1
|
Bahojb Mahdavi SZ, Jebelli A, Aghbash PS, Baradaran B, Amini M, Oroojalian F, Pouladi N, Baghi HB, de la Guardia M, Mokhtarzadeh AA. A comprehensive overview on the crosstalk between microRNAs and viral pathogenesis and infection. Med Res Rev 2025; 45:349-425. [PMID: 39185567 PMCID: PMC11796338 DOI: 10.1002/med.22073] [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: 11/06/2021] [Revised: 04/11/2023] [Accepted: 08/04/2024] [Indexed: 08/27/2024]
Abstract
Infections caused by viruses as the smallest infectious agents, pose a major threat to global public health. Viral infections utilize different host mechanisms to facilitate their own propagation and pathogenesis. MicroRNAs (miRNAs), as small noncoding RNA molecules, play important regulatory roles in different diseases, including viral infections. They can promote or inhibit viral infection and have a pro-viral or antiviral role. Also, viral infections can modulate the expression of host miRNAs. Furthermore, viruses from different families evade the host immune response by producing their own miRNAs called viral miRNAs (v-miRNAs). Understanding the replication cycle of viruses and their relation with host miRNAs and v-miRNAs can help to find new treatments against viral infections. In this review, we aim to outline the structure, genome, and replication cycle of various viruses including hepatitis B, hepatitis C, influenza A virus, coronavirus, human immunodeficiency virus, human papillomavirus, herpes simplex virus, Epstein-Barr virus, Dengue virus, Zika virus, and Ebola virus. We also discuss the role of different host miRNAs and v-miRNAs and their role in the pathogenesis of these viral infections.
Collapse
Affiliation(s)
- Seyedeh Zahra Bahojb Mahdavi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Asiyeh Jebelli
- Department of Biological Science, Faculty of Basic ScienceHigher Education Institute of Rab‐RashidTabrizIran
- Tuberculosis and Lung Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mohammad Amini
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of MedicineNorth Khorasan University of Medical SciencesBojnurdIran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic SciencesAzarbaijan Shahid Madani UniversityTabrizIran
| | | | | | | |
Collapse
|
2
|
Mishra SK, Senathilake KS, Kumar N, Patel CN, Uddin MB, Alqahtani T, Alqahtani A, Alharbi HM, Georrge JJ. Exploratory algorithms to devise multi-epitope subunit vaccine by examining HIV-1 envelope glycoprotein: An immunoinformatics and viroinformatics approach. PLoS One 2025; 20:e0318523. [PMID: 40014623 DOI: 10.1371/journal.pone.0318523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/16/2025] [Indexed: 03/01/2025] Open
Abstract
Acquired immune deficiency syndrome (AIDS), a widespread pandemic and severe health issue, is triggered by the human immunodeficiency virus (HIV); there is no specific vaccine to cure this infection, and the situation is worsening. Therefore, this research sought to develop a vaccine with multiple epitopes against this infection targeting envelope glycoprotein (vital in host-cell interaction) through the immunoinformatics and viroinformatics approach. We identified one B-cell, eight MHC-I, and four MHC-II epitopes on its immunogen-assisted screening. In addition, these putative epitopes were conjoined concurrently using a specific linker (EAAAK, KK, GPGPG), including an adjuvant and a His-Tag at the N and C terminal, respectively, to augment its immune reaction. The final constructed entity consists of 284 amino acids; immunological evaluation demonstrated that the developed vaccine possesses antigenic features with a value of 0.6222, is non-allergenic, and has prospective physiochemical characteristics. The secondary and tertiary structures were anticipated, and their quality has been evaluated. Further, docking analysis between vaccines with TLR3 shows a strong molecular interaction with a -20.0 kcal/mol binding energy, and the stability was analysed through the MD simulation (100ns). Moreover, the designed vaccine expression and immune response were analysed, and a high vaccine expression level was found (pET28a (+)) and robust immune response followed by codon adaptation index value 0.94, 58.36% GC content, and the generation of IgM + IgG, cytokines and interleukin. Based on overall investigation, the developed vaccine stimulates a robust immune response. Nevertheless, laboratory analysis is needed to confirm the protective potency of the vaccine.
Collapse
Affiliation(s)
- Saurav Kumar Mishra
- Department of Bioinformatics, University of North Bengal, Darjeeling, West Bengal, India
| | | | - Neeraj Kumar
- Department of Pharmaceutical Chemistry Bhupal Nobles, College of Pharmacy, Udaipur, Rajasthan, India
| | - Chirag N Patel
- Department of Botany, Bioinformatics and Climate Change Impacts Management, School of Science, Gujarat University, Ahmedabad, India
- Biotechnology Research Center, Technology Innovation Institute, Abu Dhabi, United Arab Emirates
| | - Mohammad Borhan Uddin
- Computational Biology Research Laboratory, Department of Pharmacy, Faculty of Health and Life Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Ali Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Hanan M Alharbi
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - John J Georrge
- Department of Bioinformatics, University of North Bengal, Darjeeling, West Bengal, India
| |
Collapse
|
3
|
Schimmich C, Vabret A, Zientara S, Valle-Casuso JC. Equine Infectious Anemia Virus Cellular Partners Along the Viral Cycle. Viruses 2024; 17:5. [PMID: 39861793 PMCID: PMC11769393 DOI: 10.3390/v17010005] [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: 11/29/2024] [Revised: 12/21/2024] [Accepted: 12/21/2024] [Indexed: 01/27/2025] Open
Abstract
Equine infectious anemia virus (EIAV) is the simplest described lentivirus within the Retroviridae family, related to the human immunodeficiency viruses (HIV-1 and HIV-2). There is an important interplay between host cells and viruses. Viruses need to hijack cellular proteins for their viral cycle completion and some cellular proteins are antiviral agents interfering with viral replication. HIV cellular partners have been extensively studied and described, with a special attention to host proteins able to inhibit specific steps of the viral cycle, called restriction factors. Viruses develop countermeasures against these restriction factors. Here, we aim to describe host cellular protein partners of EIAV viral replication, being proviral or antiviral. A comprehensive vision of the interactions between the virus and specific host's proteins can help with the discovery of new targets for the design of therapeutics. Studies performed on HIV-1 can provide insights into the functioning of EIAV, as well as differences, as both types of virus research can benefit from each other.
Collapse
Affiliation(s)
- Cécile Schimmich
- ANSES Animal Health Laboratory, PhEED Unit, 14430 Goustranville, France;
| | - Astrid Vabret
- Department of Virology, University of Caen Normandy, Dynamicure INSERM UMR 1311, Centre Hospitalo Universitaire (CHU) Caen, 14000 Caen, France;
| | - Stéphan Zientara
- UMR VIROLOGIE, INRAE, École Nationale Vétérinaire d’Alfort, ANSES Laboratoire de Santé Animale, Université Paris-Est, 94700 Maisons-Alfort, France;
| | - José Carlos Valle-Casuso
- ANSES Animal Health Laboratory, PhEED Unit, 14430 Goustranville, France;
- Mixed Technological Unit “Equine Health and Welfare—Organisation and Traceability of the Equine Industry” (UMT SABOT), 14430 Goustranville, France
| |
Collapse
|
4
|
De Clercq E. A scientific career from the early 1960s till 2023: A tale of the various protagonists. Biochem Pharmacol 2024; 228:116248. [PMID: 38701868 DOI: 10.1016/j.bcp.2024.116248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
In this era spanning more than 60 years (from the early 1960s till today (2023), a broad variety of actors played a decisive role: Piet De Somer, Tom C. Merigan, Paul A. Janssen, Maurice Hilleman, and Georges Smets. Two protagonists (Antonín Holý and John C. Martin) formed with me a unique triangle (the Holý Trinity). Walter Fiers' group (with the help of Jean Content) contributed to the cloning of human β-interferon, and Piet Herdewijn accomplished the chemical synthesis of an array of anti-HIV 2',3'-dideoxynucleoside analogues. Rudi Pauwels, Masanori Baba, Dominique Schols, Johan Neyts, Lieve Naesens, Anita Van Lierde, Graciela Andrei, Robert Snoeck and Dirk Daelemans, as members of my team, helped me in achieving the intended goal, the development of a selective therapy for virus infections. The collaboration with "Lowie" (Guangdi Li) generated a new dimension for the future.
Collapse
Affiliation(s)
- Erik De Clercq
- KU Leuven, Rega Institute for Medical Research, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
5
|
Sharafutdinov I, Friedrich B, Rottner K, Backert S, Tegtmeyer N. Cortactin: A major cellular target of viral, protozoal, and fungal pathogens. Mol Microbiol 2024; 122:165-183. [PMID: 38868928 DOI: 10.1111/mmi.15284] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024]
Abstract
Many viral, protozoal, and fungal pathogens represent major human and animal health problems due to their great potential of causing infectious diseases. Research on these pathogens has contributed substantially to our current understanding of both microbial virulence determinants and host key factors during infection. Countless studies have also shed light on the molecular mechanisms of host-pathogen interactions that are employed by these microbes. For example, actin cytoskeletal dynamics play critical roles in effective adhesion, host cell entry, and intracellular movements of intruding pathogens. Cortactin is an eminent host cell protein that stimulates actin polymerization and signal transduction, and recently emerged as fundamental player during host-pathogen crosstalk. Here we review the important role of cortactin as major target for various prominent viral, protozoal and fungal pathogens in humans, and its role in human disease development and cancer progression. Most if not all of these important classes of pathogens have been reported to hijack cortactin during infection through mediating up- or downregulation of cortactin mRNA and protein expression as well as signaling. In particular, pathogen-induced changes in tyrosine and serine phosphorylation status of cortactin at its major phospho-sites (Y-421, Y-470, Y-486, S-113, S-298, S-405, and S-418) are addressed. As has been reported for various Gram-negative and Gram-positive bacteria, many pathogenic viruses, protozoa, and fungi also control these regulatory phospho-sites, for example, by activating kinases such as Src, PAK, ERK1/2, and PKD, which are known to phosphorylate cortactin. In addition, the recruitment of cortactin and its interaction partners, like the Arp2/3 complex and F-actin, to the contact sites between pathogens and host cells is highlighted, as this plays an important role in the infection process and internalization of several pathogens. However, there are also other ways in which the pathogens can exploit the function of cortactin for their needs, as the cortactin-mediated regulation of cellular processes is complex and involves numerous different interaction partners. Here, the current state of knowledge is summarized.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Barbara Friedrich
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Braunschweig, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Ghorai S, Shand H, Patra S, Panda K, Santiago MJ, Rahman MS, Chinnapaiyan S, Unwalla HJ. Nanomedicine for the Treatment of Viral Diseases: Smaller Solution to Bigger Problems. Pharmaceutics 2024; 16:407. [PMID: 38543301 PMCID: PMC10975899 DOI: 10.3390/pharmaceutics16030407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 04/01/2024] Open
Abstract
The continuous evolution of new viruses poses a danger to world health. Rampant outbreaks may advance to pandemic level, often straining financial and medical resources to breaking point. While vaccination remains the gold standard to prevent viral illnesses, these are mostly prophylactic and offer minimal assistance to those who have already developed viral illnesses. Moreover, the timeline to vaccine development and testing can be extensive, leading to a lapse in controlling the spread of viral infection during pandemics. Antiviral therapeutics can provide a temporary fix to tide over the time lag when vaccines are not available during the commencement of a disease outburst. At times, these medications can have negative side effects that outweigh the benefits, and they are not always effective against newly emerging virus strains. Several limitations with conventional antiviral therapies may be addressed by nanotechnology. By using nano delivery vehicles, for instance, the pharmacokinetic profile of antiviral medications can be significantly improved while decreasing systemic toxicity. The virucidal or virus-neutralizing qualities of other special nanomaterials can be exploited. This review focuses on the recent advancements in nanomedicine against RNA viruses, including nano-vaccines and nano-herbal therapeutics.
