr virus diseases , *LONGITUDINAL method , *RESEARCH funding , *VIRAL physiology , *DISEASE prevalence , *PHYSIOLOGY , *DIAGNOSIS - Abstract
Background: The 2 strains of Epstein-Barr virus (EBV), EBV type 1 (EBV-1) and EBV-2, differ in latency genes, suggesting that they use distinct mechanisms to establish latency. We previously reported that EBV-2 infects T cells in vitro. In this study, we tested the possibility that EBV-2 infects T cells in vivo.Methods: Purified T-cell fractions isolated from children positive for EBV-1 or EBV-2 and their mothers were examined for the presence of EBV and for EBV type.Results: We detected EBV-2 in all T-cell samples obtained from EBV-2-infected children at 12 months of age, with some children retaining EBV-2-positive T cells through 24 months of age, suggesting that EBV-2 persists in T cells. We were unable to detect EBV-2 in T-cell samples from mothers but could detect EBV-2 in samples of their breast milk and saliva.Conclusions: These data suggest that EBV-2 uses T cells as an additional latency reservoir but that, over time, the frequency of infected T cells may drop below detectable levels. Alternatively, EBV-2 may establish a prolonged transient infection in the T-cell compartment. Collectively, these novel findings demonstrate that EBV-2 infects T cells in vivo and suggest EBV-2 may use the T-cell compartment to establish latency. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
29. Equine Arteritis Virus Has Specific Tropism for Stromal Cells and CD8+ T and CD21+ B Lymphocytes but Not for Glandular Epithelium at the Primary Site of Persistent Infection in the Stallion Reproductive Tract.
- Author
-
Carossino, Mariano, Loynachan, Alan T., Canisso, Igor F., Cook, R. Frank, Campos, Juliana R., Nam, Bora, Yun Young Go, Squires, Edward L., Troedsson, Mats H. T., Swerczek, Thomas, Del Piero, Fabio, Bailey, Ernest, Timoney, Peter J., and Balasuriya, Udeni B. R.
- Subjects
- *
ARTERITIS , *STALLIONS , *VIRAL tropism , *HORSE diseases , *IMMUNOFLUORESCENCE , *IMMUNOHISTOCHEMISTRY , *DISEASES - Abstract
Equine arteritis virus (EAV) has a global impact on the equine industry as the causative agent of equine viral arteritis (EVA), a respiratory, systemic, and reproductive disease of equids. A distinctive feature of EAV infection is that it establishes long-term persistent infection in 10 to 70% of infected stallions (carriers). In these stallions, EAV is detectable only in the reproductive tract, and viral persistence occurs despite the presence of high serum neutralizing antibody titers. Carrier stallions constitute the natural reservoir of the virus as they continuously shed EAV in their semen. Although the accessory sex glands have been implicated as the primary sites of EAV persistence, the viral host cell tropism and whether viral replication in carrier stallions occurs in the presence or absence of host inflammatory responses remain unknown. In this study, dual immunohistochemical and immunofluorescence techniques were employed to unequivocally demonstrate that the ampulla is the main EAV tissue reservoir rather than immunologically privileged tissues (i.e., testes). Furthermore, we demonstrate that EAV has specific tropism for stromal cells (fibrocytes and possibly tissue macrophages) and CD8+ T and CD21+ B lymphocytes but not glandular epithelium. Persistent EAV infection is associated with moderate, multifocal lymphoplasmacytic ampullitis comprising clusters of B (CD21+) lymphocytes and significant infiltration of T (CD3+, CD4+, CD8+, and CD25+) lymphocytes, tissue macrophages, and dendritic cells (Iba-1+ and CD83+), with a small number of tissue macrophages expressing CD163 and CD204 scavenger receptors. This study suggests that EAV employs complex immune eva- sion mechanisms that warrant further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
30. Linking nutrient sensing and gene expression in Plasmodium falciparum blood‐stage parasites
- Author
-
Manish Kumar, Manoj T. Duraisingh, and Kristen M. Skillman
- Subjects
Plasmodium falciparum ,Gene Expression ,Nutrient sensing ,Microbiology ,Plasmodium ,Article ,03 medical and health sciences ,parasitic diseases ,medicine ,Antigenic variation ,Animals ,Humans ,Epigenetics ,Malaria, Falciparum ,Molecular Biology ,030304 developmental biology ,Life Cycle Stages ,0303 health sciences ,biology ,030306 microbiology ,Transmission (medicine) ,Nutrients ,medicine.disease ,biology.organism_classification ,Malaria ,Cellular Tropism - Abstract
Malaria is one of the most life-threatening infectious diseases worldwide, caused by infection of humans with parasites of the genus Plasmodium. The complex life cycle of Plasmodium parasites is shared between two hosts, with infection of multiple cell types, and the parasite needs to adapt for survival and transmission through significantly different metabolic environments. Within the blood-stage alone, parasites encounter changing levels of key nutrients, including sugars, amino acids, and lipids, due to differences in host dietary nutrition, cellular tropism, and pathogenesis. In this review, we consider the mechanisms that the most lethal of malaria parasites, Plasmodium falciparum, uses to sense nutrient levels and elicit changes in gene expression during blood-stage infections. These changes are brought about by several metabolic intermediates and their corresponding sensor proteins. Sensing of distinct nutritional signals can drive P. falciparum to alter the key blood-stage processes of proliferation, antigenic variation, and transmission.
- Published
- 2020
- Full Text
- View/download PDF
31. Differential Tropism of SARS-CoV and SARS-CoV-2 in Bat Cells
- Author
-
Terrence Chi-Kong Lau, Kitty S. C. Fung, Joshua Fung, Hayes K.H. Luk, Susanna K. P. Lau, Patrick C. Y. Woo, Flora Ka Kei Cheng, Kam Leng Aw-Yong, Kenneth S. M. Li, Antonio C.P. Wong, Tony T.Y. Chan, Stella Chu, and Zirong He
- Subjects
Microbiology (medical) ,Epidemiology ,Middle East respiratory syndrome coronavirus ,SARS-related coronavirus ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,030231 tropical medicine ,lcsh:Medicine ,bat ,severe acute respiratory syndrome ,Virus Replication ,medicine.disease_cause ,lcsh:Infectious and parasitic diseases ,respiratory infections ,03 medical and health sciences ,0302 clinical medicine ,Chiroptera ,origin ,medicine ,Animals ,Humans ,lcsh:RC109-216 ,030212 general & internal medicine ,Differential Tropism of SARS-CoV and SARS-CoV-2 in Bat Cells ,Pandemics ,Rhinolophus sinicus ,Tropism ,SARS ,biology ,SARS-CoV-2 ,tropism ,lcsh:R ,Dispatch ,COVID-19 ,virus diseases ,biology.organism_classification ,Virology ,zoonoses ,Viral Tropism ,Infectious Diseases ,Severe acute respiratory syndrome-related coronavirus ,coronavirus disease ,Viral replication ,Cell culture ,Middle East Respiratory Syndrome Coronavirus ,Tissue tropism ,Cellular Tropism ,severe acute respiratory syndrome coronavirus 2 - Abstract
Severe acute respiratory syndrome coronavirus 2 did not replicate efficiently in 13 bat cell lines, whereas severe acute respiratory syndrome coronavirus replicated efficiently in kidney cells of its ancestral host, the Rhinolophus sinicus bat, suggesting different evolutionary origins. Structural modeling showed that RBD/RsACE2 binding may contribute to the differential cellular tropism.
