1
|
Ye W, Tang S, Wang Y, Wang W, Liu Y, Xu H, Meng H, Lu Y, Huang Z, Ge Y. SGK1 mediates herpes simplex keratitis via the PI3K/SGK1/ Wnt signaling pathways. Cell Signal 2024; 127:111566. [PMID: 39674392 DOI: 10.1016/j.cellsig.2024.111566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/26/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
Herpes simplex virus type 1 (HSV-1) is a common virus infecting the ocular tissue. It infects eye tissues, such as the eyelid, cornea, and conjunctiva. Corneal HSV-1 infection causes herpes simplex keratitis (HSK), which can induce vision loss. Current treatments for eye infections targeting HSV-1 can led to various sequelae. Antiviral drugs only work during active viral replication, and viral resistance has been recorded in numerous cases. Therefore, it is necessary to determine the molecular mechanisms underlying HSV-1 infection and identify new antiviral drugs. There are no reports on whether PI3K can regulate SGK1 to modulate HSV-1 infection in corneal epithelial cells (CECs), or how this mechanism works. This study found that HSV-1 levels and apoptosis increased in human corneal epithelial cells (HCECs) and BALB/c mice after HSV-1 infection. Serum and glucocorticoid-regulated protein kinase 1 (SGK1) were upregulated in HCECs and corneal tissues of BALB/c mice infected with HSV-1, as evidenced by whole-transcriptome sequencing, quantitative real-time polymerase chain reaction (RT-qPCR), and immunofluorescence staining experiments. An inhibitor of SGK1 (GSK 650394) reduced SGK1 expression, HSV-1 replication, and apoptosis in CECs, as evidenced by western blotting, flow cytometry, and in cell western blotting. The phosphatidylinositol 3'-kinase (PI3K) pathway was activated in CECs infected with HSV-1. After treatment with the PI3K inhibitor (LY294002), the expression of SGK1 and Wnt signaling pathway protein β-catenin were downregulated, and the replication of HSV-1 decreased in CECs; additionally, CECs apoptosis was reduced. HSV-1 replication causes CECs apoptosis. In HSV-1 infected CECs, SGK1 expression was upregulated by activated PI3K/SGK1 signaling pathway. Additionally, SGK1 activated Wnt/β-catenin signaling pathway to promote HSV-1 replication and cause CEC apoptosis. In conclusion, SGK1 is an important target for HSK treatment.
Collapse
Affiliation(s)
- Wei Ye
- Medical School, Nanjing University, Nanjing 210093, China; Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Songyi Tang
- Medical School, Nanjing University, Nanjing 210093, China; Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yue Wang
- Medical School, Nanjing University, Nanjing 210093, China; Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Wenzhe Wang
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yumeilan Liu
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Huanhuan Xu
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Hu Meng
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Yan Lu
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| | - Zhenping Huang
- Medical School, Nanjing University, Nanjing 210093, China.
| | - Yirui Ge
- Department of Ophthalmology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China.
| |
Collapse
|
2
|
Oh SI, Sheet S, Bui VN, Dao DT, Bui NA, Kim TH, Cha J, Park MR, Hur TY, Jung YH, Kim B, Lee HS, Cho A, Lim D. Transcriptome profiles of organ tissues from pigs experimentally infected with African swine fever virus in early phase of infection. Emerg Microbes Infect 2024; 13:2366406. [PMID: 38847223 PMCID: PMC11210422 DOI: 10.1080/22221751.2024.2366406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
African swine fever, caused by African swine fever virus (ASFV), is a highly contagious and fatal disease that poses a significant threat to the global pig industry. The limited information on ASFV pathogenesis and ASFV-host interactions has recently prompted numerous transcriptomic studies. However, most of these studies have focused on elucidating the transcriptome profiles of ASFV-infected porcine alveolar macrophages in vitro. Here, we analyzed dynamic transcriptional patterns in vivo in nine organ tissues (spleen, submandibular lymph node, mesenteric lymph node, inguinal lymph node, tonsils, lungs, liver, kidneys, and heart) obtained from pigs in the early stages of ASFV infection (1 and 3 d after viremia). We observed rapid spread of ASFV to the spleen after viremia, followed by broad transmission to the liver and lungs and subsequently, the submandibular and inguinal lymph nodes. Profound variations in gene expression patterns were observed across all organs and at all time-points, providing an understanding of the distinct defence strategies employed by each organ against ASFV infection. All ASFV-infected organs exhibited a collaborative response, activating immune-associated genes such as S100A8, thereby triggering a pro-inflammatory cytokine storm and interferon activation. Functional analysis suggested that ASFV exploits the PI3K-Akt signalling pathway to evade the host immune system. Overall, our findings provide leads into the mechanisms underlying pathogenesis and host immune responses in different organs during the early stages of infection, which can guide further explorations, aid the development of efficacious antiviral strategies against ASFV, and identify valuable candidate gene targets for vaccine development.
Collapse
Affiliation(s)
- Sang-Ik Oh
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
- Laboratory of Veterinary Pathology and Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - Sunirmal Sheet
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Vuong Nghia Bui
- Virology Department, National Institute of Veterinary Research, Hanoi, Vietnam
| | - Duy Tung Dao
- Virology Department, National Institute of Veterinary Research, Hanoi, Vietnam
| | - Ngoc Anh Bui
- Virology Department, National Institute of Veterinary Research, Hanoi, Vietnam
| | - Tae-Hun Kim
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
- TNT Research. Co., Ltd., R&D center, Sejong-si, Republic of Korea
| | - Jihye Cha
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Mi-Rim Park
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Tai-Young Hur
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Young-Hun Jung
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Bumseok Kim
- Laboratory of Veterinary Pathology and Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea
| | - Hu Suk Lee
- International Livestock Research Institute, Hanoi, Vietnam
- College of Veterinary Medicine, Chungnam National University, Daejoen, Republic of Korea
| | - Ara Cho
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
| | - Dajeong Lim
- National Institute of Animal Science, Rural Development Administration, Wanju, Republic of Korea
- Department of Animal Resources Science, College of Agriculture and Life Sciences, Chungnam National University, Daejoen, Republic of Korea
| |
Collapse
|
3
|
da S Santos FR, Valadão DF, Bambirra JL, Moreira TP, de Souza CDF, Passos IBS, Queiroz-Junior CM, Fagundes CT, Teixeira MM, Costa VV, Souza DG. Targeting PI3Kγ Pathway for Treating Dengue virus Infection. Microb Pathog 2024; 197:107060. [PMID: 39490942 DOI: 10.1016/j.micpath.2024.107060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/02/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Dengue disease is a major problem worldwide, impacting millions of people annually with no specific approved treatments. The pathogenesis of dengue is a complex interplay of viral and host factors, driven in particular by an excessive inflammatory response triggered by the infection. While it has been observed that various viruses can modulate the PI3K/Akt signaling pathway to aid replication and theunderlying mechanisms remainunclear. The study aims to explore the impact of PI3Kγ inhibition during Dengue virus (DENV) infection in vivo. Experiments were performed using both wild-type (WT) and PI3Kγ knockout mice inoculated with DENV. Parameters, including survival rates, hematologic, virologic, histopathologic, and inflammatory analyzes, were evaluated. Additionally, the therapeutic potential of a selective PI3Kγ inhibitor (AS605240) was investigated in DENV-infected A129 mice. PI3Kγ deficiency resulted in lower lethality and provided protection against DENV-induced thrombocytopenia, decreased hemoconcentration, vascular permeability, and liver damage compared to DENV-infected WT littermates. In addition, PI3Kγ deficiency correlated with reduced viral replication in the blood, spleen and liver alongside decreased production of inflammatory mediators in plasma and spleen. Pharmacologic inhibition of PI3Kγ not only ameliorated DENV-induced thrombocytopenia and liver injury, but also reduced DENV replication in target organs. Treatment with AS605240 reduced the concentration of IL-6 in the spleen and plasma.This study sheds light on the significant pro-viral effects of the PI3Kγ signaling pathway during DENV infection and its central role in pathogenesis by curbing excessive DENV-induced inflammation. Inhibition of PI3Kγ shows promising host-directed target for developing novel Dengue disease therapies, offering substantial benefits to hosts.
Collapse
Affiliation(s)
- Felipe R da S Santos
- Departament of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Departament of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Deborah F Valadão
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Jordana L Bambirra
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thaiane P Moreira
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Carla D F de Souza
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ingredy B S Passos
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Celso M Queiroz-Junior
- Departament of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caio T Fagundes
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro M Teixeira
- Departament of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vivian V Costa
- Departament of Morphology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Daniele G Souza
- Departament of Microbiology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
4
|
Amiri F, Ziaei Chamgordani S, Ghourchian H. An in vitro study for reducing the cytotoxicity and dose dumping risk of remdesivir via entrapment in nanostructured lipid carriers. Sci Rep 2024; 14:19360. [PMID: 39169059 PMCID: PMC11339451 DOI: 10.1038/s41598-024-70003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 08/12/2024] [Indexed: 08/23/2024] Open
Abstract
The aim of this study was to synthesize and evaluate nanostructured lipid carriers (NLCs) loaded with Remdesivir (RDV) to control its side effects in COVID-19 patients. Due to the low solubility and short half-life of RDV in the blood, an injectable formulation was prepared using sulphobutylether-beta-cyclodextrin. However, it can accumulate in the kidney and cause renal impairment. NLCs improve the parenteral delivery of hydrophobic drugs such as RDV by increasing drug solubility and bioavailability. For the synthesis of RDV-NLCs, the aqueous phase containing Tween 80 was injected into the lipid phase under rapid stirring and was sonicated. The experimental conditions were optimized using Box-Behnken design and Design Expert software. The optimum formulation contained a total lipid of 2.13%, a total surfactant of 1%, and a hot bath time of 71 min. The optimum formulation showed particle size, polydispersity index, zeta potential, and entrapment efficiency values of 151.0 ± 1.7 nm (from 149.1 to 152.1), 0.4 ± 0.1 (from 0.3 to 0.5), -43.8 ± 1.2 mV (from -42.4 to -44.7), and 81.34 ± 1.57% (from 79.52 to 82.33%), respectively. RDV-NLCs showed acceptable stability for 30 days at 25 ℃ and were compatible with commonly used intravenous infusion fluids for 48 h. FE-SEM images of RDV-NLC showed spherical particles with a mean diameter of 207 nm. The NLC-RDV formulation showed a sustained release of RDV with a low risk of dose-dumping, minimizing potential side effects. In addition, RDV in the form of RDV-NLC causes less cytotoxicity to healthy normal kidney cells, which is expected to reduce renal impairment in COVID-19 patients.
Collapse
Affiliation(s)
- Fatemeh Amiri
- Laboratory of Bioanalysis, Institute of Biochemistry & Biophysics, University of Tehran, Enghelab Ave, P.O. Box: 13145-1384, Tehran, 1417614411, Iran
| | - Sepideh Ziaei Chamgordani
- Laboratory of Bioanalysis, Institute of Biochemistry & Biophysics, University of Tehran, Enghelab Ave, P.O. Box: 13145-1384, Tehran, 1417614411, Iran
| | - Hedayatollah Ghourchian
- Laboratory of Bioanalysis, Institute of Biochemistry & Biophysics, University of Tehran, Enghelab Ave, P.O. Box: 13145-1384, Tehran, 1417614411, Iran.
| |
Collapse
|
5
|
Kataria R, Duhan N, Kaundal R. Navigating the human-monkeypox virus interactome: HuPoxNET atlas reveals functional insights. Front Microbiol 2024; 15:1399555. [PMID: 39155985 PMCID: PMC11327128 DOI: 10.3389/fmicb.2024.1399555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
Monkeypox virus, a close relative of variola virus, has significantly increased the incidence of monkeypox disease in humans, with several clinical symptoms. The sporadic spread of the disease outbreaks has resulted in the need for a comprehensive understanding of the molecular mechanisms underlying disease infection and potential therapeutic targets. Protein-protein interactions play a crucial role in various cellular processes and regulate different immune signals during virus infection. Computational algorithms have gained high significance in the prediction of potential protein interaction pairs. Here, we developed a comprehensive database called HuPoxNET (https://kaabil.net/hupoxnet/) using the state-of-the-art MERN stack technology. The database leverages two sequence-based computational models to predict strain-specific protein-protein interactions between human and monkeypox virus proteins. Furthermore, various protein annotations of the human and viral proteins such as gene ontology, KEGG pathways, subcellular localization, protein domains, and novel drug targets identified from our study are also available on the database. HuPoxNET is a user-friendly platform for the scientific community to gain more insights into the monkeypox disease infection and aid in the development of therapeutic drugs against the disease.
