1
|
Bouzari M, Ait Mahiout L, Mozokhina A, Volpert V. Infection propagation in a tissue with resident macrophages. Math Biosci 2025; 381:109399. [PMID: 39954943 DOI: 10.1016/j.mbs.2025.109399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
The progression of viral infection within the human body is governed by a complex interplay between the pathogen and the immune response. The initial phase of the innate immune response is driven by inflammatory cytokines and interferons produced by infected target cells and tissue-resident macrophages. These inflammatory cytokines not only amplify the immune response but also initiate programmed cell death, which helps slow the spread of the infection. The propagation of the infection within tissues can be modeled as a reaction-diffusion wave, where the speed of this wave is linked to the virus virulence, and the overall viral load determines its infectivity. In this study, we demonstrate that inflammation reduces both the speed and viral load of the infection wave, and we establish the conditions necessary to halt the spread of the infection. Depending on the relative strength of the infection and the immune response, there are three possible outcomes of infection progression. If the virus replication number is sufficiently low, the infection does not develop. For intermediate values of this parameter, the infection spreads within the affected tissue at a decreasing speed and amplitude before ultimately being eliminated. However, if the virus replication number is high, the infection propagates as a reaction-diffusion wave with a constant speed and amplitude. These findings are derived using analytical methods and are corroborated by numerical simulations. Additionally, we explore viral diffusion, comparing the conventional parabolic diffusion model with the hyperbolic diffusion model, which is introduced to address the limitation of infinite propagation speed. Our results show that while the viral load remains the same across both models, the wave speed in the hyperbolic model is smaller and approaches that of the parabolic model as the relaxation time decreases.
Collapse
Affiliation(s)
- M Bouzari
- Laboratoire d'équations aux dérivées partielles non linéaires et histoire des mathématiques, École Normale Supérieure, B.P. 92, Vieux Kouba, 16050 Algiers, Algeria
| | - L Ait Mahiout
- Laboratoire d'équations aux dérivées partielles non linéaires et histoire des mathématiques, École Normale Supérieure, B.P. 92, Vieux Kouba, 16050 Algiers, Algeria
| | - A Mozokhina
- Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation
| | - V Volpert
- Peoples' Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya St, Moscow 117198, Russian Federation; Universite Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, 69622 Villeurbanne, France.
| |
Collapse
|
2
|
Kufera JT, Armstrong C, Wu F, Singhal A, Zhang H, Lai J, Wilkins HN, Simonetti FR, Siliciano JD, Siliciano RF. CD4+ T cells with latent HIV-1 have reduced proliferative responses to T cell receptor stimulation. J Exp Med 2024; 221:e20231511. [PMID: 38270554 PMCID: PMC10818065 DOI: 10.1084/jem.20231511] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/04/2023] [Accepted: 01/09/2024] [Indexed: 01/26/2024] Open
Abstract
The latent reservoir for HIV-1 in resting CD4+ T cells persists despite antiretroviral therapy as a barrier to cure. The antigen-driven proliferation of infected cells is a major mechanism of reservoir persistence. However, activation through the T cell antigen receptor (TCR) can induce latent proviruses, leading to viral cytopathic effects and immune clearance. In single-cell studies, we show that, relative to uninfected cells or cells with a defective provirus, CD4+ T cells with an intact provirus have a profound proliferative defect in response to TCR stimulation. Virion production was observed in only 16.5% of cultures with an intact provirus, but proliferation was reduced even when no virion production was detected. Proliferation was inversely correlated with in vivo clone size. These results may reflect the effects of previous in vivo proliferation and do not support attempts to reduce the reservoir with antiproliferative agents, which may have greater effects on normal T cell responses.
Collapse
Affiliation(s)
- Joshua T. Kufera
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ciara Armstrong
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fengting Wu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anushka Singhal
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hao Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jun Lai
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hannah N. Wilkins
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Janet D. Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F. Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Howard Hughes Medical Institute, Baltimore, MD, USA
| |
Collapse
|
3
|
Clarke C, Pankavich S. Three-stage modeling of HIV infection and implications for antiretroviral therapy. J Math Biol 2024; 88:34. [PMID: 38418658 DOI: 10.1007/s00285-024-02056-1] [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: 07/25/2023] [Revised: 12/01/2023] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
We consider a deterministic model of HIV infection that involves macrophages as a long-term active reservoir to describe all three stages of the disease process: the acute stage, chronic infection, and the transition to AIDS. The proposed model is shown to retain crucial properties, such as the positivity of solutions, regardless of variations in model parameters. A dynamical analysis is performed to identify the local stability properties of the viral clearance steady state. This analysis illustrates how chronically infected macrophages can explain the progression to AIDS and provoke viral explosion, while previous models do not. We further demonstrate that the infected T-cell population, even if not responsible for the majority of new infections that lead to viral explosion, may contribute significantly to the transition amongst the three stages of infection. Moreover, we explore the implications of the model for the administration of antiretroviral therapy (ART) and provide quantitative estimates that emphasize the time sensitive nature of treatment initiation and the level of drug efficacy. Finally, we study the effects of treatment interruption on the disease dynamics predicted by the model and elucidate the influence of both interruption time and duration.
