1
|
Raballah E, Anyona SB, Osata SW, Wasena SA, Onyango C, Hurwitz I, Cheng Q, Seidenberg PD, McMahon BH, Ouma C, Ong'echa JM, Schneider KA, Perkins DJ. Impact of age, HIV1, sickle-cell genotypes, and interferon-gamma gene upstream variants on malaria disease outcomes in a longitudinal pediatric cohort. Sci Rep 2025; 15:13043. [PMID: 40234522 PMCID: PMC12000329 DOI: 10.1038/s41598-025-97267-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 04/17/2025] Open
Abstract
This prospective cohort study explored the association between two upstream IFN-γ variants (rs2069709: G > T and rs2069705: A > G) and hazard factors for malaria outcomes in a longitudinal cohort of children (n = 941, 3-36 mos.), followed for three years. The impact of age, sex, previous malaria exposure, HIV1 infection, and sickle-cell genotypes (HbAA, HbAS, and HbSS) was also investigated. Reduced malaria episodes were associated with older age at enrollment [HR = 0.957 (95% CI = 0.953-0.961) per month, P < 2.2e-16], HIV1 infection [0.687 (0.545-0.866), P = 0.001], being female [0.910 (0.859-0.964), P = 0.040], and HbAS [0.823 (0.754-0.898), P = 0.005]. The GA/TA diplotype [0.376 (0.230-0.614), P = 0.002] also reduced the hazard of malaria, while TA haplotype increased susceptibility [1.749 (1.159-2.640), P = 0.029]. Factors protecting against the development of SMA [Hemoglobin (Hb < 6.0 g/dL)] included older age [0.927 (0.913-0.942) per month, P < 2.2e-16], previous malaria episodes [0.576 (0.542-0.614, P = 9.5e-32)], HbAS [0.553 (0.400-0.766), P = 0.015]. The rs2069705AG genotype increased the hazard of SMA [1.697 (1.002-2.875), P = 0.042]. Reduced hazard of mortality was observed for older children [0.898 (0.857-0.941), P < 2.2e-16], while a higher hazard was present in HIV-infected children [12.475 (6.380-24.392), P < 2.2e-16], and in those with HbSS [6.341 (1.944-20.686), P = 0.007]. The GG haplotype increased the mortality hazard [1.817 (0.936-3.527), P = 0.078]. The results here highlight critical factors influencing the hazard of malaria, SMA, and mortality.
Collapse
Affiliation(s)
- Evans Raballah
- Department of Medical Laboratory Sciences, School of Public Health Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, Kakamega, Kenya
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
| | - Samuel B Anyona
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Medical Biochemistry, School of Medicine, Maseno University, Maseno, Kenya
| | - Shamim W Osata
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Sharley A Wasena
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Clinton Onyango
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Ivy Hurwitz
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Qiuying Cheng
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Philip D Seidenberg
- Department of Emergency Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Benjamin H McMahon
- Theoretical Division, Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Collins Ouma
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - John M Ong'echa
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Kristan A Schneider
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA
| | - Douglas J Perkins
- Global Health Programs, University of New Mexico, Kisumu and Siaya, Kenya.
- Department of Internal Medicine, Center for Global Health, University of New Mexico, 913 Camino de Salud, IDTC 3140, Albuquerque, NM, 87131, USA.
| |
Collapse
|
2
|
Koka PS, Ramdass B. ISG15-LFA1 interactions in latent HIV clearance: mechanistic implications in designing antiviral therapies. Front Cell Dev Biol 2024; 12:1497964. [PMID: 39810915 PMCID: PMC11729345 DOI: 10.3389/fcell.2024.1497964] [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: 09/18/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Interferon types-I/II (IFN-αβ/γ) secretions are well-established antiviral host defenses. The human immunodeficiency virus (HIV) particles are known to prevail following targeted cellular interferon secretion. CD4+ T-lymphocytes are the primary receptor targets for HIV entry, but the virus has been observed to hide (be latent) successfully in these cells through an alternate entry route via interactions with LFA1. HIV facilitates its post-entry latency-driven mode of hiding through these interactions to displace or inhibit ISG15 by forming the HIV1-LFA1 complex in lieu of ISG15-LFA1, which would at least transiently halt and bypass type-I IFN secretion. This could explain why the elimination of HIV from cellular hideouts is difficult. Hence, HIV clearance needs to be addressed to reverse its latency in LFA1+ T-lymphocytes and CD34+/CD133+ early progenitor stem cells. In the context of hematopoietic or endothelial stem-progenitor cells (HSPC/ESPC), we discuss the potential role of LFA1 in HIV permissiveness and latency in LFA1-CD34+/CD133+ versus LFA1+CD34+/CD133+ HSPCs/ESPCs. In HIV latency, the viral particles may remain engaged on the naïve-resting cells' LFA1, which are then unable to accommodate the ISG15 molecules owing to conformational changes induced upon occupation by the virus at the ISG15-LFA1 binding or interaction sites through halting of the subsequent downstream type-II IFN secretion. Viral binding to LFA1, including its transfer through activated-naïve cell-cell contacts may be a key step that needs to be addressed to prevent "transient or partial" virus-induced shutdown of type-I IFN secretion. This process allows an alternate viral entry and hideout site via LFA1. The subsequent administration of recombinant ISG15 may ensure sufficient type I/II IFN release to promote, enhance, or sustain the innate immune responses. Thus, combination antiviral therapies could potentially include exogenous ISG15 to maintain or sustain biologically and clinically relevant ISG15-LFA1 interactions. In addition to alternating with co-challenges of PKC-pro-LRA-drug modulators, this is administered post (antiretroviral therapy) and continued with periodic ART until permanent elimination of viral resurgence and latency is achieved in patients with HIV/AIDS. This triple-combination drug regimen is expected to pave the path for systemic virus clearance in vivo.