Collapse
Affiliation(s)
- Raghav Kataria
- Department of Plants, Soils, and Climate, College of Agriculture and Applied Sciences, Logan, UT, United States
- Bioinformatics Facility, Center for Integrated BioSystems, Logan, UT, United States
| | - Naveen Duhan
- Department of Plants, Soils, and Climate, College of Agriculture and Applied Sciences, Logan, UT, United States
- Bioinformatics Facility, Center for Integrated BioSystems, Logan, UT, United States
| | - Rakesh Kaundal
- Department of Plants, Soils, and Climate, College of Agriculture and Applied Sciences, Logan, UT, United States
- Bioinformatics Facility, Center for Integrated BioSystems, Logan, UT, United States
- Department of Computer Science, College of Science, Utah State University, Logan, UT, United States
| |
Collapse
|
6
|
Chauhan M, Martinak PE, Hollenberg BM, Goodman AG. Drosophila melanogaster Toll-9 elicits antiviral immunity against Drosophila C virus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.19.599730. [PMID: 38948804 PMCID: PMC11212974 DOI: 10.1101/2024.06.19.599730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The Toll pathway plays a pivotal role in innate immune responses against pathogens. The evolutionary conserved pathogen recognition receptors (PRRs), including Toll like receptors (TLRs), play a crucial role in recognition of pathogen associated molecular patterns (PAMPs). The Drosophila genome encodes nine Toll receptors that are orthologous to mammalian TLRs. While mammalian TLRs directly recognize PAMPs, most Drosophila Tolls recognize the proteolytically cleaved ligand Spätzle to activate downstream signaling cascades. In this study, we demonstrated that Toll-9 is crucial for antiviral immunity against Drosophila C virus (DCV), a natural pathogen of Drosophila . A transposable element insertion in the Toll-9 gene renders the flies more susceptible to DCV. The stable expression of Toll-9 in S2 cells confers resistance against DCV infection by upregulation of the RNAi pathway. Toll-9 promotes the dephosphorylation of AKT, resulting in the induction of antiviral RNAi genes to inhibit DCV replication. Toll-9 localizes to the endosome where it binds dsRNA, suggesting its role to detect viral dsRNA. Toll-9 also induces apoptosis during DCV infection, contributing to its antiviral role. Together, this work identifies the role of Toll-9 in antiviral immunity against DCV infection through its ability to bind dsRNA and induce AKT-mediated RNAi antiviral immunity. IMPORTANCE Insects rely on innate immunity and RNA interference (RNAi) to combat viral infections. Our study underscores the pivotal role of Drosophila Toll-9 in antiviral immunity, aligning with findings in Bombyx mori , where Toll-9 activation upregulates the RNAi component Dicer2 . We demonstrate that Drosophila Toll-9 functions as a pattern recognition receptor (PRR) for double-stranded RNA (dsRNA) during Drosophila C virus (DCV) infection, akin to mammalian TLRs. Toll-9 activation leads to the upregulation of key RNAi components, Dicer2 and Argonaute2 , and dephosphorylation of AKT triggers apoptosis via induction of proapoptotic genes Hid and Reaper . This study also reveals that Toll-9 localizes in endosomal compartments where it interacts with dsRNA. These insights enhance our understanding of Drosophila innate immune mechanisms, reflecting the evolutionary conservation of immune responses across diverse species and providing impetus for further research into the conserved roles of TLRs across the animal kingdom.
Collapse
|
7
|
Tai LT, Yeh CY, Chang YJ, Liu JF, Hsu KC, Cheng JC, Lu CH. Discovery of Novel Spike Inhibitors against SARS-CoV-2 Infection. Int J Mol Sci 2024; 25:6105. [PMID: 38892294 PMCID: PMC11172837 DOI: 10.3390/ijms25116105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is responsible for the current coronavirus disease pandemic. With the rapid evolution of variant strains, finding effective spike protein inhibitors is a logical and critical priority. Angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor for SARS-CoV-2 viral entry, and thus related therapeutic approaches associated with the spike protein-ACE2 interaction show a high degree of feasibility for inhibiting viral infection. Our computer-aided drug design (CADD) method meticulously analyzed more than 260,000 compound records from the United States National Cancer Institute (NCI) database, to identify potential spike inhibitors. The spike protein receptor-binding domain (RBD) was chosen as the target protein for our virtual screening process. In cell-based validation, SARS-CoV-2 pseudovirus carrying a reporter gene was utilized to screen for effective compounds. Ultimately, compounds C2, C8, and C10 demonstrated significant antiviral activity against SARS-CoV-2, with estimated EC50 values of 8.8 μM, 6.7 μM, and 7.6 μM, respectively. Using the above compounds as templates, ten derivatives were generated and robust bioassay results revealed that C8.2 (EC50 = 5.9 μM) exhibited the strongest antiviral efficacy. Compounds C8.2 also displayed inhibitory activity against the Omicron variant, with an EC50 of 9.3 μM. Thus, the CADD method successfully discovered lead compounds binding to the spike protein RBD that are capable of inhibiting viral infection.
Collapse
Affiliation(s)
- Li-Te Tai
- Industrial Development Graduate Program of College of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300193, Taiwan
| | - Cheng-Yun Yeh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
| | - Yu-Jen Chang
- The Ph.D. Program of Biotechnology and Biomedical Industry, China Medical University, Taichung 404333, Taiwan
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei 110301, Taiwan
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404333, Taiwan
| | - Chih-Hao Lu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu 300193, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu 300193, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300193, Taiwan
| |
Collapse
|
8
|
Jung J, Bae J, Park JS, Lee SW, Jeong JH, Park SJ. In Vitro Anti-Rotaviral Activity of Bavachin Isolated from Psoralea corylifolia L. ( Fabaceae). Vet Sci 2024; 11:188. [PMID: 38787160 PMCID: PMC11126063 DOI: 10.3390/vetsci11050188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/12/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Rotavirus is the main causative agent of viral gastroenteritis among young animals worldwide. Currently, no clinically approved or effective antiviral drugs are available to combat rotavirus infections. Herein, we evaluated the anti-rotaviral activities of extracts and bavachin isolated from Psoralea corylifolia L. (Fabaceae) (P. corylifolia) against the bovine rotavirus G8P[7] and porcine rotavirus G5P[7] in vitro. Two assay strategies were performed: (1) a virucidal assay to reduce viral infectivity by virus neutralization and (2) a post-treatment assay to assess viral replication suppression. The results from the virucidal assay showed that the extracts and bavachin did not exert anti-rotaviral activities. In the follow-up analysis after treatment, bavachin exhibited robust antiviral efficacy, with 50% effective concentration (EC50) values of 10.6 μM (selectivity index [SI] = 2.38) against bovine rotavirus G8P[7] and 13.0 μM (SI = 1.94) against porcine rotavirus G5P[7]. Bavachin strongly suppressed viral RNA synthesis in the early (6 h) and late stages (18 h) after rotaviral infection. These findings strongly suggest that bavachin may have hindered the virions by effectively inhibiting the early stages of the virus replication cycle after rotaviral infection. Furthermore, confocal imaging showed that bavachin suppressed viral protein synthesis, notably that of the rotaviral protein (VP6). These results suggest that bavachin has strong antiviral activity against rotaviruses, inhibits viral replication, and is a candidate natural therapeutic drug targeting rotaviral infection. The utilization of bavachin isolated from P. corylifolia may contribute to decreased mortality rates, lower medication expenses, and enhanced economic viability in domestic farms.
Collapse
Affiliation(s)
- Jinseok Jung
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea; (J.J.); (J.B.); (J.S.P.); (S.W.L.)
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Jaehoon Bae
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea; (J.J.); (J.B.); (J.S.P.); (S.W.L.)
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Ji Sun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea; (J.J.); (J.B.); (J.S.P.); (S.W.L.)
| | - Seung Woong Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea; (J.J.); (J.B.); (J.S.P.); (S.W.L.)
| | - Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea; (J.J.); (J.B.); (J.S.P.); (S.W.L.)
| |
Collapse
|
9
|
Maroui MA, Odongo GA, Mundo L, Manara F, Mure F, Fusil F, Jay A, Gheit T, Michailidis TM, Ferrara D, Leoncini L, Murray P, Manet E, Ohlmann T, De Boevre M, De Saeger S, Cosset FL, Lazzi S, Accardi R, Herceg Z, Gruffat H, Khoueiry R. Aflatoxin B1 and Epstein-Barr virus-induced CCL22 expression stimulates B cell infection. Proc Natl Acad Sci U S A 2024; 121:e2314426121. [PMID: 38574017 PMCID: PMC11032484 DOI: 10.1073/pnas.2314426121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 02/20/2024] [Indexed: 04/06/2024] Open
Abstract
Epstein-Barr Virus (EBV) infects more than 90% of the adult population worldwide. EBV infection is associated with Burkitt lymphoma (BL) though alone is not sufficient to induce carcinogenesis implying the involvement of co-factors. BL is endemic in African regions faced with mycotoxins exposure. Exposure to mycotoxins and oncogenic viruses has been shown to increase cancer risks partly through the deregulation of the immune response. A recent transcriptome profiling of B cells exposed to aflatoxin B1 (AFB1) revealed an upregulation of the Chemokine ligand 22 (CCL22) expression although the underlying mechanisms were not investigated. Here, we tested whether mycotoxins and EBV exposure may together contribute to endemic BL (eBL) carcinogenesis via immunomodulatory mechanisms involving CCL22. Our results revealed that B cells exposure to AFB1 and EBV synergistically stimulated CCL22 secretion via the activation of Nuclear Factor-kappa B pathway. By expressing EBV latent genes in B cells, we revealed that elevated levels of CCL22 result not only from the expression of the latent membrane protein LMP1 as previously reported but also from the expression of other viral latent genes. Importantly, CCL22 overexpression resulting from AFB1-exposure in vitro increased EBV infection through the activation of phosphoinositide-3-kinase pathway. Moreover, inhibiting CCL22 in vitro and in humanized mice in vivo limited EBV infection and decreased viral genes expression, supporting the notion that CCL22 overexpression plays an important role in B cell infection. These findings unravel new mechanisms that may underpin eBL development and identify novel pathways that can be targeted in drug development.
Collapse
Affiliation(s)
- Mohamed Ali Maroui
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Grace Akinyi Odongo
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Lucia Mundo
- Limerick Digital Cancer Research Centre, Health Research Institute, Bernal Institute and School of Medicine, University of Limerick, LimerickV94 T9PX, Ireland
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Francesca Manara
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Fabrice Mure
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Floriane Fusil
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Antonin Jay
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Tarik Gheit
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Thanos M. Michailidis
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
| | - Domenico Ferrara
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Lorenzo Leoncini
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Paul Murray
- Limerick Digital Cancer Research Centre, Health Research Institute, Bernal Institute and School of Medicine, University of Limerick, LimerickV94 T9PX, Ireland
| | - Evelyne Manet
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Théophile Ohlmann
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Marthe De Boevre
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
| | - Sarah De Saeger
- Centre of Excellence in Mycotoxicology and Public Health, Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent9000, Belgium
- Department of Biotechnology and Food Technology, Faculty of Science, University of Johannesburg, Gauteng2028, South Africa
| | - François-Loïc Cosset
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Stefano Lazzi
- Department of Medical Biotechnology, Section of Pathology, University of Siena, Siena53100, Italy
| | - Rosita Accardi
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Zdenko Herceg
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| | - Henri Gruffat
- Centre International de Recherche en Infectiologie, University Claude Bernard Lyon I, INSERM U1111, CNRS UMR5308, Ecole Normale Supérieure, Lyon69366 Cedex 07, France
| | - Rita Khoueiry
- Epigenomics and Mechanisms Branch, International Agency for Research on Cancer, World Health Organization, Lyon69366 Cedex 07, France
| |
Collapse
|
10
|
Nhau PT, Gamede M, Sibiya N. COVID-19-Induced Diabetes Mellitus: Comprehensive Cellular and Molecular Mechanistic Insights. PATHOPHYSIOLOGY 2024; 31:197-209. [PMID: 38651404 PMCID: PMC11036300 DOI: 10.3390/pathophysiology31020016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/06/2024] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
Despite evidence demonstrating the risks of developing diabetes mellitus because of SARS-CoV-2, there is, however, insufficient scientific data available to elucidate the relationship between diabetes mellitus and COVID-19. Research indicates that SARS-CoV-2 infection is associated with persistent damage to organ systems due to the systemic inflammatory response. Since COVID-19 is known to induce these conditions, further investigation is necessary to fully understand its long-term effects on human health. Consequently, it is essential to consider the effect of the COVID-19 pandemic when predicting the prevalence of diabetes mellitus in the future, especially since the incidence of diabetes mellitus was already on the rise before the pandemic. Additional research is required to fully comprehend the impact of SARS-CoV-2 infection on glucose tolerance and insulin sensitivity. Therefore, this article delves deeper into the current literature and links the perceived relationship between SARS-CoV-2 and diabetes. In addition, the article highlights the necessity for further research to fully grasp the mechanisms that SARS-CoV-2 utilises to induce new-onset diabetes. Where understanding and consensus are reached, therapeutic interventions to prevent the onset of diabetes could be proposed. Lastly, we propose advocating for the regular screening of diabetes and pre-diabetes, particularly for the high-risk population with a history of COVID-19 infection.
Collapse
Affiliation(s)
- Praise Tatenda Nhau
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa;
| | - Mlindeli Gamede
- Human Physiology Department, University of Pretoria, Pretoria 0028, South Africa;
| | - Ntethelelo Sibiya
- Pharmacology Division, Faculty of Pharmacy, Rhodes University, Makhanda 6139, South Africa;
| |
Collapse
|
11
|
He H, Li Y, Chen Y, Chen J, Li Z, Li L, Shi D, Zhang X, Shi H, Xue M, Feng L. NLRP1 restricts porcine deltacoronavirus infection via IL-11 inhibiting the phosphorylation of the ERK signaling pathway. J Virol 2024; 98:e0198223. [PMID: 38411106 PMCID: PMC10949457 DOI: 10.1128/jvi.01982-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/08/2024] [Indexed: 02/28/2024] Open
Abstract
Continuously emerging highly pathogenic coronaviruses remain a major threat to human and animal health. Porcine deltacoronavirus (PDCoV) is a newly emerging enterotropic swine coronavirus that causes large-scale outbreaks of severe diarrhea disease in piglets. Unlike other porcine coronaviruses, PDCoV has a wide range of species tissue tropism, including primary human cells, which poses a significant risk of cross-species transmission. Nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 1 (NLRP1) has a key role in linking host innate immunity to microbes and the regulation of inflammatory pathways. We now report a role for NLRP1 in the control of PDCoV infection. Overexpression of NLRP1 remarkably suppressed PDCoV infection, whereas knockout of NLRP1 led to a significant increase in PDCoV replication. A mechanistic study revealed that NLRP1 suppressed PDCoV replication in cells by upregulating IL-11 expression, which in turn inhibited the phosphorylation of the ERK signaling pathway. Furthermore, the ERK phosphorylation inhibitor U0126 effectively hindered PDCoV replication in pigs. Together, our results demonstrated that NLRP1 exerted an anti-PDCoV effect by IL-11-mediated inhibition of the phosphorylation of the ERK signaling pathway, providing a novel antiviral signal axis of NLRP1-IL-11-ERK. This study expands our understanding of the regulatory network of NLRP1 in the host defense against virus infection and provides a new insight into the treatment of coronaviruses and the development of corresponding drugs.IMPORTANCECoronavirus, which mainly infects gastrointestinal and respiratory epithelial cells in vivo, poses a huge threat to both humans and animals. Although porcine deltacoronavirus (PDCoV) is known to primarily cause fatal diarrhea in piglets, reports detected in plasma samples from Haitian children emphasize the potential risk of animal-to-human spillover. Finding effective therapeutics against coronaviruses is crucial for controlling viral infection. Nucleotide-binding oligomerization-like receptor (NLR) family pyrin domain-containing 1 (NLRP1), a key regulatory factor in the innate immune system, is highly expressed in epithelial cells and associated with the pathogenesis of viruses. We demonstrate here that NLRP1 inhibits the infection of the intestinal coronavirus PDCoV through IL-11-mediated phosphorylation inhibition of the ERK signaling pathway. Furthermore, the ERK phosphorylation inhibitor can control the infection of PDCoV in pigs. Our study emphasizes the importance of NLRP1 as an immune regulatory factor and may open up new avenues for the treatment of coronavirus infection.
