1
|
Al Meanazel O, Alshbool FZ, Khasawneh FT. Design and Pharmacological Characterization of a Novel Antithrombotic P2Y 1 Receptor-Based Vaccine. Int J Mol Sci 2025; 26:4383. [PMID: 40362620 PMCID: PMC12072916 DOI: 10.3390/ijms26094383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Platelet activation processes begin when injury to blood vessels exposes the subendothelial matrix, leading platelets to attach to it, where they become activated and exert their hemostatic function. Excessive platelet aggregation is associated with thrombotic disorders such as arterial thrombosis. To manage such diseases, medications that inhibit thrombosis are continuously sought, despite potential drawbacks that include hemorrhage. This study described the development of a novel peptide-based vaccine that targets the purinergic ADP P2Y1 receptor (abbreviated EL2Vac) and its pharmacological characterization. Thus, we designed and developed an EL2Vac that targets the ligand-binding domain of the P2Y1 receptor protein, which is located in its second extracellular loop (EL2). We then evaluated the vaccine's ability to trigger an immune response (antibody production) in immunized mice, modulate platelet function, its antithrombotic activity, and any effects on hemostasis, by employing a thrombosis model and the tail bleeding time assay. Results showed significant levels of antibody production in mice treated with EL2Vac, in comparison with the random peptide vaccine control (EL2rVac), which persisted at least up to six months post vaccination. Moreover, we observed significant inhibition of the ADP-induced aggregation response in platelets from EL2Vac-treated mice, relative to those from EL2rVac controls. This inhibition was selective for ADP, as other agonists, such as the thromboxane A2 receptor (TPR) agonist U46619 or high-dose collagen, had no detectable effect on aggregation. As for its capacity to protect against thrombosis, our data showed a significant delay in the occlusion time of the EL2Vac mice when compared with the random peptide control vaccine, which was also observed (at least) six months post vaccination. Interestingly, EL2Vac did not appear to prolong the tail bleeding time, supporting the notion that it is devoid of a bleeding diathesis. As a conclusion, this study documents the design and evaluation of a novel peptide-based vaccine, EL2Vac, which appears to selectively target the P2Y1 receptor and protect against thrombus formation without impairing hemostasis. Thus, EL2Vac may provide a promising clinical option to treat thromboembolic disorders.
Collapse
Affiliation(s)
- Osaid Al Meanazel
- Department of Pharmacy Practice, College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (O.A.M.); (F.Z.A.)
| | - Fatima Z. Alshbool
- Department of Pharmacy Practice, College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA; (O.A.M.); (F.Z.A.)
| | - Fadi T. Khasawneh
- Department of Pharmaceutical Sciences, College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| |
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 PMCID: PMC11867397 DOI: 10.1371/journal.pone.0318523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [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
|
Kalams SA, Felber BK, Mullins JI, Scott HM, Allen MA, De Rosa SC, Heptinstall J, Tomaras GD, Hu J, DeCamp AC, Rosati M, Bear J, Pensiero MN, Eldridge J, Egan MA, Hannaman D, McElrath MJ, Pavlakis GN, HIV Vaccine Trials Network 119(HVTN 119) Study Team. Focusing HIV-1 Gag T cell responses to highly conserved regions by DNA vaccination in HVTN 119. JCI Insight 2024; 9:e180819. [PMID: 39088271 PMCID: PMC11466283 DOI: 10.1172/jci.insight.180819] [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: 03/04/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUNDAn HIV-1 DNA vaccine composed of 7 highly conserved, structurally important elements (conserved elements, CE) of p24Gag was tested in a phase I randomized, double-blind clinical trial (HVTN 119, NCT03181789) in people without HIV. DNA vaccination of CE prime/CE+p55Gag boost was compared with p55Gag.METHODSTwo groups (n = 25) received 4 DNA vaccinations (CE/CE+p55Gag or p55Gag) by intramuscular injection/electroporation, including IL-12 DNA adjuvant. The placebo group (n = 6) received saline. Participants were followed for safety and tolerability. Immunogenicity was assessed for T cell and antibody responses.RESULTSBoth regimens were safe and generally well tolerated. The p24CE vaccine was immunogenic and significantly boosted by CE+p55Gag (64% CD4+, P = 0.037; 42% CD8+, P = 0.004). CE+p55Gag induced responses to 5 of 7 CE, compared with only 2 CE by p55Gag DNA, with a higher response to CE5 in 30% of individuals (P = 0.006). CE+p55Gag induced significantly higher CD4+ CE T cell breadth (0.68 vs. 0.22 CE; P = 0.029) and a strong trend for overall T cell breadth (1.14 vs. 0.52 CE; P = 0.051). Both groups developed high cellular and humoral responses. p24CE vaccine-induced CD4+ CE T cell responses correlated (P = 0.007) with p24Gag antibody responses.CONCLUSIONThe CE/CE+p55Gag DNA vaccine induced T cell responses to conserved regions in p24Gag, increasing breadth and epitope recognition throughout p55Gag compared with p55Gag DNA. Vaccines focusing immune responses by priming responses to highly conserved regions could be part of a comprehensive HIV vaccine strategy.TRIAL REGISTRATIONClinical Trials.gov NCT03181789FUNDINGHVTN, NIAID/NIH.
Collapse
Affiliation(s)
- Spyros A. Kalams
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee, USA
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - James I. Mullins
- Departments of Microbiology, Medicine and Global Health, University of Washington, Seattle, Washington, USA
| | - Hyman M. Scott
- San Francisco Department of Public Health, San Francisco, California, USA
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mary A. Allen
- Division of AIDS, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - Stephen C. De Rosa
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jack Heptinstall
- Duke Center for Human Systems Immunology, Departments of Surgery, Integrative Immunobiology, Molecular Genetics, and Microbiology, Durham, North Carolina, USA
| | - Georgia D. Tomaras
- Duke Center for Human Systems Immunology, Departments of Surgery, Integrative Immunobiology, Molecular Genetics, and Microbiology, Durham, North Carolina, USA
| | - Jiani Hu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Allan C. DeCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Margherita Rosati
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | - Michael N. Pensiero
- Division of AIDS, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Rockville, Maryland, USA
| | - John Eldridge
- Auro Vaccines LLC (formerly Profectus BioSciences, Inc.), Pearl River, New York, USA
| | - Michael A. Egan
- Auro Vaccines LLC (formerly Profectus BioSciences, Inc.), Pearl River, New York, USA
| | | | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, USA
| | | |
Collapse
|
4
|
Valentin A, Bergamaschi C, Rosati M, Angel M, Burns R, Agarwal M, Gergen J, Petsch B, Oostvogels L, Loeliger E, Chew KW, Deeks SG, Mullins JI, Pavlakis GN, Felber BK. Comparative immunogenicity of an mRNA/LNP and a DNA vaccine targeting HIV gag conserved elements in macaques. Front Immunol 2022; 13:945706. [PMID: 35935984 PMCID: PMC9355630 DOI: 10.3389/fimmu.2022.945706] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/24/2022] [Indexed: 01/14/2023] Open
Abstract
Immunogenicity of HIV-1 mRNA vaccine regimens was analyzed in a non-human primate animal model. Rhesus macaques immunized with mRNA in lipid nanoparticle (mRNA/LNP) formulation expressing HIV-1 Gag and Gag conserved regions (CE) as immunogens developed robust, durable antibody responses but low adaptive T-cell responses. Augmentation of the dose resulted in modest increases in vaccine-induced cellular immunity, with no difference in humoral responses. The gag mRNA/lipid nanoparticle (LNP) vaccine provided suboptimal priming of T cell responses for a heterologous DNA booster vaccination regimen. In contrast, a single immunization with gag mRNA/LNP efficiently boosted both humoral and cellular responses in macaques previously primed by a gag DNA-based vaccine. These anamnestic cellular responses were mediated by activated CD8+ T cells with a phenotype of differentiated T-bet+ cytotoxic memory T lymphocytes. The heterologous prime/boost regimens combining DNA and mRNA/LNP vaccine modalities maximized vaccine-induced cellular and humoral immune responses. Analysis of cytokine responses revealed a transient systemic signature characterized by the release of type I interferon, IL-15 and IFN-related chemokines. The pro-inflammatory status induced by the mRNA/LNP vaccine was also characterized by IL-23 and IL-6, concomitant with the release of IL-17 family of cytokines. Overall, the strong boost of cellular and humoral immunity induced by the mRNA/LNP vaccine suggests that it could be useful as a prophylactic vaccine in heterologous prime/boost modality and in immune therapeutic interventions against HIV infection or other chronic human diseases.
Collapse
Affiliation(s)
- Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Matthew Angel
- Vaccine Branch, Center for Cancer Research, National Cncer Institute, Bethesda, MD, United States
- Center for Cancer Research Collaborative Bioinformatics Resource, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Robert Burns
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Mahesh Agarwal
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | | | | | | | | | - Kara W. Chew
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, CA, United States
| | - James I. Mullins
- Department of Microbiology, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
- *Correspondence: Barbara K. Felber,
| |
Collapse
|
5
|
Zhang H, He C, Jiang F, Cao S, Zhao B, Ding H, Dong T, Han X, Shang H. A longitudinal analysis of immune escapes from HLA-B*13-restricted T-cell responses at early stage of CRF01_AE subtype HIV-1 infection and implications for vaccine design. BMC Immunol 2022; 23:15. [PMID: 35366796 PMCID: PMC8976269 DOI: 10.1186/s12865-022-00491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/24/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Identifying immunogens which can elicit effective T cell responses against human immunodeficiency virus type 1 (HIV-1) is important for developing a T-cell based vaccine. It has been reported that human leukocyte antigen (HLA)-B*13-restricted T-cell responses contributed to HIV control in subtype B' and C infected individuals. However, the kinetics of B*13-restricted T-cell responses, viral evolution within epitopes, and the impact on disease progression in CRF01_AE subtype HIV-1-infected men who have sex with men (MSM) are not known. RESULTS Interferon-γ ELISPOT assays and deep sequencing of viral RNAs were done in 14 early HLA-B*13-positive CRF01_AE subtype HIV-1-infected MSM. We found that responses to RQEILDLWV (Nef106-114, RV9), GQMREPRGSDI (Gag226-236, GI11), GQDQWTYQI (Pol487-498, GI9), and VQNAQGQMV (Gag135-143, VV9) were dominant. A higher relative magnitude of Gag-specific T-cell responses, contributed to viral control, whereas Nef-specific T-cell responses were associated with rapid disease progression. GI11 (Gag) was conserved and strong GI11 (Gag)-specific T-cell responses showed cross-reactivity with a dominant variant, M228I, found in 3/12 patients; GI11 (Gag)-specific T-cell responses were positively associated with CD4 T-cell counts (R = 0.716, P = 0.046). Interestingly, the GI9 (Pol) epitope was also conserved, but GI9 (Pol)-specific T-cell responses did not influence disease progression (P > 0.05), while a D490G variant identified in one patient did not affect CD4 T-cell counts. All the other epitopes studied [VV9 (Gag), RQYDQILIEI (Pol113-122, RI10), HQSLSPRTL (Gag144-152, HL9), and RQANFLGRL (Gag429-437, RL9)] developed escape mutations within 1 year of infection, which may have contributed to overall disease progression. Intriguingly, we found early RV9 (Nef)-specific T-cell responses were associated with rapid disease progression, likely due to escape mutations. CONCLUSIONS Our study strongly suggested the inclusion of GI11 (Gag) and exclusion of RV9 (Nef) for T-cell-based vaccine design for B*13-positive CRF01_AE subtype HIV-1-infected MSM and high-risk individuals.