Collapse
Affiliation(s)
- Suvankar Ghorai
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
- Department of Microbiology, Raiganj University, Raiganj 733134, India; (H.S.); (S.P.)
| | - Harshita Shand
- Department of Microbiology, Raiganj University, Raiganj 733134, India; (H.S.); (S.P.)
| | - Soumendu Patra
- Department of Microbiology, Raiganj University, Raiganj 733134, India; (H.S.); (S.P.)
| | - Kingshuk Panda
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
| | - Maria J. Santiago
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
- Department of Chemistry and Biochemistry, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Md. Sohanur Rahman
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
| | - Srinivasan Chinnapaiyan
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
| | - Hoshang J. Unwalla
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA; (S.G.); (K.P.); (M.J.S.); (M.S.R.); (S.C.)
| |
Collapse
|
7
|
Linkner TR, Ambrus V, Kunkli B, Szojka ZI, Kalló G, Csősz É, Kumar A, Emri M, Tőzsér J, Mahdi M. Comparative Analysis of Differential Cellular Transcriptome and Proteome Regulation by HIV-1 and HIV-2 Pseudovirions in the Early Phase of Infection. Int J Mol Sci 2023; 25:380. [PMID: 38203551 PMCID: PMC10779251 DOI: 10.3390/ijms25010380] [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: 10/30/2023] [Revised: 12/18/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
In spite of the similar structural and genomic organization of human immunodeficiency viruses type 1 and 2 (HIV-1 and HIV-2), striking differences exist between them in terms of replication dynamics and clinical manifestation of infection. Although the pathomechanism of HIV-1 infection is well characterized, relatively few data are available regarding HIV-2 viral replication and its interaction with host-cell proteins during the early phase of infection. We utilized proteo-transcriptomic analyses to determine differential genome expression and proteomic changes induced by transduction with HIV-1/2 pseudovirions during 8, 12 and 26 h time-points in HEK-293T cells. We show that alteration in the cellular milieu was indeed different between the two pseudovirions. The significantly higher number of genes altered by HIV-2 in the first two time-points suggests a more diverse yet subtle effect on the host cell, preparing the infected cell for integration and latency. On the other hand, GO analysis showed that, while HIV-1 induced cellular oxidative stress and had a greater effect on cellular metabolism, HIV-2 mostly affected genes involved in cell adhesion, extracellular matrix organization or cellular differentiation. Proteomics analysis revealed that HIV-2 significantly downregulated the expression of proteins involved in mRNA processing and translation. Meanwhile, HIV-1 influenced the cellular level of translation initiation factors and chaperones. Our study provides insight into the understudied replication cycle of HIV-2 and enriches our knowledge about the use of HIV-based lentiviral vectors in general.
Collapse
Affiliation(s)
- Tamás Richárd Linkner
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary;
| | - Viktor Ambrus
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary;
| | - Balázs Kunkli
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary;
| | - Zsófia Ilona Szojka
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
- Division of Medical Microbiology, Department of Laboratory Medicine, Lund University, 22100 Lund, Sweden
| | - Gergő Kalló
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.K.); (É.C.)
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.K.); (É.C.)
| | - Ajneesh Kumar
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary;
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.K.); (É.C.)
| | - Miklós Emri
- Department of Medical Imaging, Division of Nuclear Medicine and Translational Imaging, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
| | - József Tőzsér
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (G.K.); (É.C.)
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (T.R.L.); (V.A.); (B.K.); (Z.I.S.)
| |
Collapse
|
8
|
Sun L, Nie P, Luan L, Herdewijn P, Wang YT. Synthetic approaches and application of clinically approved small-molecule Anti-HIV drugs: An update. Eur J Med Chem 2023; 261:115847. [PMID: 37801826 DOI: 10.1016/j.ejmech.2023.115847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/18/2023] [Accepted: 09/29/2023] [Indexed: 10/08/2023]
Abstract
Application of chemotherapeutic agents to inhibit the HIV replication process has brought about a significant metamorphosis in the landscape of AIDS. Substantial declines in morbidity and mortality rates have been attained, accompanied by notable decreases in healthcare resource utilization. However, treatment modalities do not uniformly inhibit HIV replication in every patient, while the emergence of drug-resistant viral strains poses a substantial obstacle to subsequent therapeutic interventions. Furthermore, chronic administration of therapy may lead to the manifestation of toxicities. These challenges necessitate the exploration of novel pharmacological agents and innovative therapeutic approaches aimed at effectively managing the persistent viral replication characteristic of chronic infection. This review examines the role of clinically approved small-molecule drugs in the treatment of HIV/AIDS, which provides an in-depth analysis of the major classes of small-molecule drugs, including nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), integrase inhibitors, entry inhibitors, and pharmacokinetic enhancers. The review mainly discusses the application, synthetic routes, and mechanisms of action of small-molecule drugs employed in the treatment of HIV, as well as their use in combination with antiretroviral therapy, presenting viewpoints on forthcoming avenues in the development of novel anti-HIV drugs.
Collapse
Affiliation(s)
- Lu Sun
- Zhongshan Hospital Affiliated to Dalian University, Dalian, 116001, China
| | - Peng Nie
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Li Luan
- Zhongshan Hospital Affiliated to Dalian University, Dalian, 116001, China.
| | - Piet Herdewijn
- Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Ya-Tao Wang
- First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China; Medicinal Chemistry, Rega Institute of Medical Research, KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| |
Collapse
|
9
|
Huang Y, Zhu X, Guo X, Zhou Y, Liu D, Mao J, Xiong Y, Deng Y, Gao X. Advances in mRNA vaccines for viral diseases. J Med Virol 2023; 95:e28924. [PMID: 37417396 DOI: 10.1002/jmv.28924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/08/2023]
Abstract
Since the onset of the pandemic caused by severe acute respiratory syndrome coronavirus 2, messenger RNA (mRNA) vaccines have demonstrated outstanding performance. mRNA vaccines offer significant advantages over conventional vaccines in production speed and cost-effectiveness, making them an attractive option against other viral diseases. This article reviewed recent advances in viral mRNA vaccines and their delivery systems to provide references and guidance for developing mRNA vaccines for new viral diseases.
Collapse
Affiliation(s)
- Yukai Huang
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xuerui Zhu
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiao Guo
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuancheng Zhou
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu, China
| | - Dongying Liu
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jingrui Mao
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yongai Xiong
- Department of Pharmaceutics, School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Youcai Deng
- Department of Hematology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xinghong Gao
- Department of Microbiology, School of Basic Medicine, Zunyi Medical University, Zunyi, Guizhou, China
- Provincial Department of Education, Key Laboratory of Infectious Disease & Bio-Safety, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
10
|
Asl FD, Mousazadeh M, Taji S, Bahmani A, Khashayar P, Azimzadeh M, Mostafavi E. Nano drug-delivery systems for management of AIDS: liposomes, dendrimers, gold and silver nanoparticles. Nanomedicine (Lond) 2023; 18:279-302. [PMID: 37125616 PMCID: PMC10242436 DOI: 10.2217/nnm-2022-0248] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/08/2023] [Indexed: 05/02/2023] Open
Abstract
AIDS causes increasing mortality every year. With advancements in nanomedicine, different nanomaterials (NMs) have been applied to treat AIDS and overcome its limitations. Among different NMs, nanoparticles (NPs) can act as nanocarriers due to their enhanced solubility, sustained release, targeting abilities and facilitation of drug-dose reductions. This review discusses recent advancements in therapeutics for AIDS/HIV using various NMs, mainly focused on three classifications: polymeric, liposomal and inorganic NMs. Polymeric dendrimers, polyethylenimine-NPs, poly(lactic-co-glycolic acid)-NPs, chitosan and the use of liposomal-based delivery systems and inorganic NPs, including gold and silver NPs, are explored. Recent advances, current challenges and future perspectives on the use of these NMs for better management of HIV/AIDS are also discussed.
Collapse
Affiliation(s)
- Fateme Davarani Asl
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, 88138-33435, Iran
| | - Marziyeh Mousazadeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
| | - Shirinsadat Taji
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-154, Iran
- Institute for Genetics, University of Cologne, Cologne, D-50674, Germany
| | - Abbas Bahmani
- Institute for Nanoscience & Nanotechnology (INST), Sharif University of Technology, Tehran, 14588-89694, Iran
| | - Patricia Khashayar
- Center for Microsystems Technology, Imec & Ghent University, Ghent, 9050, Belgium
| | - Mostafa Azimzadeh
- Medical Nanotechnology & Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, 89195-999, Iran
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Peng S, Wang H, Wang Z, Wang Q. Progression of Antiviral Agents Targeting Viral Polymerases. Molecules 2022; 27:7370. [PMID: 36364196 PMCID: PMC9654062 DOI: 10.3390/molecules27217370] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 08/08/2023] Open
Abstract
Viral DNA and RNA polymerases are two kinds of very important enzymes that synthesize the genetic materials of the virus itself, and they have become extremely favorable targets for the development of antiviral drugs because of their relatively conserved characteristics. There are many similarities in the structure and function of different viral polymerases, so inhibitors designed for a certain viral polymerase have acted as effective universal inhibitors on other types of viruses. The present review describes the development of classical antiviral drugs targeting polymerases, summarizes a variety of viral polymerase inhibitors from the perspective of chemically synthesized drugs and natural product drugs, describes novel approaches, and proposes promising development strategies for antiviral drugs.
Collapse
Affiliation(s)
| | | | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingzhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
12
|
Dominant Negative Mutants of Human Immunodeficiency Virus Type 1 Viral Infectivity Factor (Vif) Disrupt Core-Binding Factor Beta-Vif Interaction. J Virol 2022; 96:e0055522. [PMID: 35950859 PMCID: PMC9472641 DOI: 10.1128/jvi.00555-22] [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: 11/20/2022] Open
Abstract
Apolipoprotein B mRNA-editing catalytic polypeptide-like 3 family members (APOBEC3s) are host restriction factors that inhibit viral replication. Viral infectivity factor (Vif), a human immunodeficiency virus type 1 (HIV-1) accessory protein, mediates the degradation of APOBEC3s by forming the Vif-E3 complex, in which core-binding factor beta (CBFβ) is an essential molecular chaperone. Here, we screened nonfunctional Vif mutants with high affinity for CBFβ to inhibit HIV-1 in a dominant negative manner. We applied the yeast surface display technology to express Vif random mutant libraries, and mutants showing high CBFβ affinity were screened using flow cytometry. Most of the screened Vif mutants containing random mutations of different frequencies were able to rescue APOBEC3G (A3G). In the subsequent screening, three of the mutants restricted HIV-1, recovered G-to-A hypermutation, and rescued APOBEC3s. Among them, Vif-6M showed a cross-protection effect toward APOBEC3C, APOBEC3F, and African green monkey A3G. Stable expression of Vif-6M in T lymphocytes inhibited the viral replication in newly HIV-1-infected cells and the chronically infected cell line H9/HXB2. Furthermore, the expression of Vif-6M provided a survival advantage to T lymphocytes infected with HIV-1. These results suggest that dominant negative Vif mutants acting on the Vif-CBFβ target potently restrict HIV-1. IMPORTANCE Antiviral therapy cannot eliminate HIV and exhibits disadvantages such as drug resistance and toxicity. Therefore, novel strategies for inhibiting viral replication in patients with HIV are urgently needed. APOBEC3s in host cells are able to inhibit viral replication but are antagonized by HIV-1 Vif-mediated degradation. Therefore, we screened nonfunctional Vif mutants with high affinity for CBFβ to compete with the wild-type Vif (wtVif) as a potential strategy to assist with HIV-1 treatment. Most screened mutants rescued the expression of A3G in the presence of wtVif, especially Vif-6M, which could protect various APOBEC3s and improve the incorporation of A3G into HIV-1 particles. Transduction of Vif-6M into T lymphocytes inhibited the replication of the newly infected virus and the chronically infected virus. These data suggest that Vif mutants targeting the Vif-CBFβ interaction may be promising in the development of a new AIDS therapeutic strategy.
Collapse
|
13
|
Rajan A, Shrivastava S, Janhawi, Kumar A, Singh AK, Arora PK. CRISPR-Cas system: from diagnostic tool to potential antiviral treatment. Appl Microbiol Biotechnol 2022; 106:5863-5877. [PMID: 36008567 PMCID: PMC9411046 DOI: 10.1007/s00253-022-12135-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 11/27/2022]
Abstract
This mini review focuses on the diagnosis and treatment of virus diseases using Crisper-Cas technology. The present paper describes various strategies involved in diagnosing diseases using Crispr-Cas-based assays. Additionally, CRISPR-Cas systems offer great potential as new therapeutic tools for treating viral infections including HIV, Influenza, and SARS-CoV-2. There are several major challenges to be overcome before this technology can be applied routinely in clinical settings, such as finding a suitable delivery tool, toxicity, and immunogenicity, as well as off-target effects. This review also discusses ways to deal with the challenges associated with Crisper-Cas technology. KEY POINTS: • Crisper technology is being applied to diagnose infectious and non-infectious diseases. • A new generation of CRISPR-Cas-based assays has been developed which detect pathogens within minutes, providing rapid diagnosis of diseases. • Crispr-Cas tools can be used to combat viral infections, specifically HIV, influenza, and SARS-CoV-2.