- Published
- 2020
- Full Text
- View/download PDF
32. The conserved L1089 in the S2 subunit of avian infectious bronchitis virus determines viral kidney tropism by disrupting virus-cell fusion.
- Author
-
Li, Shu-Yun, Shen, Yu-Xi, Xiang, Xue-Lian, Li, Yong-Xin, Li, Nian-Ling, Wang, An-Dong, Cui, Min, Han, Xin-Feng, Huang, Yong, and Xia, Jing
- Subjects
- *
VIRAL tropism , *AVIAN infectious bronchitis virus , *CORONAVIRUSES , *AMINO acid sequence , *VACCINE development - Abstract
The virulence of avian gamma-coronavirus infectious bronchitis viruses (IBV) for the kidney has led to high mortality in dominant-genotype isolations, but the key sites of viral protein that determine kidney tropism are still not fully clear. In this study, the amino acid sequences of the S2 subunit of IBVs with opposing adaptivity to chicken embryonic kidney cells (CEKs) were aligned to identify putative sites associated with differences in viral adaptability. The S2 gene and the putative sites of the non-adapted CN strain were introduced into the CEKs-adapted SczyC30 strain to rescue seven mutants. Analysis of growth characteristics showed that the replacement of the entire S2 subunit and the L1089I substitution in the S2 subunit entirely abolished the proliferation of recombinant IBV in CEKs as well as in primary chicken oviduct epithelial cells. Pathogenicity assays also support the decisive role of this L1089 for viral nephrotropism, and this non-nephrotropic L1089I substitution significantly attenuates pathogenicity. Analysis of the putative cause of proliferation inhibition in CEKs suggests that the L1089I substitution affects neither virus attachment nor endocytosis, but instead fails to form double-membrane vesicles to initiate the viral replication and translation. Position 1089 of the IBV S2 subunit is conservative and predicted to lie in heptad repeat 2 domains. It is therefore reasonable to conclude that the L1089I substitution alters the nephrotropism of parent strain by affecting virus-cell fusion. These findings provide crucial insights into the adaptive mechanisms of IBV and have applications in the development of vaccines and drugs against IB. • The S2 subunit is responsible for IBV tropism to CEKs and kidneys. • The L1089I substitution abolished the IBV tropism to CEKs and kidneys. • The L1089I substitution significantly attenuated pathogenicity. • The L1089 was predicted to lie in HR2 domains on the S2 subunit. • The L1089I substitution affected virus-cell fusion. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
33. In-Vitro Infection of Peripheral Blood Dendritic Cells with Human Immunodeficiency Virus-1 Causes Impairment of Accessory Functions
- Author
-
Chehimi, Jihed, Prakash, Kesh, Shanmugam, Vedapuri, Jackson, Stephanie J., Bandyopadhyay, Santu, Starr, Stuart E., Kamperdijk, Eduard W. A., editor, Nieuwenhuis, Paul, editor, and Hoefsmit, Elisabeth C. M., editor
- Published
- 1993
- Full Text
- View/download PDF
34. Tick Cell Culture Analysis of Growth Dynamics and Cellular Tropism of Rickettsia buchneri, an Endosymbiont of the Blacklegged Tick, Ixodes scapularis
- Author
-
Xin-Ru Wang, Timothy J. Kurtti, Ulrike G. Munderloh, and Cody J. Thorpe
- Subjects
Ixodes ricinus ,Science ,Tick ,Article ,growth dynamics ,Microbiology ,blacklegged tick ,23S ribosomal RNA ,parasitic diseases ,RNA FISH ,Rickettsia buchneri ,Ixodes ricinus cell line IRE11 ,tick cell lines ,Ixodes scapularis endosymbiont ,biology ,fungi ,biology.organism_classification ,bacterial infections and mycoses ,Ixodes scapularis ,Cell culture ,Insect Science ,Tissue tropism ,bacteria ,Ixodes ,Cellular Tropism - Abstract
The blacklegged tick, Ixodes scapularis, a species of significant importance to human and animal health, harbors an endosymbiont Rickettsia buchneri sensu stricto. The symbiont is largely restricted to the ovaries, but all life stages can harbor various quantities or lack R. buchneri entirely. The endosymbiont is cultivable in cell lines isolated from embryos of Ixodes ticks. Rickettsia buchneri most readily grows and is maintained in the cell line IRE11 from the European tick, Ixodes ricinus. The line was characterized by light and electron microscopy and used to analyze the growth dynamics of wildtype and GFPuv-expressing R. buchneri. qPCR indicated that the genome copy doubling time in IRE11 was >, 7 days. Measurements of fluorescence using a plate reader indicated that the amount of green fluorescent protein doubled every 11 days. Two 23S rRNA probes were tested via RNA FISH on rickettsiae grown in vitro and adapted to evaluate the tissue tropism of R. buchneri in field-collected female I. scapularis. We observed strong positive signals of R. buchneri in the ovaries and surrounding the nucleus of the developing oocytes. Tissue tropism in I. scapularis and in vitro growth dynamics strengthen the contemporary understanding of R. buchneri as a transovarially transmitted, non-pathogenic endosymbiont.