Collapse
Affiliation(s)
- Hui Zhang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Chuan He
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412636.40000 0004 1757 9485Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Fanming Jiang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412636.40000 0004 1757 9485Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| | - Shuang Cao
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China ,grid.412449.e0000 0000 9678 1884Department of Laboratory Medicine, China Medical University Shengjing Hospital Nanhu Branch, Shenyang, 110001 China
| | - Bin Zhao
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Haibo Ding
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Tao Dong
- grid.4991.50000 0004 1936 8948Nuffield Department of Medicine, Chinese Academy of Medical Sciences Oxford Institute, Oxford University, Oxford, UK ,grid.4991.50000 0004 1936 8948Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK
| | - Xiaoxu Han
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| | - Hong Shang
- grid.412636.40000 0004 1757 9485NHC Key Laboratory of AIDS Immunology (China Medical University), National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001 Liaoning Province China ,Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001 China ,Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001 China ,grid.13402.340000 0004 1759 700XCollaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Street, Hangzhou, 310003 China
| |
Collapse
|
6
|
Nkone P, Loubser S, Quinn TC, Redd AD, Ismail A, Tiemessen CT, Mayaphi SH. Deep sequencing of the HIV-1 polymerase gene for characterisation of cytotoxic T-lymphocyte epitopes during early and chronic disease stages. Virol J 2022; 19:56. [PMID: 35346259 PMCID: PMC8959563 DOI: 10.1186/s12985-022-01772-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/07/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Despite multiple attempts, there is still no effective HIV-1 vaccine available. The HIV-1 polymerase (pol) gene is highly conserved and encodes cytotoxic T-lymphocyte (CTL) epitopes. The aim of the study was to characterise HIV-1 Pol CTL epitopes in mostly sample pairs obtained during early and chronic stages of infection. METHODS Illumina deep sequencing was performed for all samples while Sanger sequencing was only performed on baseline samples. Codons under immune selection pressure were assessed by computing nonsynonymous to synonymous mutation ratios using MEGA. Minority CTL epitope variants occurring at [Formula: see text] 5% were detected using low-frequency variant tool in CLC Genomics. Los Alamos HIV database was used for mapping mutations to known HIV-1 CTL epitopes. RESULTS Fifty-two participants were enrolled in the study. Their median age was 28 years (interquartile range: 24-32 years) and majority of participants (92.3%) were female. Illumina minority variant analysis identified a significantly higher number of CTL epitopes (n = 65) compared to epitopes (n = 8) identified through Sanger sequencing. Most of the identified epitopes mapped to reverse transcriptase (RT) and integrase (IN) regardless of sequencing method. There was a significantly higher proportion of minority variant epitopes in RT (n = 39, 60.0%) compared to IN (n = 17, 26.2%) and PR (n = 9, 13.8%), p = 0.002 and < 0.0001, respectively. However, no significant difference was observed between the proportion of minority variant epitopes in IN versus PR, p = 0.06. Some epitopes were detected in either early or chronic HIV-1 infection whereas others were detected in both stages. Different distribution patterns of minority variant epitopes were observed in sample pairs; with some increasing or decreasing over time, while others remained constant. Some of the identified epitopes have not been previously reported for HIV-1 subtype C. There were also variants that could not be mapped to reported CTL epitopes in the Los Alamos HIV database. CONCLUSION Deep sequencing revealed many Pol CTL epitopes, including some not previously reported for HIV-1 subtype C. The findings of this study support the inclusion of RT and IN epitopes in HIV-1 vaccine candidates as these proteins harbour many CTL epitopes.
Collapse
Affiliation(s)
- Paballo Nkone
- Department of Medical Virology, University of Pretoria, Private Bag X323, Gezina, 0031, South Africa
| | - Shayne Loubser
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Thomas C Quinn
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew D Redd
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.,Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Arshad Ismail
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Caroline T Tiemessen
- National Institute for Communicable Diseases and Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Simnikiwe H Mayaphi
- Department of Medical Virology, University of Pretoria, Private Bag X323, Gezina, 0031, South Africa. .,National Health Laboratory Service-Tshwane Academic Division (NHLS-TAD), Tshwane, South Africa.
| |
Collapse
|
7
|
Identification of HIV Rapid Mutations Using Differences in Nucleotide Distribution over Time. Genes (Basel) 2022; 13:genes13020170. [PMID: 35205215 PMCID: PMC8872422 DOI: 10.3390/genes13020170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 02/07/2023] Open
Abstract
Mutation is the driving force of species evolution, which may change the genetic information of organisms and obtain selective competitive advantages to adapt to environmental changes. It may change the structure or function of translated proteins, and cause abnormal cell operation, a variety of diseases and even cancer. Therefore, it is particularly important to identify gene regions with high mutations. Mutations will cause changes in nucleotide distribution, which can be characterized by natural vectors globally. Based on natural vectors, we propose a mathematical formula for measuring the difference in nucleotide distribution over time to investigate the mutations of human immunodeficiency virus. The studied dataset is from public databases and includes gene sequences from twenty HIV-infected patients. The results show that the mutation rate of the nine major genes or gene segment regions in the genome exhibits discrepancy during the infected period, and the Env gene has the fastest mutation rate. We deduce that the peak of virus mutation has a close temporal relationship with viral divergence and diversity. The mutation study of HIV is of great significance to clinical diagnosis and drug design.
Collapse
|
8
|
Mohanty E, Mohanty A. Role of artificial intelligence in peptide vaccine design against RNA viruses. INFORMATICS IN MEDICINE UNLOCKED 2021; 26:100768. [PMID: 34722851 PMCID: PMC8536498 DOI: 10.1016/j.imu.2021.100768] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/16/2021] [Accepted: 10/16/2021] [Indexed: 01/18/2023] Open
Abstract
RNA viruses have high rate of replication and mutation that help them adapt and change according to their environmental conditions. Many viral mutants are the cause of various severe and lethal diseases. Vaccines, on the other hand have the capacity to protect us from infectious diseases by eliciting antibody or cell-mediated immune responses that are pathogen-specific. While there are a few reviews pertaining to the use of artificial intelligence (AI) for SARS-COV-2 vaccine development, none focus on peptide vaccination for RNA viruses and the important role played by AI in it. Peptide vaccine which is slowly coming to be recognized as a safe and effective vaccination strategy has the capacity to overcome the mutant escape problem which is also being currently faced by SARS-COV-2 vaccines in circulation.Here we review the present scenario of peptide vaccines which are developed using mathematical and computational statistics methods to prevent the spread of disease caused by RNA viruses. We also focus on the importance and current stage of AI and mathematical evolutionary modeling using machine learning tools in the establishment of these new peptide vaccines for the control of viral disease.
Collapse
Affiliation(s)
- Eileena Mohanty
- Trident School of Biotech Sciences, Trident Academy of Creative Technology (TACT), Bhubaneswar, Odisha, 751024, India
| | - Anima Mohanty
- School of Biotechnology (KSBT), KIIT University-2, Bhubaneswar, 751024, India
| |
Collapse
|
9
|
Khan S, Shaker B, Ahmad S, Abbasi SW, Arshad M, Haleem A, Ismail S, Zaib A, Sajjad W. Towards a novel peptide vaccine for Middle East respiratory syndrome coronavirus and its possible use against pandemic COVID-19. J Mol Liq 2021; 324:114706. [PMID: 33173250 PMCID: PMC7644433 DOI: 10.1016/j.molliq.2020.114706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/05/2020] [Accepted: 11/02/2020] [Indexed: 12/02/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is an emerging health concern due to its high mortality rate of 35%. At present, no vaccine is available to protect against MERS-CoV infections. Therefore, an in silico search for potential antigenic epitopes in the non-redundant proteome of MERS-CoV was performed herein. First, a subtractive proteome-based approach was employed to look for the surface exposed and host non-homologous proteins. Following, immunoinformatics analysis was performed to predict antigenic B and T cell epitopes that were used in the design of a multi-epitopes peptide. Molecular docking study was carried out to predict vaccine construct affinity of binding to Toll-like receptor 3 (TLR3) and understand its binding conformation to extract ideas about its processing by the host immune system. We identified membrane protein, envelope small membrane protein, non-structural protein ORF3, non-structural protein ORF5, and spike glycoprotein as potential candidates for subunit vaccine designing. The designed multi-epitope peptide then linked to β-defensin adjuvant is showing high antigenicity. Further, the sequence of the designed vaccine construct is optimized for maximum expression in the Escherichia coli expression system. A rich pattern of hydrogen and hydrophobic interactions of the construct was observed with the TLR3 allowing stable binding of the construct at the docked site as predicted by the molecular dynamics simulation and MM-PBSA binding energies. We expect that the panel of subunit vaccine candidates and the designed vaccine construct could be highly effective in immunizing populations from infections caused by MERS-CoV and could possible applied on the current pandemic COVID-19.
Collapse
Affiliation(s)
- Salman Khan
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu 73000, PR China
| | - Bilal Shaker
- School of Integrative Engineering, Chung ANG University, Seoul, South Korea
| | - Sajjad Ahmad
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Sumra Wajid Abbasi
- Department of Biological Sciences, National University of Medical Sciences, the Mall, Rawalpindi 46000, Pakistan
| | - Muhammad Arshad
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Abdul Haleem
- Department of Microbiology, Quaid-i-Azam University, Islamabad, Pakistan
| | - Saba Ismail
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Anita Zaib
- National Center for Bioinformatics, Quaid-i-Azam University, Islamabad, Pakistan
| | - Wasim Sajjad
- Department of Biological Sciences, National University of Medical Sciences, the Mall, Rawalpindi 46000, Pakistan
| |
Collapse
|
10
|
Forni D, Cagliani R, Pontremoli C, Mozzi A, Pozzoli U, Clerici M, Sironi M. Antigenic variation of SARS-CoV-2 in response to immune pressure. Mol Ecol 2020; 30:3548-3559. [PMID: 33289207 PMCID: PMC7753431 DOI: 10.1111/mec.15730] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Analysis of the bat viruses most closely related to SARS-CoV-2 indicated that the virus probably required limited adaptation to spread in humans. Nonetheless, since its introduction in human populations, SARS-CoV-2 must have been subject to the selective pressure imposed by the human immune system. We exploited the availability of a large number of high-quality SARS-CoV-2 genomes, as well as of validated epitope predictions, to show that B cell epitopes in the spike glycoprotein (S) and in the nucleocapsid protein (N) have higher diversity than nonepitope positions. Similar results were obtained for other human coronaviruses and for sarbecoviruses sampled in bats. Conversely, in the SARS-CoV-2 population, epitopes for CD4+ and CD8+ T cells were not more variable than nonepitope positions. A significant reduction in epitope variability was instead observed for some of the most immunogenic proteins (S, N, ORF8 and ORF3a). Analysis over longer evolutionary time frames indicated that this effect is not due to differential constraints. These data indicate that SARS-CoV-2 evolves to elude the host humoral immune response, whereas recognition by T cells is not actively avoided by the virus. However, we also found a trend of lower diversity of T cell epitopes for common cold coronaviruses, indicating that epitope conservation per se is not directly linked to disease severity. We suggest that conservation serves to maintain epitopes that elicit tolerizing T cell responses or induce T cells with regulatory activity.
Collapse
Affiliation(s)
- Diego Forni
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| | - Rachele Cagliani
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| | - Chiara Pontremoli
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| | - Alessandra Mozzi
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| | - Uberto Pozzoli
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| | - Mario Clerici
- Department of Physiopathology and TransplantationUniversity of MilanMilanItaly
- Don C. Gnocchi Foundation ONLUSIRCCSMilanItaly
| | - Manuela Sironi
- Scientific Institute IRCCS E. MEDEABioinformaticsBosisio PariniItaly
| |
Collapse
|
11
|
Beretta M, Migraine J, Moreau A, Essat A, Goujard C, Chaix ML, Drouin A, Bouvin-Pley M, Meyer L, Barin F, Braibant M. Common evolutionary features of the envelope glycoprotein of HIV-1 in patients belonging to a transmission chain. Sci Rep 2020; 10:16744. [PMID: 33028961 PMCID: PMC7541522 DOI: 10.1038/s41598-020-73975-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/23/2020] [Indexed: 11/09/2022] Open
Abstract
The diversity of the HIV-1 envelope glycoproteins (Env) is largely a consequence of the pressure exerted by the adaptive immune response to infection. While it was generally assumed that the neutralizing antibody (NAb) response depended mainly on the infected individual, the concept that virus-related factors could be important in inducing this response has recently emerged. Here, we analyzed the influence of the infecting viral strain in shaping NAb responses in four HIV-1 infected subjects belonging to a transmission chain. We also explored the impact of NAb responses on the functional evolution of the viral quasispecies. The four patients developed a strong autologous neutralizing antibody response that drove viral escape and coincided with a parallel evolution of their infecting quasispecies towards increasing infectious properties, increasing susceptibility to T20 and increasing resistance to both CD4 analogs and V3 loop-directed NAbs. This evolution was associated with identical Env sequence changes at several positions in the V3 loop, the fusion peptide and the HR2 domain of gp41. The common evolutionary pattern of Env in different hosts suggests that the capacity of a given Env to adapt to changing environments may be restricted by functional constraints that limit its evolutionary landscape.