Collapse
Affiliation(s)
- Cameron Clarke
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, 80403, USA
| | - Stephen Pankavich
- Department of Applied Mathematics and Statistics, Colorado School of Mines, Golden, CO, 80403, USA.
| |
Collapse
|
4
|
Zhitkevich A, Bayurova E, Avdoshina D, Zakirova N, Frolova G, Chowdhury S, Ivanov A, Gordeychuk I, Palefsky JM, Isaguliants M. HIV-1 Reverse Transcriptase Expression in HPV16-Infected Epidermoid Carcinoma Cells Alters E6 Expression and Cellular Metabolism, and Induces a Hybrid Epithelial/Mesenchymal Cell Phenotype. Viruses 2024; 16:193. [PMID: 38399969 PMCID: PMC10892743 DOI: 10.3390/v16020193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The high incidence of epithelial malignancies in HIV-1 infected individuals is associated with co-infection with oncogenic viruses, such as high-risk human papillomaviruses (HR HPVs), mostly HPV16. The molecular mechanisms underlying the HIV-1-associated increase in epithelial malignancies are not fully understood. A collaboration between HIV-1 and HR HPVs in the malignant transformation of epithelial cells has long been anticipated. Here, we delineated the effects of HIV-1 reverse transcriptase on the in vitro and in vivo properties of HPV16-infected cervical cancer cells. A human cervical carcinoma cell line infected with HPV16 (Ca Ski) was made to express HIV-1 reverse transcriptase (RT) by lentiviral transduction. The levels of the mRNA of the E6 isoforms and of the factors characteristic to the epithelial/mesenchymal transition were assessed by real-time RT-PCR. The parameters of glycolysis and mitochondrial respiration were determined using Seahorse technology. RT expressing Ca Ski subclones were assessed for the capacity to form tumors in nude mice. RT expression increased the expression of the E6*I isoform, modulated the expression of E-CADHERIN and VIMENTIN, indicating the presence of a hybrid epithelial/mesenchymal phenotype, enhanced glycolysis, and inhibited mitochondrial respiration. In addition, the expression of RT induced phenotypic alterations impacting cell motility, clonogenic activity, and the capacity of Ca Ski cells to form tumors in nude mice. These findings suggest that HIV-RT, a multifunctional protein, affects HPV16-induced oncogenesis, which is achieved through modulation of the expression of the E6 oncoprotein. These results highlight a complex interplay between HIV antigens and HPV oncoproteins potentiating the malignant transformation of epithelial cells.
Collapse
Affiliation(s)
- Alla Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 119991 Moscow, Russia; (E.B.); (D.A.); (G.F.); (I.G.)
| | - Ekaterina Bayurova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 119991 Moscow, Russia; (E.B.); (D.A.); (G.F.); (I.G.)
- Gamaleya National Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia;
| | - Darya Avdoshina
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 119991 Moscow, Russia; (E.B.); (D.A.); (G.F.); (I.G.)
| | - Natalia Zakirova
- Centre for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia;
| | - Galina Frolova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 119991 Moscow, Russia; (E.B.); (D.A.); (G.F.); (I.G.)
| | - Sona Chowdhury
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA 94143, USA; (S.C.); (J.M.P.)
| | - Alexander Ivanov
- Gamaleya National Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia;
- Centre for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, 119991 Moscow, Russia;
| | - Ilya Gordeychuk
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 119991 Moscow, Russia; (E.B.); (D.A.); (G.F.); (I.G.)
- Gamaleya National Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia;
| | - Joel M. Palefsky
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA 94143, USA; (S.C.); (J.M.P.)
| | - Maria Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
5
|
Valdebenito S, Ono A, Rong L, Eugenin EA. The role of tunneling nanotubes during early stages of HIV infection and reactivation: implications in HIV cure. NEUROIMMUNE PHARMACOLOGY AND THERAPEUTICS 2023; 2:169-186. [PMID: 37476291 PMCID: PMC10355284 DOI: 10.1515/nipt-2022-0015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 11/30/2022] [Indexed: 07/22/2023]
Abstract
Tunneling nanotubes (TNTs), also called cytonemes or tumor microtubes, correspond to cellular processes that enable long-range communication. TNTs are plasma membrane extensions that form tubular processes that connect the cytoplasm of two or more cells. TNTs are mostly expressed during the early stages of development and poorly expressed in adulthood. However, in disease conditions such as stroke, cancer, and viral infections such as HIV, TNTs proliferate, but their role is poorly understood. TNTs function has been associated with signaling coordination, organelle sharing, and the transfer of infectious agents such as HIV. Here, we describe the critical role and function of TNTs during HIV infection and reactivation, as well as the use of TNTs for cure strategies.