- Published
- 2021
- Full Text
- View/download PDF
35. In vitro and in vivo evaluation of human adenovirus type 49 as a vector for therapeutic applications
- Author
-
Angela C. Bradshaw, Sophie Alizert, Emily A. Bates, Simon N. Waddington, Natalie Suff, Ashley Boyle, Stuart A. Nicklin, Andrew H. Baker, Alan L. Parker, Alexander T. Baker, and John R. Counsell
- Subjects
0301 basic medicine ,Cell type ,Genetic enhancement ,Genetic Vectors ,Biology ,Microbiology ,Article ,Cell Line ,Viral vector ,ADENOVIRUS ,Mice ,03 medical and health sciences ,Transduction (genetics) ,0302 clinical medicine ,Immune system ,Genes, Reporter ,Transduction, Genetic ,In vivo ,Virology ,Animals ,Humans ,biochemistry ,Lung ,Phylogeny ,Vaccines ,Adenoviruses, Human ,viral vector ,Genetic Therapy ,adenovirus ,vaccines ,gene therapy ,QR1-502 ,Cell biology ,030104 developmental biology ,Infectious Diseases ,Liver ,030220 oncology & carcinogenesis ,Spleen ,Cellular Tropism ,Ex vivo - Abstract
The human adenovirus phylogenetic tree is split across seven species (A–G). Species D adenoviruses offer potential advantages for gene therapy applications, with low rates of pre-existing immunity detected across screened populations. However, many aspects of the basic virology of species D—such as their cellular tropism, receptor usage, and in vivo biodistribution profile—remain unknown. Here, we have characterized human adenovirus type 49 (HAdV-D49)—a relatively understudied species D member. We report that HAdV-D49 does not appear to use a single pathway to gain cell entry, but appears able to interact with various surface molecules for entry. As such, HAdV-D49 can transduce a broad range of cell types in vitro, with variable engagement of blood coagulation FX. Interestingly, when comparing in vivo biodistribution to adenovirus type 5, HAdV-D49 vectors show reduced liver targeting, whilst maintaining transduction of lung and spleen. Overall, this presents HAdV-D49 as a robust viral vector platform for ex vivo manipulation of human cells, and for in vivo applications where the therapeutic goal is to target the lung or gain access to immune cells in the spleen, whilst avoiding liver interactions, such as intravascular vaccine applications.
- Published
- 2021
- Full Text
- View/download PDF
36. The Influence of Murine Genetic Background in Adeno-Associated Virus Transduction of the Mouse Brain
- Author
-
Chengwen Li, Nikita Elexa Hall, Michelle S. Itano, Ting He, R. Jude Samulski, Natallia V. Riddick, and Lauriel F. Earley
- Subjects
viruses ,Genetic Vectors ,Green Fluorescent Proteins ,Mutant ,Central nervous system ,Mice, Inbred Strains ,Nerve Tissue Proteins ,Biology ,Serogroup ,medicine.disease_cause ,Virus ,Green fluorescent protein ,Mice ,Transduction (genetics) ,Transduction, Genetic ,medicine ,Animals ,Gene ,Adeno-associated virus ,Genetics (clinical) ,Brain ,Original Articles ,Dependovirus ,Virology ,Corpus Striatum ,DNA-Binding Proteins ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Rotarod Performance Test ,Female ,Microglia ,Genetic Background ,Cellular Tropism - Abstract
Adeno-associated virus (AAV) vectors have become an important tool for delivering therapeutic genes for a wide range of neurological diseases. AAV serotypes possess differential cellular tropism in the central nervous system. Although several AAV serotypes or mutants have been reported to transduce the brain efficiently, conflicting data occur across studies with the use of various rodent strains from different genetic backgrounds. Herein, we performed a systematic comparison of the brain transduction properties among five AAV serotypes (AAV2, 5, 7, 8, and 9) in two common rodent strains (C57BL/6J and FVB/N), following local intrastriatal injection of AAV vectors encoding enhanced green fluorescent protein (EGFP) driven by the CBh promoter. Important differences were found regarding overall cellular tropism and transduction efficiency, including contralateral transduction among the AAV serotypes and between the mouse strains. We have further found loss of NeuN-immunoreactivity and microglial activation from AAV transduction in the different mouse strains. The important strain-specific differences from our study suggest that the genetic background of the mouse may affect AAV serotype transduction properties in the brain. These data can provide valuable information about how to choose an effective AAV vector for clinical application and interpret the data obtained from preclinical studies and clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
37. Replication kinetics and cellular tropism of emerging reoviruses in sheep and swine respiratory ex vivo organ cultures
- Author
-
Alessandra Leone, Maria Loredana Colaianni, Antonio Petrini, Giovanni Di Teodoro, Alessio Lorusso, Giovanni Savini, Nicola D'Alterio, Francesco Bonfante, Alfonso Rosamilia, Alessio Bortolami, Liana Teodori, Calogero Terregino, and Daniela Malatesta
- Subjects
Swine ,viruses ,Bronchi ,Biology ,Virus Replication ,Microbiology ,Virus ,03 medical and health sciences ,Organ Culture Techniques ,In vivo ,Animals ,Respiratory system ,Tropism ,030304 developmental biology ,Infectivity ,0303 health sciences ,Sheep ,General Veterinary ,030306 microbiology ,General Medicine ,Virology ,Kinetics ,Orthoreovirus ,Viral Tropism ,Alveolar Epithelial Cells ,Respiratory virus ,Bluetongue virus ,Cellular Tropism ,Ex vivo - Abstract
Ex vivo organ cultures (EVOCs) are extensively used to study the cellular tropism and infectivity of different pathogens. In this study, we used ovine and porcine respiratory EVOCs to investigate the replication kinetics and cellular tropism of selected emerging reoviruses namely Pteropine orthoreovirus, an emerging bat-borne zoonotic respiratory virus, and atypical Bluetongue virus (BTV) serotypes which, unlike classical serotypes, do not cause Bluetongue, a major OIE-listed disease of ruminants. BTV failed to replicate in ovine EVOCs. Instead, PRV showed slight replication in porcine lower respiratory EVOCs and a more sustained replication in all ovine respiratory tissues. By confocal laser scanning microscopy, PRV was demonstrated to infect bronchiolar and type I pneumocytes of ovine tissues. Overall, respiratory EVOCs from different animal species, eventually obtained at slaughterhouse, are a useful tool for testing and preliminarily characterize novel and emerging viruses addressing the essential in vivo animal work. Further experiments are, indeed, warranted in order to characterize the pathogenesis and transmission of these emerging reoviruses.