Collapse
Affiliation(s)
- Aishwarya Rajan
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Stuti Shrivastava
- Electronics and Communication, Jaypee Institute of Information Technology, Noida, India
| | - Janhawi
- Department of Zoology, Kalindi College, University of Delhi, Delhi, India
| | - Akhilesh Kumar
- Department of Botany, Banaras Hindu University, Varanasi, India.
| | - Alok Kumar Singh
- Department of Biochemistry, Shaheed Rajguru College of Applied Sciences for Women, University of Delhi, Delhi, India.
| | - Pankaj Kumar Arora
- Department of Environmental Microbiology, Babasaheb Bhimrao Ambedkar University, Lucknow, India.
| |
Collapse
|
14
|
Vergni D, Santoni D, Bouba Y, Lemme S, Fabeni L, Carioti L, Bertoli A, Gennari W, Forbici F, Perno CF, Gagliardini R, Ceccherini-Silberstein F, Santoro MM. Evaluation of HIV-1 integrase variability by combining computational and probabilistic approaches. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 101:105294. [PMID: 35513162 DOI: 10.1016/j.meegid.2022.105294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/24/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
This study aimed at updating previous data on HIV-1 integrase variability, by using effective bioinformatics methods combining different statistical instruments from simple entropy and mutation rate to more specific approaches such as Hellinger distance. A total of 2133 HIV-1 integrase sequences were analyzed in: i) 1460 samples from drug-naïve [DN] individuals; ii) 386 samples from drug-experienced but INI-naïve [IN] individuals; iii) 287 samples from INI-experienced [IE] individuals. Within the three groups, 76 amino acid positions were highly conserved (≤0.2% variation, Hellinger distance: <0.25%), with 35 fully invariant positions; while, 80 positions were conserved (>0.2% to <1% variation, Hellinger distance: <1%). The H12-H16-C40-C43 and D64-D116-E152 motifs were all well conserved. Some residues were affected by dramatic changes in their mutation distributions, especially between DN and IE samples (Hellinger distance ≥1%). In particular, 15 positions (D6, S24, V31, S39, L74, A91, S119, T122, T124, T125, V126, K160, N222, S230, C280) showed a significant decrease of mutation rate in IN and/or IE samples compared to DN samples. Conversely, 8 positions showed significantly higher mutation rate in samples from treated individuals (IN and/or IE) compared to DN. Some of these positions, such as E92, T97, G140, Y143, Q148 and N155, were already known to be associated with resistance to integrase inhibitors; other positions including S24, M154, V165 and D270 are not yet documented to be associated with resistance. Our study confirms the high conservation of HIV-1 integrase and identified highly invariant positions using robust and innovative methods. The role of novel mutations located in the critical region of HIV-1 integrase deserves further investigation.
Collapse
Affiliation(s)
- Davide Vergni
- Istituto per le Applicazioni del Calcolo "Mauro Picone" - CNR, Rome, Italy.
| | - Daniele Santoni
- Istituto di Analisi dei Sistemi ed. Informatica "Antonio Ruberti" - CNR, Rome, Italy
| | - Yagai Bouba
- Chantal BIYA International Reference Centre for research on HIV/AIDS prevention and management (CIRCB), Yaoundé, Cameroon; Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Saverio Lemme
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Lavinia Fabeni
- Laboratory of Virology, IRCCS, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Luca Carioti
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Ada Bertoli
- Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy; Laboratory of Virology, University Hospital "Tor Vergata", Rome, Italy
| | - William Gennari
- Microbiology and Virology Unit, University Hospital, University of Modena and Reggio Emilia, Modena, Italy
| | - Federica Forbici
- Laboratory of Virology, IRCCS, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | - Carlo Federico Perno
- Multimodal Laboratory Research Department, Children Hospital Bambino Gesù, IRCCS, Rome, Italy
| | - Roberta Gagliardini
- HIV/AIDS Department, IRCCS, National Institute for Infectious Diseases "Lazzaro Spallanzani", Rome, Italy
| | | | | | | |
Collapse
|
15
|
Yue T, Zhang P, Hao Y, He J, Zheng J, De Clercq E, Li G, Huang Y, Zheng F. Epidemiology and Clinical Outcomes of HIV Infection in South-Central China: A Retrospective Study From 2003 to 2018. Front Public Health 2022; 10:902537. [PMID: 35757651 PMCID: PMC9218543 DOI: 10.3389/fpubh.2022.902537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Objective HIV epidemiology in South-Central China is rarely reported. This study aims to characterize epidemiological and clinical features of HIV-infected patients in Hunan Province, located in South-Central China, for better management of HIV infections. Methods This retrospective study retrieved multi-center records of laboratory-confirmed HIV-infected patients in Hunan province. Information on HIV-associated mortality and antiretroviral therapies was also collected. Results Among 34,297 patients diagnosed with HIV infections from 2003 to 2018, 73.9% were males, 41.3% were older adults (≥50 years), and 71.2% were infected by heterosexual transmission. Despite a slow growth of new HIV infections in the overall population, annual percentages of HIV infections increased in older males (85.3% through heterosexual transmission) and young patients <30 years (39.9% through homosexual transmission). At baseline, serum levels of CD4+ T-cell counts were lower in older adults (191.0 cells/μl) than in young patients (294.6 cells/μl, p-value < 0.0001). A large proportion (47.2%, N = 16,165) of HIV-infected patients had advanced HIV disease (CD4+ T-cell counts < 200 cells/μl) from 2003 to 2018. All-cause mortality (57.0% due to AIDS-related illnesses) was reported among 4411 HIV-infected patients, including 2619 older adults. The 10-year survival rate was significantly lower in elderly males than in other patients (59.0 vs. 78.4%, p-value < 0.05). Conclusions Elderly males are prone to HIV infections with a high risk of HIV-associated fatality. Our findings support early prevention and critical care for elderly populations to control HIV infections.
Collapse
Affiliation(s)
- Tingting Yue
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Pan Zhang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
| | - Yuantao Hao
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Jianmei He
- Hunan Center for Disease Control and Prevention, Changsha, China
| | - Jun Zheng
- Hunan Center for Disease Control and Prevention, Changsha, China
| | - Erik De Clercq
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Guangdi Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, China
- Hunan Children's Hospital, Changsha, China
| | - Yaxiong Huang
- Department of Infectious Disease, The First Hospital of Changsha, Changsha, China
| | - Fang Zheng
- Department of Infectious Disease, The First Hospital of Changsha, Changsha, China
| |
Collapse
|
16
|
Urvashi, Senthil Kumar JB, Das P, Tandon V. Development of Azaindole-Based Frameworks as Potential Antiviral Agents and Their Future Perspectives. J Med Chem 2022; 65:6454-6495. [PMID: 35477274 PMCID: PMC9063994 DOI: 10.1021/acs.jmedchem.2c00444] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Indexed: 11/29/2022]
Abstract
The azaindole (AI) framework continues to play a significant role in the design of new antiviral agents. Modulating the position and isosteric replacement of the nitrogen atom of AI analogs notably influences the intrinsic physicochemical properties of lead compounds. The intra- and intermolecular interactions of AI derivatives with host receptors or viral proteins can also be fine tuned by carefully placing the nitrogen atom in the heterocyclic core. This wide-ranging perspective article focuses on AIs that have considerable utility in drug discovery programs against RNA viruses. The inhibition of influenza A, human immunodeficiency, respiratory syncytial, neurotropic alpha, dengue, ebola, and hepatitis C viruses by AI analogs is extensively reviewed to assess their plausible future potential in antiviral drug discovery. The binding interaction of AIs with the target protein is examined to derive a structural basis for designing new antiviral agents.
Collapse
Affiliation(s)
- Urvashi
- Drug Discovery Laboratory, Special Centre for
Molecular Medicine, Jawaharlal Nehru University, New Delhi 110
067, India
- Department of Chemistry, University of
Delhi, New Delhi 110007, India
| | - J. B. Senthil Kumar
- Drug Discovery Laboratory, Special Centre for
Molecular Medicine, Jawaharlal Nehru University, New Delhi 110
067, India
| | - Parthasarathi Das
- Department of Chemistry, Indian Institute
of Technology (ISM), Dhanbad 826004, India
| | - Vibha Tandon
- Drug Discovery Laboratory, Special Centre for
Molecular Medicine, Jawaharlal Nehru University, New Delhi 110
067, India
| |
Collapse
|
17
|
Climaco-Arvizu S, Flores-López V, González-Torres C, Gaytán-Cervantes FJ, Hernández-García MC, Zárate-Segura PB, Chávez-Torres M, Tesoro-Cruz E, Pinto-Cardoso SM, Bekker-Méndez VC. Protease and gag diversity and drug resistance mutations among treatment-naive Mexican people living with HIV. BMC Infect Dis 2022; 22:447. [PMID: 35538426 PMCID: PMC9088029 DOI: 10.1186/s12879-022-07446-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/29/2022] [Indexed: 09/17/2024] Open
Abstract
Introduction In Mexico, HIV genotyping is performed in people living with HIV (PLWH) failing their first-line antiretroviral (ARV) regimen; it is not routinely done for all treatment-naive PLWH before ARV initiation. The first nationally representative survey published in 2016 reported that the prevalence of pretreatment drug mutations in treatment-naive Mexican PLWH was 15.5% to any antiretroviral drug and 10.6% to non-nucleoside reverse transcriptase inhibitors (NNRTIs) using conventional Sanger sequencing. Most reports in Mexico focus on HIV pol gene and nucleoside and non-nucleoside reverse transcriptase inhibitor (NRTI and NNRTI) drug resistance mutations (DRMs) prevalence, using Sanger sequencing, next-generation sequencing (NGS) or both. To our knowledge, NGS has not be used to detect pretreatment drug resistance mutations (DRMs) in the HIV protease (PR) gene and its substrate the Gag polyprotein. Methods Treatment-naive adult Mexican PLWH were recruited between 2016 and 2019. HIV Gag and protease sequences were obtained by NGS and DRMs were identified using the WHO surveillance drug resistance mutation (SDRM) list. Results One hundred PLWH attending a public national reference hospital were included. The median age was 28 years-old, and most were male. The median HIV viral load was 4.99 [4.39–5.40] log copies/mL and median CD4 cell count was 150 [68.0–355.78] cells/mm3. As expected, most sequences clustered with HIV-1 subtype B (97.9%). Major PI resistance mutations were detected: 8 (8.3%) of 96 patients at a detection threshold of 1% and 3 (3.1%) at a detection threshold of 20%. A total of 1184 mutations in Gag were detected, of which 51 have been associated with resistance to PI, most of them were detected at a threshold of 20%. Follow-up clinical data was available for 79 PLWH at 6 months post-ART initiation, seven PLWH failed their first ART regimen; however no major PI mutations were identified in these individuals at baseline. Conclusions The frequency of DRM in the HIV protease was 7.3% at a detection threshold of 1% and 3.1% at a detection threshold of 20%. NGS-based HIV drug resistance genotyping provide improved detection of DRMs. Viral load was used to monitor ARV response and treatment failure was 8.9%. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07446-8.