Collapse
Affiliation(s)
- Haojie He
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Yongfeng Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Yunyan Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Jianfei Chen
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Zhongyuan Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Liang Li
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Da Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xin Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Hongyan Shi
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Mei Xue
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Li Feng
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| |
Collapse
|
12
|
Khaparkhuntikar K, Maji I, Gupta SK, Mahajan S, Aalhate M, Sriram A, Gupta U, Guru SK, Kulkarni P, Singh PK. Acalabrutinib as a novel hope for the treatment of breast and lung cancer: an in-silico proof of concept. J Biomol Struct Dyn 2024; 42:1469-1484. [PMID: 37272883 DOI: 10.1080/07391102.2023.2217923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/01/2023] [Indexed: 06/06/2023]
Abstract
Drug repurposing is proved to be a groundbreaking concept in the field of cancer research, accelerating the pace of de novo drug discovery by investigating the anti-cancer activity of the already approved drugs. On the other hand, it got highly benefitted from the advancement in the in-silico tools and techniques, which are used to build up the initial "proof of concept" based on the drug-target interaction. Acalabrutinib (ACL) is a well-known drug for the treatment of hematological malignancies. But, the therapeutic ability of ACL against solid tumors is still unexplored. Thereby, the activity of ACL on breast cancer and lung cancer was evaluated utilizing different computational methods. A series of proteins such as VEGFR1, ALK, BCL2, CXCR-4, mTOR, AKT, PI3K, HER-2, and Estrogen receptors were selected based on their involvement in the progression of the breast as well as lung cancer. A multi-level computational study starting from protein-ligand docking to molecular dynamic (MD) simulations were performed to detect the binding potential of ACL towards the selected proteins. Results of the study led to the identification of ACL as a ligand that showed a high docking score and binding energy with HER-2, mTOR, and VEGFR-1 successively. Whereas, the MD simulations study has also shown good docked complex stability of ACL with HER2 and VEGFR1. Our findings suggest that interaction with those receptors can lead to preventive action on both breast and lung cancer, thus it can be concluded that ACL could be a potential molecule for the same purpose.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Kedar Khaparkhuntikar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Indrani Maji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sunil Kumar Gupta
- Department of Bioinformatics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Anitha Sriram
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ujala Gupta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Prachi Kulkarni
- Department of Physiology, Shri B. M. Patil Medical College, Hospital & Research Centre BLDE (Deemed to be University), Vijayapura, Karnataka, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
13
|
Kang H, Kang T, Jackson L, Murphy A, Nitta T. Evidence for Involvement of ADP-Ribosylation Factor 6 in Intracellular Trafficking and Release of Murine Leukemia Virus Gag. Cells 2024; 13:270. [PMID: 38334661 PMCID: PMC10854678 DOI: 10.3390/cells13030270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024] Open
Abstract
Murine leukemia viruses (MuLVs) are simple retroviruses that cause several diseases in mice. Retroviruses encode three basic genes: gag, pol, and env. Gag is translated as a polyprotein and moves to assembly sites where viral particles are shaped by cleavage of poly-Gag. Viral release depends on the intracellular trafficking of viral proteins, which is determined by both viral and cellular factors. ADP-ribosylation factor 6 (Arf6) is a small GTPase that regulates vesicular trafficking and recycling of different types of cargo in cells. Arf6 also activates phospholipase D (PLD) and phosphatidylinositol-4-phosphate 5-kinase (PIP5K) and produces phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2). We investigated how Arf6 affected MuLV release with a constitutively active form of Arf6, Arf6Q67L. Expression of Arf6Q67L impaired Gag release by accumulating Gag at PI(4,5)P2-enriched compartments in the cytoplasm. Treatment of the inhibitors for PLD and PIP5K impaired or recovered MuLV Gag release in the cells expressing GFP (control) and Arf6Q67L, implying that regulation of PI(4,5)P2 through PLD and PIP5K affected MuLV release. Interference with the phosphoinositide 3-kinases, mammalian target of rapamycin (mTOR) pathway, and vacuolar-type ATPase activities showed further impairment of Gag release from the cells expressing Arf6Q67L. In contrast, mTOR inhibition increased Gag release in the control cells. The proteasome inhibitors reduced viral release in the cells regardless of Arf6Q67L expression. These data outline the differences in MuLV release under the controlled and overactivated Arf6 conditions and provide new insight into pathways for MuLV release.
Collapse
Affiliation(s)
- Hyokyun Kang
- Department of Biology, Savannah State University, Savannah, GA 31404, USA; (H.K.); (T.K.); (L.J.); (A.M.)
| | - Taekwon Kang
- Department of Biology, Savannah State University, Savannah, GA 31404, USA; (H.K.); (T.K.); (L.J.); (A.M.)
| | - Lauryn Jackson
- Department of Biology, Savannah State University, Savannah, GA 31404, USA; (H.K.); (T.K.); (L.J.); (A.M.)
| | - Amaiya Murphy
- Department of Biology, Savannah State University, Savannah, GA 31404, USA; (H.K.); (T.K.); (L.J.); (A.M.)
| | - Takayuki Nitta
- Department of Biology, Savannah State University, Savannah, GA 31404, USA; (H.K.); (T.K.); (L.J.); (A.M.)
- Department of Molecular Biology and Biochemistry, Cancer Research Institute, University of California, Irvine, CA 92697, USA
| |
Collapse
|
14
|
Chen H, Zhao P, Zhang C, Ming X, Zhang C, Jung YS, Qian Y. Veratramine inhibits porcine epidemic diarrhea virus entry through macropinocytosis by suppressing PI3K/Akt pathway. Virus Res 2024; 339:199260. [PMID: 37923169 PMCID: PMC10661853 DOI: 10.1016/j.virusres.2023.199260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/07/2023]
Abstract
Porcine epidemic diarrhea (PED) is a contagious intestinal disease caused by α-coronavirus porcine epidemic diarrhea virus (PEDV). At present, no effective vaccine is available to prevent the disease. Therefore, research for novel antivirals is important. This study aimed to identify the antiviral mechanism of Veratramine (VAM), which actively inhibits PEDV replication with a 50 % inhibitory concentration (IC50) of ∼5 µM. Upon VAM treatment, both PEDV-nucleocapsid (N) protein level and virus titer decreased significantly. The time-of-addition assay results showed that VAM could inhibit PEDV replication by blocking viral entry. Importantly, VAM could inhibit PEDV-induced phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) activity and further suppress micropinocytosis, which is required for PEDV entry. In addition, PI3K inhibitor LY294002 showed anti-PEDV activity by blocking viral entry as well. Taken together, VAM possessed anti-PEDV properties against the entry stage of PEDV by inhibiting the macropinocytosis pathway by suppressing the PI3K/Akt pathway. VAM could be considered as a lead compound for the development of anti-PEDV drugs and may be used during the viral entry stage of PEDV infection.
Collapse
Affiliation(s)
- Huan Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Pu Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Caisheng Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xin Ming
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chaofeng Zhang
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicine, China Pharmaceutical University, Nanjing, China
| | - Yong-Sam Jung
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| | - Yingjuan Qian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, China; One Health Laboratory, Jiangsu Province Foreign Expert Workstation, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China; Jiangsu Agri-Animal Husbandry Vocational College, Veterinary Bio-Pharmaceutical, Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Taizhou, Jiangsu, China.
| |
Collapse
|
15
|
Duggan NN, Dragic T, Chanda SK, Pache L. Breaking the Silence: Regulation of HIV Transcription and Latency on the Road to a Cure. Viruses 2023; 15:2435. [PMID: 38140676 PMCID: PMC10747579 DOI: 10.3390/v15122435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Antiretroviral therapy (ART) has brought the HIV/AIDS epidemic under control, but a curative strategy for viral eradication is still needed. The cessation of ART results in rapid viral rebound from latently infected CD4+ T cells, showing that control of viral replication alone does not fully restore immune function, nor does it eradicate viral reservoirs. With a better understanding of factors and mechanisms that promote viral latency, current approaches are primarily focused on the permanent silencing of latently infected cells ("block and lock") or reactivating HIV-1 gene expression in latently infected cells, in combination with immune restoration strategies to eliminate HIV infected cells from the host ("shock and kill"). In this review, we provide a summary of the current, most promising approaches for HIV-1 cure strategies, including an analysis of both latency-promoting agents (LPA) and latency-reversing agents (LRA) that have shown promise in vitro, ex vivo, and in human clinical trials to reduce the HIV-1 reservoir.
Collapse
Affiliation(s)
- Natasha N. Duggan
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Tatjana Dragic
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Sumit K. Chanda
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, USA
| | - Lars Pache
- NCI Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
16
|
Wrobel ER, Jackson J, Abraham M, He B. Regulation of host gene expression by J paramyxovirus. PLoS One 2023; 18:e0294173. [PMID: 37963152 PMCID: PMC10645344 DOI: 10.1371/journal.pone.0294173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
Paramyxoviruses are negative-sense, single-stranded RNA viruses that are associated with numerous diseases in humans and animals. J paramyxovirus (JPV) was first isolated from moribund mice (Mus musculus) with hemorrhagic lung lesions in Australia in 1972. In 2016, JPV was classified into the newly established genus Jeilongvirus. Novel jeilongviruses are being discovered worldwide in wildlife populations. However, the effects of jeilongvirus infection on host gene expression remains uncharacterized. To address this, cellular RNA from JPV-infected mouse fibroblasts was collected at 2, 4, 8, 12, 16, 24, and 48 hours post-infection (hpi) and were sequenced using single-end 75 base pairs (SE75) sequencing chemistry on an Illumina NextSeq platform. Differentially expressed genes (DEGs) between the virus-infected replicates and mock replicates at each timepoint were identified using the Tophat2-Cufflinks-Cuffdiff protocol. At 2 hpi, 11 DEGs were identified in JPV-infected cells, while 1,837 DEGs were detected at 48 hpi. A GO analysis determined that the genes at the earlier timepoints were involved in interferon responses, while there was a shift towards genes that are involved in antigen processing and presentation processes at the later timepoints. At 48 hpi, a KEGG analysis revealed that many of the DEGs detected were involved in pathways that are important for immune responses. qRT-PCR verified that Rtp4, Ifit3, Mx2, and Stat2 were all upregulated during JPV infection, while G0s2 was downregulated. After JPV infection, the expression of inflammatory and antiviral factors in mouse fibroblasts changes significantly. This study provides crucial insight into the different arms of host immunity that mediate Jeilongvirus infection. Understanding the pathogenic mechanisms of Jeilongvirus will lead to better strategies for the prevention and control of potential diseases that may arise from this group of viruses.
Collapse
Affiliation(s)
- Elizabeth R. Wrobel
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Jared Jackson
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Mathew Abraham
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Biao He
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
17
|
Su M, Ren X, Du D, He H, Zhang D, Xie R, Deng X, Zou C, Zou H. Curcumol β-cyclodextrin inclusion complex enhances radiosensitivity of esophageal cancer under hypoxic and normoxic condition. Jpn J Radiol 2023; 41:1275-1289. [PMID: 37227584 PMCID: PMC10613597 DOI: 10.1007/s11604-023-01446-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023]
Abstract
PURPOSE Radiotherapy is an indispensable treatment for esophageal cancer (EC), but radioresistance is not uncommon. Curcumol, as an active extract from traditional Chinese medicines, has been reported to have antitumor activity in various types of human tumor cells. However, its reversal of radioresistance has been rarely reported. MATERIALS AND METHODS In the present study, curcumol was prepared as an inclusion complex with β-cyclodextrin. EC cell lines were treated with radiation and curcumol β-cyclodextrin inclusion complex (CβC), and the effect of radiosensitization of CβC was investigated in vitro and in vivo. The in vitro experiments included cell proliferation assay, clonogenic survival assay, apoptosis assay, cell cycle assay, and western blot assay. RESULTS The in vitro data revealed that CβC and irradiation synergistically inhibited the proliferation, reduced the colony formation, promoted the apoptosis, increased the G2/M phase, inhibited DNA damage repair, and reversed the hypoxia-mediated radioresistance of EC cells to a greater extent than did CβC alone or irradiation alone. The sensitization enhancement ratios (SERs) were 1.39 for TE-1 and 1.48 for ECA109 under hypoxia. The SERs were 1.25 for TE-1 and 1.32 for ECA109 under normoxia. The in vivo data demonstrated that the combination of CβC and irradiation could inhibit tumor growth to the greatest extent compared with either monotherapy alone. The enhancement factor was 2.45. CONCLUSION This study demonstrated that CβC could enhance radiosensitivity of EC cells under hypoxic and normoxic condition. Thus, CβC can be used as an effective radiosensitizer for EC.