Collapse
Affiliation(s)
- Silvana Valdebenito
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Akira Ono
- Department of Microbiology & Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Libin Rong
- Department of Mathematics, University of Florida, Gainesville, FL, USA
| | - Eliseo A. Eugenin
- Department of Neurobiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| |
Collapse
|
6
|
Murray JM. Dynamics of latent HIV under clonal expansion. PLoS Pathog 2021; 17:e1010165. [PMID: 34929000 PMCID: PMC8722732 DOI: 10.1371/journal.ppat.1010165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/03/2022] [Accepted: 12/02/2021] [Indexed: 11/23/2022] Open
Abstract
The HIV latent reservoir exhibits slow decay on antiretroviral therapy (ART), impacted by homeostatic proliferation and activation. How these processes contribute to the total dynamic while also producing the observed profile of sampled latent clone sizes is unclear. An agent-based model was developed that tracks individual latent clones, incorporating homeostatic proliferation of cells and activation of clones. The model was calibrated to produce observed latent reservoir dynamics as well as observed clonal size profiles. Simulations were compared to previously published latent HIV integration data from 5 adults and 3 children. The model simulations reproduced reservoir dynamics as well as generating residual plasma viremia levels (pVL) consistent with observations on ART. Over 382 Latin Hypercube Sample simulations, the median latent reservoir grew by only 0.3 log10 over the 10 years prior to ART initiation, after which time it decreased with a half-life of 15 years, despite number of clones decreasing at a faster rate. Activation produced a maximum size of genetically intact clones of around one million cells. The individual simulation that best reproduced the sampled clone profile, produced a reservoir that decayed with a 13.9 year half-life and where pVL, produced mainly from proliferation, decayed with a half-life of 10.8 years. These slow decay rates were achieved with mean cell life-spans of only 14.2 months, due to expansion of the reservoir through proliferation and activation. Although the reservoir decayed on ART, a number of clones increased in size more than 4,000-fold. While small sampled clones may have expanded through proliferation, the large sizes exclusively arose from activation. Simulations where homeostatic proliferation contributed more to pVL than activation, produced pVL that was less variable over time and exhibited fewer viral blips. While homeostatic proliferation adds to the latent reservoir, activation can both add and remove latent cells. Latent activation can produce large clones, where these may have been seeded much earlier than when first sampled. Elimination of the reservoir is complicated by expanding clones whose dynamic differ considerably to that of the entire reservoir. The HIV latent reservoir decreases slowly on antiretroviral therapy (ART). However there are cellular processes operating within this reservoir that can expand or contract subpopulations. This means that what is happening at the macro level may not be reflected at the micro level. To investigate this, we analysed published data on HIV latent clone sizes. By constructing an agent model incorporating the processes of cellular activation and proliferation, we were able to show that activation can expand clone sizes significantly even while on ART. Homeostatic proliferation also plays a role in maintaining the reservoir but these clones, though more frequent, are much smaller in size. Our calculations also show that activation and proliferation of the intact latent reservoir can lead to some of these cells becoming virally productive to a level consistent with observed residual viremia during ART. This analysis explains how normal cellular processes restructure the make-up of the latent reservoir and contribute to residual viremia.
Collapse
Affiliation(s)
- John M. Murray
- School of Mathematics and Statistics, UNSW Sydney, Australia
- * E-mail:
| |
Collapse
|
7
|
A Comprehensive Analysis of Human Endogenous Retroviruses HERV-K (HML.2) from Teratocarcinoma Cell Lines and Detection of Viral Cargo in Microvesicles. Int J Mol Sci 2021; 22:ijms222212398. [PMID: 34830279 PMCID: PMC8619701 DOI: 10.3390/ijms222212398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
About 8% of our genome is composed of sequences from Human Endogenous Retroviruses (HERVs). The HERV-K (HML.2) family, here abbreviated HML.2, is able to produce virus particles that were detected in cell lines, malignant tumors and in autoimmune diseases. Parameters and properties of HML.2 released from teratocarcinoma cell lines GH and Tera-1 were investigated in detail. In most experiments, analyzed viruses were purified by density gradient centrifugation. HML.2 structural proteins, reverse transcriptase (RT) activity, viral RNA (vRNA) and particle morphology were analyzed. The HML.2 markers were predominantly detected in fractions with a buoyant density of 1.16 g/cm3. Deglycosylation of TM revealed truncated forms of transmembrane (TM) protein. Free virions and extracellular vesicles (presumably microvesicles—MVs) with HML.2 elements, including budding intermediates, were detected by electron microscopy. Viral elements and assembled virions captured and exported by MVs can boost specific immune responses and trigger immunomodulation in recipient cells. Sequencing of cDNA clones demonstrated exclusive presence of HERV-K108 env in HML.2 from Tera-1 cells. Not counting two recombinant variants, four known env sequences were found in HML.2 from GH cells. Obtained results shed light on parameters and morphology of HML.2. A possible mechanism of HML.2-induced diseases is discussed.
Collapse
|
8
|
A diffusive virus model with a fixed intracellular delay and combined drug treatments. J Math Biol 2021; 83:19. [PMID: 34324062 DOI: 10.1007/s00285-021-01646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 04/26/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022]
Abstract
The method of administration of an effective drug treatment to eradicate viruses within a host is an important issue in studying viral dynamics. Overuse of a drug can lead to deleterious side effects, and overestimating the efficacy of a drug can result in failure to treat infection. In this study, we proposed a reaction-diffusion within-host virus model which incorporated information regarding spatial heterogeneity, drug treatment, and the intracellular delay to produce productively infected cells and viruses. We also calculated the basic reproduction number [Formula: see text] under the assumptions of spatial heterogeneity. We have shown that the infection-free periodic solution is globally asymptotically stable when [Formula: see text], whereas when [Formula: see text] there is an infected periodic solution and the infection is uniformly persistent. By conducting numerical simulations, we also revealed the effects of various parameters on the value of [Formula: see text]. First, we showed that the dependence of [Formula: see text] on the intracellular delay could be monotone or non-monotone, depending on the death rate of infected cells in the immature stage. Second, we found that the mobility of infected cells or virions could facilitate the virus clearance. Third, we found that the spatial fragmentation of the virus environment enhanced viral infection. Finally, we showed that the combination of spatial heterogeneity and different sets of diffusion rates resulted in complicated viral dynamic outcomes.