Collapse
Affiliation(s)
- Maxime Beretta
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France.,Laboratory of Humoral Immunology, Department of Immunology, Institut Pasteur, Paris, France
| | - Julie Migraine
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France
| | - Alain Moreau
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France
| | - Asma Essat
- Université Paris Sud, Université Paris Saclay, CESP Inserm U1018, Le Kremlin-Bicêtre, France
| | - Cécile Goujard
- Université Paris Sud, Université Paris Saclay, CESP Inserm U1018, Le Kremlin-Bicêtre, France.,AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - Marie-Laure Chaix
- Université de Paris, Inserm U944, Paris, France.,Laboratoire de Virologie, AP-HP, Hôpital Saint Louis, Paris, France
| | - Aurélie Drouin
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France
| | | | - Laurence Meyer
- Université Paris Sud, Université Paris Saclay, CESP Inserm U1018, Le Kremlin-Bicêtre, France.,AP-HP Hôpital de Bicêtre, Le Kremlin-Bicêtre, France
| | - Francis Barin
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France.,CHRU de Tours, CNR VIH, Tours, France
| | - Martine Braibant
- Université de Tours et CHRU de Tours, Inserm U1259, Tours, France.
| |
Collapse
|
12
|
Madhavan M, Mustafa S. En route to Peptide Therapeutics for COVID 19: Harnessing Potential Antigenic Mimicry Between Viral and Human Proteins. TRANSACTIONS OF THE INDIAN NATIONAL ACADEMY OF ENGINEERING : AN INTERNATIONAL JOURNAL OF ENGINEERING AND TECHNOLOGY 2020; 5:411-415. [PMID: 38624407 PMCID: PMC7306100 DOI: 10.1007/s41403-020-00132-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 11/23/2022]
Abstract
Molecular mimicry is a general strategy used by pathogens to infect the host cells. The emergence of SARS-CoV-2 virus has resulted in more than 6,700,000 infections and 390,000 deaths worldwide. Coronavirus disease (COVID-19) is an infectious disease caused by this virus. In this project concept, we aim to focus on the peptide-protein interaction analysis using two important drug targets in SARS-CoV-2 such as spike (S) protein and nucleocapsid (N) protein. These proteins play an important role in the virus entry and encapsidation of the viral particles. Motifs or functional regions in these two proteins must be sharing sequence homology with human protein (ACE2) which may be involved in the binding mechanism. The results will show a set of motif regions which can disrupt the viral infection. Once we identify these sets of antigenic determinant regions, antibody binding activity studies can be performed by in vitro methods. Our results from this study may suggest the existence of antigenic mimicry between SARS-CoV-2 and host proteins. The hit peptide components will have therapeutic applications to be developed into a wide variety of medicinal formulations against SARS-CoV-2 such as vaccine, intranasal and inhalation formulations. Also, the choice of conserved regions will lead to development of cross protective therapeutics against wide range of coronaviruses.
Collapse
Affiliation(s)
- Maya Madhavan
- Department of Biochemistry, Govt Arts and Science College, Kulathoor, Neyyattinkara, Thiruvananthapuram, India
| | - Sabeena Mustafa
- Department of Biostatistics and Bioinformatics, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard Health Affairs (MNGHA), Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
13
|
Ndekezi C, Nkamwesiga J, Ochwo S, Kimuda MP, Mwiine FN, Tweyongyere R, Amanyire W, Muhanguzi D. Identification of Ixodid Tick-Specific Aquaporin-1 Potential Anti-tick Vaccine Epitopes: An in-silico Analysis. Front Bioeng Biotechnol 2019; 7:236. [PMID: 31612130 PMCID: PMC6775757 DOI: 10.3389/fbioe.2019.00236] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022] Open
Abstract
Ticks are arthropod vectors of pathogens of both Veterinary and Public health importance. Acaricide application, which is currently the mainstay of tick control, is hampered by high cost, the need for regular application and a selection of multi-acaricide resistant tick populations. In light of this, future tick control approaches are poised to rely on the integration of rational acaricide application and other methods, such as vaccination. To contribute to systematic research-guided efforts to produce anti-tick vaccines, we carried out an in-silico analysis of tick aquaporin-1 (AQP1) protein in order to identify tick-specific AQP1 peptide motifs that can be used in future peptide anti-tick vaccine development. We carried out multiple sequence alignment (MSA), motif analysis, homology modeling, and structural analysis to identify tick-specific AQP1 peptide motifs. BepiPred, Chou and Fasman-Turn, Karplus and Schulz Flexibility, and Parker-Hydrophilicity prediction models were used to predict these motifs' potential to induce B cell mediated immune responses. The tick AQP1 (GenBankID: QDO67142.1) protein was largely similar to the bovine AQP1 (PDB:1J4N) (23 % sequence similarity; Structural superimposition of the homology model and 14JN homotetramers gave an RMSD = 3.269 while superimposition of a single chain gave an RMSD = 1.475). Tick and bovine AQP1 transmembrane domains were largely similar while their extracellular and cytoplasmic domain loops showed variation. Two tick-specific AQP1 peptide motifs; M7 (residues 106-125, p = 5.4e-25), and M8 (residues 85-104, p = 3.3e-24) were identified. These two motifs are located on the extra-cellular AQP1 domain. Motifs; M7 and M8 showed the highest Parker-Hydrophilicity prediction immunogenicity scores of 1.784 and 1.536, respectively. These two motifs can be a good starting point for the development of potential tick AQP1 peptide-based anti-tick vaccines. Further analyses such as molecular dynamics, in vitro assays, and in vivo immunization assays are required to validate these findings.
Collapse
Affiliation(s)
- Christian Ndekezi
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Joseph Nkamwesiga
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Sylvester Ochwo
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Magambo Phillip Kimuda
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
- Research Unit in Bioinformatics, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown, South Africa
| | - Frank Norbert Mwiine
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Robert Tweyongyere
- School of Veterinary Medicine and Animal Resources, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Wilson Amanyire
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
| | - Dennis Muhanguzi
- School of Biosecurity, Biotechnical and Laboratory Science, College of Veterinary Medicine Animal Resources and Biosecurity, Makerere University, Kampala, Uganda
- Makerere University/Uganda Virus Research Institute Centre of Excellence in Infection and Immunity Research and Training, Entebbe, Uganda
| |
Collapse
|
14
|
Palmer DS, Turner I, Fidler S, Frater J, Goedhals D, Goulder P, Huang KHG, Oxenius A, Phillips R, Shapiro R, Vuuren CV, McLean AR, McVean G. Mapping the drivers of within-host pathogen evolution using massive data sets. Nat Commun 2019; 10:3017. [PMID: 31289267 PMCID: PMC6616926 DOI: 10.1038/s41467-019-10724-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 05/20/2019] [Indexed: 11/09/2022] Open
Abstract
Differences among hosts, resulting from genetic variation in the immune system or heterogeneity in drug treatment, can impact within-host pathogen evolution. Genetic association studies can potentially identify such interactions. However, extensive and correlated genetic population structure in hosts and pathogens presents a substantial risk of confounding analyses. Moreover, the multiple testing burden of interaction scanning can potentially limit power. We present a Bayesian approach for detecting host influences on pathogen evolution that exploits vast existing data sets of pathogen diversity to improve power and control for stratification. The approach models key processes, including recombination and selection, and identifies regions of the pathogen genome affected by host factors. Our simulations and empirical analysis of drug-induced selection on the HIV-1 genome show that the method recovers known associations and has superior precision-recall characteristics compared to other approaches. We build a high-resolution map of HLA-induced selection in the HIV-1 genome, identifying novel epitope-allele combinations.
Collapse
Affiliation(s)
- Duncan S Palmer
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK.
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK.
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK.
| | - Isaac Turner
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
| | - Sarah Fidler
- Division of Medicine, Wright Fleming Institute, Imperial College, London, W2 1PG, UK
| | - John Frater
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7LE, UK
| | - Dominique Goedhals
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, 4013, South Africa
| | - Philip Goulder
- Division of Infectious Diseases, University of the Free State, and 3 Military Hospital, Bloemfontein, 9300, South Africa
- Department of Paediatrics, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
| | - Kuan-Hsiang Gary Huang
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Einstein Medical Center Philadelphia, 5501 Old York Road, PA, 19141, USA
| | - Annette Oxenius
- Institute of Microbiology, Swiss Federal Institute of Technology Zurich, 8093, Zurich, Switzerland
| | - Rodney Phillips
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Peter Medawar Building for Pathogen Research, Oxford, OX1 3SY, UK
- Oxford NIHR Biomedical Research Centre, Oxford, OX3 7LE, UK
- Faculty of Medicine, UNSW Sydney, NSW, 2052, Australia
| | - Roger Shapiro
- Botswana Harvard AIDS Institute Partnership, Gaborone, BO 320, Botswana
- Department of Immunology and Infectious Diseases, Harvard TH Chan School of Public Health, Boston, MA, 02215, USA
| | - Cloete van Vuuren
- HIV Pathogenesis Programme, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, 4013, South Africa
| | - Angela R McLean
- Institute for Emerging Infections, The Oxford Martin School, Oxford, OX1 3BD, UK
- Zoology Department, University of Oxford, South Parks Road, Oxford, OX1 3PS, UK
| | - Gil McVean
- Department of Statistics, University of Oxford, 24-29 St Giles', Oxford, OX1 3LB, UK
- Wellcome Trust Centre for Human Genetics, Roosevelt Drive, Oxford, OX3 7BN, UK
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| |
Collapse
|
15
|
Bazmara S, Shadmani M, Ghasemnejad A, Aghazadeh H, Pooshang Bagheri K. In silico rational design of a novel tetra-epitope tetanus vaccine with complete population coverage using developed immunoinformatics and surface epitope mapping approaches. Med Hypotheses 2019; 130:109267. [PMID: 31383332 DOI: 10.1016/j.mehy.2019.109267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 06/03/2019] [Accepted: 06/08/2019] [Indexed: 01/01/2023]
Abstract
Presentation of many unwanted epitopes within tetanus toxoid vaccine to lymphocyte clones may lead to production of many unwanted antibodies. Moreover an ideal vaccine must cover all individuals in a population that is dependent to the kinds of human leukocyte antigen alleles. Concerning these issues, our study was aimed to in silico design of a multi-epitope tetanus vaccine (METV) in order to improve population coverage and protectivity of tetanus vaccine as well as reduction of complications. Concerning these issues, a novel rational filtration was implemented to design a novel METV using immunoinformatics and surface epitope mapping approaches. Prediction of epitopes for tetanus toxin was performed in the candidate country in which the frequency had been gathered from almost all geographical distributions. The most strong binder epitopes for major histocompatibility complex class II were selected and among them the surface epitopes of native toxin were selected. The population coverage of the selected epitopes was estimated. The final candidate epitopes had highly population coverage. Molecular docking was performed to prediction of binding affinity of our candidate epitopes to the HLA-DRB1 alleles. At first, 680 strong binder epitopes were predicted. Among them 11 epitopes were selected. Finally, 4 epitopes had the most population coverage and suggested as a tetra-epitope tetanus vaccine. 99.41% of inessential strong binders were deleted using our tree steps filtration. HLA-DP had the most roles in epitope presentation. Molecular docking analysis proved the strong binding affinity of candidate epitopes to the HLA-DRB1 alleles. In conclusion, we theoretically reduced 99.41% of unwanted antibodies using our novel filtration strategies. Our tetra-epitope tetanus vaccine showed 100% population coverage in the candidate country. Furthermore, it was demonstrated that HLA-DP and HLA-DQ had more potential in epitope presentation in comparison to HLA-DRB1.
Collapse
Affiliation(s)
- Samira Bazmara
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mahsa Shadmani
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Atefeh Ghasemnejad
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Hossein Aghazadeh
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Kamran Pooshang Bagheri
- Venom and Biotherapeutics Molecules Lab., Biotechnology Dept., Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
16
|
In silico homology modelling and prediction of novel epitopic peptides from P24 protein of Haemonchus contortus. Gene 2019; 703:102-111. [DOI: 10.1016/j.gene.2019.03.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/23/2019] [Accepted: 03/25/2019] [Indexed: 01/02/2023]
|
17
|
Lima ENDC, Piqueira JRC, Camargo M, Galinskas J, Sucupira MC, Diaz RS. Impact of antiretroviral resistance and virological failure on HIV-1 informational entropy. J Antimicrob Chemother 2019; 73:1054-1059. [PMID: 29373694 DOI: 10.1093/jac/dkx508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/07/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives The present study investigated the relationship between genomic variability and resistance of HIV-1 sequences in protease (PR) and reverse transcriptase (RT) regions of the pol gene. In addition, we analysed the resistance among 651 individuals presenting antiretroviral virological failure, from 2009 to 2011, in the state of São Paulo, Brazil. Methods The genomic variability was quantified by using informational entropy methods and the relationship between resistance and replicative fitness, as inferred by the residual viral load and CD4+ T cell count. Results The number of antiretroviral schemes is related to the number of resistance mutations in the HIV-1 PR (α = 0.2511, P = 0.0003, R2 = 0.8672) and the RT (α = 0.7892, P = 0.0001, R2 = 0.9141). Increased informational entropy rate is related to lower levels of HIV-1 viral loads (α = -0.0121, P = 0.0471, R2 = 0.7923), lower levels of CD4+ T cell counts (α = -0.0120, P = 0.0335, R2 = 0.8221) and a higher number of antiretroviral resistance-related mutations. Conclusions Less organized HIV genomes as inferred by higher levels of informational entropy relate to less competent host immune systems, lower levels of HIV replication and HIV genetic evolution as a consequence of antiretroviral resistance.