- Published
- 2019
- Full Text
- View/download PDF
38. Molecular mechanism by which residues at position 481 and 546 of measles virus hemagglutinin protein define CD46 receptor binding using a molecular docking approach
- Author
-
Amruta Shirode, Sara Sajjadi, Sarah S. Cherian, and Sunil R. Vaidya
- Subjects
0301 basic medicine ,Viral pathogenesis ,viruses ,Ligand (biochemistry) ,Hemagglutinins, Viral ,Hemagglutinin (influenza) ,Wild type ,Biochemistry ,Membrane Cofactor Protein ,Measles virus ,Docking (molecular) ,03 medical and health sciences ,Residue (chemistry) ,0302 clinical medicine ,Structural Biology ,Genetics ,Humans ,Amino Acid Sequence ,Receptor ,Biology ,CD46 ,Binding Sites ,biology ,Chemistry ,Organic Chemistry ,Hydrogen Bonding ,biology.organism_classification ,female genital diseases and pregnancy complications ,3. Good health ,Molecular Docking Simulation ,Computational Mathematics ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Receptors, Virus ,Cellular Tropism ,Protein Binding - Abstract
The hemagglutinin (H) protein of measles viruses (MeV) mediates binding to the cellular receptors, CD46,human signaling lymphocyte activation molecule and nectin-4. Vaccine strains primarily contain H-proteins possessing MeV-H: Y481 and can utilize CD46. Reports suggest that a single amino acid change in MeV-H at position 481 in wild type strains renders them inefficient in utilizing CD46. The in-depth molecular mechanism by which substitutions at 481 and another reported critical residue position 546 affects CD46 binding affinity however remains elusive. We used molecular docking studies of CD46 with MeV-H possessing Y481 N/D to understand the in-depth molecular mechanism involved. It was found that loss in either of the hydrogen bond (H-bond) contacts (MeV-H:481–CD46:65, MeV-H:546–CD46:63) in the central contact region prevented efficient CD46 binding. Y481 N could form the specific H-bond, while G546S H-bond could be formed only in conjunction with Y481, revealing the significance of these residues in determining CD46 receptor binding potential. Elucidating the underlying molecular mechanism of receptor usage by the MeV has implications to understanding cellular tropism, viral pathogenesis and therapy.
- Published
- 2019
39. Evolutionary aspects of Parvovirus B-19V associated diseases and their pathogenesis patterns with an emphasis on vaccine development
- Author
-
Piyanki Das, Tathagata Choudhuri, Nabanita Roy Chattopadhyay, and Koustav Chatterjee
- Subjects
Parvoviridae ,biology ,Parvovirus ,viruses ,Review Article ,Disease ,biology.organism_classification ,Virology ,Virus ,Pathogenesis ,Vaccination ,Infectious Diseases ,Erythroid Progenitor Cells ,Cellular Tropism - Abstract
Parvovirus B-19, a single human pathogenic member of the Parvoviridae family with it's small ssDNA and non-enveloped structure, is known to cause diseases in erythroid progenitor cells. But a wide range of clinical association of this virus with cells of non-erythroid origins has recently been discovered and many of those are being investigated for such association. Higher substitution rates in due course of evolution has been suggested for this cellular tropism and persistence. In this report, we have summarized the different disease manifestations of B-19 virus and have tried to find out a pattern of pathogenesis. Finally, we have focused on the vaccination strategies available against B-19 virus to correlate these with the mechanisms involved in various diseases caused by this virus.
- Published
- 2019
- Full Text
- View/download PDF
40. Cell-type specific innate immune responses shape rabies virus tropism
- Author
-
Hervé Bourhy, G. Dias De Melo, Lena Feige, Vincent Guillemot, Florence Larrous, Florent Ginhoux, and T. Kozaki
- Subjects
Immune system ,Innate immune system ,Viral replication ,Rabies virus ,Tissue tropism ,medicine ,Biology ,medicine.disease_cause ,Virology ,Cellular Tropism ,Virus ,Tropism - Abstract
Viral tropism, or the specificity of a particular virus to infect a certain cell type, is crucial in determining virus replication, viral spread, and ultimately host survival. Rabies, one of the deadliest known zoonotic diseases, is still causing 60.000 human deaths annually. Upon central nervous system (CNS) entry, neurotropic rabies virus (RABV) preserves the neural network by limiting apoptosis and inflammation. To date, we do not fully understand the factors determining RABV tropism and why glial cells are unable to clear RABV from the infected brain. Here, we compare susceptibilities and innate immune responses of CNS cell types towards infection with virulent dog RABV Tha and less virulent Th2P-4M in vitro, highlighting differences in cellular susceptibility and antiviral responses. Less virulent Th2P-4M bears mutations introduced in viral phosphoprotein (P-protein) and matrix protein (M-protein) thereby hindering viral immune evasion of the host nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and Janus kinase (JAK) - signal transducer and activator of transcription protein (STAT) pathways. Our results reveal that human neural stem cell (hNSC)-derived neurons and astrocytes, in contrast to human iPSC-derived microglia, are highly susceptible to Tha and Th2P-4M infection in vitro. Surprisingly, Th2P-4M presents a stronger neurotropism in hNSC-derived CNS cultures compared to Tha suggesting that NF-κB- and JAK-STAT-mediated antiviral host responses are defining RABV replication and thereby its tropism. Further, we show that astrocyte-like (SVGp12) and microglia-like (HMC3) cells protect neuroblastoma cells (SK-N-SH) from Tha infection in vitro. Transcription profiles and quantification of intracellular protein levels revealed major differences in antiviral immune responses mediated by neurons, astrocytes (IFNB1, CCL5, CXCL10, IL1B, IL6, LIF), and microglia (CCL5, CXCL10, ISG15, MX1, IL6) upon virulent Tha infection. Overall, we provide evidence that RABV tropism depends on its capability to evade cell-type specific immune responses via P- and M-proteins.Author summaryRabies virus (RABV) neurotropism is widely reported as a unique feature of rabies, and still the exact mechanism underlying RABV susceptibility remains to be elucidated. Several receptors are known to accelerate RABV entry to the cell (1–4) and yet, none of them seems to be essential for successful infection (5,6) questioning a marked cellular tropism. Although RABV is classically reported as strictly neurotropic (7), recent studies report profound infection of glial cells in vivo depending on the viral strain and the infection route used (8,9). Here, we provide evidence that human neural stem cell (hNSC)-derived neurons (hiNeurons) and astrocytes (hiAstrocytes) are highly susceptible towards infection with the virulent field RABV strain Tha and less virulent Th2P-4M. In contrast, human iPSC-derived microglia-like cells (hiMicros) are resistant to viral replication in vitro. Whereas hiNeurons are immunologically quiescent upon Tha infection, fetal astrocytes and hiMicros establish strong antiviral responses. In contrast to Tha, Th2P-4M, which is unable to evade NF-κB and JAK-STAT pathways (10), shows a more profound neurotropism suggesting that cell-type specific responses shape RABV tropism. Hence, we conclude that viral evasion mechanisms mediated by P- and M-proteins partly determine Tha tropism of human CNS cell types in vitro.