Collapse
Affiliation(s)
- Prasad S. Koka
- Biomedical Research Institute of Southern California, Oceanside, CA, United States
| | | |
Collapse
|
3
|
Chathuranga K, Shin Y, Uddin MB, Paek J, Chathuranga WAG, Seong Y, Bai L, Kim H, Shin JH, Chang YH, Lee JS. The novel immunobiotic Clostridium butyricum S-45-5 displays broad-spectrum antiviral activity in vitro and in vivo by inducing immune modulation. Front Immunol 2023; 14:1242183. [PMID: 37881429 PMCID: PMC10595006 DOI: 10.3389/fimmu.2023.1242183] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Clostridium butyricum is known as a probiotic butyric acid bacterium that can improve the intestinal environment. In this study, we isolated a new strain of C. butyricum from infant feces and evaluated its physiological characteristics and antiviral efficacy by modulating the innate immune responses in vitro and in vivo. The isolated C. butyricum S-45-5 showed typical characteristics of C. butyricum including bile acid resistance, antibacterial ability, and growth promotion of various lactic acid bacteria. As an antiviral effect, C. butyricum S-45-5 markedly reduced the replication of influenza A virus (PR8), Newcastle Disease Virus (NDV), and Herpes Simplex Virus (HSV) in RAW264.7 cells in vitro. This suppression can be explained by the induction of antiviral state in cells by the induction of antiviral, IFN-related genes and secretion of IFNs and pro-inflammatory cytokines. In vivo, oral administration of C. butyricum S-45-5 exhibited prophylactic effects on BALB/c mice against fatal doses of highly pathogenic mouse-adapted influenza A subtypes (H1N1, H3N2, and H9N2). Before challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed increased levels of IFN-β, IFN-γ, IL-6, and IL-12 in serum, the small intestine, and bronchoalveolar lavage fluid (BALF), which correlated with observed prophylactic effects. Interestingly, after challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed reduced levels of pro-inflammatory cytokines and relatively higher levels of anti-inflammatory cytokines at day 7 post-infection. Taken together, these findings suggest that C. butyricum S-45-5 plays an antiviral role in vitro and in vivo by inducing an antiviral state and affects immune modulation to alleviate local and systemic inflammatory responses caused by influenza virus infection. Our study provides the beneficial effects of the new C. butyricum S-45-5 with antiviral effects as a probiotic.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yeseul Shin
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medicine, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jayoung Paek
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | - Yebin Seong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Lu Bai
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hongik Kim
- Research and Development Division, Vitabio, Inc., Daejeon, Republic of Korea
| | - Jeong Hwan Shin
- Department of Laboratory Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Young-Hyo Chang
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
4
|
Fleitas-Salazar N, Lamazares E, Pedroso-Santana S, Kappes T, Pérez-Alonso A, Hidalgo Á, Altamirano C, Sánchez O, Fernández K, Toledo JR. Long-term release of bioactive interferon-alpha from PLGA-chitosan microparticles: in vitro and in vivo studies. BIOMATERIALS ADVANCES 2022; 143:213167. [PMID: 36356469 DOI: 10.1016/j.bioadv.2022.213167] [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: 05/16/2022] [Revised: 08/12/2022] [Accepted: 10/18/2022] [Indexed: 06/16/2023]
Abstract
Effective cytokine treatments often require high- and multiple-dose due to the short half-life of these molecules. Here, porcine interferon-alpha (IFNα) is encapsulated in PLGA-chitosan microparticles (IFNα-MPs) to accomplish both slow drug release and drug protection from degradation. A procedure that combines emulsion and spray-drying techniques yielded almost spherical microspheres with an average diameter of 3.00 ± 1.50 μm. SEM, Microtrac, and Z-potential analyses of three IFNα-MP batches showed similar results, indicating the process is reproducible. These studies supported molecular evidence obtained in FTIR analysis, which indicated a compact structure of IFNα-MPs. Consistently, IFNα release kinetics assessed in vitro followed a zero-order behavior typical of sustained release from a polymeric matrix. This study showed that IFNα-MPs released bioactive molecules for at least 15 days, achieving IFNα protection. In addition, pigs treated with IFNα-MPs exhibited overexpression of IFNα-stimulated genes 16 days after treatment. Instead, the expression levels of these genes decreased after day 4th in pigs treated with non-encapsulated IFNα. In vitro and in vivo experiments demonstrated that the formulation improved the prophylactic and therapeutic potential of IFNα, accomplishing molecule protection and long-term release for at least two weeks. The procedure used to obtain IFNα-MPs is reproducible, scalable, and suitable for encapsulating other drugs.
Collapse
Affiliation(s)
- Noralvis Fleitas-Salazar
- Biotecnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Emilio Lamazares
- Biotecnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Seidy Pedroso-Santana
- Biotecnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Tomás Kappes
- Laboratory of Biomaterials, Departamento de Ingeniería Química, Facultad de Ingeniería, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Alain Pérez-Alonso
- Departamento de Electrónica e Informática, Universidad Técnica Federico Santa María, Concepción CP. 4030000, Chile
| | - Ángela Hidalgo
- Biotecnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Claudia Altamirano
- Laboratorio de Cultivos Celulares, Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, 2362803 Valparaíso, Chile
| | - Oliberto Sánchez
- Recombinant Biopharmaceuticals Laboratory, Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Katherina Fernández
- Laboratory of Biomaterials, Departamento de Ingeniería Química, Facultad de Ingeniería, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile
| | - Jorge R Toledo
- Biotecnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Barrio Universitario s/n, Concepción CP. 4030000, Chile.
| |
Collapse
|
5
|
Song Q, Zhao X, Cao C, Duan M, Shao C, Jiang S, Zhou B, Zhou Y, Dong W, Yang Y, Wang X, Song H. Research advances on interferon (IFN) response during BVDV infection. Res Vet Sci 2022; 149:151-158. [DOI: 10.1016/j.rvsc.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/21/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
|
6
|
Kooti W, Esmaeili Gouvarchin Ghaleh H, Farzanehpour M, Dorostkar R, Jalali Kondori B, Bolandian M. Oncolytic Viruses and Cancer, Do You Know the Main Mechanism? Front Oncol 2022; 11:761015. [PMID: 35004284 PMCID: PMC8728693 DOI: 10.3389/fonc.2021.761015] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 11/22/2021] [Indexed: 12/28/2022] Open
Abstract
The global rate of cancer has increased in recent years, and cancer is still a threat to human health. Recent developments in cancer treatment have yielded the understanding that viruses have a high potential in cancer treatment. Using oncolytic viruses (OVs) is a promising approach in the treatment of malignant tumors. OVs can achieve their targeted treatment effects through selective cell death and induction of specific antitumor immunity. Targeting tumors and the mechanism for killing cancer cells are among the critical roles of OVs. Therefore, evaluating OVs and understanding their precise mechanisms of action can be beneficial in cancer therapy. This review study aimed to evaluate OVs and the mechanisms of their effects on cancer cells.