Collapse
Affiliation(s)
- Elidamar Nunes de Carvalho Lima
- Division of Infectious Diseases, Paulista School of Medicine, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil.,Telecommunication and Control Engineering Department, Engineering School, University of São Paulo, São Paulo, SP, Brazil
| | - José Roberto Castilho Piqueira
- Telecommunication and Control Engineering Department, Engineering School, University of São Paulo, São Paulo, SP, Brazil
| | - Michelle Camargo
- Division of Infectious Diseases, Paulista School of Medicine, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Juliana Galinskas
- Division of Infectious Diseases, Paulista School of Medicine, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Maria Cecilia Sucupira
- Division of Infectious Diseases, Paulista School of Medicine, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil
| | - Ricardo Sobhie Diaz
- Division of Infectious Diseases, Paulista School of Medicine, Federal University of São Paulo-UNIFESP, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Timilsina U, Ghimire D, Sharma S, Gaur R. Role of APOBEC3 proteins in proteasome inhibitor-mediated reactivation of latent HIV-1 viruses. J Gen Virol 2019; 100:523-532. [PMID: 30566069 DOI: 10.1099/jgv.0.001205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Proteasome inhibitors (PIs) have been identified as an emerging class of HIV-1 latency-reversing agents (LRAs). These inhibitors can reactivate latent HIV-1 to produce non-infectious viruses. The mechanism underlying reduced infectivity of reactivated viruses is unknown. In this study, we analysed PI-reactivated viruses using biochemical and virological assays and demonstrated that these PIs stabilized the cellular expression of HIV-1 restriction factor, APOBEC3G, facilitating its packaging in the released viruses. Using infectivity assay and immunoblotting, we observed that the reduction in viral infectivity was due to enhanced levels of functionally active APOBEC3 proteins packaged in the virions. Sequencing of the proviral genome in the target cells revealed the presence of APOBEC3 signature hypermutations. Our study strengthens the role of PIs as bifunctional LRAs and demonstrates that the loss of infectivity of reactivated HIV-1 virions may be due to the increased packaging of APOBEC3 proteins in the virus.
Collapse
Affiliation(s)
- Uddhav Timilsina
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, Delhi
| | - Dibya Ghimire
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, Delhi
| | - Shilpa Sharma
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, Delhi
| | - Ritu Gaur
- Faculty of Life Sciences and Biotechnology, South Asian University, New Delhi-110021, Delhi
| |
Collapse
|
19
|
Damilano GD, Sued O, Ruiz MJ, Ghiglione Y, Canitano F, Pando M, Turk G, Cahn P, Salomón H, Dilernia D. Computational comparison of availability in CTL/gag epitopes among patients with acute and chronic HIV-1 infection. Vaccine 2018; 36:4142-4151. [PMID: 29802001 DOI: 10.1016/j.vaccine.2018.04.086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/23/2018] [Accepted: 04/29/2018] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent studies indicate that there is selection bias for transmission of viral polymorphisms associated with higher viral fitness. Furthermore, after transmission and before a specific immune response is mounted in the recipient, the virus undergoes a number of reversions which allow an increase in their replicative capacity. These aspects, and others, affect the viral population characteristic of early acute infection. METHODS 160 singlegag-gene amplifications were obtained by limiting-dilution RT-PCR from plasma samples of 8 ARV-naïve patients with early acute infection (<30 days, 22 days average) and 8 ARV-naive patients with approximately a year of infection (10 amplicons per patient). Sanger sequencing and NGS SMRT technology (Pacific Biosciences) were implemented to sequence the amplicons. Phylogenetic analysis was performed by using MEGA 6.06. HLA-I (A and B) typing was performed by SSOP-PCR method. The chromatograms were analyzed with Sequencher 4.10. Epitopes and immune-proteosomal cleavages prediction was performed with CBS prediction server for the 30 HLA-A and -B alleles most prevalent in our population with peptide lengths from 8 to 14 mer. Cytotoxic response prediction was performed by using IEDB Analysis Resource. RESULTS After implementing epitope prediction analysis, we identified a total number of 325 possible viral epitopes present in two or more acute or chronic patients. 60.3% (n = 196) of them were present only in acute infection (prevalent acute epitopes) while 39.7% (n = 129) were present only in chronic infection (prevalent chronic epitopes). Within p24, the difference was equally dramatic with 59.4% (79/133) being acute epitopes (p < 0.05). This is consistent with progressive viral adaptation to immune response in time and further supported by the fact that cytotoxic responses prediction showed that acute epitopes are more likely to generate immune response than chronic epitopes. Interestingly, only 27.5% of acute epitopes match the population-level consensus sequence of the virus. CONCLUSIONS Our results indicate that certain non-consensus viral residues might be transmitted more frequently than consensus-residues when located in immunological relevant positions (epitopes). This observation might be relevant to the rationale behind development of an effective vaccineto reduce viral reservoir and induce functional cure of HIV infection based in prevalent acute epitopes.
Collapse
Affiliation(s)
- Gabriel Dario Damilano
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Omar Sued
- Fundación Huésped, Buenos Aires, Argentina.
| | - Maria Julia Ruiz
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Yanina Ghiglione
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Flavia Canitano
- Instituto de Investigaciones Médicas Alfredo Lanari, Buenos Aires, Argentina.
| | - Maria Pando
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Gabriela Turk
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Pedro Cahn
- Fundación Huésped, Buenos Aires, Argentina.
| | - Horacio Salomón
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina.
| | - Dario Dilernia
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA Facultad de Medicina, Universidad de Buenos Aires, Argentina; Emory University, Atlanta, USA.
| |
Collapse
|
20
|
Peptide based therapeutics and their use for the treatment of neurodegenerative and other diseases. Biomed Pharmacother 2018; 103:574-581. [PMID: 29677544 DOI: 10.1016/j.biopha.2018.04.025] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/21/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022] Open
Abstract
Bioactive peptides are actively involved in different biological functions and importantly contribute to human health, and the use of peptides as therapeutics has a long successful history in disease management. A number of peptides have wide-ranging therapeutic effects, such as antioxidant, antimicrobial, and antithrombotic effects. Neurodegenerative diseases are typically caused by abnormal aggregations of proteins or peptides, and the depositions of these aggregates in or on neurons, disrupt signaling and eventually kill neurons. During recent years, research on short peptides has advanced tremendously. This review offers a brief introduction to peptide based therapeutics and their application in disease management and provides an overview of peptide vaccines, and toxicity related issues. In addition, the importance of peptides in the management of different neurodegenerative diseases and their therapeutic applications is discussed. The present review provides an understanding of peptides and their applications for the management of different diseases, but with focus on neurodegenerative diseases. The role of peptides as anti-cancer, antimicrobial and antidiabetic agents has also been discussed.
Collapse
|
21
|
Kamori D, Ueno T. HIV-1 Tat and Viral Latency: What We Can Learn from Naturally Occurring Sequence Variations. Front Microbiol 2017; 8:80. [PMID: 28194140 PMCID: PMC5276809 DOI: 10.3389/fmicb.2017.00080] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/11/2017] [Indexed: 01/25/2023] Open
Abstract
Despite the effective use of antiretroviral therapy, the remainder of a latently HIV-1-infected reservoir mainly in the resting memory CD4+ T lymphocyte subset has provided a great setback toward viral eradication. While host transcriptional silencing machinery is thought to play a dominant role in HIV-1 latency, HIV-1 protein such as Tat, may affect both the establishment and the reversal of latency. Indeed, mutational studies have demonstrated that insufficient Tat transactivation activity can result in impaired transcription of viral genes and the establishment of latency in cell culture experiments. Because Tat protein is one of highly variable proteins within HIV-1 proteome, it is conceivable that naturally occurring Tat mutations may differentially modulate Tat functions, thereby influencing the establishment and/or the reversal of viral latency in vivo. In this mini review, we summarize the recent findings of Tat naturally occurring polymorphisms associating with host immune responses and we highlight the implication of Tat sequence variations in relation to HIV latency.
Collapse
Affiliation(s)
- Doreen Kamori
- Center for AIDS Research, Kumamoto University Kumamoto, Japan
| | - Takamasa Ueno
- Center for AIDS Research, Kumamoto UniversityKumamoto, Japan; International Research Center for Medical Sciences, Kumamoto UniversityKumamoto, Japan
| |
Collapse
|
22
|
|
23
|
Kitchen SG, Zack JA. Engineering HIV-Specific Immunity with Chimeric Antigen Receptors. AIDS Patient Care STDS 2016; 30:556-561. [PMID: 27905838 DOI: 10.1089/apc.2016.0239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
HIV remains a highly important public health and clinical issue despite many recent advances in attempting to develop a cure, which has remained elusive for most people infected with HIV. HIV disease can be controlled with pharmacologic therapies; however, these treatments are expensive, may have severe side effects, and are not curative. Consequently, an improved means to control or eliminate HIV replication is needed. Cytotoxic T lymphocytes (CTLs) play a critical role in controlling viral replication and are an important part in the ability of the immune response to eradicate most viral infections. There are considerable efforts to enhance CTL responses in HIV-infected individuals in hopes of providing the immune response with armaments to more effectively control viral replication. In this review, we discuss some of these efforts and focus on the development of a gene therapy-based approach to engineer hematopoietic stem cells with an HIV-1-specific chimeric antigen receptor, which seeks to provide an inexhaustible source of HIV-1-specific immune cells that are MHC unrestricted and superior to natural antiviral T cell responses. These efforts provide the basis for further development of T cell functional enhancement to target and treat chronic HIV infection in hopes of eradicating the virus from the body.
Collapse
Affiliation(s)
- Scott G. Kitchen
- Division of Hematology/Oncology, Department of Medicine, and UCLA Center for AIDS Research, UCLA AIDS Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Jerome A. Zack
- Division of Hematology/Oncology, Department of Medicine, and UCLA Center for AIDS Research, UCLA AIDS Institute, David Geffen School of Medicine at UCLA, Los Angeles, California
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
24
|
Acevedo-Sáenz L, Carmona-Pérez L, Velilla-Hernández PA, Delgado JC, Rugeles L MT. The APPEESFRS Peptide, Restricted by the HLA-B*35:01 Molecule, and the APPEESFRF Variant Derived from an Autologous HIV-1 Strain Induces Polyfunctional Responses in CD8+ T Cells. Biores Open Access 2015; 4:115-20. [PMID: 26309788 PMCID: PMC4497628 DOI: 10.1089/biores.2014.0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Numerous reports have focused on consensus peptides to determine CD8+ T-cell responses; however, few studies evaluated the functional profile using peptides derived from circulating strains of a specific region. We determined the effector profile and maturation phenotype of CD8+ T-cells targeting the consensus APPEESFRS (AS9) epitope and its variant APPEESFRF (AF9), previously identified. The free energy of binding, maturation phenotype, and polyfunctional profile of both peptides were similar. The magnitude of CD8+ T-cell responses to AF9 was greater than the one elicited by AS9, although the difference was not significant. The polyfunctional profile of AF9 was characterized by CD107a/interleukin-2 (IL-2)/macrophage inflammatory protein beta (MIP1β) and by interferon gamma (IFNγ)/MIP1β/tumor necrosis factor alpha (TNFα) in response to AS9. TNFα production was significantly higher in response to AF9 than to AS9, and there was a negative correlation between the absolute number of CD8+ T-cell-producing TNFα and the plasma human immunodeficiency virus (HIV) load, suggesting a role of this cytokine in the control of HIV replication.