- Published
- 2021
- Full Text
- View/download PDF
41. A Multifaceted Approach to Optimizing AAV Delivery to the Brain for the Treatment of Neurodegenerative Diseases
- Author
-
Jonathan Fischell and Paul S. Fishman
- Subjects
Intracerebral injection ,Transgene ,Genetic enhancement ,viruses ,Neurosciences. Biological psychiatry. Neuropsychiatry ,adeno-associated virus (AAV) ,Bioinformatics ,Focused ultrasound ,capsid engineering ,Transduction (genetics) ,neurodegenerative disease ,Hypothesis and Theory ,Medicine ,Significant risk ,Vector (molecular biology) ,blood–brain barrier disruption ,business.industry ,General Neuroscience ,intra-arterial (IA) delivery ,gene therapy ,genetic vectors ,business ,Cellular Tropism ,RC321-571 ,Neuroscience ,intra-thecal drug delivery systems - Abstract
Despite major advancements in gene therapy technologies, there are no approved gene therapies for diseases which predominantly effect the brain. Adeno-associated virus (AAV) vectors have emerged as the most effective delivery vector for gene therapy owing to their simplicity, wide spread transduction and low immunogenicity. Unfortunately, the blood–brain barrier (BBB) makes IV delivery of AAVs, to the brain highly inefficient. At IV doses capable of widespread expression in the brain, there is a significant risk of severe immune-mediated toxicity. Direct intracerebral injection of vectors is being attempted. However, this method is invasive, and only provides localized delivery for diseases known to afflict the brain globally. More advanced methods for AAV delivery will likely be required for safe and effective gene therapy to the brain. Each step in AAV delivery, including delivery route, BBB transduction, cellular tropism and transgene expression provide opportunities for innovative solutions to optimize delivery efficiency. Intra-arterial delivery with mannitol, focused ultrasound, optimized AAV capsid evolution with machine learning algorithms, synthetic promotors are all examples of advanced strategies which have been developed in pre-clinical models, yet none are being investigated in clinical trials. This manuscript seeks to review these technological advancements, and others, to improve AAV delivery to the brain, and to propose novel strategies to build upon this research. Ultimately, it is hoped that the optimization of AAV delivery will allow for the human translation of many gene therapies for neurodegenerative and other neurologic diseases.
- Published
- 2021
42. Nasal ciliated cells are primary targets for SARS-CoV-2 replication in the early stage of COVID-19
- Author
-
Ji-Hoon Ahn, M D Tazikur Rahman, Chang-Seop Lee, Young Seok Ju, JungMo Kim, Sang Duk Hong, Ho Min Kim, Seon Pyo Hong, Man Ki Chung, Young Tae Kim, Hosung Bae, Gou Young Koh, Myung Jin Yang, and Sung Yong Choi
- Subjects
0301 basic medicine ,viruses ,Cellular differentiation ,Mucous membrane of nose ,Biology ,Virus Replication ,Models, Biological ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Cilia ,RNA, Messenger ,RNA-Seq ,Pandemics ,Tropism ,Furin ,Host Microbial Interactions ,SARS-CoV-2 ,Stem Cells ,Cilium ,Serine Endopeptidases ,COVID-19 ,Cell Differentiation ,General Medicine ,Virus Internalization ,Epithelium ,Cell biology ,Nasal Mucosa ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Macaca ,Angiotensin-Converting Enzyme 2 ,Stem cell ,Cellular Tropism ,Research Article ,Respiratory tract - Abstract
The upper respiratory tract is compromised in the early period of COVID-19, but SARS-CoV-2 tropism at the cellular level is not fully defined. Unlike recent single-cell RNA-Seq analyses indicating uniformly low mRNA expression of SARS-CoV-2 entry-related host molecules in all nasal epithelial cells, we show that the protein levels are relatively high and that their localizations are restricted to the apical side of multiciliated epithelial cells. In addition, we provide evidence in patients with COVID-19 that SARS-CoV-2 is massively detected and replicated within the multiciliated cells. We observed these findings during the early stage of COVID-19, when infected ciliated cells were rapidly replaced by differentiating precursor cells. Moreover, our analyses revealed that SARS-CoV-2 cellular tropism was restricted to the nasal ciliated versus oral squamous epithelium. These results imply that targeting ciliated cells of the nasal epithelium during the early stage of COVID-19 could be an ideal strategy to prevent SARS-CoV-2 propagation.
- Published
- 2021
- Full Text
- View/download PDF
43. Viruses Like Sugars: How to Assess Glycan Involvement in Viral Attachment
- Author
-
Gregory Mathez and Valeria Cagno
- Subjects
0301 basic medicine ,Microbiology (medical) ,Glycan ,2019-20 coronavirus outbreak ,QH301-705.5 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,viruses ,030106 microbiology ,Computational biology ,Review ,Biology ,Microbiology ,Viral infection ,Glycocalyx ,03 medical and health sciences ,chemistry.chemical_compound ,EV-D68 ,Virology ,Biology (General) ,glycan ,SARS-CoV-2 ,Heparan sulfate ,HBGA ,3. Good health ,030104 developmental biology ,chemistry ,attachment receptor ,sialic acid ,biology.protein ,heparan sulfate ,Enterovirus D68 ,Cellular Tropism - Abstract
The first step of viral infection requires interaction with the host cell. Before finding the specific receptor that triggers entry, the majority of viruses interact with the glycocalyx. Identifying the carbohydrates that are specifically recognized by different viruses is important both for assessing the cellular tropism and for identifying new antiviral targets. Advances in the tools available for studying glycan–protein interactions have made it possible to identify them more rapidly; however, it is important to recognize the limitations of these methods in order to draw relevant conclusions. Here, we review different techniques: genetic screening, glycan arrays, enzymatic and pharmacological approaches, and surface plasmon resonance. We then detail the glycan interactions of enterovirus D68 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), highlighting the aspects that need further clarification.
- Published
- 2021
44. Cellular tropism and antigenicity of mink-derived SARS-CoV-2 variants
- Author
-
Xiaoming Yang, Youchun Wang, Qianqian Li, Xuguang Li, Jianhui Nie, Haixin Wang, Li Zhang, Ruxia Ding, Weijin Huang, and Jiajing Wu
- Subjects
Cancer Research ,Antigenicity ,2019-20 coronavirus outbreak ,Letter ,Glycosylation ,Coronavirus disease 2019 (COVID-19) ,Swine ,QH301-705.5 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Madin Darby Canine Kidney Cells ,Betacoronavirus ,Mice ,Dogs ,Cricetinae ,biology.animal ,Chlorocebus aethiops ,Genetics ,Animals ,Humans ,Biology (General) ,Mink ,Antigens, Viral ,Vero Cells ,Tropism ,Vaccines ,Binding Sites ,Virulence ,biology ,SARS-CoV-2 ,Macaca mulatta ,Virology ,HEK293 Cells ,RAW 264.7 Cells ,A549 Cells ,Mutation ,Spike Glycoprotein, Coronavirus ,Mutation (genetic algorithm) ,Medicine ,Cattle ,Infection ,Cellular Tropism ,HeLa Cells - Abstract
The spike protein of SARS-CoV-2 has been undergoing mutations and is highly glycosylated. It is critically important to investigate the biological significance of these mutations. Here, we investigated 80 variants and 26 glycosylation site modifications for the infectivity and reactivity to a panel of neutralizing antibodies and sera from convalescent patients. D614G, along with several variants containing both D614G and another amino acid change, were significantly more infectious. Most variants with amino acid change at receptor binding domain were less infectious, but variants including A475V, L452R, V483A, and F490L became resistant to some neutralizing antibodies. Moreover, the majority of glycosylation deletions were less infectious, whereas deletion of both N331 and N343 glycosylation drastically reduced infectivity, revealing the importance of glycosylation for viral infectivity. Interestingly, N234Q was markedly resistant to neutralizing antibodies, whereas N165Q became more sensitive. These findings could be of value in the development of vaccine and therapeutic antibodies.