Collapse
Affiliation(s)
- Wesam Kooti
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Mahdieh Farzanehpour
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ruhollah Dorostkar
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Bahman Jalali Kondori
- Department of Anatomical Sciences, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.,Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoumeh Bolandian
- Applied Virology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Ye YZ, Dou YL, Hao JH, Zhou L, Lin AW, Wang SN, Deng JK, Lei M, Luo RP, Liao YN, Chen Y, Long YY, Chen BQ, Yang Z, Gan L, Nong GM, Yan WL, Yu H. Efficacy and safety of interferon α-2b spray for herpangina in children: A randomized, controlled trial. Int J Infect Dis 2021; 107:62-68. [PMID: 33878461 DOI: 10.1016/j.ijid.2021.04.049] [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: 11/30/2020] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES The treatment of acute herpangina is inconsistent. We aim to evaluate the effectiveness and safety of interferon α-2b spray versus Ribavirin for this disease. METHODS A randomized, controlled trial was conducted in eight hospitals in China between 2016 and 2018. 668 patients (1-7 years old) were randomized into an experimental group (treated with Interferon α-2b spray) or control group (received Ribavirin Aerosol). Body temperature returning to normal within 72 h and remaining so for 24 h was the primary outcome; release of oral herpes and adverse events were the secondary outcomes. RESULTS (1) The average age of onset was 2.5 years old. (2) After 72 h treatment, body temperature of 98.5% patients in experimental group and 94.3% in control group returned to normal and remained so for 24 h (P = 0.004). The differences were greater at 48 h treatment (95.2% vs. 85.9%, P < 0.001) and at 24 h (77.5% vs. 66.5%, P = 0.001). (3) The rate of improved oral herpes in the experimental group was higher than that in control group (46.7% vs.37.1%, P = 0.011). No adverse reaction occurred. CONCLUSIONS Local application of recombinant interferon α-2b spray showed better efficacy for acute herpangina in children. It was safe for use.
Collapse
Affiliation(s)
- Ying-Zi Ye
- Department of Infectious Diseases, Children's Hospital of Fudan University, Shanghai, China
| | - Ya-Lan Dou
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China
| | - Jian-Hua Hao
- Department of Internal Medicine, Children's Hospital of Kaifeng City, Kaifeng, China
| | - Li Zhou
- Department of Internal Medicine, Children's Hospital of Kaifeng City, Kaifeng, China
| | - Ai-Wei Lin
- Department of Infectious Diseases, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Shao-Ning Wang
- Department of Infectious Diseases, Qilu Children's Hospital of Shandong University, Jinan, China
| | - Ji-Kui Deng
- Department of Infectious Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Min Lei
- Department of Infectious Diseases, Shenzhen Children's Hospital, Shenzhen, China
| | - Ru-Ping Luo
- Department of Infectious Diseases, Hunan Children's Hospital, Changsha, China
| | - Yi-Nan Liao
- Department of Infectious Diseases, Hunan Children's Hospital, Changsha, China
| | - Yan Chen
- Department of Pediatrics Internal Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuan-Yuan Long
- Department of Pediatrics Internal Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Bi-Quan Chen
- Department of Infectious Diseases, Anhui Provincial Children'S Hospital, Hefei, China
| | - Zhi Yang
- Department of Infectious Diseases, Anhui Provincial Children'S Hospital, Hefei, China
| | - Lu Gan
- Department of Pediatrics, Changhai Hospital, Shanghai, China
| | - Guang-Min Nong
- Department of Pediatrics, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei-Li Yan
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, Shanghai, China.
| | - Hui Yu
- Department of Infectious Diseases, Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Lo CW, Takeshima SN, Okada K, Saitou E, Fujita T, Matsumoto Y, Wada S, Inoko H, Aida Y. Association of Bovine Leukemia Virus-Induced Lymphoma with BoLA-DRB3 Polymorphisms at DNA, Amino Acid, and Binding Pocket Property Levels. Pathogens 2021; 10:pathogens10040437. [PMID: 33917549 PMCID: PMC8067502 DOI: 10.3390/pathogens10040437] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/02/2021] [Accepted: 04/02/2021] [Indexed: 01/01/2023] Open
Abstract
Bovine leukemia virus (BLV) causes enzootic bovine leucosis, a malignant B-cell lymphoma in cattle. The DNA sequence polymorphisms of bovine leukocyte antigen (BoLA)-DRB3 have exhibited a correlation with BLV-induced lymphoma in Holstein cows. However, the association may vary between different cattle breeds. Furthermore, little is known about the relationship between BLV-induced lymphoma and DRB3 at the amino acid and structural diversity levels. Here, we comprehensively analyzed the correlation between BLV-induced lymphoma and DRB3 at DNA, amino acid, and binding pocket property levels, using 106 BLV-infected asymptomatic and 227 BLV-induced lymphoma Japanese black cattle samples. DRB3*011:01 was identified as a resistance allele, whereas DRB3*005:02 and DRB3*016:01 were susceptibility alleles. Amino acid association studies showed that positions 9, 11, 13, 26, 30, 47, 57, 70, 71, 74, 78, and 86 were associated with lymphoma susceptibility. Structure and electrostatic charge modeling further indicated that binding pocket 9 of resistance DRB3 was positively charged. In contrast, alleles susceptible to lymphoma were neutrally charged. Altogether, this is the first association study of BoLA-DRB3 polymorphisms with BLV-induced lymphoma in Japanese black cattle. In addition, our results further contribute to understanding the mechanisms regarding how BoLA-DRB3 polymorphisms mediate susceptibility to BLV-induced lymphoma.
Collapse
Affiliation(s)
- Chieh-Wen Lo
- Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (C.-W.L.); (Y.M.)
- Laboratory of Global Infectious Diseases Control Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Shin-nosuke Takeshima
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan;
- Department of Food and Nutrition, Jumonji University, Niiza, Saitama 352-8510, Japan
| | - Kosuke Okada
- Iwate University, 7-360 Mukai-shinden Ukai, Takizawa, Iwate 020-0667, Japan;
| | - Etsuko Saitou
- Hyogo Prefectural Awaji Meat Inspection Center, 49-18 Shitoorinagata, Minamiawaji, Hyogo 656-0152, Japan;
| | - Tatsuo Fujita
- Livestock Research Institute of Oita Prefectural Agriculture, Forestry and Fisheries, Research Center, Kuju, Taketa, Oita 878-0201, Japan;
| | - Yasunobu Matsumoto
- Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (C.-W.L.); (Y.M.)
- Laboratory of Global Infectious Diseases Control Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Satoshi Wada
- Photonics Control Technology Team, RIKEN Center for Advanced Photonics, Wako 351-0198, Japan;
| | - Hidetoshi Inoko
- Genome Analysis Division, GenoDive Pharma Inc., 4-14-1 Nakamachi, Atsugi-shi, Kanagawa 243-0018, Japan;
| | - Yoko Aida
- Laboratory of Global Animal Resource Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan; (C.-W.L.); (Y.M.)