Collapse
|
9
|
Ratti V, Nanda S, Eszterhas SK, Howell AL, Wallace DI. A mathematical model of HIV dynamics treated with a population of gene-edited haematopoietic progenitor cells exhibiting threshold phenomenon. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2021; 37:212-242. [PMID: 31265056 DOI: 10.1093/imammb/dqz011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 04/03/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
The use of gene-editing technology has the potential to excise the CCR5 gene from haematopoietic progenitor cells, rendering their differentiated CD4-positive (CD4+) T cell descendants HIV resistant. In this manuscript, we describe the development of a mathematical model to mimic the therapeutic potential of gene editing of haematopoietic progenitor cells to produce a class of HIV-resistant CD4+ T cells. We define the requirements for the permanent suppression of viral infection using gene editing as a novel therapeutic approach. We develop non-linear ordinary differential equation models to replicate HIV production in an infected host, incorporating the most appropriate aspects found in the many existing clinical models of HIV infection, and extend this model to include compartments representing HIV-resistant immune cells. Through an analysis of model equilibria and stability and computation of $R_0$ for both treated and untreated infections, we show that the proposed therapy has the potential to suppress HIV infection indefinitely and return CD4+ T cell counts to normal levels. A computational study for this treatment shows the potential for a successful 'functional cure' of HIV. A sensitivity analysis illustrates the consistency of numerical results with theoretical results and highlights the parameters requiring better biological justification. Simulations of varying level production of HIV-resistant CD4+ T cells and varying immune enhancements as the result of these indicate a clear threshold response of the model and a range of treatment parameters resulting in a return to normal CD4+ T cell counts.
Collapse
Affiliation(s)
| | - Seema Nanda
- Department of Mathematics, Dartmouth College, Hanover, USA
| | - Susan K Eszterhas
- Veterans Affairs Medical Center, White River Junction, USA.,Departments of Microbiology and Immunology, and Medicine, Geisel School of Medicine at Dartmouth, Lebanon, USA
| | - Alexandra L Howell
- Veterans Affairs Medical Center, White River Junction, USA.,Departments of Microbiology and Immunology, and Medicine, Geisel School of Medicine at Dartmouth, Lebanon, USA
| | | |
Collapse
|
10
|
Mechanistic basis of post-treatment control of SIV after anti-α4β7 antibody therapy. PLoS Comput Biol 2021; 17:e1009031. [PMID: 34106916 PMCID: PMC8189501 DOI: 10.1371/journal.pcbi.1009031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Treating macaques with an anti-α4β7 antibody under the umbrella of combination antiretroviral therapy (cART) during early SIV infection can lead to viral remission, with viral loads maintained at < 50 SIV RNA copies/ml after removal of all treatment in a subset of animals. Depletion of CD8+ lymphocytes in controllers resulted in transient recrudescence of viremia, suggesting that the combination of cART and anti-α4β7 antibody treatment led to a state where ongoing immune responses kept the virus undetectable in the absence of treatment. A previous mathematical model of HIV infection and cART incorporates immune effector cell responses and exhibits the property of two different viral load set-points. While the lower set-point could correspond to the attainment of long-term viral remission, attaining the higher set-point may be the result of viral rebound. Here we expand that model to include possible mechanisms of action of an anti-α4β7 antibody operating in these treated animals. We show that the model can fit the longitudinal viral load data from both IgG control and anti-α4β7 antibody treated macaques, suggesting explanations for the viral control associated with cART and an anti-α4β7 antibody treatment. This effective perturbation to the virus-host interaction can also explain observations in other nonhuman primate experiments in which cART and immunotherapy have led to post-treatment control or resetting of the viral load set-point. Interestingly, because the viral kinetics in the various treated animals differed—some animals exhibited large fluctuations in viral load after cART cessation—the model suggests that anti-α4β7 treatment could act by different primary mechanisms in different animals and still lead to post-treatment viral control. This outcome is nonetheless in accordance with a model with two stable viral load set-points, in which therapy can perturb the system from one set-point to a lower one through different biological mechanisms. Some macaques treated with an anti-α4β7 monoclonal antibody along with antiretroviral therapy during the early stages of simian immunodeficiency virus infection had their viral load become undetectable (below 50 SIV RNA copies/ml) after all treatment was stopped, whereas animals not given the antibody all had their viral loads rebound to high levels. Using a mathematical model, we examined four potential ways in which the antibody could have altered the balance between viral growth and immune control to maintain an undetectable viral load. We show that a shift to controlled infection can occur through multiple biologically reasonable mechanisms of action of the anti-α4β7 antibody.
Collapse
|
11
|
Santos-Beneit F, Raškevičius V, Skeberdis VA, Bordel S. A metabolic modeling approach reveals promising therapeutic targets and antiviral drugs to combat COVID-19. Sci Rep 2021; 11:11982. [PMID: 34099831 PMCID: PMC8184994 DOI: 10.1038/s41598-021-91526-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
In this study we have developed a method based on Flux Balance Analysis to identify human metabolic enzymes which can be targeted for therapeutic intervention against COVID-19. A literature search was carried out in order to identify suitable inhibitors of these enzymes, which were confirmed by docking calculations. In total, 10 targets and 12 bioactive molecules have been predicted. Among the most promising molecules we identified Triacsin C, which inhibits ACSL3, and which has been shown to be very effective against different viruses, including positive-sense single-stranded RNA viruses. Similarly, we also identified the drug Celgosivir, which has been successfully tested in cells infected with different types of viruses such as Dengue, Zika, Hepatitis C and Influenza. Finally, other drugs targeting enzymes of lipid metabolism, carbohydrate metabolism or protein palmitoylation (such as Propylthiouracil, 2-Bromopalmitate, Lipofermata, Tunicamycin, Benzyl Isothiocyanate, Tipifarnib and Lonafarnib) are also proposed.
Collapse
Affiliation(s)
| | - Vytautas Raškevičius
- Cell Culture Laboratory, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Vytenis A Skeberdis
- Cell Culture Laboratory, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Sergio Bordel
- Institute of Sustainable Processes, Universidad de Valladolid, Valladolid, Spain.