Collapse
Affiliation(s)
- Meng Su
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Xiaolin Ren
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Dexi Du
- Department of Radiation Oncology, Lishui Central Hospital, Lishui, Zhejiang, People's Republic of China
| | - Huijuan He
- Department of Radiation Oncology, Quzhou People's Hospital, Quzhou, Zhejiang, People's Republic of China
| | - Dahai Zhang
- Department of Radiation Oncology, Dongyang People's Hospital, Jinhua, Zhejiang, People's Republic of China
| | - Raoying Xie
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Xia Deng
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Changlin Zou
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Wenzhou, 325000, Zhejiang, People's Republic of China.
| | - Haizhou Zou
- Derpartment of Medical Oncology, Wenzhou Hospital of Chinese Medicine, No. 9 Jiaowei Street, Wenzhou, 325000, Zhejiang, People's Republic of China.
| |
Collapse
|
18
|
Kang S, Vu TH, Heo J, Kim C, Lillehoj HS, Hong YH. Analysis of miRNA expression in the trachea of Ri chicken infected with the highly pathogenic avian influenza H5N1 virus. J Vet Sci 2023; 24:e73. [PMID: 38031652 PMCID: PMC10556288 DOI: 10.4142/jvs.23141] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/13/2023] [Accepted: 08/17/2023] [Indexed: 12/01/2023] Open
Abstract
BACKGROUND Highly pathogenic avian influenza virus (HPAIV) is considered a global threat to both human health and the poultry industry. MicroRNAs (miRNA) can modulate the immune system by affecting gene expression patterns in HPAIV-infected chickens. OBJECTIVES To gain further insights into the role of miRNAs in immune responses against H5N1 infection, as well as the development of strategies for breeding disease-resistant chickens, we characterized miRNA expression patterns in tracheal tissues from H5N1-infected Ri chickens. METHODS miRNAs expression was analyzed from two H5N1-infected Ri chicken lines using small RNA sequencing. The target genes of differentially expressed (DE) miRNAs were predicted using miRDB. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis were then conducted. Furthermore, using quantitative real-time polymerase chain reaction, we validated the expression levels of DE miRNAs (miR-22-3p, miR-146b-3p, miR-27b-3p, miR-128-3p, miR-2188-5p, miR-451, miR-205a, miR-203a, miR-21-3p, and miR-200a-3p) from all comparisons and their immune-related target genes. RESULTS A total of 53 miRNAs were significantly expressed in the infection samples of the resistant compared to the susceptible line. Network analyses between the DE miRNAs and target genes revealed that DE miRNAs may regulate the expression of target genes involved in the transforming growth factor-beta, mitogen-activated protein kinase, and Toll-like receptor signaling pathways, all of which are related to influenza A virus progression. CONCLUSIONS Collectively, our results provided novel insights into the miRNA expression patterns of tracheal tissues from H5N1-infected Ri chickens. More importantly, our findings offer insights into the relationship between miRNA and immune-related target genes and the role of miRNA in HPAIV infections in chickens.
Collapse
Affiliation(s)
- Suyeon Kang
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Thi Hao Vu
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Jubi Heo
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Chaeeun Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea
| | - Hyun S Lillehoj
- Animal Biosciences and Biotechnology Laboratory, Agricultural Research Services, United States Department of Agriculture, Beltsville, MD 20705, USA
| | - Yeong Ho Hong
- Department of Animal Science and Technology, Chung-Ang University, Anseong 17546, Korea.
| |
Collapse
|
19
|
Bruce JW, Park E, Magnano C, Horswill M, Richards A, Potts G, Hebert A, Islam N, Coon JJ, Gitter A, Sherer N, Ahlquist P. HIV-1 virological synapse formation enhances infection spread by dysregulating Aurora Kinase B. PLoS Pathog 2023; 19:e1011492. [PMID: 37459363 PMCID: PMC10374047 DOI: 10.1371/journal.ppat.1011492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 07/27/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
HIV-1 spreads efficiently through direct cell-to-cell transmission at virological synapses (VSs) formed by interactions between HIV-1 envelope proteins (Env) on the surface of infected cells and CD4 receptors on uninfected target cells. Env-CD4 interactions bring the infected and uninfected cellular membranes into close proximity and induce transport of viral and cellular factors to the VS for efficient virion assembly and HIV-1 transmission. Using novel, cell-specific stable isotope labeling and quantitative mass spectrometric proteomics, we identified extensive changes in the levels and phosphorylation states of proteins in HIV-1 infected producer cells upon mixing with CD4+ target cells under conditions inducing VS formation. These coculture-induced alterations involved multiple cellular pathways including transcription, TCR signaling and, unexpectedly, cell cycle regulation, and were dominated by Env-dependent responses. We confirmed the proteomic results using inhibitors targeting regulatory kinases and phosphatases in selected pathways identified by our proteomic analysis. Strikingly, inhibiting the key mitotic regulator Aurora kinase B (AURKB) in HIV-1 infected cells significantly increased HIV activity in cell-to-cell fusion and transmission but had little effect on cell-free infection. Consistent with this, we found that AURKB regulates the fusogenic activity of HIV-1 Env. In the Jurkat T cell line and primary T cells, HIV-1 Env:CD4 interaction also dramatically induced cell cycle-independent AURKB relocalization to the centromere, and this signaling required the long (150 aa) cytoplasmic C-terminal domain (CTD) of Env. These results imply that cytoplasmic/plasma membrane AURKB restricts HIV-1 envelope fusion, and that this restriction is overcome by Env CTD-induced AURKB relocalization. Taken together, our data reveal a new signaling pathway regulating HIV-1 cell-to-cell transmission and potential new avenues for therapeutic intervention through targeting the Env CTD and AURKB activity.
Collapse
Affiliation(s)
- James W. Bruce
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Eunju Park
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Chris Magnano
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mark Horswill
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Alicia Richards
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Gregory Potts
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Alexander Hebert
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Nafisah Islam
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Joshua J. Coon
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Anthony Gitter
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Nathan Sherer
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Paul Ahlquist
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
20
|
Zhang X, Li A, Li T, Shou Z, Li Y, Qiao X, Zhou R, Zhong X, Li S, Li L. A potential anti-HIV-1 compound, Q308, inhibits HSV-2 infection and replication in vitro and in vivo. Biomed Pharmacother 2023; 162:114595. [PMID: 36989723 DOI: 10.1016/j.biopha.2023.114595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
HSV-2 is a common human pathogen worldwide that causes genital herpes. Due to the lack of an effective HSV-2 vaccine in the foreseeable future, there is an urgent need to develop effective, safe and affordable anti-HSV-2 agents. Our previous studies confirmed that a small-molecule compound, Q308, effectively inhibits the reactivation of latent HIV and might be developed as an anti-HIV-1 agent. Patients infected with HSV-2 are generally more susceptible to HIV-1 infection than normal humans. In this study, we found that Q308 treatment had strong inhibitory activity against both HSV-2 and acyclovir-resistant HSV-2 strains in vitro and reduced the viral titers in tissue. And this treatment effectively ameliorated the cytokine storm and pathohistological changes caused by HSV-2 infection in HSV-2-infected mice. Unlike nucleoside analogs such as acyclovir, Q308 inhibited post-viral entry events by attenuating the synthesis of viral proteins. Furthermore, Q308 treatment blocked HSV-2-induced PI3K/AKT phosphorylation due to its inhibition on viral infection and replication. Overall, Q308 treatment exhibits potent anti-HSV-2 activity by inhibiting viral replication both in vitro and in vivo. Q308 is a promising lead compound for the development of new anti-HSV-2/HIV-1 therapies, particularly against acyclovir-resistant HSV-2 strains.
Collapse
|
21
|
Lekshmi VS, Asha K, Sanicas M, Asi A, Arya UM, Kumar B. PI3K/Akt/Nrf2 mediated cellular signaling and virus-host interactions: latest updates on the potential therapeutic management of SARS-CoV-2 infection. Front Mol Biosci 2023; 10:1158133. [PMID: 37325475 PMCID: PMC10267462 DOI: 10.3389/fmolb.2023.1158133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
The emergence and re-emergence of viral diseases, which cause significant global mortality and morbidity, are the major concerns of this decade. Of these, current research is focused majorly on the etiological agent of the COVID-19 pandemic, SARS-CoV-2. Understanding the host response and metabolic changes during viral infection may provide better therapeutic targets for the proper management of pathophysiological conditions associated with SARS-CoV-2 infection. We have achieved control over most emerging viral diseases; however, a lack of understanding of the underlying molecular events prevents us from exploring novel therapeutic targets, leaving us forced to witness re-emerging viral infections. SARS-CoV-2 infection is usually accompanied by oxidative stress, which leads to an overactive immune response, the release of inflammatory cytokines, increasing lipid production, and also alterations in the endothelial and mitochondrial functions. PI3K/Akt signaling pathway confers protection against oxidative injury by various cell survival mechanisms including Nrf2-ARE mediated antioxidant transcriptional response. SARS-CoV-2 is also reported to hijack this pathway for its survival within host and few studies have suggested the role of antioxidants in modulating the Nrf2 pathway to manage disease severity. This review highlights the interrelated pathophysiological conditions associated with SARS-CoV-2 infection and the host survival mechanisms mediated by PI3K/Akt/Nrf2 signaling pathways that can help ameliorate the severity of the disease and provide effective antiviral targets against SARS-CoV-2.
Collapse
Affiliation(s)
- V. S. Lekshmi
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - Kumari Asha
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | | | - Abhila Asi
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - U. M. Arya
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| | - Binod Kumar
- Department of Antiviral Research, Institute of Advanced Virology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
22
|
Hu K, Onintsoa Diarimalala R, Yao C, Li H, Wei Y. EV-A71 Mechanism of Entry: Receptors/Co-Receptors, Related Pathways and Inhibitors. Viruses 2023; 15:785. [PMID: 36992493 PMCID: PMC10051052 DOI: 10.3390/v15030785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 03/31/2023] Open
Abstract
Enterovirus A71, a non-enveloped single-stranded (+) RNA virus, enters host cells through three stages: attachment, endocytosis and uncoating. In recent years, receptors/co-receptors anchored on the host cell membrane and involved in this process have been continuously identified. Among these, hSCARB-2 was the first receptor revealed to specifically bind to a definite site of the EV-A71 viral capsid and plays an indispensable role during viral entry. It actually acts as the main receptor due to its ability to recognize all EV-A71 strains. In addition, PSGL-1 is the second EV-A71 receptor discovered. Unlike hSCARB-2, PSGL-1 binding is strain-specific; only 20% of EV-A71 strains isolated to date are able to recognize and bind it. Some other receptors, such as sialylated glycan, Anx 2, HS, HSP90, vimentin, nucleolin and fibronectin, were discovered successively and considered as "co-receptors" because, without hSCARB-2 or PSGL-1, they are not able to mediate entry. For cypA, prohibitin and hWARS, whether they belong to the category of receptors or of co-receptors still needs further investigation. In fact, they have shown to exhibit an hSCARB-2-independent entry. All this information has gradually enriched our knowledge of EV-A71's early stages of infection. In addition to the availability of receptors/co-receptors for EV-A71 on host cells, the complex interaction between the virus and host proteins and various intracellular signaling pathways that are intricately connected to each other is critical for a successful EV-A71 invasion and for escaping the attack of the immune system. However, a lot remains unknown about the EV-A71 entry process. Nevertheless, researchers have been continuously interested in developing EV-A71 entry inhibitors, as this study area offers a large number of targets. To date, important progress has been made toward the development of several inhibitors targeting: receptors/co-receptors, including their soluble forms and chemically designed compounds; virus capsids, such as capsid inhibitors designed on the VP1 capsid; compounds potentially interfering with related signaling pathways, such as MAPK-, IFN- and ATR-inhibitors; and other strategies, such as siRNA and monoclonal antibodies targeting entry. The present review summarizes these latest studies, which are undoubtedly of great significance in developing a novel therapeutic approach against EV-A71.
Collapse
Affiliation(s)
| | | | | | | | - Yanhong Wei
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (K.H.); (R.O.D.); (C.Y.); (H.L.)
| |
Collapse
|
23
|
Wang J, Zhu HD, Wang YX, Guo ZX, Liu YX, Huang ZH, Zhu LB, Liu MH, Liu SH, Xu JP. Trehalose hydrolysis and transport-related genes promote Bombyx mori nucleopolyhedrovirus proliferation through the phosphoinositide 3-kinase-Akt signalling pathway in BmN cell. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 140:104625. [PMID: 36572165 DOI: 10.1016/j.dci.2022.104625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 06/17/2023]
Abstract
The reprogramming of host physiology has been considered an essential process for baculovirus propagation. Trehalose, the main sugar in insect blood, plays a crucial role as an instant energy source. Although the trehalose level is modulated following infection with Bombyx mori nucleopolyhedrovirus (BmNPV), the mechanism of trehalose metabolism in response to BmNPV infection is still unclear. In this study, we demonstrated that the trehalose level tended to be lower in BmNPV-infected hemolymph and higher in the midgut. The omics analysis revealed that two trehalose transporters, BmTret1-1 and BmTret1-2, and trehalase, BmTRE1 and BmTRE2, were differentially expressed in the midgut after BmNPV infection. BmTret1-1 and BmTret1-2 had the ability to transport trehalose into the cell and promoted cellular absorption of trehalose. Furthermore, the functions of BmTret1-1, BmTret1-2, BmTRE1 and BmTRE2 in BmNPV infection were analyzed. These genes were upregulated in the midgut after BmNPV infection. Virus amplification analysis revealed that these genes could promote BmNPV proliferation in BmN cells. In addition, these genes could promote the expression of BmPI3K, BmPDK1 and BmAkt and inhibit the expression of BmFoxO in the phosphoinositide 3-kinase (PI3K)-Akt signalling pathway. Similarly, the increased trehalose level in BmN cells could promote the expression of BmPI3K, BmPDK1 and BmAkt and inhibit the expression of BmFoxO. Taken together, BmNPV infection promote the expression of trehalose hydrolysis and transport-related genes. These changes affect the PI3K-Akt signalling pathway to facilitate BmNPV proliferation. These findings help clarify the relationship between trehalose metabolism and BmNPV infection.