- Laboratory of Global Infectious Diseases Control Science, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
- Viral Infectious Diseases Unit, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan;
- Benno Laboratory, Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
- Correspondence:
| |
Collapse
|
9
|
Loganathan T, Ramachandran S, Shankaran P, Nagarajan D, Mohan S S. Host transcriptome-guided drug repurposing for COVID-19 treatment: a meta-analysis based approach. PeerJ 2020; 8:e9357. [PMID: 32566414 PMCID: PMC7293190 DOI: 10.7717/peerj.9357] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/24/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been declared a pandemic by the World Health Organization, and the identification of effective therapeutic strategy is a need of the hour to combat SARS-CoV-2 infection. In this scenario, the drug repurposing approach is widely used for the rapid identification of potential drugs against SARS-CoV-2, considering viral and host factors. METHODS We adopted a host transcriptome-based drug repurposing strategy utilizing the publicly available high throughput gene expression data on SARS-CoV-2 and other respiratory infection viruses. Based on the consistency in expression status of host factors in different cell types and previous evidence reported in the literature, pro-viral factors of SARS-CoV-2 identified and subject to drug repurposing analysis based on DrugBank and Connectivity Map (CMap) using the web tool, CLUE. RESULTS The upregulated pro-viral factors such as TYMP, PTGS2, C1S, CFB, IFI44, XAF1, CXCL2, and CXCL3 were identified in early infection models of SARS-CoV-2. By further analysis of the drug-perturbed expression profiles in the connectivity map, 27 drugs that can reverse the expression of pro-viral factors were identified, and importantly, twelve of them reported to have anti-viral activity. The direct inhibition of the PTGS2 gene product can be considered as another therapeutic strategy for SARS-CoV-2 infection and could suggest six approved PTGS2 inhibitor drugs for the treatment of COVID-19. The computational study could propose candidate repurposable drugs against COVID-19, and further experimental studies are required for validation.
Collapse
Affiliation(s)
- Tamizhini Loganathan
- School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Srimathy Ramachandran
- School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Prakash Shankaran
- School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Devipriya Nagarajan
- School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Suma Mohan S
- School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| |
Collapse
|
10
|
Newcastle Disease Virus (NDV) Oncolytic Activity in Human Glioma Tumors Is Dependent on CDKN2A-Type I IFN Gene Cluster Codeletion. Cells 2020; 9:cells9061405. [PMID: 32516884 PMCID: PMC7349162 DOI: 10.3390/cells9061405] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor in adults with a median overall survival of 15 months. Tumor recurrence and poor prognosis are related to cancer stem cells (CSCs), which drive resistance to therapies. A common characteristic in GBM is CDKN2A gene loss, located close to the cluster of type I IFN genes at Ch9p21. Newcastle disease virus (NDV) is an avian paramyxovirus with oncolytic and immunostimulatory properties that has been proposed for the treatment of GBM. We have analyzed the CDKN2A-IFN I gene cluster in 1018 glioma tumors and evaluated the NDV oncolytic effect in six GBM CSCs ex vivo and in a mouse model. Our results indicate that more than 50% of GBM patients have some IFN deletion. Moreover, GBM susceptibility to NDV is dependent on the loss of the type I IFN. Infection of GBM with an NDV-expressing influenza virus NS1 protein can overcome the resistance to oncolysis by NDV of type I-competent cells. These results highlight the potential of using NDV vectors in antitumor therapies.
Collapse
|
11
|
O’Connell P, Amalfitano A, Aldhamen YA. SLAM Family Receptor Signaling in Viral Infections: HIV and Beyond. Vaccines (Basel) 2019; 7:E184. [PMID: 31744090 PMCID: PMC6963180 DOI: 10.3390/vaccines7040184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023] Open
Abstract
The signaling lymphocytic activation molecule (SLAM) family of receptors are expressed on the majority of immune cells. These receptors often serve as self-ligands, and play important roles in cellular communication and adhesion, thus modulating immune responses. SLAM family receptor signaling is differentially regulated in various immune cell types, with responses generally being determined by the presence or absence of two SLAM family adaptor proteins-Ewing's sarcoma-associated transcript 2 (EAT-2) and SLAM-associated adaptor protein (SAP). In addition to serving as direct regulators of the immune system, certain SLAM family members have also been identified as direct targets for specific microbes and viruses. Here, we will discuss the known roles for these receptors in the setting of viral infection, with special emphasis placed on HIV infection. Because HIV causes such complex dysregulation of the immune system, studies of the roles for SLAM family receptors in this context are particularly exciting.
Collapse
Affiliation(s)
- Patrick O’Connell
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
| | - Andrea Amalfitano
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
- Department of Pediatrics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Yasser A. Aldhamen
- Department of Microbiology and Molecular Genetics, College of Osteopathic Medicine, Michigan State University, East Lansing, MI 48824, USA, (A.A.)
| |
Collapse
|
12
|
Zhao Y, Ran Z, Jiang Q, Hu N, Yu B, Zhu L, Shen L, Zhang S, Chen L, Chen H, Jiang J, Chen D. Vitamin D Alleviates Rotavirus Infection through a Microrna-155-5p Mediated Regulation of the TBK1/IRF3 Signaling Pathway In Vivo and In Vitro. Int J Mol Sci 2019; 20:ijms20143562. [PMID: 31330869 PMCID: PMC6678911 DOI: 10.3390/ijms20143562] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022] Open
Abstract
(1) Background: Vitamin D (VD) plays a vital role in anti-viral innate immunity. However, the role of VD in anti-rotavirus and its mechanism is still unclear. The present study was performed to investigate whether VD alleviates rotavirus (RV) infection through a microRNA-155-5p (miR-155-5p)-mediated regulation of TANK-binding kinase 1 (TBK1)/interferon regulatory factors 3 (IRF3) signaling pathway in vivo and in vitro. (2) Methods: The efficacy of VD treatment was evaluated in DLY pig and IPEC-J2. Dual-luciferase reporter activity assay was performed to verify the role of miR-155-5p in 1α,25-dihydroxy-VD3 (1,25D3) mediating the regulation of the TBK1/IRF3 signaling pathway. (3) Results: A 5000 IU·kg–1 dietary VD3 supplementation attenuated RV-induced the decrease of the villus height and crypt depth (p < 0.05), and up-regulated TBK1, IRF3, and IFN-β mRNA expressions in the jejunum (p < 0.05). Incubation with 1,25D3 significantly decreased the RV mRNA expression and the RV antigen concentration, and increased the TBK1 mRNA and protein levels, and the phosphoprotein IRF3 (p-IRF3) level (p < 0.05). The expression of miR-155-5p was up-regulated in response to an RV infection in vivo and in vitro (p < 0.05). 1,25D3 significantly repressed the up-regulation of miR-155-5p in vivo and in vitro (p < 0.05). Overexpression of miR-155-5p remarkably suppressed the mRNA and protein levels of TBK1 and p-IRF3 (p < 0.01), while the inhibition of miR-155-5p had an opposite effect. Luciferase activity assays confirmed that miR-155-5p regulated RV replication by directly targeting TBK1, and miR-155-5p suppressed the TBK1 protein level (p < 0.01). (4) Conclusions: These results indicate that miR-155-5p is involved in 1,25D3 mediating the regulation of the TBK1/IRF3 signaling pathway by directly targeting TBK1.