- Cell Culture Laboratory, Institute of Cardiology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| |
Collapse
|
12
|
Romano A, Casazza M, Gonella F. Addressing Non-linear System Dynamics of Single-Strand RNA Virus-Host Interaction. Front Microbiol 2021; 11:600254. [PMID: 33519741 PMCID: PMC7843927 DOI: 10.3389/fmicb.2020.600254] [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: 08/29/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
Positive single-strand ribonucleic acid [(+)ssRNA] viruses can cause multiple outbreaks, for which comprehensive tailored therapeutic strategies are still missing. Virus and host cell dynamics are tightly connected, generating a complex dynamics that conveys in virion assembly to ensure virus spread in the body. Starting from the knowledge of relevant processes in (+ss)RNA virus replication, transcription, translation, virions budding and shedding, and their respective energy costs, we built up a systems thinking (ST)-based diagram of the virus-host interaction, comprehensive of stocks, flows, and processes as well-described in literature. In ST approach, stocks and flows are expressed by a proxy of the energy embedded and transmitted, respectively, whereas processes are referred to the energy required for the system functioning. In this perspective, healthiness is just a particular configuration, in which stocks relevant for the system (equivalent but not limited to proteins, RNA, DNA, and all metabolites required for the survival) are constant, and the system behavior is stationary. At time of infection, the presence of additional stocks (e.g., viral protein and RNA and all metabolites required for virion assembly and spread) confers a complex network of feedbacks leading to new configurations, which can evolve to maximize the virions stock, thus changing the system structure, output, and purpose. The dynamic trajectories will evolve to achieve a new stationary status, a phenomenon described in microbiology as integration and symbiosis when the system is resilient enough to the changes, or the system may stop functioning and die. Application of external driving forces, acting on processes, can affect the dynamic trajectories adding a further degree of complexity, which can be captured by ST approach, used to address these new configurations. Investigation of system configurations in response to external driving forces acting is developed by computational analysis based on ST diagrams, with the aim at designing novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Romano
- Sezione di Ematologia, Dipartimento di Chirurgia Generale e Specialità Medico Chirurgiche (CHIRMED), Università degli Studi di Catania, Catania, Italy
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Marco Casazza
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Francesco Gonella
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca’ Foscari Venezia, Venezia, Italy
| |
Collapse
|
13
|
Environmental Restrictions: A New Concept Governing HIV-1 Spread Emerging from Integrated Experimental-Computational Analysis of Tissue-Like 3D Cultures. Cells 2020; 9:cells9051112. [PMID: 32365826 PMCID: PMC7291240 DOI: 10.3390/cells9051112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
HIV-1 can use cell-free and cell-associated transmission modes to infect new target cells, but how the virus spreads in the infected host remains to be determined. We recently established 3D collagen cultures to study HIV-1 spread in tissue-like environments and applied iterative cycles of experimentation and computation to develop a first in silico model to describe the dynamics of HIV-1 spread in complex tissue. These analyses (i) revealed that 3D collagen environments restrict cell-free HIV-1 infection but promote cell-associated virus transmission and (ii) defined that cell densities in tissue dictate the efficacy of these transmission modes for virus spread. In this review, we discuss, in the context of the current literature, the implications of this study for our understanding of HIV-1 spread in vivo, which aspects of in vivo physiology this integrated experimental-computational analysis takes into account, and how it can be further improved experimentally and in silico.
Collapse
|
14
|
Petravic J, Wilson DP. Simulating the entire natural course of HIV infection by extending the basic viral dynamics equations to include declining viral clearance. Pathog Dis 2020; 77:5545593. [PMID: 31397848 DOI: 10.1093/femspd/ftz043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 08/08/2019] [Indexed: 12/21/2022] Open
Abstract
The basic model of viral dynamics is a relatively simple set of equations describing the most essential features of the host-pathogen interactions. Coupled with data, it has been used extensively and successfully to reproduce and explain the features of the early acute phase of HIV infection and the effects of antiretroviral treatment, as well as to estimate the lifespan of infected cells, viral growth and clearance rates and predict early outcomes under different circumstances. However, it cannot reproduce the entire natural course of untreated HIV infection consistently with constant parameters. Here we show that it is possible to qualitatively reproduce the whole course of untreated HIV infection within the general framework of the basic model by assuming progressively declining viral clearance coupled with viral load. We discuss the interpretation of this model as proof-of-concept that may inspire further research into the role of viral clearance in HIV infection.
Collapse
Affiliation(s)
- Janka Petravic
- Burnet Institute, 85 Commercial Rd, Melbourne, VIC 3004, Australia
| | - David P Wilson
- Burnet Institute, 85 Commercial Rd, Melbourne, VIC 3004, Australia
| |
Collapse
|
15
|
Chen SS, Cheng CY, Rong L. Within-Host Viral Dynamics in a Multi-compartmental Environment. Bull Math Biol 2019; 81:4271-4308. [DOI: 10.1007/s11538-019-00658-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/09/2019] [Indexed: 11/29/2022]
|
16
|
Bons E, Regoes RR. Virus dynamics and phyloanatomy: Merging population dynamic and phylogenetic approaches. Immunol Rev 2019; 285:134-146. [PMID: 30129202 DOI: 10.1111/imr.12688] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In evolutionary biology and epidemiology, phylodynamic methods are widely used to infer population biological characteristics, such as the rates of replication, death, migration, or, in the epidemiological context, pathogen spread. More recently, these methods have been used to elucidate the dynamics of viruses within their hosts. Especially the application of phylogeographic approaches has the potential to shed light on anatomical colonization pathways and the exchange of viruses between distinct anatomical compartments. We and others have termed this phyloanatomy. Here, we review the promise and challenges of phyloanatomy, and compare them to more classical virus dynamics and population genetic approaches. We argue that the extremely strong selection pressures that exist within the host may represent the main obstacle to reliable phyloanatomic analysis.