Collapse
Affiliation(s)
- Jie Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Institute of Sericulture, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Han-Dan Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Yan-Xiang Wang
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Zhe-Xiao Guo
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Ying-Xue Liu
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Zhi-Hao Huang
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Lin-Bao Zhu
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China
| | - Ming-Hui Liu
- Institute of Sericulture, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Shi-Huo Liu
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China.
| | - Jia-Ping Xu
- School of Life Sciences, Anhui Agricultural University, Hefei, China; Anhui International Joint Research and Developmental Center of Sericulture Resources Utilization, Hefei, China.
| |
Collapse
|
24
|
Gamil H, Assaf M, Khater M, Fawzy M. Abnormal nuclear expression of aquaporin-3 in lesional and perilesional skin of vitiligo patients: A novel immunohistochemical finding. J Cosmet Dermatol 2023; 22:1063-1070. [PMID: 36437598 DOI: 10.1111/jocd.15518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Vitiligo is a skin disease characterized by a complex etiopathogenesis. Keratinocyte apoptosis may play a role in vitiligo pathogenesis. Aquaporin-3 (AQP-3) is an aqua-glyceroporin that controls keratinocyte proliferation and differentiation. AIM To assess the immunohistochemical expression of AQP-3 in lesional and perilesional skin of vitiligo patients compared to healthy control skin. METHODS A total of 20 patients with generalized non-segmental vitiligo and 20 age- and sex-matched healthy controls were included. Lesional and perilesional skin of vitiligo patients, as well as normal skin of control subjects, were biopsied. The immunohistochemical expression of AQP3 in the epidermis was examined. RESULTS Compared to control skin, both lesional and perilesional skin showed a significant reduction in the intensity of membranous staining of AQP-3 (p < 0.001, p = 0.002, respectively). Moreover, the membrano-cytoplasmic pattern of AQP-3 staining was significantly detected in 80% of lesions and 85% of perilesional biopsies, while it was absent in control skin (p < 0.001). Additionally, nuclear AQP-3 expression was significantly detected in 35% of lesions and 55% of perilesional biopsies, while it was not detected in control skin (p = 0.012, p < 0.001, respectively). No statistically significant difference was detected between lesional and perilesional skin. CONCLUSIONS To our knowledge, this is the first immunohistochemical research to show a significant abnormal nuclear expression of AQP-3 in lesional and perilesional skin of vitiligo patients. This abnormality may reflect impaired functions of AQP-3, leading to keratinocyte apoptosis with subsequent melanocyte death and development of vitiligo.
Collapse
Affiliation(s)
- Hend Gamil
- Department of Dermatology, Venereology & Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Magda Assaf
- Department of Pathology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohamad Khater
- Department of Dermatology, Venereology & Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Manal Fawzy
- Department of Dermatology, Venereology & Andrology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
25
|
Chakraborty C, Bhattacharya M, Dhama K, Lee SS. Evaluation of differentially expressed genes during replication using gene expression landscape of monkeypox-infected MK2 cells: A bioinformatics and systems biology approach to understanding the genomic pattern of viral replication. J Infect Public Health 2023; 16:399-409. [PMID: 36724696 PMCID: PMC9874307 DOI: 10.1016/j.jiph.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
PURPOSE The current outbreak of monkeypox (MPX) has created colossal concerns. However, immense research gaps have been noted in our understanding of the replication process, machinery, and genomic landscape during host cell infection. To fill this gap, differentially expressed genes (DEGs) were comprehensively analyzed during viral replication in host (MK2) cells. METHODS We used a microarray GEO dataset which was divided into three groups: control, MPXV-infected MK2 cells at 3 h, and MPXV-infected MK2 cells at 7 h. Using the dataset, DEG analysis, PPI network analysis, co-expression, and pathway analysis were conducted using bioinformatics, systems biology, and statistical approaches. RESULTS We identified 250 DEGs and 24 top-ranked genes. During the DEG analysis, we identified eight up-regulated genes (LOC695323, TMEM107, LOC695427, HIST1H2AD, LOC705469, PMAIP1, HIST1H2BJ, and HIST1H3D) and 16 down-regulated genes (HOXA9, BAMBI, LMO4, PAX6, AJUBA, CREBRF, CD24, JADE1, SLC7A11, EID2, SOX4, B4GALT5, PPARGC1A, BUB3, SOS2, and CDK19). We also developed PPI networks and performed co-expression analyses using the top-ranked genes. Furthermore, five genes were listed for co-expression pattern analysis. CONCLUSIONS This study will help in better understanding the replication process, machinery, and genomic landscape of the virus. This will further aid the discovery and development of therapeutics against viruses.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India.
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore 756020, Odisha, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon 24252, Gangwon-Do, Republic of Korea.
| |
Collapse
|
26
|
Yang LH, Dong RJ, Lu YW, Wang HM, Kuang YQ, Wang RR, Li YY. Integration of metabolomics and transcriptomics analyses reveals sphingosine-1-phosphate-mediated S1PR2/PI3K/Akt pathway involved in Talaromyces marneffei infection of macrophages. Microb Pathog 2023; 175:105985. [PMID: 36638850 DOI: 10.1016/j.micpath.2023.105985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Talaromycosis is a fatal mycosis caused by the thermally dimorphic fungus Talaromyces marneffei (T. marneffei). The pathogenic mechanisms of talaromycosis are still poorly understood. This work combined metabolomics, transcriptomics, and verification experiments in vivo and in vitro to detect metabolic profiles and differentially expressed genes (DEGs) in T. marneffei infected and uninfected macrophages to explore possible pathogenesis and underlying mechanisms. A total of 256 differential metabolites (117 up-regulated and 148 down-regulated) and 1320 DEGs (1286 up-regulated and 34 down-regulated) were identified between the two groups. Integrative metabolomics and transcriptomics analysis showed sphingolipid signaling pathway is the most influential. Verification experiments showed that compared with the control group, the production of sphingosine-1-phosphate (S1P) and the expression of the S1PR1, S1PR2, phosphor-PI3K, and phosphor-Akt genes involved in the sphingolipid signaling pathway have significantly increased in the T. marneffei infection group (p < 0.05). T. marneffei activates the S1PR2/PI3K/Akt pathways in J774A.1 macrophage, regulation of the S1P singling might serve as a promising therapeutic strategy for talaromycosis.
Collapse
Affiliation(s)
- Lu-Hui Yang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Rong-Jing Dong
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - You-Wang Lu
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - Hong-Mei Wang
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi-Qun Kuang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, China; Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Rui-Rui Wang
- School of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, China.
| | - Yu-Ye Li
- Department of Dermatology and Venereology, First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
27
|
Mardi N, Haiaty S, Rahbarghazi R, Mobarak H, Milani M, Zarebkohan A, Nouri M. Exosomal transmission of viruses, a two-edged biological sword. Cell Commun Signal 2023; 21:19. [PMID: 36691072 PMCID: PMC9868521 DOI: 10.1186/s12964-022-01037-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
As a common belief, most viruses can egress from the host cells as single particles and transmit to uninfected cells. Emerging data have revealed en bloc viral transmission as lipid bilayer-cloaked particles via extracellular vesicles especially exosomes (Exo). The supporting membrane can be originated from multivesicular bodies during intra-luminal vesicle formation and autophagic response. Exo are nano-sized particles, ranging from 40-200 nm, with the ability to harbor several types of signaling molecules from donor to acceptor cells in a paracrine manner, resulting in the modulation of specific signaling reactions in target cells. The phenomenon of Exo biogenesis consists of multiple and complex biological steps with the participation of diverse constituents and molecular pathways. Due to similarities between Exo biogenesis and virus replication and the existence of shared pathways, it is thought that viruses can hijack the Exo biogenesis machinery to spread and evade immune cells. To this end, Exo can transmit complete virions (as single units or aggregates), separate viral components, and naked genetic materials. The current review article aims to scrutinize challenges and opportunities related to the exosomal delivery of viruses in terms of viral infections and public health. Video Abstract.
Collapse
Affiliation(s)
- Narges Mardi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sanya Haiaty
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Halimeh Mobarak
- Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St., Tabriz, Iran
| | - Morteza Milani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Zarebkohan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
Ferrara G, Longobardi C, Damiano S, Ciarcia R, Pagnini U, Montagnaro S. Modifications of the PI3K/Akt/mTOR axis during FeHV-1 infection in permissive cells. Front Vet Sci 2023; 10:1157350. [PMID: 37026095 PMCID: PMC10072329 DOI: 10.3389/fvets.2023.1157350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/02/2023] [Indexed: 04/08/2023] Open
Abstract
FeHV-1 is the causative agent of infectious rhinotracheitis in cats. The relationship between viral infection and the PI3K/Akt/mTOR pathway, as well as its function in crucial physiological processes like as autophagy, apoptosis or the IFN induction cascade is known for other varicelloviruses. However, there is no information on whether autophagy is activated during FeHV-1 infection nor on how this infection modifies PI3K/Akt/mTOR pathway. In this work, we aim to elucidate the involvement of this pathway during cytolytic infection by FeHV-1 in permissive cell lines. Using a phenotypic approach, the expression of proteins involved in the PI3K/Akt/mTOR pathway was examined by Western blot analysis. The findings demonstrated the lack of modifications in relation to viral dose (except for phospho-mTOR), whereas there were changes in the expression of several markers in relation to time as well as a mismatch in the time of activation of this axis. These results suggest that FeHV-1 may interact independently with different autophagic signaling pathways. In addition, we found an early phosphorylation of Akt, approximately 3 h after infection, without a concomitant decrease in constitutive Akt. This result suggests a possible role for this axis in viral entry. In a second phase, the use of early autophagy inhibitors was examined for viral yield, cytotoxic effects, viral glycoprotein expression, and autophagy markers and resulted in inefficient inhibition of viral replication (12 h post-infection for LY294002 and 48 h post-infection for 3-methyladenine). The same markers were examined during Akt knockdown, and we observed no differences in viral replication. This result could be explained by the presence of a protein kinase in the FeHV-1 genome (encoded by the Us3 gene) that can phosphorylate various Akt substrates as an Akt surrogate, as has already been demonstrated in genetically related viruses (HSV-1, PRV, etc.). For the same reasons, the use of LY294002 at the beginning of infection did not affect FeHV-1-mediated Akt phosphorylation. Our findings highlight changes in the PI3K/Akt/mTOR pathway during FeHV-1 infection, although further research is needed to understand the importance of these changes and how they affect cellular processes and viral propagation.
Collapse
Affiliation(s)
- Gianmarco Ferrara
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
- *Correspondence: Gianmarco Ferrara
| | - Consiglia Longobardi
- Department of Mental, Physical Health and Preventive Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Sara Damiano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Roberto Ciarcia
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Ugo Pagnini
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Serena Montagnaro
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| |
Collapse
|
29
|
Tumolo MR, Scoditti E, Guarino R, Grassi T, Bagordo F, Sabina S. MIR-29A-3P, MIR-29C-3P, MIR-146B-5P AND MIR-150-5P, Their Target Genes and lncrnas in HIV Infection: A Bioinformatic Study. Curr HIV Res 2023; 21:128-139. [PMID: 37226785 DOI: 10.2174/1570162x21666230524151328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/12/2023] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
INTRODUCTION Increasing evidence suggests that microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) have emerged as attractive targets in viral infections, including Human immunodeficiency virus (HIV). OBJECTIVE To deepen the understanding of the molecular mechanisms that lead to HIV and provide potential targets for the future development of molecular therapies for its treatment. METHODS Four miRNAs were selected as candidates based on a previous systematic review. A combination of bioinformatic analyses was performed to identify their target genes, lncRNAs and biological processes that regulate them. RESULTS In the constructed miRNA-mRNA network, 193 gene targets are identified. These miRNAs potentially control genes from several important processes, including signal transduction and cancer. LncRNA-XIST, lncRNA-NEAT1 and lncRNA-HCG18 interact with all four miRNAs. CONCLUSION This preliminary result forms the basis for improving reliability in future studies to fully understand the role these molecules and their interactions play in HIV.
Collapse
Affiliation(s)
- Maria Rosaria Tumolo
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Egeria Scoditti
- Institute of Clinical Physiology, National Research Council, Branch of Lecce, Lecce, Italy
| | - Roberto Guarino
- Institute of Clinical Physiology, National Research Council, Branch of Lecce, Lecce, Italy
| | - Tiziana Grassi
- Department of Biological and Environmental Sciences and Technology, University of Salento, Lecce, Italy
| | - Francesco Bagordo
- Department of Pharmacy- Pharmaceutical Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Saverio Sabina
- Institute of Clinical Physiology, National Research Council, Branch of Lecce, Lecce, Italy
| |
Collapse
|
30
|
PIK-24 Inhibits RSV-Induced Syncytium Formation via Direct Interaction with the p85α Subunit of PI3K. J Virol 2022; 96:e0145322. [PMID: 36416586 PMCID: PMC9749462 DOI: 10.1128/jvi.01453-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Phosphoinositide-3 kinase (PI3K) signaling regulates many cellular processes, including cell survival, differentiation, proliferation, cytoskeleton reorganization, and apoptosis. The actin cytoskeleton regulated by PI3K signaling plays an important role in plasma membrane rearrangement. Currently, it is known that respiratory syncytial virus (RSV) infection requires PI3K signaling. However, the regulatory pattern or corresponding molecular mechanism of PI3K signaling on cell-to-cell fusion during syncytium formation remains unclear. This study synthesized a novel PI3K inhibitor PIK-24 designed with PI3K as a target and used it as a molecular probe to investigate the involvement of PI3K signaling in syncytium formation during RSV infection. The results of the antiviral mechanism revealed that syncytium formation required PI3K signaling to activate RHO family GTPases Cdc42, to upregulate the inactive form of cofilin, and to increase the amount of F-actin in cells, thereby causing actin cytoskeleton reorganization and membrane fusion between adjacent cells. PIK-24 treatment significantly abolished the generation of these events by blocking the activation of PI3K signaling. Moreover, PIK-24 had an obvious binding activity with the p85α regulatory subunit of PI3K. The anti-RSV effect similar to PIK-24 was obtained after knockdown of p85α in vitro or knockout of p85α in vivo, suggesting that PIK-24 inhibited RSV infection by targeting PI3K p85α. Most importantly, PIK-24 exerted a potent anti-RSV activity, and its antiviral effect was stronger than that of the classic PI3K inhibitor LY294002, PI-103, and broad-spectrum antiviral drug ribavirin. Thus, PIK-24 has the potential to be developed into a novel anti-RSV agent targeting cellular PI3K signaling. IMPORTANCE PI3K protein has many functions and regulates various cellular processes. As an important regulatory subunit of PI3K, p85α can regulate the activity of PI3K signaling. Therefore, it serves as the key target for virus infection. Indeed, p85α-regulated PI3K signaling facilitates various intracellular plasma membrane rearrangement events by modulating the actin cytoskeleton, which may be critical for RSV-induced syncytium formation. In this study, we show that a novel PI3K inhibitor inhibits RSV-induced PI3K signaling activation and actin cytoskeleton reorganization by targeting the p85α protein, thereby inhibiting syncytium formation and exerting a potent antiviral effect. Respiratory syncytial virus (RSV) is one of the most common respiratory pathogens, causing enormous morbidity, mortality, and economic burden. Currently, no effective antiviral drugs or vaccines exist for RSV infection. This study contributes to understanding the molecular mechanism by which PI3K signaling regulates syncytium formation and provides a leading compound for anti-RSV infection drug development.