- Published
- 2021
- Full Text
- View/download PDF
45. Cellular Tropism of SARS-CoV-2 across Human Tissues and Age-related Expression of ACE2 and TMPRSS2 in Immune-inflammatory Stromal Cells
- Author
-
Ming Zheng
- Subjects
Cell type ,Opinion ,Stromal cell ,SARS-CoV-2 ,immune-inflammatory stromal cells ,Viral pathogenesis ,viruses ,human tissues ,ACE2 ,Cell Biology ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Viral entry ,Immunology ,medicine ,Tissue tropism ,Neurology (clinical) ,Geriatrics and Gerontology ,Tropism ,Cellular Tropism ,TMPRSS2 ,Coronavirus - Abstract
Recently, emerging evidence has indicated that COVID-19 represents a major threat to older populations, but the underlying mechanisms remain unclear. The pathogen causing COVID-19 is acute respiratory syndrome coronavirus 2 (SARS-CoV-2). SARS-CoV-2 infection depends on the key entry factors, angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSS2). Recognizing the importance of ACE2 and TMPRSS2 for the cellular tropism of SARS-CoV-2, we analyzed and presented the landscape of cell-type identities for ACE2+ TMPRSS2+ cells across different human tissues and the age-related alterations in ACE2 and TMPRSS2 expression across different cell types. Additionally, most of the post-acute COVID-19 sequelae could attribute to the ACE2-expressing organ systems. Therefore, these SARS-CoV-2 tropism data should be an essential resource for guiding clinical treatment and pathological studies, which should draw attention toward the prioritization of COVID-19 research in the future. Notably, we discovered the age-related expression of ACE2 and TMPRSS2 in the immune-inflammatory stromal cells, implying the potential interplay between COVID-19, stromal cells, and aging. In this study, we developed a novel and practical analysis framework for mapping the cellular tropism of SARS-CoV-2. This approach was built to aid the identification of viral-specific cell types and age-related alterations of viral tropism, highlighting the power of single-cell RNA sequencing (scRNA-seq) to address viral pathogenesis systematically. With the rapid accumulation of scRNA-seq data and the continuously increasing insight into viral entry factors, we anticipate that this scRNA-seq-based approach will attract broader interest in the virus research communities.
- Published
- 2021
46. Contributions of the Four Essential Entry Glycoproteins to HSV-1 Tropism and the Selection of Entry Routes
- Author
-
Adam T. Hilterbrand, Raecliffe Daly, and Ekaterina E. Heldwein
- Subjects
Cell type ,viruses ,Cell ,Herpesvirus 1, Human ,HSL and HSV ,Biology ,Endocytosis ,Microbiology ,Cell Line ,Host-Microbe Biology ,Viral Envelope Proteins ,Viral envelope ,Virology ,Chlorocebus aethiops ,inhibitors ,medicine ,Animals ,Humans ,endocytosis ,herpes simplex viruses ,Vero Cells ,cell entry ,glycoproteins ,Tropism ,chemistry.chemical_classification ,Virus Internalization ,biology.organism_classification ,Phenotype ,HSV-1 ,QR1-502 ,Cell biology ,Molecular Typing ,Viral Tropism ,HEK293 Cells ,pseudotypes ,medicine.anatomical_structure ,chemistry ,Vesicular stomatitis virus ,Cell culture ,Gain of Function Mutation ,Pseudotyping ,biology.protein ,Antibody ,Glycoprotein ,Cellular Tropism ,HeLa Cells ,Research Article - Abstract
Herpes simplex viruses (HSV-1 and HSV-2) contain up to 16 different proteins in their envelopes. Four of these, glycoproteins gB, gD, gH, and gL, are termed essential with regard to entry, whereas the rest are typically referred to as nonessential based on the entry phenotypes of the respective single genetic deletions., Herpes simplex viruses (HSV-1 and HSV-2) encode up to 16 envelope proteins, four of which are essential for entry. However, whether these four proteins alone are sufficient to dictate the broad cellular tropism of HSV-1 and the selection of different cell type-dependent entry routes is unknown. To begin addressing this, we previously pseudotyped vesicular stomatitis virus (VSV), lacking its native glycoprotein G, with only the four essential entry glycoproteins of HSV-1: gB, gH, gL, and gD. This novel VSVΔG-BHLD pseudotype recapitulated several important features of HSV-1 entry: the requirement for gB, gH, gL, gD, and a cellular receptor and sensitivity to anti-gB and anti-gH/gL neutralizing antibodies. However, due to the use of a single cell type in that study, the tropism of the VSVΔG-BHLD pseudotype was not investigated. Here, we show that the cellular tropism of the pseudotype is severely limited compared to that of wild-type HSV-1 and that its entry pathways differ from the native HSV-1 entry pathways. To test the hypothesis that other HSV-1 envelope proteins may contribute to HSV-1 tropism, we generated a derivative pseudotype containing the HSV-1 glycoprotein C (VSVΔG-BHLD-gC) and observed a gC-dependent increase in entry efficiency in two cell types. We propose that the pseudotyping platform developed here has the potential to uncover functional contributions of HSV-1 envelope proteins to entry in a gain-of-function manner.