Collapse
Affiliation(s)
- Eva Bons
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Roland R Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
GARIRA WINSTON, MAFUNDA MARTINCANAAN. FROM INDIVIDUAL HEALTH TO COMMUNITY HEALTH: TOWARDS MULTISCALE MODELING OF DIRECTLY TRANSMITTED INFECTIOUS DISEASE SYSTEMS. J BIOL SYST 2019. [DOI: 10.1142/s0218339019500074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In this paper, we present a new method for developing a class of nested multiscale models for directly transmitted infectious disease systems that integrates within-host scale and between-host scale using community pathogen load (CPL) as a new public health measure of a community’s level of infectiousness and as an indicator of the effectiveness of health interventions. The approach develops a multiscale modeling science base for directly transmitted infectious disease systems (where the inside-host environment’s biological entities such as cells, tissues, organs, body fluids, whole body are the reservoir of infective pathogen in the community) that is comparable to an existing multiscale modeling science base for environmentally transmitted infectious diseases (where the outside-host geographical environment’s physical entities such as soil, air, formites/contact surfaces, food and water are the reservoir of infective pathogen in the community) where pathogen load in the environment is explicitly incorporated into the model. This is achieved by assuming that infected hosts in the community are homogeneous and unevenly distributed microbial habitats. We illustrate the utility of this multiscale modeling methodology by evaluating the comparative effectiveness of HIV/AIDS preventive and treatment interventions as a case study.
Collapse
Affiliation(s)
- WINSTON GARIRA
- Modelling Health and Environmental Linkages Research Group (MHELRG), Department of Mathematics and Applied Mathematics, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa
| | - MARTIN CANAAN MAFUNDA
- Modelling Health and Environmental Linkages Research Group (MHELRG), Department of Mathematics and Applied Mathematics, University of Venda, Private Bag X5050, Thohoyandou 0950, South Africa
| |
Collapse
|
18
|
Spouge JL. An accurate approximation for the expected site frequency spectrum in a Galton-Watson process under an infinite sites mutation model. Theor Popul Biol 2019; 127:7-15. [PMID: 30876864 DOI: 10.1016/j.tpb.2019.03.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 01/26/2023]
Abstract
If viruses or other pathogens infect a single host, the outcome of infection often hinges on the fate of the initial invaders. The initial basic reproduction number R0, the expected number of cells infected by a single infected cell, helps determine whether the initial viruses can establish a successful beachhead. To determine R0, the Kingman coalescent or continuous-time birth-and-death process can be used to infer the rate of exponential growth in an historical population. Given M sequences sampled in the present, the two models can make the inference from the site frequency spectrum (SFS), the count of mutations that appear in exactly k sequences (k=1,2,…,M). In the case of viruses, however, if R0 is large and an infected cell bursts while propagating virus, the two models are suspect, because they are Markovian with only binary branching. Accordingly, this article develops an approximation for the SFS of a discrete-time branching process with synchronous generations (i.e., a Galton-Watson process). When evaluated in simulations with an asynchronous, non-Markovian model (a Bellman-Harris process) with parameters intended to mimic the bursting viral reproduction of HIV, the approximation proved superior to approximations derived from the Kingman coalescent or continuous-time birth-and-death process. This article demonstrates that in analogy to methods in human genetics, the SFS of viral sequences sampled well after latent infection can remain informative about the initial R0. Thus, it suggests the utility of analyzing the SFS of sequences derived from patient and animal trials of viral therapies, because in some cases, the initial R0 may be able to indicate subtle therapeutic progress, even in the absence of statistically significant differences in the infection of treatment and control groups.
Collapse
Affiliation(s)
- John L Spouge
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Hill AL, Rosenbloom DIS, Nowak MA, Siliciano RF. Insight into treatment of HIV infection from viral dynamics models. Immunol Rev 2018; 285:9-25. [PMID: 30129208 PMCID: PMC6155466 DOI: 10.1111/imr.12698] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The odds of living a long and healthy life with HIV infection have dramatically improved with the advent of combination antiretroviral therapy. Along with the early development and clinical trials of these drugs, and new field of research emerged called viral dynamics, which uses mathematical models to interpret and predict the time-course of viral levels during infection and how they are altered by treatment. In this review, we summarize the contributions that virus dynamics models have made to understanding the pathophysiology of infection and to designing effective therapies. This includes studies of the multiphasic decay of viral load when antiretroviral therapy is given, the evolution of drug resistance, the long-term persistence latently infected cells, and the rebound of viremia when drugs are stopped. We additionally discuss new work applying viral dynamics models to new classes of investigational treatment for HIV, including latency-reversing agents and immunotherapy.