Collapse
Affiliation(s)
- Liliana Acevedo-Sáenz
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA) , Medellín, Colombia
| | - Liseth Carmona-Pérez
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA) , Medellín, Colombia
| | | | - Julio C Delgado
- ARUP Institute for Clinical and Experimental Pathology, Department of Pathology, University of Utah School of Medicine , Salt Lake City, Utah
| | - María Teresa Rugeles L
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia (UdeA) , Medellín, Colombia
| |
Collapse
|
25
|
Jung U, Urak K, Veillette M, Nepveu-Traversy MÉ, Pham QT, Hamel S, Rossi JJ, Berthoux L. Preclinical Assessment of Mutant Human TRIM5α as an Anti-HIV-1 Transgene. Hum Gene Ther 2015; 26:664-79. [PMID: 26076730 DOI: 10.1089/hum.2015.059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Current HIV-1 gene therapy approaches aim at stopping the viral life cycle at its earliest steps, such as entry or immediate postentry events. Among the most widely adopted strategies are CCR5 downregulation/knockout and the use of broadly neutralizing antibodies. However, the long-term efficacy and side effects are still unclear. TRIM5α is an interferon-stimulated restriction factor that can intercept incoming retroviruses within one hour of cytosolic entry and potently inhibit the infectivity of restriction-sensitive viruses. The human TRIM5α (TRIM5αhu) generally does not efficiently target HIV-1, but point mutations in its capsid-binding domain can confer anti-HIV-1 activity. Although the mechanisms by which TRIM5αhu mutants inhibit HIV-1 are relatively well understood, their characterization as potential transgenes for gene therapy is lacking. Additionally, previous reports of general immune activation by overexpression of TRIM5α have hindered its broad adoption as a potential transgene. Here we demonstrate the ability of the R332G-R335G TRIM5αhu mutant to efficiently restrict highly divergent HIV-1 strains, including Group O, as well as clinical isolates bearing cytotoxic T lymphocyte escape mutations. R332G-R335G TRIM5αhu efficiently protected human lymphocytes against HIV-1 infection, even when expressed at relatively low levels following lentiviral transduction. Most importantly, under these conditions Rhesus macaque TRIM5α (TRIM5αRh) and TRIM5αhu (wild-type or mutated) had no major effects on the NF-κB pathway. Transgenic TRIM5α did not modulate the kinetics of IκBα, JunB, and TNFAIP3 expression following TNF-α treatment. Finally, we show that human lymphocytes expressing R332G-R335G TRIM5αhu have clear survival advantages over unmodified parental cells in the presence of pathogenic, replication-competent HIV-1. These results support the relevance of R332G-R335G and other mutants of TRIM5αhu as candidate effectors for HIV-1 gene therapy.
Collapse
Affiliation(s)
- Ulrike Jung
- 1 Division of Molecular & Cell Biology, Beckman Research Institute of the City of Hope , Duarte, California
| | - Kevin Urak
- 1 Division of Molecular & Cell Biology, Beckman Research Institute of the City of Hope , Duarte, California
| | - Maxime Veillette
- 2 Laboratory of Retrovirology, Department of Medical Biology, Université du Québec, Trois-Rivières, Canada
| | | | - Quang Toan Pham
- 2 Laboratory of Retrovirology, Department of Medical Biology, Université du Québec, Trois-Rivières, Canada
| | - Sophie Hamel
- 2 Laboratory of Retrovirology, Department of Medical Biology, Université du Québec, Trois-Rivières, Canada
| | - John J Rossi
- 1 Division of Molecular & Cell Biology, Beckman Research Institute of the City of Hope , Duarte, California.,3 Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, California
| | - Lionel Berthoux
- 2 Laboratory of Retrovirology, Department of Medical Biology, Université du Québec, Trois-Rivières, Canada
| |
Collapse
|
26
|
Fitness-Balanced Escape Determines Resolution of Dynamic Founder Virus Escape Processes in HIV-1 Infection. J Virol 2015. [PMID: 26223634 DOI: 10.1128/jvi.01876-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED To understand the interplay between host cytotoxic T-lymphocyte (CTL) responses and the mechanisms by which HIV-1 evades them, we studied viral evolutionary patterns associated with host CTL responses in six linked transmission pairs. HIV-1 sequences corresponding to full-length p17 and p24 gag were generated by 454 pyrosequencing for all pairs near the time of transmission, and seroconverting partners were followed for a median of 847 days postinfection. T-cell responses were screened by gamma interferon/interleukin-2 (IFN-γ/IL-2) FluoroSpot using autologous peptide sets reflecting any Gag variant present in at least 5% of sequence reads in the individual's viral population. While we found little evidence for the occurrence of CTL reversions, CTL escape processes were found to be highly dynamic, with multiple epitope variants emerging simultaneously. We found a correlation between epitope entropy and the number of epitope variants per response (r = 0.43; P = 0.05). In cases in which multiple escape mutations developed within a targeted epitope, a variant with no fitness cost became fixed in the viral population. When multiple mutations within an epitope achieved fitness-balanced escape, these escape mutants were each maintained in the viral population. Additional mutations found to confer escape but undetected in viral populations incurred high fitness costs, suggesting that functional constraints limit the available sites tolerable to escape mutations. These results further our understanding of the impact of CTL escape and reversion from the founder virus in HIV infection and contribute to the identification of immunogenic Gag regions most vulnerable to a targeted T-cell attack. IMPORTANCE Rapid diversification of the viral population is a hallmark of HIV-1 infection, and understanding the selective forces driving the emergence of viral variants can provide critical insight into the interplay between host immune responses and viral evolution. We used deep sequencing to comprehensively follow viral evolution over time in six linked HIV transmission pairs. We then mapped T-cell responses to explore if mutations arose due to adaption to the host and found that escape processes were often highly dynamic, with multiple mutations arising within targeted epitopes. When we explored the impact of these mutations on replicative capacity, we found that dynamic escape processes only resolve with the selection of mutations that conferred escape with no fitness cost to the virus. These results provide further understanding of the complicated viral-host interactions that occur during early HIV-1 infection and may help inform the design of future vaccine immunogens.
Collapse
|
27
|
Shytaj IL, Savarino A. Cell-mediated anti-Gag immunity in pharmacologically induced functional cure of simian AIDS: a 'bottleneck effect'? J Med Primatol 2015; 44:227-40. [PMID: 26058990 DOI: 10.1111/jmp.12176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND Administration of antiretroviral therapy and two experimental drugs, auranofin and buthionine sulfoximine (BSO), was previously shown to be followed by drug-free control of chronic SIVmac251 infection, decreased immune activation and increased cell-mediated anti-Gag responses. METHODS Phylogeny was analysed with Phylogeny.fr. Entropy was calculated with the specific tool of the HIV Sequence Database. The capsid Gag structure was computed using SPDBV. The bottleneck effect was simulated through an appropriate online tool. RESULTS The region of Gag predominantly targeted during control of SIVmac251 infection is highly conserved in primate lentiviruses and plays an important role in capsid architecture. Computer-aided simulations support the view that the preferential development of immune responses against this region is derived from a 'bottleneck effect' after restriction, by auranofin and BSO, of the activated lymphocyte pool. CONCLUSIONS Restriction of immune activation through auranofin/BSO may result in stochastic selection of cell clones targeting conserved epitopes leading to a functional cure-like condition.
Collapse
|
28
|
Manocheewa S, Lanxon-Cookson EC, Liu Y, Swain JV, McClure J, Rao U, Maust B, Deng W, Sunshine JE, Kim M, Rolland M, Mullins JI. Pairwise growth competition assay for determining the replication fitness of human immunodeficiency viruses. J Vis Exp 2015:e52610. [PMID: 25993602 PMCID: PMC4542137 DOI: 10.3791/52610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In vitro fitness assays are essential tools for determining viral replication fitness for viruses such as HIV-1. Various measurements have been used to extrapolate viral replication fitness, ranging from the number of viral particles per infectious unit, growth rate in cell culture, and relative fitness derived from multiple-cycle growth competition assays. Growth competition assays provide a particularly sensitive measurement of fitness since the viruses are competing for cellular targets under identical growth conditions. There are several experimental factors to consider when conducting growth competition assays, including the multiplicity of infection (MOI), sampling times, and viral detection and fitness calculation methods. Each factor can affect the end result and hence must be considered carefully during the experimental design. The protocol presented here includes steps from constructing a new recombinant HIV-1 clone to performing growth competition assays and analyzing the experimental results. This protocol utilizes experimental parameter values previously shown to yield consistent and robust results. Alternatives are discussed, as some parameters need to be adjusted according to the cell type and viruses being studied. The protocol contains two alternative viral detection methods to provide flexibility as the availability of instruments, reagents and expertise varies between laboratories.
Collapse
Affiliation(s)
| | | | - Yi Liu
- Department of Microbiology, University of Washington
| | | | - Jan McClure
- Department of Microbiology, University of Washington
| | - Ushnal Rao
- Department of Microbiology, University of Washington
| | - Brandon Maust
- Department of Microbiology, University of Washington
| | - Wenjie Deng
- Department of Microbiology, University of Washington
| | | | - Moon Kim
- Department of Microbiology, University of Washington
| | - Morgane Rolland
- U.S Military HIV Research Program, Walter Reed Army Institute of Research; Henry M. Jackson Foundation
| | - James I Mullins
- Department of Microbiology, University of Washington; Departments of Medicine and Laboratory Medicine, University of Washington;
| |
Collapse
|
29
|
de Goede AL, Vulto AG, Osterhaus ADME, Gruters RA. Understanding HIV infection for the design of a therapeutic vaccine. Part I: Epidemiology and pathogenesis of HIV infection. ANNALES PHARMACEUTIQUES FRANÇAISES 2014; 73:87-99. [PMID: 25496723 DOI: 10.1016/j.pharma.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 11/01/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023]
Abstract
HIV infection leads to a gradual loss CD4+ T lymphocytes comprising immune competence and progression to AIDS. Effective treatment with combined antiretroviral drugs (cART) decreases viral load below detectable levels but is not able to eliminate the virus from the body. The success of cART is frustrated by the requirement of expensive life-long adherence, accumulating drug toxicities and chronic immune activation resulting in increased risk of several non-AIDS disorders, even when viral replication is suppressed. Therefore there is a strong need for therapeutic strategies as an alternative to cART. Immunotherapy, or therapeutic vaccination, aims to increase existing immune responses against HIV or induce de novo immune responses. These immune responses should provide a functional cure by controlling viral replication and preventing disease progression in the absence of cART. The key difficulty in the development of an HIV vaccine is our ignorance of the immune responses that control of viral replication, and thus how these responses can be elicited and how they can be monitored. Part one of this review provides an extensive overview of the (patho-) physiology of HIV infection. It describes the structure and replication cycle of HIV, the epidemiology and pathogenesis of HIV infection and the innate and adaptive immune responses against HIV. Part two of this review discusses therapeutic options for HIV. Prevention modalities and antiretroviral therapy are briefly touched upon, after which an extensive overview on vaccination strategies for HIV is provided, including the choice of immunogens and delivery strategies.
Collapse
Affiliation(s)
- A L de Goede
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands; Department of Hospital Pharmacy, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands.
| | - A G Vulto
- Department of Hospital Pharmacy, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| | - A D M E Osterhaus
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| | - R A Gruters
- Department of Viroscience, Erasmus MC 's-Gravendijkwal 230, 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
30
|
Peptide Vaccine: Progress and Challenges. Vaccines (Basel) 2014; 2:515-36. [PMID: 26344743 PMCID: PMC4494216 DOI: 10.3390/vaccines2030515] [Citation(s) in RCA: 479] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/10/2014] [Accepted: 06/13/2014] [Indexed: 12/17/2022] Open
Abstract
Conventional vaccine strategies have been highly efficacious for several decades in reducing mortality and morbidity due to infectious diseases. The bane of conventional vaccines, such as those that include whole organisms or large proteins, appear to be the inclusion of unnecessary antigenic load that, not only contributes little to the protective immune response, but complicates the situation by inducing allergenic and/or reactogenic responses. Peptide vaccines are an attractive alternative strategy that relies on usage of short peptide fragments to engineer the induction of highly targeted immune responses, consequently avoiding allergenic and/or reactogenic sequences. Conversely, peptide vaccines used in isolation are often weakly immunogenic and require particulate carriers for delivery and adjuvanting. In this article, we discuss the specific advantages and considerations in targeted induction of immune responses by peptide vaccines and progresses in the development of such vaccines against various diseases. Additionally, we also discuss the development of particulate carrier strategies and the inherent challenges with regard to safety when combining such technologies with peptide vaccines.