Collapse
|
31
|
Lu MY, Chtarbanova S. The role of micro RNAs (miRNAs) in the regulation of Drosophila melanogaster's innate immunity. Fly (Austin) 2022; 16:382-396. [PMID: 36412256 PMCID: PMC9683055 DOI: 10.1080/19336934.2022.2149204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs ~19-22 nt long which post-transcriptionally regulate gene expression. Their ability to exhibit dynamic expression patterns coupled with their wide variety of targets allows miRNAs to regulate many processes, including the innate immune response of Drosophila melanogaster. Recent studies have identified miRNAs in Drosophila which are differentially expressed during infection with different pathogens as well as miRNAs that may affect immune signalling when differentially expressed. This review provides an overview of miRNAswhich have been identified to play a role in the immune response of Drosophila through targeting of the Toll and IMD signalling pathways and other immune processes. It will also explore the role of miRNAs in fine-tuning the immune response in Drosophila and highlight current gaps in knowledge regarding the role of miRNAs in immunity and areas for further research.
Collapse
Affiliation(s)
- Max Yang Lu
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA,Center for Convergent Bioscience & Medicine, University of Alabama, Tuscaloosa, AL, USA,Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, USA,CONTACT Stanislava Chtarbanova Department of Biological Sciences, the University of Alabama, 300, Hackberry Ln, Tuscaloosa, AL-35487, USA
| |
Collapse
|
32
|
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogen responsible for the coronavirus disease 2019 (COVID-19) pandemic. Of particular interest for this topic are the signaling cascades that regulate cell survival and death, two opposite cell programs whose control is hijacked by viral infections. The AKT and the Unfolded Protein Response (UPR) pathways, which maintain cell homeostasis by regulating these two programs, have been shown to be deregulated during SARS-CoVs infection as well as in the development of cancer, one of the most important comorbidities in relation to COVID-19. Recent evidence revealed two way crosstalk mechanisms between the AKT and the UPR pathways, suggesting that they might constitute a unified homeostatic control system. Here, we review the role of the AKT and UPR pathways and their interaction in relation to SARS-CoV-2 infection as well as in tumor onset and progression. Feedback regulation between AKT and UPR pathways emerges as a master control mechanism of cell decision making in terms of survival or death and therefore represents a key potential target for developing treatments for both viral infection and cancer. In particular, drug repositioning, the investigation of existing drugs for new therapeutic purposes, could significantly reduce time and costs compared to de novo drug discovery.
Collapse
|
33
|
Antiviral perspectives of economically important Indian medicinal plants and spices. PROCEEDINGS OF THE INDIAN NATIONAL SCIENCE ACADEMY 2022. [PMCID: PMC9422945 DOI: 10.1007/s43538-022-00099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human respiratory diseases caused by viral infections leads to morbidity. Among infectious diseases, viral infections associated with the respiratory tract remain the primary reason for global deaths due to their transmissibility. Since immemorial, traditional Indian medicinal plants, their extracts, and several phytochemicals can treat various diseases. Sources for this review paper are data derived from a peer-reviewed journal that emphasizes the economic importance of medicinal plants. Several plant-based medicines have been reported to be effective against multiple viral infections, including the Human Adenovirus, Enterovirus, Influenza virus, Hepatitis virus, etc. This review emphasizes use of the Indian medicinal plants like as Withania somnifera (Ashwagandha, Winter Cherry), Moringa oleifera (Drumstick), Ocimum tenuiflorum (Tulsi), Azadirachta indica (Neem), Curcuma longa (Turmeric), Terminalia chebula (Chebulic Myrobalan), Punica granatum (Pomegranate) and the Indian household spices (ginger, garlic and black pepper). It further describes their secondary phytoconstituents extraction procedure, mode of action and the potential application to improve clinical outcomes of neutraceuticals against various viral infections.
Collapse
|
34
|
Du X, Zhou D, Zhou J, Xue J, Cheng Z. RIOK3-mediated Akt phosphorylation facilitates synergistic replication of Marek's disease and reticuloendotheliosis viruses. Virulence 2022; 13:1184-1198. [PMID: 35795905 PMCID: PMC9331201 DOI: 10.1080/21505594.2022.2096247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Co-infection of Marek’s disease virus (MDV) and reticuloendotheliosis virus (REV) synergistically drives disease progression, yet little is known about the mechanism of the synergism. Here, we found that co-infection of REV and MDV increased their replication via the RIOK3-Akt pathway. Initially, we noticed that the viral titres of MDV and REV significantly increased in REV and MDV co-infected cells compared with single-infected cells. Furthermore, tandem mass tag peptide labelling coupled with LC/MS analysis showed that Akt was upregulated in REV and MDV co-infected cells. Overexpression of Akt promoted synergistic replication of MDV and REV. Conversely, inhibition of Akt suppressed synergistic replication of MDV and REV. However, PI3K inhibition did not affect synergistic replication of MDV and REV, suggesting that the PI3K/Akt pathway is not involved in the synergism of MDV and REV. In addition, we revealed that RIOK3 was recruited to regulate Akt in REV and MDV co-infected cells. Moreover, wild-type RIOK3, but not kinase-dead RIOK3, mediated Akt phosphorylation and promoted synergistic replication of MDV and REV. Our results illustrate that MDV and REV activated a novel RIOK3-Akt signalling pathway to facilitate their synergistic replication.
Collapse
Affiliation(s)
- Xusheng Du
- College of Veterinary Medicine, Shandong Agricultural University, China
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, China
| | - Jing Zhou
- College of Veterinary Medicine, Shandong Agricultural University, China
| | - Jingwen Xue
- College of Veterinary Medicine, Shandong Agricultural University, China
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, China
| |
Collapse
|
35
|
Tang Q, Luan F, Yuan A, Sun J, Rao Z, Wang B, Liu Y, Zeng N. Sophoridine Suppresses Herpes Simplex Virus Type 1 Infection by Blocking the Activation of Cellular PI3K/Akt and p38 MAPK Pathways. Front Microbiol 2022; 13:872505. [PMID: 35756044 PMCID: PMC9229184 DOI: 10.3389/fmicb.2022.872505] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a ubiquitous and important human pathogen capable of causing significant clinical diseases ranging from skin damage to encephalitis, particularly in immunocompromised and neonatal hosts. Currently, widely used nucleoside analogs, including acyclovir and penciclovir, have some limitations in their use due to side effects and drug resistance. Herein, we report sophoridine's (SRI) dramatic inhibition of HSV-1 replication in vitro. SRI exhibited a remarkable inhibitory influence on HSV-1 virus-induced cytopathic effect and plaque formation, as well as on progeny viruses in Vero and HeLa cells, with selection indexes (SI) of 38.96 and 22.62, respectively. Moreover, SRI also considerably suppressed HSV-1 replication by hindering the expression of viral immediate-early (ICP0 and ICP22), early (ICP8 and TK), and late (gB and gD) genes and the expression of viral proteins ICP0, gB, and gD. We suggest that SRI can directly inactivate viral particles and block some stages in the life cycle of HSV-1 after adsorption. Further experiments showed that SRI downregulated the cellular PI3K/Akt signaling pathway and obstructed HSV-1 replication even more. Most importantly, SRI markedly repressed HSV-1-induced p38 MAPK pathway activation. Collectively, this natural bioactive alkaloid could be a promising therapeutic candidate against HSV-1 via the modulation of cellular PI3K/Akt and p38 MAPK pathways.
Collapse
Affiliation(s)
- Qiong Tang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Luan
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - An Yuan
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhili Rao
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Baojun Wang
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yao Liu
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Nan Zeng
- State Key Laboratory of South Western Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Rastegarpanah M, Azadmanesh K, Negahdari B, Asgari Y, Mazloomi M. Screening of candidate genes associated with high titer production of oncolytic measles virus based on systems biology approach. Virus Genes 2022; 58:270-283. [PMID: 35477822 DOI: 10.1007/s11262-022-01902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/01/2022] [Indexed: 10/18/2022]
Abstract
The number of viral particles required for oncolytic activity of measles virus (MV) can be more than a million times greater than the reported amount for vaccination. The aim of the current study is to find potential genes and signaling pathways that may be involved in the high-titer production of MV. In this study, a systems biology approach was considered including collection of gene expression profiles from the Gene Expression Omnibus (GEO) database, obtaining differentially expressed genes (DEGs), performing gene ontology, functional enrichment analyses, and topological analyses on the protein-protein interaction (PPI) network. Then, to validate the in-silico data, total RNA was isolated from five cell lines, and full-length cDNA from template RNA was synthesized. Subsequently, quantitative reverse transcription-PCR (RT-qPCR) was employed. We identified five hub genes, including RAC1, HSP90AA1, DNM1, LTBP1, and FSTL1 associated with the enhancement in MV titer. Pathway analysis indicated enrichment in PI3K-Akt signaling pathway, axon guidance, proteoglycans in cancer, regulation of actin cytoskeleton, focal adhesion, and calcium signaling pathways. Upon verification by RT-qPCR, the relative expression of candidate genes was generally consistent with our bioinformatics analysis. Hub genes and signaling pathways may be involved in understanding the pathological mechanisms by which measles virus manipulates host factors in order to facilitate its replication. RAC1, HSP90AA1, DNM1, LTBP1, and FSTL1 genes, in combination with genetic engineering techniques, will allow the direct design of high-throughput cell lines to answer the required amounts for the oncolytic activity of MV.
Collapse
Affiliation(s)
- Malihe Rastegarpanah
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Kayhan Azadmanesh
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Yazdan Asgari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammadali Mazloomi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
37
|
Bruno C, Paparo L, Pisapia L, Romano A, Cortese M, Punzo E, Berni Canani R. Protective effects of the postbiotic deriving from cow's milk fermentation with L. paracasei CBA L74 against Rotavirus infection in human enterocytes. Sci Rep 2022; 12:6268. [PMID: 35428750 PMCID: PMC9012738 DOI: 10.1038/s41598-022-10083-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/11/2022] [Indexed: 01/17/2023] Open
Abstract
Rotavirus (RV) is the leading cause of acute gastroenteritis-associated mortality in early childhood. Emerging clinical evidence suggest the efficacy of the postbiotic approach based on cow's milk fermentation with the probiotic Lacticaseibacillus paracasei CBAL74 (FM-CBAL74) in preventing pediatric acute gastroenteritis, but the mechanisms of action are still poorly characterized. We evaluated the protective action of FM-CBAL74 in an in vitro model of RV infection in human enterocytes. The number of infected cells together with the relevant aspects of RV infection were assessed: epithelial barrier damage (tight-junction proteins and transepithelial electrical resistance evaluation), and inflammation (reactive oxygen species, pro-inflammatory cytokines IL-6, IL-8 and TNF-α, and mitogen-activated protein kinase pathway activation). Pre-incubation with FM-CBA L74 resulted in an inhibition of epithelial barrier damage and inflammation mediated by mitogen-activated protein kinase pathway activation induced by RV infection. Modulating several protective mechanisms, the postbiotic FM-CBAL74 exerted a preventive action against RV infection. This approach could be a disrupting nutritional strategy against one of the most common killers for the pediatric age.
Collapse
Affiliation(s)
- Cristina Bruno
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Laura Pisapia
- Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Alessia Romano
- ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Maddalena Cortese
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Erika Punzo
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy.,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy. .,ImmunoNutritionLab at CEINGE Advanced Biotechnologies, University of Naples Federico II, Naples, Italy. .,European Laboratory for the Investigation of Food-Induced Diseases, University of Naples Federico II, Naples, Italy. .,Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy. .,Task Force for Nutraceuticals and Functional Foods, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
38
|
Vasileva NS, Ageenko AB, Richter VA, Kuligina EV. The Signaling Pathways Controlling the Efficacy of Glioblastoma Therapy. Acta Naturae 2022; 14:62-70. [PMID: 35923561 PMCID: PMC9307987 DOI: 10.32607/actanaturae.11623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/16/2022] [Indexed: 11/20/2022] Open
Abstract
The resistance of glioblastoma to existing therapies puts limits on quality-of-life improvements and patient survival with a glioblastoma diagnosis. The development of new effective glioblastoma therapies is based on knowledge about the mechanisms governing tumor resistance to therapeutic agents. Virotherapy is one of the most actively developing approaches to the treatment of malignant neoplasms: glioblastoma in particular. Previously, we demonstrated that the recombinant vaccinia virus VV-GMCSF-Lact exhibits in vitro cytotoxic activity and in vivo antitumor efficacy against human glioblastoma. However, the studied glioblastoma cell cultures had different sensitivities to the oncotoxic effect of the virus. In this study, we investigated cancer stem cell (CSC) surface markers in glioblastoma cells with different sensitivities to VV-GMCSFLact using flow cytometry and we assessed the levels of proteins affecting viral entry into cells and virus infection efficiency by western blotting. We showed that cell cultures more sensitive to VV-GMCSF-Lact are characterized by a greater number of cells with CSC markers and a lower level of activated Akt kinase. Akt probably inhibits lactaptin-induced apoptosis in virus-resistant cells. Hence, we suggest that the sensitivity of glioblastoma cells to the oncotoxic effect of VV-GMCSF-Lact is determined by the nature and extent of the disturbances in cell death regulation in various cultures. Further investigation of the factors affecting glioblastoma resistance to virotherapy will test this hypothesis and identify targets for antitumor therapy, combined with VV-GMCSF-Lact.