Collapse
Affiliation(s)
- Alison L. Hill
- Program for Evolutionary DynamicsHarvard UniversityCambridgeMassachusetts
| | - Daniel I. S. Rosenbloom
- Department of PharmacokineticsPharmacodynamics, & Drug MetabolismMerck Research LaboratoriesKenilworthNew Jersey
| | - Martin A. Nowak
- Program for Evolutionary DynamicsHarvard UniversityCambridgeMassachusetts
| | - Robert F. Siliciano
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMaryland
- Howard Hughes Medical InstituteBaltimoreMaryland
| |
Collapse
|
20
|
Exploring an alternative explanation for the second phase of viral decay: Infection of short-lived cells in a drug-limited compartment during HAART. PLoS One 2018; 13:e0198090. [PMID: 30016329 PMCID: PMC6049925 DOI: 10.1371/journal.pone.0198090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/14/2018] [Indexed: 11/19/2022] Open
Abstract
Most HIV-infected patients who initiate combination antiretroviral therapy experience a viral load decline in several phases. These phases are characterized by different rates of viral load decay that decrease when transitioning from one phase to the next. There is no consensus as to the origin of these phases. One hypothesis put forward is that short- and long-lived infected cells are responsible for the first and second phases of decay, respectively. However, significant differences in drug concentrations are observed in monocytes from various tissues, suggesting the first two phases of decay in viral loads could instead be attributed to short-lived cells being differently exposed to drugs. Compared to a well-exposed compartment, new cell infection can be expected in a compartment with limited drug exposure, thus leading to a slower viral load decay with potential virologic failure and drug resistance. In the current study, the latter hypothesis was investigated using a model of viral kinetics. Empirical datasets were involved in model elaboration and parameter estimation. In particular, susceptibility assay data was used for an in vitro to in vivo extrapolation based on the expected drug concentrations inside physiological compartments. Results from numerical experiments of the short-term evolution of viral loads can reproduce the first two phases of viral decay when allowing new short-lived cell infections in an unidentified drug-limited compartment. Model long-term predictions are however less consistent with clinical observations. For the hypothesis to hold, efavirenz, tenofovir and emtricitabine drug exposure in the drug-limited compartment would have to be very low compared to exposure in peripheral blood. This would lead to significant long-term viral growth and the frequent development of resistant strains, a prediction not supported by clinical observations. This suggests that the existence of a drug-limited anatomical compartment is unlikely, by itself, to explain the second phase of viral load decay.
Collapse
|
21
|
Chopard C, Tong PBV, Tóth P, Schatz M, Yezid H, Debaisieux S, Mettling C, Gross A, Pugnière M, Tu A, Strub JM, Mesnard JM, Vitale N, Beaumelle B. Cyclophilin A enables specific HIV-1 Tat palmitoylation and accumulation in uninfected cells. Nat Commun 2018; 9:2251. [PMID: 29884859 PMCID: PMC5993824 DOI: 10.1038/s41467-018-04674-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 05/15/2018] [Indexed: 12/21/2022] Open
Abstract
Most HIV-1 Tat is unconventionally secreted by infected cells following Tat interaction with phosphatidylinositol (4,5) bisphosphate (PI(4,5)P2) at the plasma membrane. Extracellular Tat is endocytosed by uninfected cells before escaping from endosomes to reach the cytosol and bind PI(4,5)P2. It is not clear whether and how incoming Tat concentrates in uninfected cells. Here we show that, in uninfected cells, the S-acyl transferase DHHC-20 together with the prolylisomerases cyclophilin A (CypA) and FKBP12 palmitoylate Tat on Cys31 thereby increasing Tat affinity for PI(4,5)P2. In infected cells, CypA is bound by HIV-1 Gag, resulting in its encapsidation and CypA depletion from cells. Because of the lack of this essential cofactor, Tat is not palmitoylated in infected cells but strongly secreted. Hence, Tat palmitoylation specifically takes place in uninfected cells. Moreover, palmitoylation is required for Tat to accumulate at the plasma membrane and affect PI(4,5)P2-dependent membrane traffic such as phagocytosis and neurosecretion. It is not clear whether and how incoming HIV-1 Tat accumulates in uninfected cells. Here, Chopard et al. show that, in uninfected cells, incoming Tat is palmitoylated on Cys31 by DHHC-20, which increases its affinity for PI(4,5)P2 and results in its accumulation at the plasma membrane.
Collapse
Affiliation(s)
- Christophe Chopard
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Phuoc Bao Viet Tong
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Petra Tóth
- INCI, UPR 3212 CNRS, 5 rue Blaise Pascal, 67084, Strasbourg, France.
| | - Malvina Schatz
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Hocine Yezid
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Solène Debaisieux
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Clément Mettling
- IGH, UPR 1142 CNRS, 141 Rue de la Cardonille, 34396, Montpellier, France
| | - Antoine Gross
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Martine Pugnière
- IRCM, INSERM U 1194, 208 Rue des Apothicaires, 34298, Montpellier, France
| | - Annie Tu
- INCI, UPR 3212 CNRS, 5 rue Blaise Pascal, 67084, Strasbourg, France
| | - Jean-Marc Strub
- CNRS, IPHC UMR 7178, Université de Strasbourg, 67000, Strasbourg, France
| | - Jean-Michel Mesnard
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France
| | - Nicolas Vitale
- INCI, UPR 3212 CNRS, 5 rue Blaise Pascal, 67084, Strasbourg, France.,INSERM, 75654, Paris Cedex 13, France
| | - Bruno Beaumelle
- IRIM, UMR 9004, Université de Montpellier-CNRS, 1919 Route de Mende, 34293, Montpellier, France.
| |
Collapse
|
22
|
Are microRNAs Important Players in HIV-1 Infection? An Update. Viruses 2018; 10:v10030110. [PMID: 29510515 PMCID: PMC5869503 DOI: 10.3390/v10030110] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 02/21/2018] [Accepted: 02/25/2018] [Indexed: 12/15/2022] Open
Abstract
HIV-1 has already claimed over 35 million human lives globally. No curative treatments are currently available, and the only treatment option for over 36 million people currently living with HIV/AIDS are antiretroviral drugs that disrupt the function of virus-encoded proteins. However, such virus-targeted therapeutic strategies are constrained by the ability of the virus to develop drug-resistance. Despite major advances in HIV/AIDS research over the years, substantial knowledge gaps exist in many aspects of HIV-1 replication, especially its interaction with the host. Hence, understanding the mechanistic details of virus–host interactions may lead to novel therapeutic strategies for the prevention and/or management of HIV/AIDS. Notably, unprecedented progress in deciphering host gene silencing processes mediated by several classes of cellular small non-coding RNAs (sncRNA) presents a promising and timely opportunity for developing non-traditional antiviral therapeutic strategies. Cellular microRNAs (miRNA) belong to one such important class of sncRNAs that regulate protein synthesis. Evidence is mounting that cellular miRNAs play important roles in viral replication, either usurped by the virus to promote its replication or employed by the host to control viral infection by directly targeting the viral genome or by targeting cellular proteins required for productive virus replication. In this review, we summarize the findings to date on the role of miRNAs in HIV-1 biology.