Collapse
|
31
|
Liu Y, Rao U, McClure J, Konopa P, Manocheewa S, Kim M, Chen L, Troyer RM, Tebit DM, Holte S, Arts EJ, Mullins JI. Impact of mutations in highly conserved amino acids of the HIV-1 Gag-p24 and Env-gp120 proteins on viral replication in different genetic backgrounds. PLoS One 2014; 9:e94240. [PMID: 24713822 PMCID: PMC3979772 DOI: 10.1371/journal.pone.0094240] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 03/14/2014] [Indexed: 11/24/2022] Open
Abstract
It has been hypothesized that a single mutation at a highly conserved amino acid site (HCS) can be severely deleterious to HIV in most if not all isolate-specific genetic backgrounds. Consequently, potentially universal HIV-1 vaccines exclusively targeting highly conserved regions of the viral proteome have been proposed. To test this hypothesis, we examined the impact of 10 Gag-p24 and 9 Env-gp120 HCS single mutations on viral fitness. In the original founder sequence of the subject in whom these mutations were identified, all Gag-p24 HCS mutations significantly reduced viral replication fitness, including 7 that were lethal. Similar results were obtained at 9/10 sites when the same mutations were introduced into the founder sequences of two epidemiologically unlinked subjects. In contrast, none of the 9 Env-gp120 HCS mutations were lethal in the original founder sequence, and four had no fitness cost. Hence, HCS mutations in Gag-p24 are likely to be severely deleterious in different HIV-1 subtype B backgrounds; however, some HCS mutations in both Gag-p24 and Env-gp120 fragments can be well tolerated. Therefore, when designing HIV-1 immunogens that are intended to force the virus to nonviable escape pathways, the fitness constraints on the HIV segments included should be considered beyond their conservation level.
Collapse
Affiliation(s)
- Yi Liu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| | - Ushnal Rao
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Jan McClure
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Philip Konopa
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Siriphan Manocheewa
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Moon Kim
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Lennie Chen
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Ryan M. Troyer
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Denis M. Tebit
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Sarah Holte
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Eric J. Arts
- Division of Infectious Diseases, Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - James I. Mullins
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
| |
Collapse
|
32
|
The impact of viral evolution and frequency of variant epitopes on primary and memory human immunodeficiency virus type 1-specific CD8⁺ T cell responses. Virology 2013; 450-451:34-48. [PMID: 24503065 DOI: 10.1016/j.virol.2013.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 09/11/2013] [Accepted: 10/08/2013] [Indexed: 12/18/2022]
Abstract
It is unclear if HIV-1 variants lose the ability to prime naïve CD8(+) cytotoxic T lymphocytes (CTL) during progressive, untreated infection. We conducted a comprehensive longitudinal analysis of viral evolution and its impact on primary and memory CD8(+) T cell responses pre-seroconversion (SC), post-SC, and during combination antiretroviral therapy (cART). Memory T cell responses targeting autologous virus variants reached a nadir by 8 years post-SC with development of AIDS, followed by a transient enhancement of anti-HIV-1 CTL responses upon initiation of cART. We show broad and high magnitude primary T cell responses to late variants in pre-SC T cells, comparable to primary anti-HIV-1 responses induced in T cells from uninfected persons. Despite evolutionary changes, CD8(+) T cells could still be primed to HIV-1 variants. Hence, vaccination against late, mutated epitopes could be successful in enhancing primary reactivity of T cells for control of the residual reservoir of HIV-1 during cART.
Collapse
|
33
|
Mailliard RB, Smith KN, Fecek RJ, Rappocciolo G, Nascimento EJM, Marques ET, Watkins SC, Mullins JI, Rinaldo CR. Selective induction of CTL helper rather than killer activity by natural epitope variants promotes dendritic cell-mediated HIV-1 dissemination. THE JOURNAL OF IMMUNOLOGY 2013; 191:2570-80. [PMID: 23913962 DOI: 10.4049/jimmunol.1300373] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The ability of HIV-1 to rapidly accumulate mutations provides the virus with an effective means of escaping CD8(+) CTL responses. In this study, we describe how subtle alterations in CTL epitopes expressed by naturally occurring HIV-1 variants can result in an incomplete escape from CTL recognition, providing the virus with a selective advantage. Rather than paralyzing the CTL response, these epitope modifications selectively induce the CTL to produce proinflammatory cytokines in the absence of target killing. Importantly, instead of dampening the immune response through CTL elimination of variant Ag-expressing immature dendritic cells (DC), a positive CTL-to-DC immune feedback loop dominates whereby the immature DC differentiate into mature proinflammatory DC. Moreover, these CTL-programmed DC exhibit a superior capacity to mediate HIV-1 trans-infection of T cells. This discordant induction of CTL helper activity in the absence of killing most likely contributes to the chronic immune activation associated with HIV-1 infection, and can be used by HIV-1 to promote viral dissemination and persistence. Our findings highlight the need to address the detrimental potential of eliciting dysfunctional cross-reactive memory CTL responses when designing and implementing anti-HIV-1 immunotherapies.
Collapse
Affiliation(s)
- Robbie B Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sanjuán R, Nebot MR, Peris JB, Alcamí J. Immune activation promotes evolutionary conservation of T-cell epitopes in HIV-1. PLoS Biol 2013; 11:e1001523. [PMID: 23565057 PMCID: PMC3614509 DOI: 10.1371/journal.pbio.1001523] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 02/14/2013] [Indexed: 01/06/2023] Open
Abstract
HIV, unlike other viruses, may benefit from immune recognition by preserving the sequence of its T cell epitopes, thereby enhancing transmission between cells. The immune system should constitute a strong selective pressure promoting viral genetic diversity and evolution. However, HIV shows lower sequence variability at T-cell epitopes than elsewhere in the genome, in contrast with other human RNA viruses. Here, we propose that epitope conservation is a consequence of the particular interactions established between HIV and the immune system. On one hand, epitope recognition triggers an anti-HIV response mediated by cytotoxic T-lymphocytes (CTLs), but on the other hand, activation of CD4+ helper T lymphocytes (TH cells) promotes HIV replication. Mathematical modeling of these opposite selective forces revealed that selection at the intrapatient level can promote either T-cell epitope conservation or escape. We predict greater conservation for epitopes contributing significantly to total immune activation levels (immunodominance), and when TH cell infection is concomitant to epitope recognition (trans-infection). We suggest that HIV-driven immune activation in the lymph nodes during the chronic stage of the disease may offer a favorable scenario for epitope conservation. Our results also support the view that some pathogens draw benefits from the immune response and suggest that vaccination strategies based on conserved TH epitopes may be counterproductive. A key component of the immune response against viruses and other pathogens is the recognition of short foreign protein sequences called epitopes. However, viruses can escape the immune system by mutating, so epitopes should accumulate high levels of genetic variability. This has been documented in several human viruses, but in HIV, unexpectedly, epitopes tend to be relatively conserved. Here, we propose that this is a consequence of the peculiar interactions that occur between HIV and the immune system. As with other viruses, recognition of HIV epitopes promotes the activation of cytotoxic and helper T lymphocytes, which then orchestrate a cellular immune response. However, HIV infects helper T lymphocytes as their target cell in the body and does so more efficiently when these cells have been activated to participate in an immune response. Mathematical modeling showed that, in some cases, HIV may take advantage of immune activation, thus favoring epitope conservation. This should be more likely to occur with epitopes that trigger more vigorous T-cell responses, and during the process known as “trans-infection,” in which helper T lymphocytes are infected while being activated. Our results highlight the potential advantages of an HIV vaccination strategy based on epitopes that stimulate cytotoxic T lymphocytes without specifically stimulating helper T lymphocytes.
Collapse
Affiliation(s)
- Rafael Sanjuán
- Institut Cavanilles de Biodiversitat i Biologia Evolutiva, Universitat de València, València, Spain.
| | | | | | | |
Collapse
|
35
|
Kulkarni V, Rosati M, Valentin A, Ganneru B, Singh AK, Yan J, Rolland M, Alicea C, Beach RK, Zhang GM, Le Gall S, Broderick KE, Sardesai NY, Heckerman D, Mothe B, Brander C, Weiner DB, Mullins JI, Pavlakis GN, Felber BK. HIV-1 p24(gag) derived conserved element DNA vaccine increases the breadth of immune response in mice. PLoS One 2013; 8:e60245. [PMID: 23555935 PMCID: PMC3610668 DOI: 10.1371/journal.pone.0060245] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 02/24/2013] [Indexed: 11/18/2022] Open
Abstract
Viral diversity is considered a major impediment to the development of an effective HIV-1 vaccine. Despite this diversity, certain protein segments are nearly invariant across the known HIV-1 Group M sequences. We developed immunogens based on the highly conserved elements from the p24gag region according to two principles: the immunogen must (i) include strictly conserved elements of the virus that cannot mutate readily, and (ii) exclude both HIV regions capable of mutating without limiting virus viability, and also immunodominant epitopes located in variable regions. We engineered two HIV-1 p24gag DNA immunogens that express 7 highly Conserved Elements (CE) of 12–24 amino acids in length and differ by only 1 amino acid in each CE (‘toggle site’), together covering >99% of the HIV-1 Group M sequences. Altering intracellular trafficking of the immunogens changed protein localization, stability, and also the nature of elicited immune responses. Immunization of C57BL/6 mice with p55gag DNA induced poor, CD4+ mediated cellular responses, to only 2 of the 7 CE; in contrast, vaccination with p24CE DNA induced cross-clade reactive, robust T cell responses to 4 of the 7 CE. The responses were multifunctional and composed of both CD4+ and CD8+ T cells with mature cytotoxic phenotype. These findings provide a method to increase immune response to universally conserved Gag epitopes, using the p24CE immunogen. p24CE DNA vaccination induced humoral immune responses similar in magnitude to those induced by p55gag, which recognize the virus encoded p24gag protein. The inclusion of DNA immunogens composed of conserved elements is a promising vaccine strategy to induce broader immunity by CD4+ and CD8+ T cells to additional regions of Gag compared to vaccination with p55gag DNA, achieving maximal cross-clade reactive cellular and humoral responses.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Brunda Ganneru
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Ashish K. Singh
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jian Yan
- University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Morgane Rolland
- Departments of Microbiology Medicine and Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Rachel Kelly Beach
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Gen-Mu Zhang
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sylvie Le Gall
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Kate E. Broderick
- Inovio Pharmaceuticals, Inc., Blue Bell, Pennsylvania, United States of America
| | | | - David Heckerman
- Microsoft Research, Redmond, Washington, United States of America
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Autonomous University of Barcelona, Barcelona, Spain
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Autonomous University of Barcelona, Barcelona, Spain
- Institucio Catalana de Recerca i Estudis Avancats (ICREA), Barcelona, Spain
| | - David B. Weiner
- University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - James I. Mullins
- Departments of Microbiology Medicine and Laboratory Medicine, University of Washington, Seattle, Washington, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- * E-mail: (BKF); (GNP)
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
- * E-mail: (BKF); (GNP)
| |
Collapse
|
36
|
Liu Y, Holte S, Rao U, McClure J, Konopa P, Swain JV, Lanxon-Cookson E, Kim M, Chen L, Mullins JI. A sensitive real-time PCR based assay to estimate the impact of amino acid substitutions on the competitive replication fitness of human immunodeficiency virus type 1 in cell culture. J Virol Methods 2012. [PMID: 23201292 DOI: 10.1016/j.jviromet.2012.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fixation of mutations in human immunodeficiency virus type 1 (HIV-1), such as those conferring drug resistance and immune escape, can result in a change in replication fitness. To assess these changes, a real-time TaqMan PCR detection assay and statistical methods for data analysis were developed to estimate sensitively relative viral fitness in competitive viral replication experiments in cell culture. Chimeric viruses with the gene of interest in an HIV-1NL4-3 backbone were constructed in two forms, vifA (native vif gene in NL4-3) and vifB (vif gene with six synonymous nucleotide differences from vifA). Subsequently, mutations of interest were introduced into the chimeric viruses in NL4-3VifA backbones, and the mutants were competed against the chimera with the isogenic viral sequence in the NL4-3VifB backbone in cell culture. In order to assess subtle fitness differences, culture supernatants were sampled longitudinally, and the viruses differentially quantified using vifA- and vifB-specific primers in real-time PCR assays. Based on an exponential net growth model, the growth rate of each virus was determined and the fitness cost of the mutation(s) distinguishing the two viruses represented as the net growth rate difference between the mutant and the native variants. Using this assay, the fitness impact of eight amino acid substitutions was quantitated at highly conserved sites in HIV-1 Gag and Env.