Collapse
Affiliation(s)
- Ye Zhao
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China.
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China.
| | - Zhiming Ran
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Qin Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Ningming Hu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Lei Chen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
| | - Hong Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Jun Jiang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Ya'an 625014, China.
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
13
|
Ojha CR, Rodriguez M, Karuppan MKM, Lapierre J, Kashanchi F, El-Hage N. Toll-like receptor 3 regulates Zika virus infection and associated host inflammatory response in primary human astrocytes. PLoS One 2019; 14:e0208543. [PMID: 30735502 PMCID: PMC6368285 DOI: 10.1371/journal.pone.0208543] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/19/2018] [Indexed: 12/25/2022] Open
Abstract
The connection between Zika virus (ZIKV) and neurodevelopmental defects is widely recognized, although the mechanisms underlying the infectivity and pathology in primary human glial cells are poorly understood. Here we show that three isolated strains of ZIKV, an African strain MR766 (Uganda) and two closely related Asian strains R103451 (Honduras) and PRVABC59 (Puerto Rico) productively infect primary human astrocytes, although Asian strains showed a higher infectivity rate and increased cell death when compared to the African strain. Inhibition of AXL receptor significantly attenuated viral entry of MR766 and PRVABC59 and to a lesser extend R103451, suggesting an important role of TAM receptors in ZIKV cell entry, irrespective of lineage. Infection by PRVABC59 elicited the highest release of inflammatory molecules, with a 8-fold increase in the release of RANTES, 10-fold increase in secretion of IP-10 secretion and a 12-fold increase in IFN-β secretion when compared to un-infected human astrocytes. Minor changes in the release of several growth factors, endoplasmic reticulum (ER)-stress response factors and the transcription factor, NF-κB were detected with the Asian strains, while significant increases in FOXO6, MAPK10 and JNK were detected with the African strain. Activation of the autophagy pathway was evident with increased expression of the autophagy related proteins Beclin1, LC3B and p62/SQSTM1 with all three strains of ZIKV. Pharmacological inhibition of the autophagy pathway and genetic inhibition of the Beclin1 showed minimal effects on ZIKV replication. The expression of toll-like receptor 3 (TLR3) was significantly increased with all three strains of ZIKV; pharmacological and genetic inhibition of TLR3 caused a decrease in viral titers and in viral-induced inflammatory response in infected astrocytes. We conclude that TLR3 plays a vital role in both ZIKV replication and viral-induced inflammatory responses, irrespective of the strains, while the autophagy protein Beclin1 influences host inflammatory responses.
Collapse
Affiliation(s)
- Chet Raj Ojha
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Myosotys Rodriguez
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Mohan Kumar Muthu Karuppan
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Jessica Lapierre
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| | - Fatah Kashanchi
- National Center for Biodefense and Infectious Diseases, George Mason University, Manassas, Virginia, United States of America
| | - Nazira El-Hage
- Department of Immunology, Florida International University, Herbert Wertheim College of Medicine, Miami, Florida, United States of America
| |
Collapse
|
14
|
Sghaier I, Brochot E, Loueslati BY, Almawi WY. Hepatitis C virus protein interaction network for HCV clearance and association of DAA to HCC occurrence via data mining approach: A systematic review and critical analysis. Rev Med Virol 2019; 29:e2033. [PMID: 30614131 DOI: 10.1002/rmv.2033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/23/2022]
Abstract
HCV has been associated with a pro-inflammatory state, which predisposes to hepatocellular carcinoma (HCC). However, the different molecular mechanisms underlying the effect of HCV infection on HCC progression remain unclear. Although HCV infection illustrates the potential role of host genetics in the outcome of infectious diseases, there is no clear overview of some single nucleotide polymorphisms (SNPs) influencing spontaneous or treatment-induced HCV eradication. We studied the possible role of HCV infection in the processes of HCC initiation and performed a systematic analysis using data mining approaches to identify host polymorphisms associated with treatment response and HCC development using topological analysis of protein-proteins interactions (PPI) networks. On the basis of our analysis performed, we identified key hub proteins related to HCV-treatment response infection and to HCC development. Host genetic polymorphisms, such as inosine triphosphatase (ITPA), interferon, lambda 3 (IFNL3), Q5 interferon, lambda 4 (IFNL4), toll-like receptors (TLRs) and interferon-stimulated gene 15 (ISG-15), were identified as key genes for treatment prediction and HCC evolution. By comparing unique genes for HCV-treatment response and genes particular to HCV-HCC development, we found a common PPI network that may participate in more extensive signalling processes during anti-HCV treatment, which can play important roles in modulating the immune response to the occurrence of HCC. Data mining is an effective tool for identifying potential regulatory pathways involved in treatment response and HCC development. Our study may contribute to a better understanding of HCV immunopathogenesis and highlights the complex role of host genetics in HCV clearance.
Collapse
Affiliation(s)
- Ikram Sghaier
- University of Tunis El Manar, Biology department, Tunish, Tunisia
| | - Etienne Brochot
- Department of Virology, Amiens University Medical Centre, Amiens, France.,Virology Research Unit, EA 4294, Jules Verne University of Picardie, Amiens, France
| | - Besma Y Loueslati
- Faculty of Sciences of Tunis, department of Biology, Laboratory of Mycology, Pathologies and Biomarkers, Tunis, Tunisia
| | - Wassim Y Almawi
- University of Tunis El Manar, Biology department, Tunish, Tunisia.,School of Medicine, Nazarbayev University, Astana, Kazakhstan
| |
Collapse
|
15
|
Li Y, Sun B, Esser S, Jessen H, Streeck H, Widera M, Yang R, Dittmer U, Sutter K. Expression Pattern of Individual IFNA Subtypes in Chronic HIV Infection. J Interferon Cytokine Res 2018; 37:541-549. [PMID: 29252127 DOI: 10.1089/jir.2017.0076] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Interferon-α (IFN-α) plays an important role in HIV pathogenesis. IFN-α consists of 13 individual IFN-α subtypes, which exhibit individual antiviral and immunomodulatory activities in HIV infection. Here, we determined the expression profiles of all IFN-α subtypes in treated and treatment-naive HIV+ patients and their impact on the induction of distinct HIV restriction factors. We collected blood samples of chronic HIV+ patients, which underwent antiretroviral therapy or were treatment-naive, and determined the individual expression levels of different IFN-α subtypes and HIV restriction factors. HIV infection transiently enhanced the expression of IFNA mRNA. The IFN-α response was dominated by the most abundantly expressed subtypes IFNA4, A5, A7, and A14 in all individuals. HIV infection affected the expression pattern of the IFN-α response, in particular for IFNA2 and IFNA16, which were elevated by chronic HIV infection. Elevated expression of HIV restriction factors was observed in chronically HIV-infected patients, which partly decreased during successful antiretroviral treatment. In vitro stimulation of peripheral blood mononuclear cells revealed that IFN-α6, -α14, and -α21 were most effective in inducing the expression of HIV restriction factors. These results indicate that HIV infection induces a specific expression pattern of IFN-α subtypes, which in turn induce the expression of various HIV restriction factors.