Collapse
Affiliation(s)
- Samantha Climaco-Arvizu
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología "Dr Daniel Méndez Hernández", Centro Médico Nacional "La Raza", Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, C.P. 02990, México.,Laboratorio de Medicina Traslacional, Instituto Politécnico Nacional, Ciudad de México, México
| | | | - Carolina González-Torres
- División de Desarrollo de La Investigación, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | | | - María Concepción Hernández-García
- Instituto Mexicano del Seguro Social (IMSS), Hospital de Infectología "Dr Daniel Méndez Hernández", Centro Médico Nacional (CMN), La Raza", Ciudad de México, México
| | | | - Monserrat Chávez-Torres
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, C.P. 14080, México
| | - Emiliano Tesoro-Cruz
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología "Dr Daniel Méndez Hernández", Centro Médico Nacional "La Raza", Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, C.P. 02990, México
| | - Sandra María Pinto-Cardoso
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, C.P. 14080, México.
| | - Vilma Carolina Bekker-Méndez
- Unidad de Investigación Médica en Inmunología e Infectología, Hospital de Infectología "Dr Daniel Méndez Hernández", Centro Médico Nacional "La Raza", Instituto Mexicano del Seguro Social (IMSS), Ciudad de México, C.P. 02990, México.
| |
Collapse
|
18
|
Nikyar A, Bolhassani A, Rouhollah F, Heshmati M. In Vitro Delivery of HIV-1 Nef-Vpr DNA Construct Using the Human Antimicrobial Peptide LL-37. Curr Drug Deliv 2022; 19:1083-1092. [PMID: 35176981 DOI: 10.2174/1567201819666220217164055] [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: 07/25/2021] [Revised: 11/05/2021] [Accepted: 01/02/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES DNA-based therapeutic vaccines have been proposed as promising strategy for treatment of established HIV infections. However, these vaccines are often associated with certain shortcomings, such as poor immunogenicity and low transfection efficiency. In this study, we investigated ability of LL-37 to deliver a potential immunogenic fusion construct comprising HIV-1 nef and vpr genes into a mammalian cell line. METHODS First, the pEGFP-N1 eukaryotic expression vector harboring the HIV-1 nef-vpr fusion was produced free of endotoxin on large scale. Then, DNA/LL-37 complexes were prepared by co-incubation of pEGFP-nef-vpr with LL-37 for 45 minutes at different nitrogen to phosphate (N/P) ratios. Formation of DNA/peptide complexes was investigated by gel retardation assay. Next, stability and morphological characteristics of the nanoparticles were evaluated. Toxicity of LL-37 and the nanoparticles in HEK-293T cells was assessed by MTT assay. Transfection efficiency of the DNA/LL-37 complexes was studied by fluorescence microscopy, flow cytometry, and western blot analysis. RESULTS LL-37 formed stable complexes with pEGFP-nef-vpr (diameter of 150-200 nm) while providing good protection against nucleolytic and proteolytic degradation. The peptide significantly affected cell viability even at low concentrations. However, the LL-37/DNA complexes had no significant cytotoxic effect. Treatment of cells with pEGFP-N1/LL-37 and pEGFP-nef-vpr/LL-37 resulted in transfection of 36.32% ± 1.13 and 25.55% ± 2.07 of cells, respectively. CONCLUSION Given these findings and the important immunomodulatory and antiviral activities of LL-37, the use of this peptide can be further exploited in the development of novel gene delivery strategies and vaccine design.
Collapse
Affiliation(s)
- Arash Nikyar
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDs, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rouhollah
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.,Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Masoumeh Heshmati
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
19
|
van Heuvel Y, Schatz S, Rosengarten JF, Stitz J. Infectious RNA: Human Immunodeficiency Virus (HIV) Biology, Therapeutic Intervention, and the Quest for a Vaccine. Toxins (Basel) 2022; 14:toxins14020138. [PMID: 35202165 PMCID: PMC8876946 DOI: 10.3390/toxins14020138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/02/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022] Open
Abstract
Different mechanisms mediate the toxicity of RNA. Genomic retroviral mRNA hijacks infected host cell factors to enable virus replication. The viral genomic RNA of the human immunodeficiency virus (HIV) encompasses nine genes encoding in less than 10 kb all proteins needed for replication in susceptible host cells. To do so, the genomic RNA undergoes complex alternative splicing to facilitate the synthesis of the structural, accessory, and regulatory proteins. However, HIV strongly relies on the host cell machinery recruiting cellular factors to complete its replication cycle. Antiretroviral therapy (ART) targets different steps in the cycle, preventing disease progression to the acquired immunodeficiency syndrome (AIDS). The comprehension of the host immune system interaction with the virus has fostered the development of a variety of vaccine platforms. Despite encouraging provisional results in vaccine trials, no effective vaccine has been developed, yet. However, novel promising vaccine platforms are currently under investigation.
Collapse
Affiliation(s)
- Yasemin van Heuvel
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Stefanie Schatz
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Jamila Franca Rosengarten
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Institute of Technical Chemistry, Leibniz University Hannover, Callinstraße 3-9, 30167 Hannover, Germany
| | - Jörn Stitz
- Research Group Pharmaceutical Biotechnology, Faculty of Applied Natural Sciences, TH Köln—University of Applied Sciences, Chempark Leverkusen, Kaiser-Wilhelm-Allee, 51368 Leverkusen, Germany; (Y.v.H.); (S.S.); (J.F.R.)
- Correspondence:
| |
Collapse
|
20
|
Chakravarty N, Senthilnathan T, Paiola S, Gyani P, Castillo Cario S, Urena E, Jeysankar A, Jeysankar P, Ignatius Irudayam J, Natesan Subramanian S, Lavretsky H, Joshi S, Garcia G, Ramaiah A, Arumugaswami V. Neurological pathophysiology of SARS-CoV-2 and pandemic potential RNA viruses: a comparative analysis. FEBS Lett 2021; 595:2854-2871. [PMID: 34757622 PMCID: PMC8652524 DOI: 10.1002/1873-3468.14227] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/15/2021] [Accepted: 11/05/2021] [Indexed: 12/13/2022]
Abstract
SARS-CoV-2 has infected hundreds of millions of people with over four million dead, resulting in one of the worst global pandemics in recent history. Neurological symptoms associated with COVID-19 include anosmia, ageusia, headaches, confusion, delirium, and strokes. These may manifest due to viral entry into the central nervous system (CNS) through the blood-brain barrier (BBB) by means of ill-defined mechanisms. Here, we summarize the abilities of SARS-CoV-2 and other neurotropic RNA viruses, including Zika virus and Nipah virus, to cross the BBB into the CNS, highlighting the role of magnetic resonance imaging (MRI) in assessing presence and severity of brain structural changes in COVID-19 patients. We present new insight into key mutations in SARS-CoV-2 variants B.1.1.7 (P681H) and B.1.617.2 (P681R), which may impact on neuropilin 1 (NRP1) binding and CNS invasion. We postulate that SARS-CoV-2 may infect both peripheral cells capable of crossing the BBB and brain endothelial cells to traverse the BBB and spread into the brain. COVID-19 patients can be followed up with MRI modalities to better understand the long-term effects of COVID-19 on the brain.
Collapse
Affiliation(s)
| | - Thrisha Senthilnathan
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Sophia Paiola
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Priya Gyani
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | | | - Estrella Urena
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Akash Jeysankar
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Prakash Jeysankar
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | | | | | - Helen Lavretsky
- Jane and Terry Semel Institute for Neuroscience and Human BehaviorUniversity of CaliforniaLos AngelesCAUSA
| | - Shantanu Joshi
- Department of NeurologyUniversity of CaliforniaLos AngelesCAUSA
| | - Gustavo Garcia
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
| | - Arunachalam Ramaiah
- Department of Ecology and Evolutionary BiologyUniversity of CaliforniaIrvineCAUSA
- Tata Institute for Genetics and SocietyCenter at inStemBangaloreKAIndia
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical PharmacologyUniversity of CaliforniaLos AngelesCAUSA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell ResearchUniversity of CaliforniaLos AngelesCAUSA
- California NanoSystems InstituteUniversity of CaliforniaLos AngelesCAUSA
| |
Collapse
|
21
|
Antisense Oligonucleotide-Based Therapy of Viral Infections. Pharmaceutics 2021; 13:pharmaceutics13122015. [PMID: 34959297 PMCID: PMC8707165 DOI: 10.3390/pharmaceutics13122015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Nucleic acid-based therapeutics have demonstrated their efficacy in the treatment of various diseases and vaccine development. Antisense oligonucleotide (ASO) technology exploits a single-strand short oligonucleotide to either cause target RNA degradation or sterically block the binding of cellular factors or machineries to the target RNA. Chemical modification or bioconjugation of ASOs can enhance both its pharmacokinetic and pharmacodynamic performance, and it enables customization for a specific clinical purpose. ASO-based therapies have been used for treatment of genetic disorders, cancer and viral infections. In particular, ASOs can be rapidly developed for newly emerging virus and their reemerging variants. This review discusses ASO modifications and delivery options as well as the design of antiviral ASOs. A better understanding of the viral life cycle and virus-host interactions as well as advances in oligonucleotide technology will benefit the development of ASO-based antiviral therapies.
Collapse
|
22
|
Gargan S, Stevenson NJ. Unravelling the Immunomodulatory Effects of Viral Ion Channels, towards the Treatment of Disease. Viruses 2021; 13:2165. [PMID: 34834972 PMCID: PMC8618147 DOI: 10.3390/v13112165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/07/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
The current COVID-19 pandemic has highlighted the need for the research community to develop a better understanding of viruses, in particular their modes of infection and replicative lifecycles, to aid in the development of novel vaccines and much needed anti-viral therapeutics. Several viruses express proteins capable of forming pores in host cellular membranes, termed "Viroporins". They are a family of small hydrophobic proteins, with at least one amphipathic domain, which characteristically form oligomeric structures with central hydrophilic domains. Consequently, they can facilitate the transport of ions through the hydrophilic core. Viroporins localise to host membranes such as the endoplasmic reticulum and regulate ion homeostasis creating a favourable environment for viral infection. Viroporins also contribute to viral immune evasion via several mechanisms. Given that viroporins are often essential for virion assembly and egress, and as their structural features tend to be evolutionarily conserved, they are attractive targets for anti-viral therapeutics. This review discusses the current knowledge of several viroporins, namely Influenza A virus (IAV) M2, Human Immunodeficiency Virus (HIV)-1 Viral protein U (Vpu), Hepatitis C Virus (HCV) p7, Human Papillomavirus (HPV)-16 E5, Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) Open Reading Frame (ORF)3a and Polyomavirus agnoprotein. We highlight the intricate but broad immunomodulatory effects of these viroporins and discuss the current antiviral therapies that target them; continually highlighting the need for future investigations to focus on novel therapeutics in the treatment of existing and future emergent viruses.
Collapse
Affiliation(s)
- Siobhan Gargan
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland;
| | - Nigel J. Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland;
- Viral Immunology Group, Royal College of Surgeons in Ireland-Medical University of Bahrain, Manama 15503, Bahrain
| |
Collapse
|
23
|
Fiorino S, Tateo F, Biase DD, Gallo CG, Orlandi PE, Corazza I, Budriesi R, Micucci M, Visani M, Loggi E, Hong W, Pica R, Lari F, Zippi M. SARS-CoV-2: lessons from both the history of medicine and from the biological behavior of other well-known viruses. Future Microbiol 2021; 16:1105-1133. [PMID: 34468163 PMCID: PMC8412036 DOI: 10.2217/fmb-2021-0064] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2 is the etiological agent of the current pandemic worldwide and its associated disease COVID-19. In this review, we have analyzed SARS-CoV-2 characteristics and those ones of other well-known RNA viruses viz. HIV, HCV and Influenza viruses, collecting their historical data, clinical manifestations and pathogenetic mechanisms. The aim of the work is obtaining useful insights and lessons for a better understanding of SARS-CoV-2. These pathogens present a distinct mode of transmission, as SARS-CoV-2 and Influenza viruses are airborne, whereas HIV and HCV are bloodborne. However, these viruses exhibit some potential similar clinical manifestations and pathogenetic mechanisms and their understanding may contribute to establishing preventive measures and new therapies against SARS-CoV-2.