Collapse
Affiliation(s)
- N. S. Vasileva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090 Russia
| | - A. B. Ageenko
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090 Russia
| | - V. A. Richter
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090 Russia
| | - E. V. Kuligina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, 630090 Russia
| |
Collapse
|
39
|
Pasquereau S, Herbein G. CounterAKTing HIV: Toward a “Block and Clear” Strategy? Front Cell Infect Microbiol 2022; 12:827717. [PMID: 35186800 PMCID: PMC8856111 DOI: 10.3389/fcimb.2022.827717] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022] Open
Abstract
The protein kinase B or Akt is a central regulator of survival, metabolism, growth and proliferation of the cells and is known to be targeted by various viral pathogens, including HIV-1. The central role of Akt makes it a critical player in HIV-1 pathogenesis, notably by affecting viral entry, latency and reactivation, cell survival, viral spread and immune response to the infection. Several HIV proteins activate the PI3K/Akt pathway, to fuel the progression of the infection. Targeting Akt could help control HIV-1 entry, viral latency/replication, cell survival of infected cells, HIV spread from cell-to-cell, and the immune microenvironment which could ultimately allow to curtail the size of the HIV reservoir. Beside the “shock and kill” and “block and lock” strategies, the use of Akt inhibitors in combination with latency inducing agents, could favor the clearance of infected cells and be part of new therapeutic strategies with the goal to “block and clear” HIV.
Collapse
Affiliation(s)
- Sébastien Pasquereau
- Laboratory Pathogens & Inflammation-Epigenetics of Viral Infections and Inflammatory Diseases Laboratory (EPILAB), University of Franche-Comté, Bourgogne Franche-Comté University Bourgogne Franche-Comté (UBFC), Besançon, France
| | - Georges Herbein
- Laboratory Pathogens & Inflammation-Epigenetics of Viral Infections and Inflammatory Diseases Laboratory (EPILAB), University of Franche-Comté, Bourgogne Franche-Comté University Bourgogne Franche-Comté (UBFC), Besançon, France
- Laboratory of Virology, Centre Hospitalier Universitaire (CHU) Besançon University Hospital, Besançon, France
- *Correspondence: Georges Herbein,
| |
Collapse
|
40
|
Sinha M, Chakraborty U, Kool A, Chakravarti M, Das S, Ghosh S, Thakur L, Khuranna A, Nayak D, Basu B, Kar S, Ray R, Das S. In-vitro antiviral action of Eupatorium perfoliatum against dengue virus infection: Modulation of mTOR signaling and autophagy. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114627. [PMID: 34509603 DOI: 10.1016/j.jep.2021.114627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/26/2021] [Accepted: 09/08/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dengue virus (DENV) is a re-emerging mosquito-borne flavivirus that has recently engendered large epidemics around the world. Consequently antivirals with effective anti-DENV therapeutic activity are urgently required. In the 18th century, Europeans, as well as native inhabitants of North America, were known to adapt the medicinal property of the common perennial plant Eupatorium perfoliatum L. to treat fever and infections. Previous studies have shown that Eupatorium perfoliatum L. possesses anti-inflammatory, anti-oxidative, anti-plasmodial, anti-bacterial and antiviral activities. However, to the best of our knowledge, no anti-DENV activity of E. perfoliatum L. has been investigated at the molecular level so far. AIM OF STUDY Here, for the first time we have attempted to study the action of E. perfoliatum extract and its few bioactive components i.e., quercetin, caffeic acid and eupafolin against wild primary clinical isolate of DENV-2 infection in an in vitro model. MATERIALS AND METHODS The presence of the bioactive components in the E. perfoliatum extract, were analyzed by HPLC- DAD. Then, CC50 as well as IC50 values of the extract and its bioactive components were measured against DENV in HepG2 cell line. After that, the antiviral activity was studied by Time of addition assay using qRT-PCR. Further, the downstream signalling action of E. perfoliatum extract, was studied by Human phosphorylation MAPK antibody array, followed by immunofluorescence microscopy. Moreover, a molecular docking analysis was done to study the binding affinity of bioactive components of E. perfoliatum extract with TIM-1 transmembrane receptor protein, which is known for viral internalization. RESULT We found that E. perfoliatum extract has marked antiviral activity during pre-treatment against DENV infection in HepG2 cell line. The extract also significantly reduced the DENV induced autophagy in HepG2 cell line as detected by LC3 II localization. The presence of different bioactive compounds in E. perfoliatum extract were confirmed by HPLC-DAD. In the bioactive components, in parallel to earlier studies, quercetin showed the most significant preventive action against DENV infection. Further, in molecular docking analysis also, quercetin showed the strongest binding affinity towards DENV membrane receptor TIM-1 protein. CONCLUSION Our findings suggests that E. perfoliatum extract has significant potential to be an anti-DENV therapeutic agent. Moreover, among the bioactive components, quercetin may have a prophylaxis role in executing the antiviral activity of E. perfoliatum extract against DENV infection.
Collapse
Affiliation(s)
- Moonmoon Sinha
- Department of Microbiology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata-700019, India; Department of Virology, Dr. Anjali Chatterjee Regional Research Institute, Kolkata-700035, India; Department of Microbiology, Institute of Post Graduate Medical Education and Research, Kolkata-700020, India.
| | - Urmita Chakraborty
- Department of Virology, Dr. Anjali Chatterjee Regional Research Institute, Kolkata-700035, India.
| | - Anirban Kool
- Department of Virology, Dr. Anjali Chatterjee Regional Research Institute, Kolkata-700035, India.
| | - Mousumi Chakravarti
- Department of Virology, Dr. Anjali Chatterjee Regional Research Institute, Kolkata-700035, India.
| | - Souvik Das
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata-700026, India.
| | - Sandip Ghosh
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata-700026, India.
| | - Lovnish Thakur
- School of Biosciences, Apeejay Stya University, Gurugram, Haryana-122103, India.
| | - Anil Khuranna
- Central Council for Research in Homoeopathy, Ministry of AYUSH, Govt. of India, Janakpuri, New Delhi-111058, India.
| | - Debadatta Nayak
- Central Council for Research in Homoeopathy, Ministry of AYUSH, Govt. of India, Janakpuri, New Delhi-111058, India.
| | - Biswarup Basu
- Department of Neuroendocrinology and Experimental Hematology, Chittaranjan National Cancer Institute, Kolkata-700026, India.
| | - Subhabrata Kar
- School of Biosciences, Apeejay Stya University, Gurugram, Haryana-122103, India.
| | - Raja Ray
- Department of Microbiology, University of Calcutta, 35 Ballygunge Circular Road, Kolkata-700019, India; Department of Microbiology, Institute of Post Graduate Medical Education and Research, Kolkata-700020, India.
| | - Satadal Das
- Department of Virology, Dr. Anjali Chatterjee Regional Research Institute, Kolkata-700035, India.
| |
Collapse
|
41
|
Yang CY, Chen YH, Liu PJ, Hu WC, Lu KC, Tsai KW. The emerging role of miRNAs in the pathogenesis of COVID-19: Protective effects of nutraceutical polyphenolic compounds against SARS-CoV-2 infection. Int J Med Sci 2022; 19:1340-1356. [PMID: 35928726 PMCID: PMC9346380 DOI: 10.7150/ijms.76168] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/08/2022] [Indexed: 11/05/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can cause immunosuppression and cytokine storm, leading to lung damage and death. The clinical efficacy of anti-SARS-CoV-2 drugs in preventing viral entry into host cells and suppressing viral replication remains inadequate. MicroRNAs (miRNAs) are crucial to the immune response to and pathogenesis of coronaviruses, such as SARS-CoV-2. However, the specific roles of miRNAs in the life cycle of SARS-CoV-2 remain unclear. miRNAs can participate in SARS-CoV-2 infection and pathogenesis through at least four possible mechanisms: 1. host cell miRNA expression interfering with SARS-CoV-2 cell entry, 2. SARS-CoV-2-derived RNA transcripts acting as competitive endogenous RNAs (ceRNAs) that may attenuate host cell miRNA expression, 3. host cell miRNA expression modulating SARS-CoV-2 replication, and 4. SARS-CoV-2-encoded miRNAs silencing the expression of host protein-coding genes. SARS-CoV-2-related miRNAs may be used as diagnostic or prognostic biomarkers for predicting outcomes among patients with SARS-CoV-2 infection. Furthermore, accumulating evidence suggests that dietary polyphenolic compounds may protect against SARS-CoV-2 infection by modulating host cell miRNA expression. These findings have major implications for the future diagnosis and treatment of COVID-19.
Collapse
Affiliation(s)
- Chih-Yun Yang
- Division of Chest Medicine, Kaohsiung Municipal Min-Sheng Hospital, Kaohsiung, Taiwan, ROC
| | - Yu-Hsuan Chen
- Division of Chest Medicine, Department of Internal Medicine, CHENG HSIN General Hospital, Taipei, Taiwan, ROC
| | - Pei-Jung Liu
- Division of Chest Medicine, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Wan-Chung Hu
- Department of Clinical Pathology and Medical Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| | - Kuo-Cheng Lu
- Division of Nephrology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC.,Division of Nephrology, Department of Medicine, Fu-Jen Catholic University Hospital, School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan, ROC
| | - Kuo-Wang Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan, ROC
| |
Collapse
|
42
|
Liang G, Zhao J, Dou Y, Yang Y, Zhao D, Zhou Z, Zhang R, Yang W, Zeng L. Mechanism and Experimental Verification of Luteolin for the Treatment of Osteoporosis Based on Network Pharmacology. Front Endocrinol (Lausanne) 2022; 13:866641. [PMID: 35355555 PMCID: PMC8959132 DOI: 10.3389/fendo.2022.866641] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To explore the molecular mechanism of luteolin in the treatment of osteoporosis (OP) by network pharmacological prediction and experimentation. METHODS The target proteins of luteolin were obtained with the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). OP-related proteins were extracted from the Comparative Toxicogenomics Database (CTD) and GeneCards and DisGeNET databases. We imported the common protein targets of luteolin and OP into the STRING database to obtain the relationships between the targets. The common target proteins of luteolin and OP were assessed by KEGG and GO enrichment analyses with the DAVID database. Animal experiments were conducted to verify the effect of luteolin on bone mineral density in ovariectomised (OVX) rats. Finally, the effects of luteolin on key signalling pathways were verified by cell experiments in vitro. RESULTS Forty-four targets of luteolin involved in the treatment of OP, including key target proteins such as TP53, AKT1, HSP90AA1, JUN, RELA, CASP3, and MAPK1, were screened. KEGG enrichment analysis found that luteolin inhibits OP by regulating the PI3K-Akt, TNF, oestrogen and p53 signalling pathways. The results of animal experiments showed that bone mass in the low-dose luteolin group (Luteolin-L group, 10 mg/kg), high-dose luteolin group (Luteolin-H group, 50 mg/kg) and positive drug group was significantly higher than that in the OVX group (P<0.05). Western blot (WB) analysis showed that the protein expression levels of Collagen I, Osteopontin and RUNX2 in bone marrow mesenchymal stem cells (BMSCs) cultured with 0.5, 1 and 5 μM luteolin for 48 h were significantly higher than those in the dimethyl sulfoxide (DMSO) group (P<0.05). In vitro cell experiments showed that the p-PI3K/PI3K and p-Akt/Akt expression ratios in BMSCs cultured with 0.5, 1 and 5 μM luteolin for 48 h were also significantly higher than those in the DMSO group (P<0.05). CONCLUSIONS Luteolin has multitarget and multichannel effects in the treatment of OP. Luteolin could reduce bone loss in OVX rats, which may be due to its ability to promote the osteogenic differentiation of BMSCs by regulating the activity of the PI3K-Akt signalling pathway.
Collapse
Affiliation(s)
- Guihong Liang
- The 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jinlong Zhao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaoxing Dou
- The 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuan Yang
- The 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Di Zhao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhanpeng Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiyi Yang
- The 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Weiyi Yang, ; Lingfeng Zeng,
| | - Lingfeng Zeng
- The 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Weiyi Yang, ; Lingfeng Zeng,
| |
Collapse
|
43
|
Li Y, Li P, He Q, Zhang R, Li Y, Kamar N, Peppelenbosch MP, de Man RA, Wang L, Pan Q. Niclosamide inhibits hepatitis E virus through suppression of NF-kappaB signalling. Antiviral Res 2021; 197:105228. [PMID: 34929248 DOI: 10.1016/j.antiviral.2021.105228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 12/05/2021] [Accepted: 12/15/2021] [Indexed: 11/28/2022]
Abstract
Hepatitis E virus (HEV) infection can cause severe acute hepatitis in pregnant women and chronic infection in immunocompromised patients, promoting the development of effective antiviral therapies. In this study, we identified niclosamide, a widely used anthelmintic drug, as a potent inhibitor of HEV replication in a range of subgenomic and full-length HEV models, which are based on human cell lines and liver organoids harbouring genotype 1 and 3 HEV strains. Niclosamide is known to have multiple cellular targets including the inhibition of STAT3 and NFκB signaling pathways. Although HEV activates STAT3, we excluded its involvement in the anti-HEV activity of niclosamide. Interestingly, HEV infection activated NFκB and activation of NFκB promoted viral replication. Consistently, stable silencing of NFκB by lentiviral RNAi inhibited HEV replication. By targeting NFκB signaling, we further revealed its role in mediating the anti-HEV action of niclosamide. These results demonstrated that niclosamide potently inhibits HEV replication by inhibiting NFκB signaling but independent of STAT3. Our findings support the potential of repurposing niclosamide for treating HEV infection.