- Published
- 2021
- Full Text
- View/download PDF
47. The Fiber Knob Protein of Human Adenovirus Type 49 Mediates Highly Efficient and Promiscuous Infection of Cancer Cell Lines Using a Novel Cell Entry Mechanism
- Author
-
Carly M. Bliss, Pierre J. Rizkallah, James A. Davies, Emily A. Bates, Alan L. Parker, Elise Moses, Gareth Marlow, Rosie M. Mundy, David K. Cole, and Alexander T. Baker
- Subjects
Genetic Vectors ,Immunology ,Population ,Coxsackievirus ,Microbiology ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,Virology ,Humans ,anticancer therapy ,Vector (molecular biology) ,Receptor ,education ,Tropism ,030304 developmental biology ,Oncolytic Virotherapy ,0303 health sciences ,education.field_of_study ,biology ,CD46 ,Adenoviruses, Human ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Virus Internalization ,surface receptor ,biology.organism_classification ,eye diseases ,Virus-Cell Interactions ,3. Good health ,Oncolytic virus ,Cell biology ,adenoviruses ,oncolytic viruses ,030220 oncology & carcinogenesis ,Insect Science ,Receptors, Virus ,Capsid Proteins ,Cellular Tropism - Abstract
Adenoviruses are powerful tools experimentally and clinically. To maximize efficacy, the development of serotypes with low preexisting levels of immunity in the population is desirable., The human adenovirus (HAdV) phylogenetic tree is diverse, divided across seven species and comprising over 100 individual types. Species D HAdV are rarely isolated with low rates of preexisting immunity, making them appealing for therapeutic applications. Several species D vectors have been developed as vaccines against infectious diseases, where they induce robust immunity in preclinical models and early phase clinical trials. However, many aspects of the basic virology of species D HAdV, including their basic receptor usage and means of cell entry, remain understudied. Here, we investigated HAdV-D49, which previously has been studied for vaccine and vascular gene transfer applications. We generated a pseudotyped HAdV-C5 presenting the HAdV-D49 fiber knob protein (HAdV-C5/D49K). This pseudotyped vector was efficient at infecting cells devoid of all known HAdV receptors, indicating HAdV-D49 uses an unidentified cellular receptor. Conversely, a pseudotyped vector presenting the fiber knob protein of the closely related HAdV-D30 (HAdV-C5/D30K), differing in four amino acids from HAdV-D49, failed to demonstrate the same tropism. These four amino acid changes resulted in a change in isoelectric point of the knob protein, with HAdV-D49K possessing a basic apical region compared to a more acidic region in HAdV-D30K. Structurally and biologically we demonstrate that HAdV-D49 knob protein is unable to engage CD46, while potential interaction with coxsackievirus and adenovirus receptor (CAR) is extremely limited by extension of the DG loop. HAdV-C5/49K efficiently transduced cancer cell lines of pancreatic, breast, lung, esophageal, and ovarian origin, indicating it may have potential for oncolytic virotherapy applications, especially for difficult to transduce tumor types. IMPORTANCE Adenoviruses are powerful tools experimentally and clinically. To maximize efficacy, the development of serotypes with low preexisting levels of immunity in the population is desirable. Consequently, attention has focused on those derived from species D, which have proven robust vaccine platforms. This widespread usage is despite limited knowledge in their basic biology and cellular tropism. We investigated the tropism of HAdV-D49, demonstrating that it uses a novel cell entry mechanism that bypasses all known HAdV receptors. We demonstrate, biologically, that a pseudotyped HAdV-C5/D49K vector efficiently transduces a wide range of cell lines, including those presenting no known adenovirus receptor. Structural investigation suggests that this broad tropism is the result of a highly basic electrostatic surface potential, since a homologous pseudotyped vector with a more acidic surface potential, HAdV-C5/D30K, does not display a similar pantropism. Therefore, HAdV-C5/D49K may form a powerful vector for therapeutic applications capable of infecting difficult to transduce cells.
- Published
- 2021
- Full Text
- View/download PDF
48. CD300lf Conditional Knockout Mouse Reveals Strain-Specific Cellular Tropism of Murine Norovirus
- Author
-
Jin Wei, Vincent R. Graziano, Renata B. Filler, Sanghyun Lee, Craig B. Wilen, Mia Madel Alfajaro, Madison S. Strine, Timothy J. Nice, Cameron O. Schmitz, Megan T. Baldridge, Robert C. Orchard, and Leon L. Hsieh
- Subjects
Male ,Cell type ,Receptor expression ,Immunology ,ved/biology.organism_classification_rank.species ,Microbiology ,CD19 ,Mice ,03 medical and health sciences ,Virology ,Conditional gene knockout ,Animals ,Receptors, Immunologic ,Tropism ,Caliciviridae Infections ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Innate immune system ,biology ,030306 microbiology ,ved/biology ,Norovirus ,Epithelial Cells ,Immunity, Innate ,Virus-Cell Interactions ,Cell biology ,Intestines ,Viral Tropism ,Insect Science ,Host-Pathogen Interactions ,biology.protein ,Female ,Cellular Tropism ,Murine norovirus - Abstract
Noroviruses are a leading cause of gastrointestinal infection in humans and mice. Understanding human norovirus (HuNoV) cell tropism has important implications for our understanding of viral pathogenesis. Murine norovirus (MNoV) is extensively used as a surrogate model for HuNoV. We previously identified CD300lf as the receptor for MNoV. Here, we generated a Cd300lf conditional knockout (CD300lf(F/F)) mouse to elucidate the cell tropism of persistent and nonpersistent strains of murine norovirus. Using this mouse model, we demonstrated that CD300lf expression on intestinal epithelial cells (IECs), and on tuft cells in particular, is essential for transmission of the persistent MNoV strain CR6 (MNoV(CR6)) in vivo. In contrast, the nonpersistent MNoV strain CW3 (MNoV(CW3)) does not require CD300lf expression on IECs for infection. However, deletion of CD300lf in myelomonocytic cells (LysM Cre+) partially reduces CW3 viral load in lymphoid and intestinal tissues. Disruption of CD300lf expression on B cells (CD19 Cre), neutrophils (Mrp8 Cre), and dendritic cells (CD11c Cre) did not affect MNoV(CW3) viral RNA levels. Finally, we show that the transcription factor STAT1, which is critical for the innate immune response, partially restricts the cell tropism of MNoV(CW3) to LysM+ cells. Taken together, these data demonstrate that CD300lf expression on tuft cells is essential for MNoV(CR6); that myelomonocytic cells are a major, but not exclusive, target cell of MNoV(CW3); and that STAT1 signaling restricts the cellular tropism of MNoV(CW3). This study provides the first genetic system for studying the cell type-specific role of CD300lf in norovirus pathogenesis. IMPORTANCE Human noroviruses (HuNoVs) are a leading cause of gastroenteritis resulting in up to 200,000 deaths each year. The receptor and cell tropism of HuNoV in immunocompetent humans are unclear. We use murine norovirus (MNoV) as a model for HuNoV. We recently identified CD300lf as the sole physiologic receptor for MNoV. Here, we leverage this finding to generate a Cd300lf conditional knockout mouse to decipher the contributions of specific cell types to MNoV infection. We demonstrate that persistent MNoV(CR6) requires CD300lf expression on tuft cells. In contrast, multiple CD300lf+ cell types, dominated by myelomonocytic cells, are sufficient for nonpersistent MNoV(CW3) infection. CD300lf expression on epithelial cells, B cells, neutrophils, and dendritic cells is not critical for MNoV(CW3) infection. Mortality associated with the MNoV(CW3) strain in Stat1(−/−) mice does not require CD300lf expression on LysM+ cells, highlighting that both CD300lf receptor expression and innate immunity regulate MNoV cell tropism in vivo.