Collapse
Affiliation(s)
- Sirio Fiorino
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL, Bologna, 40054, Italy
| | - Fabio Tateo
- Institute of Geosciences & Earth Resources, CNR, c/o Department of Geosciences, Padova University, 35127, Italy
| | - Dario De Biase
- Department of Pharmacy & Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Claudio G Gallo
- Fisiolaserterapico Emiliano, Castel San Pietro Terme, Bologna, 40024, Italy
| | | | - Ivan Corazza
- Department of Experimental, Diagnostic & Specialty Medicine, University of Bologna, Bologna, 40126, Italy
| | - Roberta Budriesi
- Department of Pharmacy & Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, 40126, Italy
| | - Matteo Micucci
- Department of Pharmacy & Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, 40126, Italy
| | - Michela Visani
- Department of Pharmacy & Biotechnology, University of Bologna, Bologna, 40126, Italy
| | - Elisabetta Loggi
- Hepatology Unit, Department of Medical & Surgical Sciences, University of Bologna, Bologna, 40126, Italy
| | - Wandong Hong
- Department of Gastroenterology & Hepatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang, 325035, PR China
| | - Roberta Pica
- Unit of Gastroenterology & Digestive Endoscopy, Sandro Pertini Hospital, Rome, 00157, Italy
| | - Federico Lari
- Internal Medicine Unit, Budrio Hospital, Budrio (Bologna), Azienda USL, Bologna, 40054, Italy
| | - Maddalena Zippi
- Unit of Gastroenterology & Digestive Endoscopy, Sandro Pertini Hospital, Rome, 00157, Italy
| |
Collapse
|
24
|
Zhou R, Liu L, Wang Y. Viral proteins recognized by different TLRs. J Med Virol 2021; 93:6116-6123. [PMID: 34375002 DOI: 10.1002/jmv.27265] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/08/2021] [Accepted: 08/08/2021] [Indexed: 12/24/2022]
Abstract
Virus invasion activates the host's innate immune response, inducing the production of numerous cytokines and interferons to eliminate pathogens. Except for viral DNA/RNA, viral proteins are also targets of pattern recognition receptors. Membrane-bound receptors such as Toll-like receptor (TLR)1, TLR2, TLR4, TLR6, and TLR10 relate to the recognition of viral proteins. Distinct TLRs perform both protective and detrimental roles for a specific virus. Here, we review viral proteins serving as pathogen-associated molecular patterns and their corresponding TLRs. These viruses are all enveloped, including respiratory syncytial virus, hepatitis C virus, measles virus, herpesvirus human immunodeficiency virus, and coronavirus, and can encode proteins to activate innate immunity in a TLR-dependent way. The TLR-viral protein relationship plays an important role in innate immunity activation. A detailed understanding of their pathways contributes to a novel direction for vaccine development.
Collapse
Affiliation(s)
- Rui Zhou
- Department of Microbiology, Institute of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Li Liu
- Department of Microbiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yu Wang
- Department of Microbiology, Institute of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| |
Collapse
|
25
|
Gallardo CM, Wang S, Montiel-Garcia DJ, Little SJ, Smith DM, Routh AL, Torbett BE. MrHAMER yields highly accurate single molecule viral sequences enabling analysis of intra-host evolution. Nucleic Acids Res 2021; 49:e70. [PMID: 33849057 PMCID: PMC8266615 DOI: 10.1093/nar/gkab231] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022] Open
Abstract
Technical challenges remain in the sequencing of RNA viruses due to their high intra-host diversity. This bottleneck is particularly pronounced when interrogating long-range co-evolved genetic interactions given the read-length limitations of next-generation sequencing platforms. This has hampered the direct observation of these genetic interactions that code for protein-protein interfaces with relevance in both drug and vaccine development. Here we overcome these technical limitations by developing a nanopore-based long-range viral sequencing pipeline that yields accurate single molecule sequences of circulating virions from clinical samples. We demonstrate its utility in observing the evolution of individual HIV Gag-Pol genomes in response to antiviral pressure. Our pipeline, called Multi-read Hairpin Mediated Error-correction Reaction (MrHAMER), yields >1000s of viral genomes per sample at 99.9% accuracy, maintains the original proportion of sequenced virions present in a complex mixture, and allows the detection of rare viral genomes with their associated mutations present at <1% frequency. This method facilitates scalable investigation of genetic correlates of resistance to both antiviral therapy and immune pressure and enables the identification of novel host-viral and viral-viral interfaces that can be modulated for therapeutic benefit.
Collapse
Affiliation(s)
- Christian M Gallardo
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Shiyi Wang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA
| | - Daniel J Montiel-Garcia
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Susan J Little
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Davey M Smith
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA.,Veterans Affairs San Diego Healthcare System, San Diego, CA, USA
| | - Andrew L Routh
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA.,Sealy Center for Structural Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Bruce E Torbett
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA, USA.,Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
26
|
Mahoney KE, Shabanowitz J, Hunt DF. MHC Phosphopeptides: Promising Targets for Immunotherapy of Cancer and Other Chronic Diseases. Mol Cell Proteomics 2021; 20:100112. [PMID: 34129940 PMCID: PMC8724925 DOI: 10.1016/j.mcpro.2021.100112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/11/2021] [Accepted: 06/02/2021] [Indexed: 12/27/2022] Open
Abstract
Major histocompatibility complex-associated peptides have been considered as potential immunotherapeutic targets for many years. MHC class I phosphopeptides result from dysregulated cell signaling pathways that are common across cancers and both viral and bacterial infections. These antigens are recognized by central memory T cells from healthy donors, indicating that they are considered antigenic by the immune system and that they are presented across different individuals and diseases. Based on these responses and the similar dysregulation, phosphorylated antigens are promising candidates for prevention or treatment of different cancers as well as a number of other chronic diseases.
Collapse
Affiliation(s)
- Keira E Mahoney
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA.
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA; Department of Pathology, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
27
|
Al-Mozaini M, Alzahrani A, Alsharif I, Shinwari Z, Halim M, Alhokail A, Alrajhi A, Alaiya A. Quantitative proteomics analysis reveals unique but overlapping protein signatures in HIV infections. J Infect Public Health 2021; 14:795-802. [PMID: 34030014 DOI: 10.1016/j.jiph.2021.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/11/2021] [Accepted: 03/25/2021] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus-1 (HIV-1) exploits human host factors to complete its life cycle. Hence, discovery of HIV-regulated host proteins markers would better our understanding of the virus life-cycle and its contribution to pathogenesis and discovery of objective diagnostic and prognostic molecules. METHODS We conducted holistic total proteomics analysis of three closely related study populations including patients with HIV type-1 (HIV-1) and HIV type-2 (HIV-2) as well as HIV-1 elite controllers (HIV-1-EC). Peripheral blood plasma (PBP) samples were subjected to label-free quantitative liquid-chromatography tandem mass-spectrometry (LC-MS/MS). RESULTS Over 314 unique PBP protein species were identified of which 100 (approx. 32%) were significantly differentially expressed (≥2 to ∞ - fold-change; p < 0.05) between the three sample cohorts. Of the 100 proteins, 91 were significantly changed between pairs of HIV-1 versus HIV-1-EC, while 83 of the 100 proteins differed significantly between HIV-2 and HIV-1-EC. Interestingly, 76 proteins (87.5%) overlap between the two data sets indicating that majority of these proteins share similar expression changes between HIV-1 and HIV-2 sample groups. Two of the identified proteins, XRCC5 and PSME1, were implicated in the early phase of the pathway network for HIV life cycle, while others were involved in infectious disease and disease of signal transduction. Among them were MAP2K1, RPL23A, RPS3, CALR, PRDX1, SOD2, LMNB1, PHB, and FGB. Despite the high degree of similarity in protein profiles of HIV-1 and HIV-2, six proteins differed significantly including ETFB, PHB2, S100A9, LMO2, PPP3R1 and Vif, a fragment of virion infectivity factor of HIV-1. Additionally, 15 proteins were uniquely expressed, and one of them (LSP1) is present only in HIV-1-EC but absent in HIV1 and HIV-2 and vice versa for the rest 14 proteins. CONCLUSIONS Altogether, we have identified HIV-specific/related protein expression changes that might potentially be capable of early diagnosis and prognosis of HIV diseases and other related infectious diseases.
Collapse
Affiliation(s)
- Maha Al-Mozaini
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia; Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, KSA, Saudi Arabia.
| | - Alhusain Alzahrani
- College of Applied Medical Sciences, University of Hafr Al Baten, Hafr Al Baten, KSA, Saudi Arabia.
| | - Ibtihaj Alsharif
- Immunocompromised Host Research Section, Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| | - Zakia Shinwari
- Proteomics Unit, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| | - Magid Halim
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| | - Abdullah Alhokail
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| | - Abdulrahman Alrajhi
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| | - Ayodele Alaiya
- Proteomics Unit, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, KSA, Saudi Arabia.
| |
Collapse
|
28
|
Li G, Xu M, Yue T, Gu W, Tan L. Life-long passion for antiviral research and drug development: 80th birthday of Prof. Dr. Erik De Clercq. Biochem Pharmacol 2021; 185:114485. [PMID: 33617841 PMCID: PMC7895689 DOI: 10.1016/j.bcp.2021.114485] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022]
Abstract
Since the 1950s, great efforts have been made to develop antiviral agents against many infectious diseases such as human immunodeficiency virus (HIV), hepatitis B virus (HBV), hepatitis C virus (HCV), human cytomegalovirus (HCMV), herpes simplex virus (HSV), and varicella-zoster virus (VZV). Among the list of nearly 106 antiviral agents approved in the past five decades, Prof. Erik De Clercq has contributed to the development of 7 antiviral drugs: tenofovir disoproxil fumarate (Viread®) for HIV and HBV treatment, tenofovir alafenamide (Vemlidy®) for HIV and HBV treatment, brivudine (Zostex®) for HSV-1 and VZV treatment, valacyclovir (Valtrex®) for HSV and VZV treatment, adefovir dipivoxil (Hepsera®) for HBV treatment, stavudine (Zerit®) for HIV treatment, and cidofovir (Vistide®) for treating HCMV retinitis in AIDS patients. In addition to the above antiviral drugs, his contributions include two anti-cancer drugs: rabacfosadine (Tanovea®-CA1) for canine lymphoma and plerixafor (Mozobil®) for multiple myeloma and non-Hodgkin's lymphoma. These achievements are driven by his life-long passions for antiviral research and successful collaborations worldwide. To honor the 80th birthday of Prof. Erik De Clercq, this study highlights his scientific achievements and the importance of life-long passions and collaborations in the success of antiviral research and drug development.
Collapse
Affiliation(s)
- Guangdi Li
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Ming Xu
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Tingting Yue
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410078, China
| | - Weijie Gu
- Laboratory of Medicinal Chemistry, Rega Institute for Medical Research, KU Leuven, Leuven 3000, Belgium
| | - Li Tan
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
29
|
Construction of a Prokaryotic Expression Vector harboring Two HIV-1 Accessory Genes. MEDICAL LABORATORY JOURNAL 2021. [DOI: 10.52547/mlj.15.2.11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
30
|
Yadavar-Nikravesh MS, Milani A, Vahabpour R, Khoobi M, Bakhshandeh H, Bolhassani A. In vitro Anti-HIV-1 Activity of the Recombinant HIV-1 TAT Protein Along With Tenofovir Drug. Curr HIV Res 2021; 19:138-146. [PMID: 33045968 DOI: 10.2174/1570162x18666201012152600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/19/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND HIV-1 TAT protein is essential for the regulation of viral genome transcription. The first exon of TAT protein has a fundamental role in the stimulation of the extrinsic and intrinsic apoptosis pathways, but its anti-HIV activity is not clear yet. METHODS In the current study, we firstly cloned the first exon of the TAT coding sequence in the pET-24a expression vector and then protein expression was done in the Rosetta expression host. Next, the expressed TAT protein was purified by Ni-NTA column under native conditions. After that, the protein yield was determined by Bradford kit and NanoDrop spectrophotometry. Finally, the cytotoxicity effect and anti-Scr-HIV-1 activity of the recombinant TAT protein alone and along with Tenofovir drug were assessed by MTT and ELISA, respectively. RESULTS The recombinant TAT protein was successfully generated in E. coli, as confirmed by 13.5% SDS-PAGE and western blotting. The protein yield was ~150-200 μg/ml. In addition, the recombinant TAT protein at a certain dose with low toxicity could suppress Scr-HIV replication in the infected HeLa cells (~30%) that was comparable with a low toxic dose of Tenofovir drug (~40%). It was interesting that the recombinant TAT protein could enhance anti-HIV potency of Tenofovir drug up to 66%. CONCLUSION Generally, a combination of TAT protein and Tenofovir drug could significantly inhibit HIV-1 replication. It will be required to determine their mechanism of action in the next studies.