Collapse
Affiliation(s)
- Yunlong Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Qiyu He
- Department of Microbiology and Infectious Disease Centre, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Ruyi Zhang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Yang Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Nassim Kamar
- Department of Nephrology, Dialysis and Organ Transplantation, CHU Rangueil, INSERM U1043, IFR-BMT, University Paul Sabatier, Toulouse, France
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Robert A de Man
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Lin Wang
- Department of Microbiology and Infectious Disease Centre, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
44
|
Akt plays differential roles during the life cycles of acute and persistent murine norovirus strains in macrophages. J Virol 2021; 96:e0192321. [PMID: 34787460 DOI: 10.1128/jvi.01923-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Akt (Protein kinase B) is a key signaling protein in eukaryotic cells that controls many cellular processes such as glucose metabolism and cell proliferation for survival. As obligate intracellular pathogens, viruses modulate host cellular processes, including Akt signaling, for optimal replication. The mechanisms by which viruses modulate Akt and the resulting effects on the infectious cycle differ widely depending on the virus. In this study, we explored the effect of Akt serine 473 phosphorylation (p-Akt) during murine norovirus (MNV) infection. p-Akt increased during infection of murine macrophages with acute MNV-1 and persistent CR3 and CR6 strains. Inhibition of Akt with MK2206, an inhibitor of all three isoforms of Akt (Akt1/2/3), reduced infectious virus progeny of all three virus strains. This reduction was due to decreased viral genome replication (CR3), defective virus assembly (MNV-1), or diminished cellular egress (CR3 and CR6) in a virus strain-dependent manner. Collectively, our data demonstrate that Akt activation increases in macrophages during the later stages of the MNV infectious cycle, which may enhance viral infection in unique ways for different virus strains. The data, for the first time, indicate a role for Akt signaling in viral assembly and highlight additional phenotypic differences between closely related MNV strains. Importance Human noroviruses (HNoV) are a leading cause of viral gastroenteritis, resulting in high annual economic burden and morbidity; yet there are no small animal models supporting productive HNoV infection, or robust culture systems producing cell culture-derived virus stocks. As a result, research on drug discovery and vaccine development against norovirus infection has been challenging, and no targeted antivirals or vaccines against HNoV are approved. On the other hand, murine norovirus (MNV) replicates to high titers in cell culture and is a convenient and widespread model in norovirus research. Our data demonstrate the importance of Akt signaling during the late stage of the MNV life cycle. Notably, the effect of Akt signaling on genome replication, virus assembly and cellular egress is virus strain specific, highlighting the diversity of biological phenotypes despite small genetic variability among norovirus strains. This study is the first to demonstrate a role for Akt in viral assembly.
Collapse
|
45
|
Sarmiento ME, Chin KL, Lau NS, Aziah I, Ismail N, Norazmi MN, Acosta A, Yaacob NS. Comparative transcriptome profiling of horseshoe crab Tachypleus gigas hemocytes in response to lipopolysaccharides. FISH & SHELLFISH IMMUNOLOGY 2021; 117:148-156. [PMID: 34358702 DOI: 10.1016/j.fsi.2021.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/01/2021] [Accepted: 08/02/2021] [Indexed: 06/13/2023]
Abstract
Horseshoe crabs (HSCs) are living fossil species of marine arthropods with a long evolutionary history spanning approximately 500 million years. Their survival is helped by their innate immune system that comprises cellular and humoral immune components to protect them against invading pathogens. To help understand the genetic mechanisms involved, the present study utilised the Illumina HiSeq platform to perform transcriptomic analysis of hemocytes from the HSC, Tachypleus gigas, that were challenged with lipopolysaccharides (LPS). The high-throughput sequencing resulted in 352,077,208 and 386,749,136 raw reads corresponding to 282,490,910 and 305,709,830 high-quality mappable reads for the control and LPS-treated hemocyte samples, respectively. Based on the log-fold change of > 0.3 or < -0.3, 1338 genes were significantly upregulated and 215 genes were significantly downregulated following LPS stimulation. The differentially expressed genes (DEGs) were further identified to be associated with multiple pathways such as those related to immune defence, stress response, cytoskeleton function and signal transduction. This study provides insights into the underlying molecular and regulatory mechanisms in hemocytes exposed to LPS, which has relevance for the study of the immune response of HSCs to infection.
Collapse
Affiliation(s)
- Maria E Sarmiento
- School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Kai Ling Chin
- Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu, Sabah, Malaysia
| | - Nyok Sean Lau
- Centre for Chemical Biology, Universiti Sains Malaysia, Bayan Lepas, Pulau Pinang, Malaysia
| | - Ismail Aziah
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Noraznawati Ismail
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Nerus, Terengganu, Malaysia
| | - Mohd Nor Norazmi
- School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Armando Acosta
- School of Health Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia
| | - Nik Soriani Yaacob
- Department of Chemical Pathology, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, Kubang Kerian, Kelantan, Malaysia.
| |
Collapse
|
46
|
Vanderboom PM, Mun DG, Madugundu AK, Mangalaparthi KK, Saraswat M, Garapati K, Chakraborty R, Ebihara H, Sun J, Pandey A. Proteomic Signature of Host Response to SARS-CoV-2 Infection in the Nasopharynx. Mol Cell Proteomics 2021; 20:100134. [PMID: 34400346 PMCID: PMC8363427 DOI: 10.1016/j.mcpro.2021.100134] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/20/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has become a global health pandemic. COVID-19 severity ranges from an asymptomatic infection to a severe multiorgan disease. Although the inflammatory response has been implicated in the pathogenesis of COVID-19, the exact nature of dysregulation in signaling pathways has not yet been elucidated, underscoring the need for further molecular characterization of SARS-CoV-2 infection in humans. Here, we characterize the host response directly at the point of viral entry through analysis of nasopharyngeal swabs. Multiplexed high-resolution MS-based proteomic analysis of confirmed COVID-19 cases and negative controls identified 7582 proteins and revealed significant upregulation of interferon-mediated antiviral signaling in addition to multiple other proteins that are not encoded by interferon-stimulated genes or well characterized during viral infections. Downregulation of several proteasomal subunits, E3 ubiquitin ligases, and components of protein synthesis machinery was significant upon SARS-CoV-2 infection. Targeted proteomics to measure abundance levels of MX1, ISG15, STAT1, RIG-I, and CXCL10 detected proteomic signatures of interferon-mediated antiviral signaling that differentiated COVID-19-positive from COVID-19-negative cases. Phosphoproteomic analysis revealed increased phosphorylation of several proteins with known antiviral properties as well as several proteins involved in ciliary function (CEP131 and CFAP57) that have not previously been implicated in the context of coronavirus infections. In addition, decreased phosphorylation levels of AKT and PKC, which have been shown to play varying roles in different viral infections, were observed in infected individuals relative to controls. These data provide novel insights that add depth to our understanding of SARS-CoV-2 infection in the upper airway and establish a proteomic signature for this viral infection.
Collapse
Affiliation(s)
- Patrick M Vanderboom
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Dong-Gi Mun
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Anil K Madugundu
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Manipal Academy of Higher Education, Manipal, Karnataka, India; Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Kiran K Mangalaparthi
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, Kerala, India
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kishore Garapati
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA; Institute of Bioinformatics, International Technology Park, Bangalore, Karnataka, India; Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Rana Chakraborty
- Division of Pediatric Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA; Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hideki Ebihara
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jie Sun
- The Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA; Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA; Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Division of Clinical Biochemistry and Immunology, Mayo Clinic, Rochester, Minnesota, USA; Center for Molecular Medicine, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
47
|
Mudaliar P, Pradeep P, Abraham R, Sreekumar E. Targeting cap-dependent translation to inhibit Chikungunya virus replication: selectivity of p38 MAPK inhibitors to virus-infected cells due to autophagy-mediated down regulation of phospho-ERK. J Gen Virol 2021; 102. [PMID: 34328830 DOI: 10.1099/jgv.0.001629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The 5' capped, message-sense RNA genome of Chikungunya virus (CHIKV) utilizes the host cell machinery for translation. Translation is regulated by eIF2 alpha at the initiation phase and by eIF4F at cap recognition. Translational suppression by eIF2 alpha phosphorylation occurs as an early event in many alphavirus infections. We observe that in CHIKV-infected HEK293 cells, this occurs as a late event, by which time the viral replication has reached an exponential phase, implying its minimal role in virus restriction. The regulation by eIF4F is mediated through the PI3K-Akt-mTOR, p38 MAPK and RAS-RAF-MEK-ERK pathways. A kinetic analysis revealed that CHIKV infection did not modulate AKT phosphorylation, but caused a significant reduction in p38 MAPK phosphorylation. It caused degradation of phospho-ERK 1/2 by increased autophagy, leaving the PI3K-Akt-mTOR and p38 MAPK pathways for pharmacological targeting. mTOR inhibition resulted in moderate reduction in viral titre, but had no effect on CHIKV E2 protein expression, indicating a minimal role of the mTOR complex in virus replication. Inhibition of p38 MAPK using SB202190 caused a significant reduction in viral titre and CHIKV E2 and nsP3 protein expression. Furthermore, inhibiting the two pathways together did not offer any synergism, indicating that inhibiting the p38 MAPK pathway alone is sufficient to cause restriction of CHIKV replication. Meanwhile, in uninfected cells the fully functional RAS-RAF-MEK-ERK pathway can circumvent the effect of p38 MAPK inhibition on cap-dependent translation. Thus, our results show that host-directed antiviral strategies targeting cellular p38 MAPK are worth exploring against Chikungunya as they could be selective against CHIKV-infected cells with minimal effects on uninfected host cells.
Collapse
Affiliation(s)
- Prashant Mudaliar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India.,Research Centre, University of Kerala, Thiruvananthapuram 695034, Kerala, India
| | - Parvanendhu Pradeep
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India.,Research Centre, University of Kerala, Thiruvananthapuram 695034, Kerala, India
| | - Rachy Abraham
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| | - Easwaran Sreekumar
- Molecular Virology Laboratory, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram 695014, Kerala, India
| |
Collapse
|
48
|
Oh J, Garabedian E, Fuleihan R, Cunningham-Rundles C. Clinical Manifestations and Outcomes of Activated Phosphoinositide 3-Kinase δ Syndrome from the USIDNET Cohort. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2021; 9:4095-4102. [PMID: 34352450 DOI: 10.1016/j.jaip.2021.07.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/01/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Activated phosphoinosidtide 3-kinase δ syndrome (APDS) is a combined primary immunodeficiency characterized by gain-of-function mutations in PIK3CD and PIK3R1. APDS demonstrates a large range of phenotypes including respiratory and herpesvirus infections, lymphadenopathy, autoimmunity, and developmental delay. OBJECTIVE This study describes clinical phenotypes and disease outcomes of a large APDS cohort from the USIDNET Registry. METHODS 38 patients were enrolled in USIDNET and 2 additional patients were obtained from the Clinical Immunology division at Mount Sinai Hospital. Each patient's demographics, disease complications, genetic studies, laboratory data, therapeutic interventions, and clinical outcomes were reviewed. RESULTS There was a high frequency of respiratory infections (70.0% pneumonia) and herpesvirus infections (37.5%). Bronchiectasis was observed in 45.0% of patients. Lymphadenopathy was common (52.5%) and 12.5% of patients developed lymphoma. Neurological and developmental findings were common: 20.0% had developmental delay, 15.0% had seizures, and 10.0% had dysmorphic features. Asthma was more common in PIK3CD compared to PIK3R1 patients (63.6% vs 14.3%). More subjects with PIK3CD had CD3 lymphopenia compared to the PIK3R1 cohort. Seven patients underwent hematopietic stem cell transplantation. One patient died from infectious complications. CONCLUSION This is the first cohort comparing clinical manifestations in PIK3CD and PIK3R1 patients from the USIDNET Registry. Similar frequencies of respiratory and herpesvirus infections, lymphadenopathy, and developmental delay were observed compared to prior cohort studies. However, a higher frequency of asthma and CD3 lymphopenia in the PIK3CD cohort compared to the PIK3R1 cohort was observed.
Collapse
Affiliation(s)
- Jessica Oh
- Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai.
| | - Elizabeth Garabedian
- National Institute of Allergy and Infectious Diseases, National Institute of Health
| | - Ramsay Fuleihan
- Division of Pediatric Allergy, Immunology, and Rheumatology, Columbia University Irving Medical Center, New York, NY 10032
| | | |
Collapse
|
49
|
Kiser LM, Sokoloski KJ, Hardy RW. Interactions between capsid and viral RNA regulate Chikungunya virus translation in a host-specific manner. Virology 2021; 560:34-42. [PMID: 34023723 PMCID: PMC8206026 DOI: 10.1016/j.virol.2021.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/28/2021] [Indexed: 12/24/2022]
Abstract
Alphaviruses are positive sense, RNA viruses commonly transmitted by an arthropod vector to a mammalian or avian host. In recent years, a number of the Alphavirus members have reemerged as public health concerns. Transmission from mosquito vector to vertebrate hosts requires an understanding of the interaction between the virus and both vertebrate and insect hosts to develop rational intervention strategies. The current study uncovers a novel role for capsid protein during Chikungunya virus replication whereby the interaction with viral RNA in the E1 coding region regulates protein synthesis processes early in infection. Studies done in both the mammalian and mosquito cells indicate that interactions between viral RNA and capsid protein have functional consequences that are host species specific. Our data support a vertebrate-specific role for capsid:vRNA interaction in temporally regulating viral translation in a manner dependent on the PI3K-AKT-mTOR pathway.
Collapse
Affiliation(s)
- Lauren M Kiser
- Department of Biology, College of Arts and Sciences, Indiana University, Bloomington, IN, USA
| | - Kevin J Sokoloski
- Department of Microbiology and Immunology and Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville School of Medicine, Louisville, KY, USA
| | - Richard W Hardy
- Department of Biology, College of Arts and Sciences, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
50
|
The IGF-1 Signaling Pathway in Viral Infections. Viruses 2021; 13:v13081488. [PMID: 34452353 PMCID: PMC8402757 DOI: 10.3390/v13081488] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/21/2021] [Indexed: 01/29/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) and the IGF-1 receptor (IGF-1R) belong to the insulin-like growth factor family, and IGF-1 activates intracellular signaling pathways by binding specifically to IGF-1R. The interaction between IGF-1 and IGF-1R transmits a signal through a number of intracellular substrates, including the insulin receptor substrate (IRS) and the Src homology collagen (Shc) proteins, which activate two major intracellular signaling pathways: the phosphatidylinositol 3-kinase (PI3K)/AKT and mitogen-activated protein kinase (MAPK) pathways, specifically the extracellular signal-regulated kinase (ERK) pathways. The PI3K/AKT kinase pathway regulates a variety of cellular processes, including cell proliferation and apoptosis. IGF1/IGF-1R signaling also promotes cell differentiation and proliferation via the Ras/MAPK pathway. Moreover, upon IGF-1R activation of the IRS and Shc adaptor proteins, Shc stimulates Raf through the GTPase Ras to activate the MAPKs ERK1 and ERK2, phosphorylate and several other proteins, and to stimulate cell proliferation. The IGF-1 signaling pathway is required for certain viral effects in oncogenic progression and may be induced as an effect of viral infection. The mechanisms of IGF signaling in animal viral infections need to be clarified, mainly because they are involved in multifactorial signaling pathways. The aim of this review is to summarize the current data obtained from virological studies and to increase our understanding of the complex role of the IGF-1 signaling axis in animal virus infections.
Collapse
|