- Published
- 2021
- Full Text
- View/download PDF
49. Comparison of primary virus isolation in pulmonary alveolar macrophages and four different continuous cell lines for type 1 and type 2 porcine reproductive and respiratory syndrome virus
- Author
-
Nick Vereecke, Ivan Trus, Jiexiong Xie, Dayoung Oh, Sebastiaan Theuns, Jannes Sauer, Carl A. Gagnon, Nathalie Vanderheijden, Hans Nauwynck, Christian Lalonde, P Vyt, Caroline Bonckaert, Chantale Provost, and Université de Montréal. Faculté de médecine vétérinaire
- Subjects
0301 basic medicine ,INVOLVEMENT ,viruses ,0403 veterinary science ,Drug Discovery ,INFECTION ,PK15Sn-CD163 ,Pharmacology (medical) ,Receptor ,NUCLEOCAPSID PROTEIN ,virus isolation ,biology ,04 agricultural and veterinary sciences ,SYNDROME PRRS VIRUS ,macrophages ,Infectious Diseases ,Medicine ,Cellular Tropism ,Cell type ,PK15(S10-CD163) ,040301 veterinary sciences ,PK15(Sn-CD163) ,Immunology ,PK15S10-CD163 ,DENDRITIC CELLS ,Article ,Virus ,03 medical and health sciences ,NORTH-AMERICAN ,Sialoadhesin ,SIALOADHESIN ,Veterinary Sciences ,Tropism ,Pharmacology ,MARC-145 ,IDENTIFICATION ,Macrophages ,Porcine reproductive and respiratory syndrome virus ,biology.organism_classification ,EFFICACY ,Virology ,030104 developmental biology ,Cell culture ,PRRSV ,REPLICATION ,Virus isolation/production ,production - Abstract
Porcine Reproductive and Respiratory Syndrome Virus (PRRSV) has a highly restricted cellular tropism. In vivo, the virus primarily infects tissue-specific macrophages in the nose, lungs, tonsils, and pharyngeal lymphoid tissues. In vitro however, the MARC-145 cell line is one of the few PRRSV susceptible cell lines that are routinely used for in vitro propagation. Previously, several PRRSV non-permissive cell lines were shown to become susceptible to PRRSV infection upon expression of recombinant entry receptors (e.g., PK15Sn-CD163, PK15S10-CD163). In the present study, we examined the suitability of different cell lines as a possible replacement of primary pulmonary alveolar macrophages (PAM) cells for isolation and growth of PRRSV. The susceptibility of four different cell lines (PK15Sn-CD163, PK15S10-CD163, MARC-145, and MARC-145Sn) for the primary isolation of PRRSV from PCR positive sera (both PRRSV1 and PRRSV2) was compared with that of PAM. To find possible correlations between the cell tropism and the viral genotype, 54 field samples were sequenced, and amino acid residues potentially associated with the cell tropism were identified. Regarding the virus titers obtained with the five different cell types, PAM gave the highest mean virus titers followed by PK15Sn-CD163, PK15S10-CD163, MARC-145Sn, and MARC-145. The titers in PK15Sn-CD163 and PK15S10-CD163 cells were significantly correlated with virus titers in PAM for both PRRSV1 (p <, 0.001) and PRRSV2 (p <, 0.001) compared with MARC-145Sn (PRRSV1: p = 0.22 and PRRSV2: p = 0.03) and MARC-145 (PRRSV1: p = 0.04 and PRRSV2: p = 0.12). Further, a possible correlation between cell tropism and viral genotype was assessed using PRRSV whole genome sequences in a Genome-Wide-Association Study (GWAS). The structural protein residues GP2:187L and N:28R within PRRSV2 sequences were associated with their growth in MARC-145. The GP5:78I residue for PRRSV2 and the Nsp11:155F residue for PRRSV1 was linked to a higher replication on PAM. In conclusion, PK15Sn-CD163 and PK15S10-CD163 cells are phenotypically closely related to the in vivo target macrophages and are more suitable for virus isolation and titration than MARC-145/MARC-145Sn cells. The residues of PRRSV proteins that are potentially related with cell tropism will be further investigated in the future.
- Published
- 2021
50. Elevated temperature inhibits SARS-CoV-2 replication in respiratory epithelium independently of the induction of IFN-mediated innate immune defences
- Author
-
Ilaria Epifano, Agnieszka M. Szemiel, Kieran Dee, Andrew Stevenson, Meredith Stewart, Quan Gu, Rute Maria Pinto, Steven McFarlane, Daniel M. Goldfarb, Sheila V. Graham, Ruth F. Jarrett, Christoforos Rozario, Joanna K. Wojtus, Pablo R. Murcia, Chris Boutell, Andreu Masdefiol Garriga, and Vanessa Herder
- Subjects
Innate immune system ,viruses ,Inflammation ,Biology ,Transcriptome ,Immune system ,Interferon ,Immunology ,medicine ,Respiratory epithelium ,medicine.symptom ,Tropism ,Cellular Tropism ,medicine.drug - Abstract
The pandemic spread of SARS-CoV-2, the etiological agent of COVID-19, represents a significant and ongoing international health crisis. A key symptom of SARS-CoV-2 infection is the onset of fever, with a hyperthermic temperature range of 38 to 41°C. Fever is an evolutionarily conserved host response to microbial infection and inflammation that can influence the outcome of viral pathogenicity and regulation of host innate and adaptive immune responses. However, it remains to be determined what effect elevated temperature has on SARS-CoV-2 tropism and replication. Utilizing a 3D air-liquid interface (ALI) model that closely mimics the natural tissue physiology and cellular tropism of SARS-CoV-2 infection in the respiratory airway, we identify tissue temperature to play an important role in the regulation of SARS-CoV-2 infection. We show that temperature elevation induces wide-spread transcriptome changes that impact upon the regulation of multiple pathways, including epigenetic regulation and lncRNA expression, without disruption of general cellular transcription or the induction of interferon (IFN)-mediated antiviral immune defences. Respiratory tissue incubated at temperatures >37°C remained permissive to SARS-CoV-2 infection but severely restricted the initiation of viral transcription, leading to significantly reduced levels of intraepithelial viral RNA accumulation and apical shedding of infectious virus. To our knowledge, we present the first evidence that febrile temperatures associated with COVID-19 inhibit SARS-CoV-2 replication. Our data identify an important role for temperature elevation in the epithelial restriction of SARS-CoV-2 that occurs independently of the induction of canonical IFN-mediated antiviral immune defences and interferon-stimulated gene (ISG) expression.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.