Collapse
Affiliation(s)
| | - Alireza Milani
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Rouhollah Vahabpour
- Department of Medical Lab Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences; Tehran, Iran
| | - Mehdi Khoobi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Haleh Bakhshandeh
- Nanobiotechnology Department, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis, AIDS and Blood Borne Diseases, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
31
|
Seeherman S, Suzuki YJ. Viral Infection and Cardiovascular Disease: Implications for the Molecular Basis of COVID-19 Pathogenesis. Int J Mol Sci 2021; 22:ijms22041659. [PMID: 33562193 PMCID: PMC7914972 DOI: 10.3390/ijms22041659] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
The current pandemic of coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). While this respiratory virus only causes mild symptoms in younger healthy individuals, elderly people and those with cardiovascular diseases such as systemic hypertension are susceptible to developing severe conditions that can be fatal. SARS-CoV-2 infection is also associated with an increased incidence of cardiovascular diseases such as myocardial injury, acute coronary syndrome, and thromboembolism. Understanding the mechanisms of the effects of this virus on the cardiovascular system should thus help develop therapeutic strategies to reduce the mortality and morbidity associated with SARS-CoV-2 infection. Since this virus causes severe and fatal conditions in older individuals with cardiovascular comorbidities, effective therapies targeting specific populations will likely contribute to ending this pandemic. In this review article, the effects of various viruses—including other coronaviruses, influenza, dengue, and human immunodeficiency virus—on the cardiovascular system are described to help provide molecular mechanisms of pathologies associated with SARS-CoV-2 infection and COVID-19. The goal is to provide mechanistic information from the biology of other viral infections in relation to cardiovascular pathologies for the purpose of developing improved vaccines and therapeutic agents effective in preventing and/or treating the acute and long-term consequences of SARS-CoV-2 and COVID-19.
Collapse
Affiliation(s)
- Sarah Seeherman
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA;
| | - Yuichiro J. Suzuki
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20007, USA
- Correspondence:
| |
Collapse
|
32
|
Dinesh DC, Tamilarasan S, Rajaram K, Bouřa E. Antiviral Drug Targets of Single-Stranded RNA Viruses Causing Chronic Human Diseases. Curr Drug Targets 2021; 21:105-124. [PMID: 31538891 DOI: 10.2174/1389450119666190920153247] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 08/08/2019] [Accepted: 08/08/2019] [Indexed: 02/08/2023]
Abstract
Ribonucleic acid (RNA) viruses associated with chronic diseases in humans are major threats to public health causing high mortality globally. The high mutation rate of RNA viruses helps them to escape the immune response and also is responsible for the development of drug resistance. Chronic infections caused by human immunodeficiency virus (HIV) and hepatitis viruses (HBV and HCV) lead to acquired immunodeficiency syndrome (AIDS) and hepatocellular carcinoma respectively, which are one of the major causes of human deaths. Effective preventative measures to limit chronic and re-emerging viral infections are absolutely necessary. Each class of antiviral agents targets a specific stage in the viral life cycle and inhibits them from its development and proliferation. Most often, antiviral drugs target a specific viral protein, therefore only a few broad-spectrum drugs are available. This review will be focused on the selected viral target proteins of pathogenic viruses containing single-stranded (ss) RNA genome that causes chronic infections in humans (e.g. HIV, HCV, Flaviviruses). In the recent past, an exponential increase in the number of available three-dimensional protein structures (>150000 in Protein Data Bank), allowed us to better understand the molecular mechanism of action of protein targets and antivirals. Advancements in the in silico approaches paved the way to design and develop several novels, highly specific small-molecule inhibitors targeting the viral proteins.
Collapse
Affiliation(s)
| | - Selvaraj Tamilarasan
- Section of Microbial Biotechnology, Charles Tanford Protein Center, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Kaushik Rajaram
- Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| | - Evžen Bouřa
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
33
|
Abstract
Over the past 60 years, more than 100 antiviral drugs or their combinations have been approved for clinical use. Antiviral drugs can be classified according to their chemical nature (e.g., small-molecules, peptides, biologics) or mechanisms of drug actions against specific viral proteins (e.g., polymerase inhibitors, protease inhibitors, glycoprotein inhibitors). This article provides an overview of antiviral classifications in 10 important human viruses: hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), human cytomegalovirus (HCMV), herpes simplex virus (HSV), variola virus (human smallpox), varicella zoster virus (VZV), influenza virus, respiratory syncytial virus (RSV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
|
34
|
Ng'uni T, Chasara C, Ndhlovu ZM. Major Scientific Hurdles in HIV Vaccine Development: Historical Perspective and Future Directions. Front Immunol 2020; 11:590780. [PMID: 33193428 PMCID: PMC7655734 DOI: 10.3389/fimmu.2020.590780] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/05/2020] [Indexed: 12/15/2022] Open
Abstract
Following the discovery of HIV as a causative agent of AIDS, the expectation was to rapidly develop a vaccine; but thirty years later, we still do not have a licensed vaccine. Progress has been hindered by the extensive genetic variability of HIV and our limited understanding of immune responses required to protect against HIV acquisition. Nonetheless, valuable knowledge accrued from numerous basic and translational science research studies and vaccine trials has provided insight into the structural biology of the virus, immunogen design and novel vaccine delivery systems that will likely constitute an effective vaccine. Furthermore, stakeholders now appreciate the daunting scientific challenges of developing an effective HIV vaccine, hence the increased advocacy for collaborative efforts among academic research scientists, governments, pharmaceutical industry, philanthropy, and regulatory entities. In this review, we highlight the history of HIV vaccine development efforts, highlighting major challenges and future directions.
Collapse
Affiliation(s)
- Tiza Ng'uni
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Caroline Chasara
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Zaza M Ndhlovu
- KwaZulu-Natal Research Institute for Tuberculosis and HIV (K-RITH), Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA, United States
| |
Collapse
|
35
|
Szojka Z, Mótyán JA, Miczi M, Mahdi M, Tőzsér J. Y44A Mutation in the Acidic Domain of HIV-2 Tat Impairs Viral Reverse Transcription and LTR-Transactivation. Int J Mol Sci 2020; 21:ijms21165907. [PMID: 32824587 PMCID: PMC7460587 DOI: 10.3390/ijms21165907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/10/2020] [Accepted: 08/15/2020] [Indexed: 11/28/2022] Open
Abstract
HIV transactivator protein (Tat) plays a pivotal role in viral replication through modulation of cellular transcription factors and transactivation of viral genomic transcription. The effect of HIV-1 Tat on reverse transcription has long been described in the literature, however, that of HIV-2 is understudied. Sequence homology between Tat proteins of HIV-1 and 2 is estimated to be less than 30%, and the main difference lies within their N-terminal region. Here, we describe Y44A-inactivating mutation of HIV-2 Tat, studying its effect on capsid production, reverse transcription, and the efficiency of proviral transcription. Investigation of the mutation was performed using sequence- and structure-based in silico analysis and in vitro experiments. Our results indicate that the Y44A mutant HIV-2 Tat inhibited the activity and expression of RT (reverse transcriptase), in addition to diminishing Tat-dependent LTR (long terminal repeat) transactivation. These findings highlight the functional importance of the acidic domain of HIV-2 Tat in the regulation of reverse transcription and transactivation of the integrated provirions.
Collapse
Affiliation(s)
- Zsófia Szojka
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.A.M.); (M.M.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - János András Mótyán
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.A.M.); (M.M.)
| | - Márió Miczi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.A.M.); (M.M.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Mohamed Mahdi
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.A.M.); (M.M.)
- Correspondence: (M.M.); (J.T.)
| | - József Tőzsér
- Laboratory of Retroviral Biochemistry, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Z.S.); (J.A.M.); (M.M.)
- Correspondence: (M.M.); (J.T.)
| |
Collapse
|
36
|
Peixoto RT, Nogueira LFS, de Oliveira SA, Souza VD, Felipo BSL. Study of HIV Resistance Mutations Against Antiretrovirals using Bioinformatics Tools. Curr HIV Res 2020; 17:343-349. [PMID: 31629397 DOI: 10.2174/1570162x17666191019114250] [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: 04/29/2019] [Revised: 09/25/2019] [Accepted: 10/08/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Antiretroviral drugs to HIV-1 (ARV) are divided into classes: Nucleotide Reverse Transcriptase Inhibitors (NRTIs); Non-Nucleoside Reverse Transcriptase Inhibitors (NNRTIs); Protease Inhibitors (PIs); Integrase Inhibitors (INIs); fusion inhibitors and entry Inhibitors. The occurrence of mutations developing resistance to antiretroviral drugs used in HIV treatment take place in a considerable proportion and has accumulated over its long period of therapy. OBJECTIVE This study aimed to identify resistance mutations to antiretrovirals used in the treatment of HIV-1 in strains isolated from Brazilian territory deposited at Genbank, as well as to relate to the clinical significance and mechanism of action. METHODS Elucidation of these mutations was by comparative method of peptide sequence resulting from genes encoding therapeutic targets in HIV antiretroviral therapy (ART) of the strains with a reference sequence through bioinformatic genetic information manipulation techniques. RESULTS Of the 399 sequences analyzed, 121 (30.3%) had some type of mutations associated with resistance to some class of antiretroviral drug. Resistance to NNRTIs was the most prevalent, detected in 77 (63.6%) of the 121 mutated sequences, compared to NRTIs and PIs, whose resistance was detected in 60 (49.6%) and 21 (17.3%), respectively, and to INIs, only 1 (0.8%) sample showed associated resistance mutation. CONCLUSION Resistance to HIV ARV was detected at a considerable rate of 30.3%, showing some concerns about the percentage of viral strains that escape the established therapeutic regimen and that circulate currently in Brazil. The non-use of NNRTIs in Brazil is justified by the emergence of resistance mutations. The low prevalence of mutations against INIs is because drugs in this class have a high genetic barrier.
Collapse
Affiliation(s)
- Roca Tárcio Peixoto
- Faculdades Integradas Aparicio Carvalho-FIMCA, School of Biomedicine, Research Group on Health Sciences, Porto Velho, Brazil.,Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia-FIOCRUZ-RO, Porto Velho, Brazil.,Universidade Federal de Rondônia - UNIR, Porto Velho, Brazil
| | - Lima Felipe Souza Nogueira
- Faculdades Integradas Aparicio Carvalho-FIMCA, School of Biomedicine, Research Group on Health Sciences, Porto Velho, Brazil.,Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia-FIOCRUZ-RO, Porto Velho, Brazil.,Universidade Federal de Rondônia - UNIR, Porto Velho, Brazil
| | - Santos Alcione de Oliveira
- Faculdades Integradas Aparicio Carvalho-FIMCA, School of Biomedicine, Research Group on Health Sciences, Porto Velho, Brazil.,Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia-FIOCRUZ-RO, Porto Velho, Brazil.,Universidade Federal de Rondônia - UNIR, Porto Velho, Brazil
| | - Vieira Deusilene Souza
- Faculdades Integradas Aparicio Carvalho-FIMCA, School of Biomedicine, Research Group on Health Sciences, Porto Velho, Brazil.,Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia-FIOCRUZ-RO, Porto Velho, Brazil.,Universidade Federal de Rondônia - UNIR, Porto Velho, Brazil
| | - Botelho-Souza Luan Felipo
- Faculdades Integradas Aparicio Carvalho-FIMCA, School of Biomedicine, Research Group on Health Sciences, Porto Velho, Brazil.,Laboratório de Virologia Molecular, Fundação Oswaldo Cruz Rondônia-FIOCRUZ-RO, Porto Velho, Brazil.,Universidade Federal de Rondônia - UNIR, Porto Velho, Brazil.,National Institutes of Science and Technology - CNPq- INCT-EpiAmO, Porto Velho, Brazil
| |
Collapse
|
37
|
Kardani K, Bolhassani A, Agi E, Hashemi A. B1 protein: a novel cell penetrating protein for in vitro and in vivo delivery of HIV-1 multi-epitope DNA constructs. Biotechnol Lett 2020; 42:1847-1863. [PMID: 32449070 PMCID: PMC7246087 DOI: 10.1007/s10529-020-02918-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/17/2020] [Indexed: 12/02/2022]
Abstract
Objectives Enhancement of the potential ability of biomacromolecules to cross cell membranes is a critical step for development of effective therapeutic vaccine especially DNA vaccine against human immunodeficiency virus-1 (HIV-1) infection. The supercharged proteins were known as powerful weapons for delivery of different types of cargoes such as DNA and protein. Hence, we applied B1 protein with + 43 net charges obtained from a single frameshift in the gene encoding enhanced green fluorescent protein (eGFP) for delivery of two multi-epitope DNA constructs (nef-vpu-gp160-p24 and nef-vif-gp160-p24) in vitro and in vivo for the first time. For this purpose, B1 protein was generated in bacterial expression system under native conditions, and used to interact with both DNA constructs. Results Our data indicated that B1 protein (~ 27 kDa) was able to form a stable nanoparticle (~ 80–110 nm) with both DNA constructs at nitrogen: phosphate (N: P) ratio of 1:1. Moreover, the transfection efficiency of B1 protein for DNA delivery into HEK-293T cell line indicated that the cellular uptake of nef-vif-gp160-p24 DNA/ B1 and nef-vpu-gp160-p24 DNA/ B1 nanoparticles was about 32–35% with lower intensity as compared to TurboFect commercial reagent. On the other hand, immunization of BALB/c mice with different modalities demonstrated that B1 protein could enhance the levels of antibody, IFN-gamma and Granzyme B eliciting potent and strong Th1-directed cellular immunity. Conclusion Generally, our findings showed the potency of B1 protein as a promising gene delivery system to improve an effective therapeutic vaccine against HIV-1 infection.