Collapse
Affiliation(s)
- Yanpeng Li
- 1 Wuhan Institute of Virology , Chinese Academy of Sciences, Wuhan, PR China
| | - Binlian Sun
- 1 Wuhan Institute of Virology , Chinese Academy of Sciences, Wuhan, PR China
| | - Stefan Esser
- 2 Clinic of Dermatology, University Hospital Essen, University of Duisburg-Essen , Essen, Germany
| | | | - Hendrik Streeck
- 4 Institute for HIV Research, University Hospital Essen, University of Duisburg-Essen , Essen, Germany
| | - Marek Widera
- 5 Institute for Virology, University Hospital Essen, University of Duisburg-Essen , Essen, Germany
| | - Rongge Yang
- 1 Wuhan Institute of Virology , Chinese Academy of Sciences, Wuhan, PR China
| | - Ulf Dittmer
- 5 Institute for Virology, University Hospital Essen, University of Duisburg-Essen , Essen, Germany
| | - Kathrin Sutter
- 5 Institute for Virology, University Hospital Essen, University of Duisburg-Essen , Essen, Germany
| |
Collapse
|
16
|
Gao L, Wang Y, Li Y, Dong Y, Yang A, Zhang J, Li F, Zhang R. Genome-wide expression profiling analysis to identify key genes in the anti-HIV mechanism of CD4 + and CD8 + T cells. J Med Virol 2018; 90:1199-1209. [PMID: 29508932 DOI: 10.1002/jmv.25071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/07/2018] [Indexed: 11/12/2022]
Abstract
Comprehensive bioinformatics analyses were performed to explore the key biomarkers in response to HIV infection of CD4+ and CD8+ T cells. The numbers of CD4+ and CD8+ T cells of HIV infected individuals were analyzed and the GEO database (GSE6740) was screened for differentially expressed genes (DEGs) in HIV infected CD4+ and CD8+ T cells. Gene Ontology enrichment, KEGG pathway analyses, and protein-protein interaction (PPI) network were performed to identify the key pathway and core proteins in anti-HIV virus process of CD4+ and CD8+ T cells. Finally, we analyzed the expressions of key proteins in HIV-infected T cells (GSE6740 dataset) and peripheral blood mononuclear cells(PBMCs) (GSE511 dataset). 1) CD4+ T cells counts and ratio of CD4+ /CD8+ T cells decreased while CD8+ T cells counts increased in HIV positive individuals; 2) 517 DEGs were found in HIV infected CD4+ and CD8+ T cells at acute and chronic stage with the criterial of P-value <0.05 and fold change (FC) ≥2; 3) In acute HIV infection, type 1 interferon (IFN-1) pathway might played a critical role in response to HIV infection of T cells. The main biological processes of the DEGs were response to virus and defense response to virus. At chronic stage, ISG15 protein, in conjunction with IFN-1 pathway might play key roles in anti-HIV responses of CD4+ T cells; and 4) The expression of ISG15 increased in both T cells and PBMCs after HIV infection. Gene expression profile of CD4+ and CD8+ T cells changed significantly in HIV infection, in which ISG15 gene may play a central role in activating the natural antiviral process of immune cells.
Collapse
Affiliation(s)
- Lijie Gao
- Medicine College of Yan'an University, Yan'an, Shaanxi, P. R. China
| | - Yunqi Wang
- Department of Urology, Yangling Demonstration Zone Hospital, Yangling, Shaanxi, P. R. China
| | - Yi Li
- Medicine College of Yan'an University, Yan'an, Shaanxi, P. R. China
| | - Ya Dong
- Medicine College of Yan'an University, Yan'an, Shaanxi, P. R. China
| | - Aimin Yang
- School of Public Health, Brown University, Providence, Rhode Island.,School of Public Health, Lanzhou University, Lanzhou, Gansu, P. R. China
| | - Jie Zhang
- School of Public Health, Brown University, Providence, Rhode Island
| | - Fengying Li
- Immunization Department of Center for Disease Control and Prevention of Xianyang, Xianyang, Shaanxi, P. R. China
| | - Rongqiang Zhang
- School of Public Health, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, P. R. China
| |
Collapse
|
17
|
Lin H, Huang L, Zhou J, Lin K, Wang H, Xue X, Xia C. Efficacy and safety of interferon-α2b spray in the treatment of hand, foot, and mouth disease: a multicenter, randomized, double-blind trial. Arch Virol 2016; 161:3073-80. [PMID: 27518403 DOI: 10.1007/s00705-016-3012-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 08/07/2016] [Indexed: 12/22/2022]
Abstract
Hand, foot, and mouth disease (HFMD) is a common infectious enterovirus disease, occurring mostly in infants and children younger than 7 years with potentially fatal complications. Therefore, we evaluated the clinical efficacy and safety of recombinant human interferon (IFN)-α2b spray for treating mild HFMD in 400 patients in a randomized, open, controlled clinical trial. The patients were randomized to the IFN-α2b spray and placebo groups, and their temperature, skin rash, oral lesions, and appetite were monitored, while pathogen levels and safety were evaluated with a 7-day follow-up. The mean age of the patients was 20.1 ± 10.2 months. The median duration of fever, oral ulcers or vesicles (or both), and skin rash in addition to median time to regain appetite in the IFN-α2b spray group were shorter than they were in the placebo group. The number of virus-positive cases differed statistically between the two groups for the three follow-up detections. Additionally, the incidences of adverse events (AEs) and severe AEs (SAEs) were not significantly different between the two groups, and the SAEs were evidently unrelated to the IFN-α2b spray or placebo. Therefore, the IFN-α2b spray is suitable for topical treatment of HFMD, and it rapidly relieved fever, promoted oral lesions and subsidence of rash, enhanced appetite, promoted disease recovery, and was safe for application.