Collapse
Affiliation(s)
- Yi Liu
- Department of Microbiology, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Dapp MJ, Heineman RH, Mansky LM. Interrelationship between HIV-1 fitness and mutation rate. J Mol Biol 2012; 425:41-53. [PMID: 23084856 DOI: 10.1016/j.jmb.2012.10.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/09/2012] [Accepted: 10/10/2012] [Indexed: 11/16/2022]
Abstract
Differences in replication fidelity, as well as mutator and antimutator strains, suggest that virus mutation rates are heritable and prone to natural selection. Human immunodeficiency virus type 1 (HIV-1) has many distinct advantages for the study of mutation rate optimization given the wealth of structural and biochemical data on HIV-1 reverse transcriptase (RT) and mutants. In this study, we conducted parallel analyses of mutation rate and viral fitness. In particular, a panel of 10 RT mutants-most having drug resistance phenotypes-was analyzed for their effects on viral fidelity and fitness. Fidelity differences were measured using single-cycle vector assays, while fitness differences were identified using ex vivo head-to-head competition assays. As anticipated, virus mutants possessing either higher or lower fidelity had a corresponding loss in fitness. While the virus panel was not chosen randomly, it is interesting that it included more viruses possessing a mutator phenotype rather than viruses possessing an antimutator phenotype. These observations provide the first description of an interrelationship between HIV-1 fitness and mutation rate and support the conclusion that mutator and antimutator phenotypes correlate with reduced viral fitness. In addition, the findings here help support a model in which fidelity comes at a cost of replication kinetics and may help explain why retroviruses like HIV-1 and RNA viruses maintain replication fidelity near the extinction threshold.
Collapse
Affiliation(s)
- Michael J Dapp
- Institute for Molecular Virology, Academic Health Center, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
38
|
Abstract
In this Perspective, we discuss two papers that show that the breadth of the antibody response to vaccines could be manipulated by the use of particular vaccine adjuvants that stimulate innate immune mechanisms. The ability to increase the repertoire of antibodies induced by a vaccine may help to control variable pathogens that alter their exposed antigens to evade the immune system.
Collapse
Affiliation(s)
- Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford OX3 9DU, UK.
| | | |
Collapse
|
39
|
Novitsky V, Wang R, Baca J, Margolin L, McLane MF, Moyo S, van Widenfelt E, Makhema J, Essex M. Evolutionary gamut of in vivo Gag substitutions during early HIV-1 subtype C infection. Virology 2011; 421:119-28. [PMID: 22014506 DOI: 10.1016/j.virol.2011.09.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 07/18/2011] [Accepted: 09/21/2011] [Indexed: 01/03/2023]
Abstract
Two analyses of HIV-1 subtype C Gag quasispecies were performed in a prospective cohort of 42 acutely and recently infected individuals by SGA on viral RNA/proviral DNA templates. First, in vivo Gag substitutions were assessed in relation to the HIV-1C consensus sequence, which revealed that 29.3% of detected amino acid substitutions can be classified as reversions to subtype consensus, 61.3% as forward substitutions from subtype consensus, and 9.3% as polymorphisms not associated with the subtype consensus sequence. Second, the proportion, dynamics, and relationships within individual pools of viral quasispecies were analyzed. Among reverse substitutions, 16.1% were minor, 11.0% transient, 13.6% dominant, and 59.2% fixed. In contrast, 31.6% of forward substitutions were minor, 59.3% transient, 3.8% dominant, and 5.3% fixed. The distinct patterns in the spectrum and dynamics of reverse and forward Gag substitutions suggest that these differences should be considered in HIV-1 evolutionary studies and analyses of viral mutational pathways.
Collapse
Affiliation(s)
- Vladimir Novitsky
- Harvard School of Public Health AIDS Initiative, Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, Massachusetts 02115, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Gag cytotoxic T lymphocyte escape mutations can increase sensitivity of HIV-1 to human TRIM5alpha, linking intrinsic and acquired immunity. J Virol 2011; 85:11846-54. [PMID: 21917976 DOI: 10.1128/jvi.05201-11] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Although laboratory-adapted HIV-1 strains are largely resistant to the human restriction factor TRIM5α (hTRIM5α), we have recently shown that some viruses carrying capsid (CA) sequences from clinical isolates can be more sensitive to this restriction factor. In this study we evaluated the contribution to this phenotype of CA mutations known to be associated with escape from cytotoxic T lymphocyte (CTL) responses. Recombinant viruses carrying HIV-1 CA sequences from NL4-3 and three different clinical isolates were prepared, along with variants in which mutations associated with CTL resistance were modified by site-directed mutagenesis, and the infectivities of these viruses in target cells expressing hTRIM5α and cells in which TRIM5α activity had been inhibited by overexpression of TRIM5γ were compared. For both hTRIM5α-sensitive viruses studied, CTL-associated mutations were found to be responsible for this phenotype. Both CTL resistance mutations occurring within HLA-restricted CA epitopes and compensatory mutations occurring outside CTL epitopes influenced hTRIM5α sensitivity, and mutations associated with CTL resistance selected in prior hosts can contribute to this effect. The impact of CTL resistance mutations on hTRIM5α sensitivity was context dependent, because mutations shown to be responsible for the TRIM5α-sensitive phenotype in viruses from one patient could have little or no impact on this parameter when introduced into another virus. No fixed relationship between changes in hTRIM5α sensitivity and infectivity was discernible in our studies. Taken together, these findings suggest that CTL mutations may influence HIV-1 replication by modifying both viral infectivity and sensitivity to TRIM5α.
Collapse
|
41
|
Universal peptide vaccines - optimal peptide vaccine design based on viral sequence conservation. Vaccine 2011; 29:8745-53. [PMID: 21875632 DOI: 10.1016/j.vaccine.2011.07.132] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 07/28/2011] [Accepted: 07/28/2011] [Indexed: 01/06/2023]
Abstract
Rapidly mutating viruses such as the hepatitis C virus (HCV), the human immunodeficiency virus (HIV), or influenza viruses (Flu) call for highly effective universal peptide vaccines, i.e. vaccines that do not only yield broad population coverage but also broad coverage of various viral strains. The efficacy of such vaccines is determined by multiple properties of the epitopes they comprise. Beyond the specific properties of each epitope, properties of the corresponding source antigens are of great importance. If a response is mounted against viral proteins with a low copy number within the cell or against proteins expressed very late, this response may fail to induce lysis of the infected cells before budding can take place. We here propose a novel methodology to optimize the epitope composition and assembly in order to induce maximum protection. In order for a peptide vaccine to yield the best possible universal protection, several conditions should be met: (a) an optimal choice of target antigens, (b) an optimal choice of highly conserved epitopes, (c) maximum coverage of the target population, and (d) the proper ordering of the epitopes in the final vaccine to ensure favorable cleavage. We propose a mathematical formalism for epitope selection and ordering that balances the constraints imposed by these different conditions. Focusing on HCV, HIV, and Flu, we show that not all of the conditions can be satisfied for all viruses. Depending on the virus, different constraints are harder to fulfill: for Flu, the conservation constraint is violated first, while for HIV, it is difficult to focus the response at the optimal target antigens. The proposed methodology can be applied to any virus to assess the feasibility of optimally combining the above-mentioned constraints.
Collapse
|
42
|
Buranapraditkun S, Hempel U, Pitakpolrat P, Allgaier RL, Thantivorasit P, Lorenzen SI, Sirivichayakul S, Hildebrand WH, Altfeld M, Brander C, Walker BD, Phanuphak P, Hansasuta P, Rowland-Jones SL, Allen TM, Ruxrungtham K. A novel immunodominant CD8+ T cell response restricted by a common HLA-C allele targets a conserved region of Gag HIV-1 clade CRF01_AE infected Thais. PLoS One 2011; 6:e23603. [PMID: 21887282 PMCID: PMC3161737 DOI: 10.1371/journal.pone.0023603] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/20/2011] [Indexed: 01/09/2023] Open
Abstract
Background CD8+ T cell responses play an important role in the control of HIV-1. The extensive sequence diversity of HIV-1 represents a critical hurdle to developing an effective HIV-1 vaccine, and it is likely that regional-specific vaccine strains will be required to overcome the diversity of the different HIV-1 clades distributed world-wide. Unfortunately, little is known about the CD8+ T cell responses against CRF01_AE, which is responsible for the majority of infections in Southeast Asia. Methodology/Principal Findings To identify dominant CD8+ T cell responses recognized in HIV-1 clade CRF01_AE infected subjects we drew upon data from an immunological screen of 100 HIV-1 clade CRF01_AE infected subjects using IFN-gamma ELISpot to characterize a novel immunodominant CD8+ T cell response in HIV-1 Gag restricted by HLA-Cw*0102 (p24, 277YSPVSILDI285, YI9). Over 75% of Cw*0102+ve subjects targeted this epitope, representing the strongest response in more than a third of these individuals. This novel CD8 epitope was located in a highly conserved region of HIV-1 Gag known to contain immunodominant CD8 epitopes, which are restricted by HLA-B*57 and -B*27 in clade B infection. Nonetheless, viral escape in this epitope was frequently observed in Cw*0102+ve subjects, suggestive of strong selection pressure being exerted by this common CD8+ T cell response. Conclusions/Significance As HLA-Cw*0102 is frequently expressed in the Thai population (allelic frequency of 16.8%), this immunodominant Cw*0102-restricted Gag epitope may represent an attractive candidate for vaccines specific to CRF01_AE and may help facilitate further studies of immunopathogenesis in this understudied HIV-1 clade.
Collapse
Affiliation(s)
- Supranee Buranapraditkun
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Interdisciplinary Program of Medical Microbiology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Ursula Hempel
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Patrawadee Pitakpolrat
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rachel L. Allgaier
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Pattarawat Thantivorasit
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sven-Iver Lorenzen
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sunee Sirivichayakul
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - William H. Hildebrand
- Department of Microbiology and Immunology, Health Sciences Center, University of Oklahoma, Oklahoma City, Oklahoma, United States of America
| | - Marcus Altfeld
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Christian Brander
- AIDS Research Institute IrsiCaixa - HIVACAT, Hospital Germans Trias i Pujol, Badalona, and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Praphan Phanuphak
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Red Cross AIDS Research Center, Bangkok, Thailand
| | - Pokrath Hansasuta
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sarah L. Rowland-Jones
- Medical Research Council, Human Immunology Unit, Weather all Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Todd M. Allen
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, United States of America
| | - Kiat Ruxrungtham
- Vaccine and Cellular Immunology Laboratory, Chulalongkorn Medical Research Center (ChulaMRC), and Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Thai Red Cross AIDS Research Center, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
43
|
Chopera DR, Wright JK, Brockman MA, Brumme ZL. Immune-mediated attenuation of HIV-1. Future Virol 2011; 6:917-928. [PMID: 22393332 DOI: 10.2217/fvl.11.68] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immune escape mutations selected by human leukocyte antigen class I-restricted CD8(+) cytotoxic T lymphocytes (CTLs) can result in biologically and clinically relevant costs to HIV-1 replicative fitness. This phenomenon may be exploited to design an HIV-1 vaccine capable of stimulating effective CTL responses against highly conserved, mutationally constrained viral regions, where immune escape could occur only at substantial functional costs. Such a vaccine might 'channel' HIV-1 evolution towards a less-fit state, thus lowering viral load set points, attenuating the infection course and potentially reducing the risk of transmission. A major barrier to this approach, however, is the accumulation of immune escape variants at the population level, possibly leading to the loss of immunogenic CTL epitopes and diminished vaccine-induced cellular immune responses as the epidemic progresses. Here, we review the evidence supporting CTL-driven replicative defects in HIV-1 and consider the implications of this work for CTL-based vaccines designed to attenuate the infection course.