Collapse
Affiliation(s)
- Kimia Kardani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran.
| | - Elnaz Agi
- Iranian Comprehensive Hemophilia Care Center, Tehran, Iran
| | - Atieh Hashemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Barillari G. The Anti-Angiogenic Effects of Anti-Human Immunodeficiency Virus Drugs. Front Oncol 2020; 10:806. [PMID: 32528888 PMCID: PMC7253758 DOI: 10.3389/fonc.2020.00806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 04/23/2020] [Indexed: 12/17/2022] Open
Abstract
The growth and metastasis of malignant tumors benefit from the formation of blood vessels within the tumor area. There, new vessels originate from angiogenesis (the sprouting of pre-existing neighboring vessels) and/or vasculogenesis (the mobilization of bone marrow-derived endothelial cell precursors which incorporate in tumor vasculature and then differentiate into mature endothelial cells). These events are induced by soluble molecules (the angiogenic factors) and modulated by endothelial cell interactions with the perivascular matrix. Given angiogenesis/vasculogenesis relevance to tumor progression, anti-angiogenic drugs are often employed to buttress surgery, chemotherapy or radiation therapy in the treatment of a wide variety of cancers. Most of the anti-angiogenic drugs have been developed to functionally impair the angiogenic vascular endothelial growth factor: however, this leaves other angiogenic factors unaffected, hence leading to drug resistance and escape. Other anti-angiogenic strategies have exploited classical inhibitors of enzymes remodeling the perivascular matrix. Disappointingly, these inhibitors have been found toxic and/or ineffective in clinical trials, even though they block angiogenesis in pre-clinical models. These findings are stimulating the identification of other anti-angiogenic compounds. In this regard, it is noteworthy that drugs utilized for a long time to counteract human immune deficiency virus (HIV) can directly and effectively hamper molecular pathways leading to blood vessel formation. In this review the mechanisms leading to angiogenesis and vasculogenesis, and their susceptibility to anti-HIV drugs will be discussed.
Collapse
Affiliation(s)
- Giovanni Barillari
- Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
39
|
Shirvani P, Fassihi A, Saghaie L, Van Belle S, Debyser Z, Christ F. Synthesis, anti-HIV-1 and antiproliferative evaluation of novel 4-nitroimidazole derivatives combined with 5-hydroxy-4-pyridinone moiety. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
40
|
Sudderuddin H, Kinloch NN, Jin SW, Miller RL, Jones BR, Brumme CJ, Joy JB, Brockman MA, Brumme ZL. Longitudinal within-host evolution of HIV Nef-mediated CD4, HLA and SERINC5 downregulation activity: a case study. Retrovirology 2020; 17:3. [PMID: 31918727 PMCID: PMC6953280 DOI: 10.1186/s12977-019-0510-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/26/2019] [Indexed: 11/29/2022] Open
Abstract
The HIV accessory protein Nef downregulates the viral entry receptor CD4, the Human Leukocyte Antigen (HLA)-A and -B molecules, the Serine incorporator 5 (SERINC5) protein and other molecules from the infected cell surface, thereby promoting viral infectivity, replication and immune evasion. The nef locus also represents one of the most genetically variable regions in the HIV genome, and nef sequences undergo substantial evolution within a single individual over the course of infection. Few studies however have simultaneously characterized the impact of within-host nef sequence evolution on Nef protein function over prolonged timescales. Here, we isolated 50 unique Nef clones by single-genome amplification over an 11-year period from the plasma of an individual who was largely naïve to antiretroviral treatment during this time. Together, these clones harbored nonsynonymous substitutions at 13% of nef’s codons. We assessed their ability to downregulate cell-surface CD4, HLA and SERINC5 and observed that all three Nef functions declined modestly over time, where the reductions in CD4 and HLA downregulation (an average of 0.6% and 2.0% per year, respectively) achieved statistical significance. The results from this case study support all three Nef activities as being important to maintain throughout untreated HIV infection, but nevertheless suggest that, despite nef’s mutational plasticity, within-host viral evolution can compromise Nef function, albeit modestly, over prolonged periods.
Collapse
Affiliation(s)
- Hanwei Sudderuddin
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.,BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Natalie N Kinloch
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.,BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Steven W Jin
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Rachel L Miller
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | | | - Chanson J Brumme
- BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey B Joy
- BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mark A Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.,BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada
| | - Zabrina L Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada. .,BC Centre for Excellence in HIV/AIDS, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Rezaei T, Khalili S, Baradaran B, Mosafer J, Rezaei S, Mokhtarzadeh A, de la Guardia M. Recent advances on HIV DNA vaccines development: Stepwise improvements to clinical trials. J Control Release 2019; 316:116-137. [PMID: 31669566 DOI: 10.1016/j.jconrel.2019.10.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 01/10/2023]
Abstract
According to WHO (World Health Organization) reports, more than 770,000 people died from HIV and almost 1.7 million people becoming newly infected in the worldwide in 2018. Therefore, many attempts should be done to produce a forceful vaccine to control the AIDS. DNA-based vaccines have been investigated for HIV vaccination by researches during the recent 20 years. The DNA vaccines are novel approach for induction of both type of immune responses (cellular and humoral) in the host cells and have many advantages including high stability, fast and easy of fabrication and absence of severe side effects when compared with other vaccination methods. Recent studies have been focused on vaccine design, immune responses and on the use of adjuvants as a promising strategy for increased level of responses, delivery approaches by viral and non-viral methods and vector design for different antigens of HIV virus. In this review, we outlined the aforementioned advances on HIV DNA vaccines. Then we described the future trends in clinical trials as a strong strategy even in healthy volunteers and the potential developments in control and prevention of HIV.
Collapse
Affiliation(s)
- Tayebeh Rezaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajee Teacher Training University, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Mosafer
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Sarah Rezaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biotechnology, Higher Education Institute of Rab-Rashid, Tabriz, Iran.
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100, Burjassot, Valencia, Spain.
| |
Collapse
|
42
|
Wang Y, De Clercq E, Li G. Current and emerging non-nucleoside reverse transcriptase inhibitors (NNRTIs) for HIV-1 treatment. Expert Opin Drug Metab Toxicol 2019; 15:813-829. [PMID: 31556749 DOI: 10.1080/17425255.2019.1673367] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction: Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are essential components of highly active antiretroviral therapy against HIV-1 infections. Here, we provide a comprehensive overview of approved and emerging NNRTIs. Areas covered: This review covers the latest trend of NNRTIs regarding their pharmacodynamics, pharmacokinetics, mechanisms of drug action, drug resistance as well as new applications such as two-drug regimens and long-acting formulations. Expert opinion: Since the first NNRTI, nevirapine, was approved in 1996, antiviral drug discovery led to the approval of seven NNRTIs, including nevirapine, delavirdine (discontinued), etravirine, elsulfavirine, efavirenz, rilpivirine, and doravirine. The latter three compounds with favorable pharmacodynamic profiles and minimal adverse effects are often combined with one integrase inhibitor or two NRTIs in once-daily fixed-dose tablets. NNRTI-anchored regimens have been approved as initial therapies in treatment-naïve patients (efficacy: 72% to 86%) or maintaining therapies in virologically-suppressed patients (efficacy: 91% to 95%). Future development of NNRTIs includes: (i) better resistance and cross-resistance profiles; (ii) reduction of drug burden by optimizing two-drug or three-drug combinations; and (iii) improvement of patient adherence by novel long-acting formulations with weekly or monthly administration. Overall, NNRTIs play an important role in the management of HIV-1 infections, especially in resource-limited countries.
Collapse
Affiliation(s)
- Yali Wang
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University , Changsha , Hunan , China
| | - Erik De Clercq
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research , Leuven , Belgium
| | - Guangdi Li
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University , Changsha , Hunan , China
| |
Collapse
|
43
|
Abdulla F, Adhikari UK, Uddin MK. Exploring T & B-cell epitopes and designing multi-epitope subunit vaccine targeting integration step of HIV-1 lifecycle using immunoinformatics approach. Microb Pathog 2019; 137:103791. [PMID: 31606417 DOI: 10.1016/j.micpath.2019.103791] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 10/08/2019] [Accepted: 10/08/2019] [Indexed: 12/14/2022]
Abstract
Till now, AIDS, caused by the human immunodeficiency virus (HIV) is still a severe health problem worldwide. It weakens the immune system by targeting the T-helper cells. Specifically, the severity of the pandemic HIV-1 makes the emergence of an enduring effective vaccine against HIV-1. Therefore, we have applied a series of immunoinformatics approaches to the four conserved domains of HIV-1 integrase (IN) proteins to design an effective multi-epitope based subunit vaccine which might induce a competent immunity against HIV-1. Therefore, we have selected three peptide fragments that contained all overlapping epitopes (35 CD4+, 8 CD8+ T-cell epitopes, and 3 B-cell epitopes) where the epitopes had a high conservancy score. The cumulative population coverage for combined CD8+ and CD4+ T-cell epitopes and their respective HLA-alleles were found as 98.03% in the world which is also followed by East Asia (96.24%), South Asia (96.31%), North Africa (96.14%), North America (98.99%), and Europe (98.80%). The proposed vaccine composed by an adjuvant (β-defensin) at the N-terminal site of the vaccine constructs and three peptide fragments where the adjuvant was fused by EAAAK linker and the peptide fragments were fused by GPGPG linker. The designed final vaccine construct (length: 159 amino acid) was found to be antigenic and non-allergic, which indicates its safety. The vaccine construct was found as good antigenic, stable, higher thermostable, and hydrophilic in nature. The codon adaptation and in silico cloning ensured the high expression rate of the vaccine constructs in E. coli K12 with CAI value of 1.0. Finally, the binding affinity of the vaccine constructs with the immune receptor TLR3 was confirmed by the lowest energy score of -1026.8 evaluated by molecular docking. However, the proposed in silico vaccine construct needs experimental validation for assuring the safety and immunogenicity profile which will ensure an active immunity against HIV-1.
Collapse
Affiliation(s)
- Faruq Abdulla
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh
| | - Utpal Kumar Adhikari
- School of Medicine, Western Sydney University, Campbelltown, NSW-2560, Australia
| | - M Kamal Uddin
- Department of Statistics, Islamic University, Kushtia-7003, Bangladesh.
| |
Collapse
|
44
|
Yager EJ, Konan KV. Sphingolipids as Potential Therapeutic Targets against Enveloped Human RNA Viruses. Viruses 2019; 11:v11100912. [PMID: 31581580 PMCID: PMC6832137 DOI: 10.3390/v11100912] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 12/28/2022] Open
Abstract
Several notable human diseases are caused by enveloped RNA viruses: influenza, AIDS, hepatitis C, dengue hemorrhagic fever, microcephaly, and Guillain-Barré Syndrome. Being enveloped, the life cycle of this group of viruses is critically dependent on host lipid biosynthesis. Viral binding and entry involve interactions between viral envelope glycoproteins and cellular receptors localized to lipid-rich regions of the plasma membrane. Subsequent infection by these viruses leads to reorganization of cellular membranes and lipid metabolism to support the production of new viral particles. Recent work has focused on defining the involvement of specific lipid classes in the entry, genome replication assembly, and viral particle formation of these viruses in hopes of identifying potential therapeutic targets for the treatment or prevention of disease. In this review, we will highlight the role of host sphingolipids in the lifecycle of several medically important enveloped RNA viruses.
Collapse
Affiliation(s)
- Eric J Yager
- Department of Basic and Clinical Sciences, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| | - Kouacou V Konan
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208-3479, USA.
| |
Collapse
|
45
|
Wang Y, Chang J, Wang J, Zhong P, Zhang Y, Lai CC, He Y. 3D-QSAR Studies of S-DABO Derivatives as Non-nucleoside HIV-1 Reverse Transcriptase Inhibitors. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180815666180810112321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
S-dihydro-alkyloxy-benzyl-oxopyrimidines (S-DABOs) as non-nucleoside
reverse transcriptase inhibitors have received considerable attention during the last decade due to
their high potency against HIV-1.