Collapse
Affiliation(s)
- Hailong Lin
- Department of Pediatric Infection, The Second Affiliated Hospital of Wenzhou Medical University, 109th Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Leting Huang
- Department of Pediatric Infection, The Second Affiliated Hospital of Wenzhou Medical University, 109th Xueyuanxi Road, Wenzhou, Zhejiang, China
| | - Jian Zhou
- Department of Pediatrics, The first people's Hospital of Yongkang, Jinhua, China
| | - Kaichun Lin
- Department of Pediatrics, The first people's Hospital of Yongkang, Jinhua, China
| | - Hongjiao Wang
- Department of Pediatrics, The Affiliated Children's Hospital of Zhejiang University Medical College, Hangzhou, China
| | - Xia Xue
- Department of Pediatrics, The Affiliated Children's Hospital of Zhejiang University Medical College, Hangzhou, China
| | - Chan Xia
- Department of Pediatric Infection, The Second Affiliated Hospital of Wenzhou Medical University, 109th Xueyuanxi Road, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Kim JH, Weeratunga P, Kim MS, Nikapitiya C, Lee BH, Uddin MB, Kim TH, Yoon JE, Park C, Ma JY, Kim H, Lee JS. Inhibitory effects of an aqueous extract from Cortex Phellodendri on the growth and replication of broad-spectrum of viruses in vitro and in vivo. Altern Ther Health Med 2016; 16:265. [PMID: 27484768 PMCID: PMC4970287 DOI: 10.1186/s12906-016-1206-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/14/2016] [Indexed: 11/19/2022]
Abstract
Background Cortex Phellodendri (C. Phellodendri), the dried trunk bark of Phellodendron amurense Ruprecht, has been known as a traditional herbal medicine, showing several bioactivities. However, antiviral activity of C. Phellodendri aqueous extract (CP) not reported in detail, particularly aiming the prophylactic effectiveness. Methods In vitro CP antiviral activity evaluated against Influenza A virus (PR8), Vesicular Stomatitis Virus (VSV), Newcastle Disease Virus (NDV), Herpes Simplex Virus (HSV), Coxsackie Virus (H3-GFP) and Enterovirus-71 (EV-71) infection on immune (RAW264.7) and epithelial (HEK293T/HeLa) cells. Such antiviral effects were explained by the induction of antiviral state which was determined by phosphorylation of signal molecules, secretion of IFNs and cytokines, and cellular antiviral mRNA expression. Furthermore, Compounds present in the aqueous fractions confirmed by HPLC analysis and evaluated their anti-viral activities. Additionally, in vivo protective effect of CP against divergent influenza A subtypes was determined in a BALB/c mouse infection model. Results An effective dose of CP significantly reduced the virus replication both in immune and epithelial cells. Mechanically, CP induced mRNA expression of anti-viral genes and cytokine secretion in both RAW264.7 and HEK293T cells. Furthermore, the main compound identified was berberine, and shows promising antiviral properties similar to CP. Finally, BALB/c mice treated with CP displayed higher protection levels against lethal doses of highly pathogenic influenza A subtypes (H1N1, H5N2, H7N3 and H9N2). Conclusion CP including berberine play an immunomodulatory role with broad spectrum antiviral activity, due to induction of antiviral state via type I IFN stimulation mechanism. Consequently, C. Phellodendri could be a potential source for promising natural antivirals or to design other antiviral agents for animal and humans. Electronic supplementary material The online version of this article (doi:10.1186/s12906-016-1206-x) contains supplementary material, which is available to authorized users.
Collapse
|
19
|
Weeratunga P, Uddin MB, Kim MS, Lee BH, Kim TH, Yoon JE, Ma JY, Kim H, Lee JS. Interferon-mediated antiviral activities of Angelica tenuissima Nakai and its active components. J Microbiol 2016; 54:57-70. [PMID: 26727903 PMCID: PMC7091376 DOI: 10.1007/s12275-016-5555-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/03/2015] [Accepted: 12/03/2015] [Indexed: 01/07/2023]
Abstract
Angelica tenuissima Nakai is a widely used commodity in traditional medicine. Nevertheless, no study has been conducted on the antiviral and immune-modulatory properties of an aqueous extract of Angelica tenuissima Nakai. In the present study, we evaluated the antiviral activities and the mechanism of action of an aqueous extract of Angelica tenuissima Nakai both in vitro and in vivo. In vitro, an effective dose of Angelica tenuissima Nakai markedly inhibited the replication of Influenza A virus (PR8), Vesicular stomatitis virus (VSV), Herpes simplex virus (HSV), Coxsackie virus, and Enterovirus (EV-71) on epithelial (HEK293T/HeLa) and immune (RAW264.7) cells. Such inhibition can be described by the induction of the antiviral state in cells by antiviral, IFNrelated gene induction and secretion of IFNs and pro-inflammatory cytokines. In vivo, Angelica tenuissima Nakai treated BALB/c mice displayed higher survivability and lower lung viral titers when challenged with lethal doses of highly pathogenic influenza A subtypes (H1N1, H5N2, H7N3, and H9N2). We also found that Angelica tenuissima Nakai can induce the secretion of IL-6, IFN-λ, and local IgA in bronchoalveolar lavage fluid (BALF) of Angelica tenuissima Nakai treated mice, which correlating with the observed prophylactic effects. In HPLC analysis, we found the presence of several compounds in the aqueous fraction and among them; we evaluated antiviral properties of ferulic acid. Therefore, an extract of Angelica tenuissima Nakai and its components, including ferulic acid, play roles as immunomodulators and may be potential candidates for novel anti-viral/anti-influenza agents.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, 305-764, Republic of Korea
- Faculty of Veterinary & Animal Science, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Myun Soo Kim
- Vitabio Corporation, Daejeon, 305-764, Republic of Korea
| | - Byeong-Hoon Lee
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Tae-Hwan Kim
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, 305-764, Republic of Korea
| | - Ji-Eun Yoon
- Foot and Mouth Disease Division, Animal Quarantine and Inspection Agency, Anyang, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Daejeon, 305-764, Republic of Korea
| | - Hongik Kim
- Vitabio Corporation, Daejeon, 305-764, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine (BK21 Plus Program), Chungnam National University, Daejeon, 305-764, Republic of Korea.