Collapse
|
44
|
Herbeck JT, Rolland M, Liu Y, McLaughlin S, McNevin J, Zhao H, Wong K, Stoddard JN, Raugi D, Sorensen S, Genowati I, Birditt B, McKay A, Diem K, Maust BS, Deng W, Collier AC, Stekler JD, McElrath MJ, Mullins JI. Demographic processes affect HIV-1 evolution in primary infection before the onset of selective processes. J Virol 2011; 85:7523-34. [PMID: 21593162 PMCID: PMC3147913 DOI: 10.1128/jvi.02697-10] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Accepted: 05/11/2011] [Indexed: 12/12/2022] Open
Abstract
HIV-1 transmission and viral evolution in the first year of infection were studied in 11 individuals representing four transmitter-recipient pairs and three independent seroconverters. Nine of these individuals were enrolled during acute infection; all were men who have sex with men (MSM) infected with HIV-1 subtype B. A total of 475 nearly full-length HIV-1 genome sequences were generated, representing on average 10 genomes per specimen at 2 to 12 visits over the first year of infection. Single founding variants with nearly homogeneous viral populations were detected in eight of the nine individuals who were enrolled during acute HIV-1 infection. Restriction to a single founder variant was not due to a lack of diversity in the transmitter as homogeneous populations were found in recipients from transmitters with chronic infection. Mutational patterns indicative of rapid viral population growth dominated during the first 5 weeks of infection and included a slight contraction of viral genetic diversity over the first 20 to 40 days. Subsequently, selection dominated, most markedly in env and nef. Mutants were detected in the first week and became consensus as early as day 21 after the onset of symptoms of primary HIV infection. We found multiple indications of cytotoxic T lymphocyte (CTL) escape mutations while reversions appeared limited. Putative escape mutations were often rapidly replaced with mutually exclusive mutations nearby, indicating the existence of a maturational escape process, possibly in adaptation to viral fitness constraints or to immune responses against new variants. We showed that establishment of HIV-1 infection is likely due to a biological mechanism that restricts transmission rather than to early adaptive evolution during acute infection. Furthermore, the diversity of HIV strains coupled with complex and individual-specific patterns of CTL escape did not reveal shared sequence characteristics of acute infection that could be harnessed for vaccine design.
Collapse
Affiliation(s)
| | | | - Yi Liu
- Departments of Microbiology
| | | | - John McNevin
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | | | | | | | | | | | | | | | | | | | | - Ann C. Collier
- Medicine, University of Washington School of Medicine, Seattle, Washington 98195-8070
| | - Joanne D. Stekler
- Medicine, University of Washington School of Medicine, Seattle, Washington 98195-8070
| | | | - James I. Mullins
- Departments of Microbiology
- Laboratory Medicine
- Medicine, University of Washington School of Medicine, Seattle, Washington 98195-8070
| |
Collapse
|
45
|
Li JZ, Brumme ZL, Brumme CJ, Wang H, Spritzler J, Robertson MN, Lederman MM, Carrington M, Walker BD, Schooley RT, Kuritzkes DR. Factors associated with viral rebound in HIV-1-infected individuals enrolled in a therapeutic HIV-1 gag vaccine trial. J Infect Dis 2011; 203:976-83. [PMID: 21402549 DOI: 10.1093/infdis/jiq143] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1) vaccines directed to the cell-mediated immune system could have a role in lowering the plasma HIV-1 RNA set point, which may reduce infectivity and delay disease progression. METHODS Randomized, placebo-controlled trial involving HIV-1-infected participants who received a recombinant adenovirus serotype 5 (rAd5) HIV-1 gag vaccine or placebo. Sequence-based HLA typing was performed for all 110 participants who initiated analytic treatment interruption (ATI) to assess the role of HLA types previously associated with HIV prognosis. Plasma HIV-1 gag and pol RNA sequences were obtained during the ATI. Virologic endpoints and HLA groups were compared between treatment arms using the 2-sample rank sum test. A linear regression model was fitted to derive independent correlates of ATI week 16 plasma viral load (w16 PVL). RESULTS Vaccinated participants with neutral HLA alleles had lower median w16 PVLs than did vaccinated participants with protective HLA alleles (P = .01) or placebo participants with neutral HLA alleles (P = .02). Factors independently associated with lower w16 PVL included lower pre-antiretroviral therapy PVL, greater Gag sequence divergence from the vaccine sequence, decreased proportion of HLA-associated polymorphisms in Gag, and randomization to the vaccine arm. CONCLUSIONS Therapeutic vaccination with a rAd5-HIV gag vaccine was associated with lower ATI week 16 PVL even after controlling for viral and host genetic factors. CLINICAL TRIALS REGISTRATION NCT00080106.
Collapse
Affiliation(s)
- Jonathan Z Li
- Section of Retroviral Therapeutics, Brigham and Women's Hospital, Boston, Massachusetts 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mostowy R, Kouyos RD, Fouchet D, Bonhoeffer S. The role of recombination for the coevolutionary dynamics of HIV and the immune response. PLoS One 2011; 6:e16052. [PMID: 21364750 PMCID: PMC3041767 DOI: 10.1371/journal.pone.0016052] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/07/2010] [Indexed: 11/19/2022] Open
Abstract
The evolutionary implications of recombination in HIV remain not fully understood. A plausible effect could be an enhancement of immune escape from cytotoxic T lymphocytes (CTLs). In order to test this hypothesis, we constructed a population dynamic model of immune escape in HIV and examined the viral-immune dynamics with and without recombination. Our model shows that recombination (i) increases the genetic diversity of the viral population, (ii) accelerates the emergence of escape mutations with and without compensatory mutations, and (iii) accelerates the acquisition of immune escape mutations in the early stage of viral infection. We see a particularly strong impact of recombination in systems with broad, non-immunodominant CTL responses. Overall, our study argues for the importance of recombination in HIV in allowing the virus to adapt to changing selective pressures as imposed by the immune system and shows that the effect of recombination depends on the immunodominance pattern of effector T cell responses.
Collapse
Affiliation(s)
- Rafal Mostowy
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland.
| | | | | | | |
Collapse
|
47
|
Liu Y, McNevin JP, Holte S, McElrath MJ, Mullins JI. Dynamics of viral evolution and CTL responses in HIV-1 infection. PLoS One 2011; 6:e15639. [PMID: 21283794 PMCID: PMC3024315 DOI: 10.1371/journal.pone.0015639] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 11/18/2010] [Indexed: 02/06/2023] Open
Abstract
Improved understanding of the dynamics of host immune responses and viral evolution is critical for effective HIV-1 vaccine design. We comprehensively analyzed Cytotoxic T-lymphocyte (CTL)-viral epitope dynamics in an antiretroviral therapy-naïve subject over the first four years of HIV-1 infection. We found that CTL responses developed sequentially and required constant antigenic stimulation for maintenance. CTL responses exerting strong selective pressure emerged early and led to rapid escape, proliferated rapidly and were predominant during acute/early infection. Although CTL responses to a few persistent epitopes developed over the first two months of infection, they proliferated slowly. As CTL epitopes were replaced by mutational variants, the corresponding responses immediately declined, most rapidly in the cases of strongly selected epitopes. CTL recognition of epitope variants, via cross-reactivity and de novo responses, was common throughout the period of study. Our data demonstrate that HIV-specific CTL responses, especially in the critical acute/early stage, were focused on regions that are prone to escape. Failure of CTL responses to strongly target functional or structurally critical regions of the virus, as well as the sequential cascade of CTL responses, followed closely by viral escape and decline of the corresponding responses, likely contribute to a lack of sustainable viral suppression. Focusing early and rapidly proliferating CTL on persistent epitopes may be essential for durable viral control in HIV-1 infection.
Collapse
Affiliation(s)
- Yi Liu
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - John P. McNevin
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Sarah Holte
- Program in Biostatistics and Biomathematics, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - M. Juliana McElrath
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Institute, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - James I. Mullins
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
48
|
Irausquin SJ, Hughes AL. Conflicting selection pressures on T-cell epitopes in HIV-1 subtype B. INFECTION GENETICS AND EVOLUTION 2011; 11:483-8. [PMID: 21232634 DOI: 10.1016/j.meegid.2010.12.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 12/20/2010] [Accepted: 12/23/2010] [Indexed: 12/26/2022]
Abstract
Analysis of population-level polymorphism in eight coding genes of human immunodeficiency virus type 1 (HIV-1) subtype B revealed evidence not only of past purifying selection, but also of abundant slightly deleterious nonsynonymous variants subject to ongoing purifying selection. Both CD4 and CTL epitopes showed an excess of nonsynonymous variants that were singletons (occurring in just one sequence) in our dataset. Overall, median gene diversities at polymorphic nonsynonymous sites were highest at sites located in neither CD4 nor CTL epitopes, while polymorphic nonsynonymous sites in CD4 epitopes revealed the lowest median gene diversity. Our results support the hypothesis that there is an evolutionary conflict between immune escape and functional constraint on epitopes recognized by host T-cells, and suggest that amino acid sequences of CD4 epitopes are subject to particularly strong functional constraint.
Collapse
Affiliation(s)
- Stephanie Jiménez Irausquin
- Department of Biological Sciences, University of South Carolina, Coker Life Sciences Building, 700 Sumter St., Columbia, SC 29208, USA
| | | |
Collapse
|
49
|
Fluidity of HIV-1-specific T-cell responses during acute and early subtype C HIV-1 infection and associations with early disease progression. J Virol 2010; 84:12018-29. [PMID: 20826686 DOI: 10.1128/jvi.01472-10] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Deciphering immune events during early stages of human immunodeficiency virus type 1 (HIV-1) infection is critical for understanding the course of disease. We characterized the hierarchy of HIV-1-specific T-cell gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assay responses during acute subtype C infection in 53 individuals and associated temporal patterns of responses with disease progression in the first 12 months. There was a diverse pattern of T-cell recognition across the proteome, with the recognition of Nef being immunodominant as early as 3 weeks postinfection. Over the first 6 months, we found that there was a 23% chance of an increased response to Nef for every week postinfection (P = 0.0024), followed by a nonsignificant increase to Pol (4.6%) and Gag (3.2%). Responses to Env and regulatory proteins appeared to remain stable. Three temporal patterns of HIV-specific T-cell responses could be distinguished: persistent, lost, or new. The proportion of persistent T-cell responses was significantly lower (P = 0.0037) in individuals defined as rapid progressors than in those progressing slowly and who controlled viremia. Almost 90% of lost T-cell responses were coincidental with autologous viral epitope escape. Regression analysis between the time to fixed viral escape and lost T-cell responses (r = 0.61; P = 0.019) showed a mean delay of 14 weeks after viral escape. Collectively, T-cell epitope recognition is not a static event, and temporal patterns of IFN-γ-based responses exist. This is due partly to viral sequence variation but also to the recognition of invariant viral epitopes that leads to waves of persistent T-cell immunity, which appears to associate with slower disease progression in the first year of infection.
Collapse
|
50
|
Love TMT, Thurston SW, Keefer MC, Dewhurst S, Lee HY. Mathematical modeling of ultradeep sequencing data reveals that acute CD8+ T-lymphocyte responses exert strong selective pressure in simian immunodeficiency virus-infected macaques but still fail to clear founder epitope sequences. J Virol 2010; 84:5802-14. [PMID: 20335256 PMCID: PMC2876615 DOI: 10.1128/jvi.00117-10] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 03/15/2010] [Indexed: 11/20/2022] Open
Abstract
The prominent role of antiviral cytotoxic CD8(+) T-lymphocytes (CD8-TL) in containing the acute viremia of human and simian immunodeficiency viruses (HIV-1 and SIV) has rationalized the development of T-cell-based vaccines. However, the presence of escape mutations in the acute stage of infection has raised a concern that accelerated escape from vaccine-induced CD8-TL responses might undermine vaccine efficacy. We reanalyzed previously published data of 101,822 viral genomes of three CD8-TL epitopes, Nef(103-111)RM9 (RM9), Tat(28-35)SL8 (SL8), and Gag(181-189)CM9 (CM9), sampled by ultradeep pyrosequencing from eight macaques. Multiple epitope variants appeared during the resolution of acute viremia, followed by the predominance of a single mutant epitope. By fitting a mathematical model, we estimated the first acute escape rate as 0.36 day(-1) within escape-prone epitopes, RM9 and SL8, and the chronic escape rate as 0.014 day(-1) within the CM9 epitope. Our estimate of SIV acute escape rates was found to be comparable to very early HIV-1 escape rates. The timing of the first escape was more highly correlated with the timing of the peak CD8-TL response than with the magnitude of the CD8-TL response. The transmitted epitope decayed more than 400 times faster during the acute viral decline stage than predicted by a neutral evolution model. However, the founder epitope persisted as a minor population even at the viral set point; in contrast, the majority of acute escape epitopes were completely cleared. Our results suggest that a reservoir of SIV infection is preferentially formed by virus with the transmitted epitope.
Collapse
Affiliation(s)
- Tanzy M. T. Love
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York 14642, Department of Medicine, University of Rochester, Rochester, New York 14642, Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| | - Sally W. Thurston
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York 14642, Department of Medicine, University of Rochester, Rochester, New York 14642, Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| | - Michael C. Keefer
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York 14642, Department of Medicine, University of Rochester, Rochester, New York 14642, Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| | - Stephen Dewhurst
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York 14642, Department of Medicine, University of Rochester, Rochester, New York 14642, Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| | - Ha Youn Lee
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, New York 14642, Department of Medicine, University of Rochester, Rochester, New York 14642, Department of Microbiology and Immunology, University of Rochester, Rochester, New York 14642
| |
Collapse
|