Methods:
In this study, three-dimensional quantitative structure-activity relationship (3D-QSAR) of
a series of 38 S-DABO analogues developed in our lab was studied using Comparative Molecular
Field Analysis (CoMFA) and Comparative Molecular Similarity Indices Analysis (CoMSIA). The
Docking/MMFF94s computational protocol based on the co-crystallized complex (PDB ID: 1RT2)
was used to determine the most probable binding mode and to obtain reliable conformations for
molecular alignment. Statistically significant CoMFA (q2=0.766 and r2=0.949) and CoMSIA
(q2=0.827 and r2=0.974) models were generated using the training set of 30 compounds on the basis
of hybrid docking-based and ligand-based alignment.
Results:
The predictive ability of CoMFA and CoMSIA models was further validated using a test
set of eight compounds with predictive r2
pred values of 0.843 and 0.723, respectively.
Conclusion:
The information obtained from the 3D contour maps can be used in designing new SDABO
derivatives with improved HIV-1 inhibitory activity.
Collapse
Affiliation(s)
- Yueping Wang
- Department of Applied Chemistry, Faculty of Science, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Jie Chang
- Department of Applied Chemistry, Faculty of Science, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Jiangyuan Wang
- Key Laboratory of Medicinal Chemistry for Natural Resource, (Ministry of Education), School of Chemical Science and Technology, Yunnan University, Kunming Yunnan, 650091, China
| | - Peng Zhong
- Department of Applied Chemistry, Faculty of Science, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yufang Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource, (Ministry of Education), School of Chemical Science and Technology, Yunnan University, Kunming Yunnan, 650091, China
| | - Christopher Cong Lai
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Yanping He
- Key Laboratory of Medicinal Chemistry for Natural Resource, (Ministry of Education), School of Chemical Science and Technology, Yunnan University, Kunming Yunnan, 650091, China
| |
Collapse
|
46
|
Balasubramaniam M, Pandhare J, Dash C. Immune Control of HIV. JOURNAL OF LIFE SCIENCES (WESTLAKE VILLAGE, CALIF.) 2019; 1:4-37. [PMID: 31468033 PMCID: PMC6714987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The human immunodeficiency virus (HIV) infection of the immune cells expressing the cluster of differentiation 4 cell surface glycoprotein (CD4+ cells) causes progressive decline of the immune system and leads to the acquired immunodeficiency syndrome (AIDS). The ongoing global HIV/AIDS pandemic has already claimed over 35 million lives. Even after 37 years into the epidemic, neither a cure is available for the 37 million people living with HIV (PLHIV) nor is a vaccine discovered to avert the millions of new HIV infections that continue to occur each year. If left untreated, HIV infection typically progresses to AIDS and, ultimately, causes death in a majority of PLHIV. The recommended combination antiretroviral therapy (cART) suppresses virus replication and viremia, prevents or delays progression to AIDS, reduces transmission rates, and lowers HIV-associated mortality and morbidity. However, because cART does not eliminate HIV, and an enduring pool of infected resting memory CD4+ T cells (latent HIV reservoir) is established early on, any interruption to cART leads to a relapse of viremia and disease progression. Hence, strict adherence to a life-long cART regimen is mandatory for managing HIV infection in PLHIV. The HIV-1-specific cytotoxic T cells expressing the CD8 glycoprotein (CD8+ CTL) limit the virus replication in vivo by recognizing the viral antigens presented by human leukocyte antigen (HLA) class I molecules on the infected cell surface and killing those cells. Nevertheless, CTLs fail to durably control HIV-1 replication and disease progression in the absence of cART. Intriguingly, <1% of cART-naive HIV-infected individuals called elite controllers/HIV controllers (HCs) exhibit the core features that define a HIV-1 "functional cure" outcome in the absence of cART: durable viral suppression to below the limit of detection, long-term non-progression to AIDS, and absence of viral transmission. Robust HIV-1-specific CTL responses and prevalence of protective HLA alleles associated with enduring HIV-1 control have been linked to the HC phenotype. An understanding of the molecular mechanisms underlying the CTL-mediated suppression of HIV-1 replication and disease progression in HCs carrying specific protective HLA alleles may yield promising insights towards advancing the research on HIV cure and prophylactic HIV vaccine.
Collapse
Affiliation(s)
- Muthukumar Balasubramaniam
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN – 37208. USA
| | - Jui Pandhare
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN – 37208. USA
| | - Chandravanu Dash
- Center for AIDS Health Disparities Research, Meharry Medical College, Nashville, TN – 37208. USA
- Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN – 37208. USA
- School of Graduate Studies and Research, Meharry Medical College, Nashville, TN – 37208. USA
| |
Collapse
|
47
|
D Urbano V, De Crignis E, Re MC. Host Restriction Factors and Human Immunodeficiency Virus (HIV-1): A Dynamic Interplay Involving All Phases of the Viral Life Cycle. Curr HIV Res 2019; 16:184-207. [PMID: 30117396 DOI: 10.2174/1570162x16666180817115830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/31/2018] [Accepted: 08/09/2018] [Indexed: 02/08/2023]
Abstract
Mammalian cells have evolved several mechanisms to prevent or block lentiviral infection and spread. Among the innate immune mechanisms, the signaling cascade triggered by type I interferon (IFN) plays a pivotal role in limiting the burden of HIV-1. In the presence of IFN, human cells upregulate the expression of a number of genes, referred to as IFN-stimulated genes (ISGs), many of them acting as antiviral restriction factors (RFs). RFs are dominant proteins that target different essential steps of the viral cycle, thereby providing an early line of defense against the virus. The identification and characterization of RFs have provided unique insights into the molecular biology of HIV-1, further revealing the complex host-pathogen interplay that characterizes the infection. The presence of RFs drove viral evolution, forcing the virus to develop specific proteins to counteract their activity. The knowledge of the mechanisms that prevent viral infection and their viral counterparts may offer new insights to improve current antiviral strategies. This review provides an overview of the RFs targeting HIV-1 replication and the mechanisms that regulate their expression as well as their impact on viral replication and the clinical course of the disease.
Collapse
Affiliation(s)
- Vanessa D Urbano
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Elisa De Crignis
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Maria Carla Re
- Retrovirus Laboratory, Operative Unit of Clinical Microbiology, S. Orsola-Malpighi University Hospital, Bologna, Italy
| |
Collapse
|
48
|
Cat and Mouse: HIV Transcription in Latency, Immune Evasion and Cure/Remission Strategies. Viruses 2019; 11:v11030269. [PMID: 30889861 PMCID: PMC6466452 DOI: 10.3390/v11030269] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/04/2019] [Accepted: 03/13/2019] [Indexed: 12/13/2022] Open
Abstract
There is broad scientific and societal consensus that finding a cure for HIV infection must be pursued. The major barrier to achieving a cure for HIV/AIDS is the capacity of the HIV virus to avoid both immune surveillance and current antiretroviral therapy (ART) by rapidly establishing latently infected cell populations, termed latent reservoirs. Here, we provide an overview of the rapidly evolving field of HIV cure/remission research, highlighting recent progress and ongoing challenges in the understanding of HIV reservoirs, the role of HIV transcription in latency and immune evasion. We review the major approaches towards a cure that are currently being explored and further argue that small molecules that inhibit HIV transcription, and therefore uncouple HIV gene expression from signals sent by the host immune response, might be a particularly promising approach to attain a cure or remission. We emphasize that a better understanding of the game of "cat and mouse" between the host immune system and the HIV virus is a crucial knowledge gap to be filled in both cure and vaccine research.
Collapse
|
49
|
Carpi LC, Schieber TA, Pardalos PM, Marfany G, Masoller C, Díaz-Guilera A, Ravetti MG. Assessing diversity in multiplex networks. Sci Rep 2019; 9:4511. [PMID: 30872604 PMCID: PMC6418208 DOI: 10.1038/s41598-019-38869-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023] Open
Abstract
Diversity, understood as the variety of different elements or configurations that an extensive system has, is a crucial property that allows maintaining the system's functionality in a changing environment, where failures, random events or malicious attacks are often unavoidable. Despite the relevance of preserving diversity in the context of ecology, biology, transport, finances, etc., the elements or configurations that more contribute to the diversity are often unknown, and thus, they can not be protected against failures or environmental crises. This is due to the fact that there is no generic framework that allows identifying which elements or configurations have crucial roles in preserving the diversity of the system. Existing methods treat the level of heterogeneity of a system as a measure of its diversity, being unsuitable when systems are composed of a large number of elements with different attributes and types of interactions. Besides, with limited resources, one needs to find the best preservation policy, i.e., one needs to solve an optimization problem. Here we aim to bridge this gap by developing a metric between labeled graphs to compute the diversity of the system, which allows identifying the most relevant components, based on their contribution to a global diversity value. The proposed framework is suitable for large multiplex structures, which are constituted by a set of elements represented as nodes, which have different types of interactions, represented as layers. The proposed method allows us to find, in a genetic network (HIV-1), the elements with the highest diversity values, while in a European airline network, we systematically identify the companies that maximize (and those that less compromise) the variety of options for routes connecting different airports.
Collapse
Affiliation(s)
- Laura C Carpi
- Programa de Pós-Graduação em Modelagem Matemática e Computacional, PPGMMC, Centro Federal de Educação Tecnológica de Minas Gerais, CEFET-MG. Av. Amazonas, 7675. 30510-000., Belo Horizonte, MG, Brazil
| | - Tiago A Schieber
- Departamento de Ciências Administrativas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Panos M Pardalos
- Industrial and Systems Engineering, University of Florida, Gainesville, FL, USA
| | - Gemma Marfany
- Departament de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB-IRSJD), Barcelona, Spain
| | - Cristina Masoller
- Departament de Física, Universitat Politècnica de Catalunya, Rambla St. Nebridi 22, Terrassa, 08222, Barcelona, Spain
| | - Albert Díaz-Guilera
- Departament de Física de la Matèria Condensada, Universitat de Barcelona, Marti i Franques 1, Barcelona, 08028, Spain
- Universitat de Barcelona Institute of Complex Systems (UBICS), 08028, Barcelona, Spain
| | - Martín G Ravetti
- Departmento de Engenharia de Produção, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
50
|
Fleming AM, Nguyen NLB, Burrows CJ. Colocalization of m 6A and G-Quadruplex-Forming Sequences in Viral RNA (HIV, Zika, Hepatitis B, and SV40) Suggests Topological Control of Adenosine N 6-Methylation. ACS CENTRAL SCIENCE 2019; 5:218-228. [PMID: 30834310 PMCID: PMC6396389 DOI: 10.1021/acscentsci.8b00963] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Indexed: 05/09/2023]
Abstract
This Outlook calls attention to two seemingly disparate and emerging fields regarding viral genomics that may be correlated in a way previously overlooked. First, we describe identification of conserved potential G-quadruplex-forming sequences (PQSs) in viral genomes relevant to human health. Studies have demonstrated that PQSs are highly conserved and can fold to G-quadruplexes (G4s) to regulate viral processes. Key examples include G4s as a countermeasure to the host's immune system or G4-guided regulation of replication or transcription. Second, emerging data are discussed concerning the epitranscriptomic modification N 6-methyladenosine (m6A) in viral RNA installed by host proteins in a consensus sequence favoring 5'-GG(m6A)C-3'. The proposed pathways by which m6A is written, read, and erased in viral RNA genomes and the impact this has on viral replication are described. The structural reason why certain sites are selected for modification while others are not is still mysterious. Finally, we discuss our new observations regarding these previous sequencing data that identify m6A installation within the loops of two-tetrad PQSs in the RNA genomes of the Zika, HIV, hepatitis B, and SV40 viruses. We hypothesize that conserved viral PQSs can provide a framework (sequence and/or structural) for m6A installation. We also discuss literature sources suggesting that PQSs as sites of RNA modification could be a general phenomenon. We anticipate our observations will provide ample opportunities for exciting discoveries regarding the interplay between G4 structures and epitranscriptomic modifications of RNA.
Collapse
Affiliation(s)
- Aaron M. Fleming
- Department of Chemistry, University
of Utah, Salt Lake
City, Utah 84112-0850, United States
| | - Ngoc L. B. Nguyen
- Department of Chemistry, University
of Utah, Salt Lake
City, Utah 84112-0850, United States
| | - Cynthia J. Burrows
- Department of Chemistry, University
of Utah, Salt Lake
City, Utah 84112-0850, United States
| |
Collapse
|