| |
Collapse
|
20
|
Yang F, Pan Y, Chen Y, Tan S, Jin M, Wu Z, Huang J. Expression and purification of Canis interferon α in Escherichia coli using different tags. Protein Expr Purif 2015. [DOI: 10.1016/j.pep.2015.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
21
|
ISG15 expression correlates with HIV-1 viral load and with factors regulating T cell response. Immunobiology 2015; 221:282-90. [PMID: 26563749 DOI: 10.1016/j.imbio.2015.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/25/2015] [Indexed: 01/24/2023]
Abstract
Given the multifactorial nature of action of type I interferon (IFN) in HIV-1 infection and the need to firmly establish the action of key components of IFN pathways, we compared the IFN stimulated gene (ISG)15 expression with that of other well-characterized ISGs, evaluating its relationship with immunosuppressive factors regulating T-cell response in HIV-1 patients. PBMC from 225 subjects were included: healthy donors (n=30), naïve (n=93) and HAART treated HIV-1 subjects (n=102). Levels of ISG15-mRNA, ISG56-mRNA, APOBEC3G/3F-mRNA, TRAIL-mRNA, IDO-mRNA, proviral load andISG15 (rs15842 and rs1921) SNPs were evaluated by using TaqMan assays. We found that ISG15, ISG56, APOBEC3G/3F levels were increased in untreated HIV-1 patients compared to healthy donors, being ISG15 the highest ISG expressed. The amount of ISG15 correlated with viral load and with CD4+ T cell counts whereas no relationship was found between all ISGs analyzed and proviral load or HIV-1 tropism. ISG15 expression was reduced following long-term antiretroviral therapy. In addition, ISG15 levels were correlated with those of TRAIL and IDO in HIV-1 viremic patients. Lastly, ISG15 SNPs had no influence on ISG15 levels. We demonstrates that ISG15 is elevated in viremic HIV-1 patients and is associated with high TRAIL and IDO levels.
Collapse
|
22
|
Cho WK, Weeratunga P, Lee BH, Park JS, Kim CJ, Ma JY, Lee JS. Epimedium koreanum Nakai displays broad spectrum of antiviral activity in vitro and in vivo by inducing cellular antiviral state. Viruses 2015; 7:352-77. [PMID: 25609307 PMCID: PMC4306843 DOI: 10.3390/v7010352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 01/14/2015] [Indexed: 01/15/2023] Open
Abstract
Epimedium koreanum Nakai has been extensively used in traditional Korean and Chinese medicine to treat a variety of diseases. Despite the plant's known immune modulatory potential and chemical make-up, scientific information on its antiviral properties and mode of action have not been completely investigated. In this study, the broad antiviral spectrum and mode of action of an aqueous extract from Epimedium koreanum Nakai was evaluated in vitro, and moreover, the protective effect against divergent influenza A subtypes was determined in BALB/c mice. An effective dose of Epimedium koreanum Nakai markedly reduced the replication of Influenza A Virus (PR8), Vesicular Stomatitis Virus (VSV), Herpes Simplex Virus (HSV) and Newcastle Disease Virus (NDV) in RAW264.7 and HEK293T cells. Mechanically, we found that an aqueous extract from Epimedium koreanum Nakai induced the secretion of type I IFN and pro-inflammatory cytokines and the subsequent stimulation of the antiviral state in cells. Among various components present in the extract, quercetin was confirmed to have striking antiviral properties. The oral administration of Epimedium koreanum Nakai exhibited preventive effects on BALB/c mice against lethal doses of highly pathogenic influenza A subtypes (H1N1, H5N2, H7N3 and H9N2). Therefore, an extract of Epimedium koreanum Nakai and its components play roles as immunomodulators in the innate immune response, and may be potential candidates for prophylactic or therapeutic treatments against diverse viruses in animal and humans.
Collapse
Affiliation(s)
- Won-Kyung Cho
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon 305-764, Korea.
| | - Prasanna Weeratunga
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Byeong-Hoon Lee
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Jun-Seol Park
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Chul-Joong Kim
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| | - Jin Yeul Ma
- Korean Medicine (KM) Based Herbal Drug Development Group, Korea Institute of Oriental Medicine, Deajeon 305-764, Korea.
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, 220 Gung-Dong, Yuseong-Gu, Daejeon 305-764, Korea.
| |
Collapse
|
23
|
Acchioni C, Marsili G, Perrotti E, Remoli AL, Sgarbanti M, Battistini A. Type I IFN--a blunt spear in fighting HIV-1 infection. Cytokine Growth Factor Rev 2014; 26:143-58. [PMID: 25466629 DOI: 10.1016/j.cytogfr.2014.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 10/22/2014] [Indexed: 02/07/2023]
Abstract
For more than 50 years, Type I Interferon (IFN) has been recognized as critical in controlling viral infections. IFN is produced downstream germ-line encoded pattern recognition receptors (PRRs) upon engagement by pathogen-associated molecular patterns (PAMPs). As a result, hundreds of different interferon-stimulated genes (ISGs) are rapidly induced, acting in both autocrine and paracrine manner to build a barrier against viral replication and spread. ISGs encode proteins with direct antiviral and immunomodulatory activities affecting both innate and adaptive immune responses. During infection with viruses, as HIV-1, that can establish a persistent infection, IFN although produced, is not able to block the initial infection and a chronic IFN-mediated immune activation/inflammation becomes a pathogenic mechanism of disease progression. This review will briefly summarize when and how IFN is produced during HIV-1 infection and the way this innate immune response is manipulated by the virus to its own advantage to drive chronic immune activation and progression to AIDS.
Collapse
Affiliation(s)
- Chiara Acchioni
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Giulia Marsili
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Edvige Perrotti
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Anna Lisa Remoli
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Marco Sgarbanti
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy
| | - Angela Battistini
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, Rome 00161, Italy.
| |
Collapse
|
24
|
Abstract
The interferons (IFNs) are glycoproteins with strong antiviral activities that represent one of the first lines of host defense against invading pathogens. These proteins are classified into three groups, Type I, II and III IFNs, based on the structure of their receptors on the cell surface. Due to their ability to modulate immune responses, they have become attractive therapeutic options to control chronic virus infections. In combination with other drugs, Type I IFNs are considered as "standard of care" in suppressing Hepatitis C (HCV) and Hepatitis B (HBV) infections, while Type III IFN has generated encouraging results as a treatment for HCV infection in phase III clinical trials. However, though effective, using IFNs as a treatment is not without the need for caution. IFNs are such powerful cytokines that affect a wide array of cell types; as a result, patients usually experience unpleasant symptoms, with a percentage of patients suffering system wide effects. Thus, constant monitoring is required for patients treated with IFN in order to reach the treatment goals of suppressing virus infection and maintaining quality of life.
Collapse
Affiliation(s)
- Fan-ching Lin
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer, Research, National Cancer Institute, Frederick, MD 21702, USA.
| | - Howard A Young
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, Center for Cancer, Research, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|