Collapse
|
23
|
Pinkevych M, Fennessey CM, Cromer D, Tolstrup M, Søgaard OS, Rasmussen TA, Keele BF, Davenport MP. Estimating Initial Viral Levels during Simian Immunodeficiency Virus/Human Immunodeficiency Virus Reactivation from Latency. J Virol 2018; 92:e01667-17. [PMID: 29118123 PMCID: PMC5752936 DOI: 10.1128/jvi.01667-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 11/01/2017] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV) viremia rebounds rapidly after treatment interruption, and a variety of strategies are being explored to reduce or control viral reactivation posttreatment. This viral rebound arises from reactivation of individual latently infected cells, which spread during ongoing rounds of productive infection. The level of virus produced by the initial individual reactivating cells is not known, although it may have major implications for the ability of different immune interventions to control viral rebound. Here we use data from both HIV and simian immunodeficiency virus (SIV) treatment interruption studies to estimate the initial viral load postinterruption and thereby the initial individual reactivation event. Using a barcoded virus (SIVmac239M) to track reactivation from individual latent cells, we use the observed viral growth rates and frequency of reactivation to model the dynamics of reactivation to estimate that a single reactivated latent cell can produce an average viral load equivalent to ∼0.1 to 0.5 viral RNA (vRNA) copies/ml. Modeling of treatment interruption in HIV suggests an initial viral load equivalent of ∼0.6 to 1 vRNA copies/ml. These low viral loads immediately following latent cell reactivation provide a window of opportunity for viral control by host immunity, before further replication allows viral spread. This work shows the initial levels of viral production that must be controlled in order to successfully suppress HIV reactivation following treatment interruption.IMPORTANCE Current treatment for HIV is able to suppress viral replication and prevent disease progression. However, treatment cannot eradicate infection, because the virus lies silent within latently infected cells. If treatment is stopped, the virus usually rebounds above the level of detection within a few weeks. There are a number of approaches being tested aimed at either eradicating latently infected cells or controlling the virus if it returns. Studying both the small pool of latently infected cells and the early events during viral reactivation is difficult, because these involve very small levels of virus that are difficult to measure directly. Here, we combine experimental data and mathematical modeling to understand the very early events during viral reactivation from latency in both HIV infection of humans and SIV infection of monkeys. We find that the initial levels of virus are low, which may help in designing therapies to control early viral reactivation.
Collapse
Affiliation(s)
- Mykola Pinkevych
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW, Australia
| | - Christine M Fennessey
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Deborah Cromer
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW, Australia
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Ole S Søgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Thomas A Rasmussen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Miles P Davenport
- Infection Analytics Program, Kirby Institute for Infection and Immunity, UNSW Australia, Sydney, NSW, Australia
| |
Collapse
|
24
|
De Boer RJ, Perelson AS. How Germinal Centers Evolve Broadly Neutralizing Antibodies: the Breadth of the Follicular Helper T Cell Response. J Virol 2017; 91:e00983-17. [PMID: 28878083 PMCID: PMC5660473 DOI: 10.1128/jvi.00983-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/11/2017] [Indexed: 12/20/2022] Open
Abstract
Many HIV-1-infected patients evolve broadly neutralizing antibodies (bnAbs). This evolutionary process typically takes several years and is poorly understood as selection taking place in germinal centers occurs on the basis of antibody affinity. B cells with the highest-affinity receptors tend to acquire the most antigen from the follicular dendritic cell (FDC) network and present the highest density of cognate peptides to follicular helper T (Tfh) cells, which provide survival signals to the B cell. bnAbs are therefore expected to evolve only when the B cell lineage evolving breadth is consistently capturing and presenting more peptides to Tfh cells than other lineages of more specific B cells. Here we develop mathematical models of Tfh cells in germinal centers to explicitly define the mechanisms of selection in this complex evolutionary process. Our results suggest that broadly reactive B cells presenting a high density of peptides bound to major histocompatibility complex class II molecules (pMHC) are readily outcompeted by B cells responding to lineages of HIV-1 that transiently dominate the within host viral population. Conversely, if broadly reactive B cells acquire a large variety of several HIV-1 proteins from the FDC network and present a high diversity of several pMHC, they can be rescued by a large fraction of the Tfh cell repertoire in the germinal center. Under such circumstances the evolution of bnAbs is much more consistent. Increasing either the magnitude of the Tfh cell response or the breadth of the Tfh cell repertoire markedly facilitates the evolution of bnAbs. Because both the magnitude and breadth can be increased by vaccination with several HIV-1 proteins, this calls for experimental testing.IMPORTANCE Many HIV-infected patients slowly evolve antibodies that can neutralize a large variety of viruses. Such broadly neutralizing antibodies (bnAbs) could in the future become therapeutic agents. bnAbs appear very late, and patients are typically not protected by them. At the moment, we fail to understand why this takes so long and how the immune system selects for broadly neutralizing capacity. Typically, antibodies are selected based on affinity and not on breadth. We developed mathematical models to study two different mechanisms by which the immune system can select for broadly neutralizing capacity. One of these is based upon the repertoire of different follicular helper T (Tfh) cells in germinal centers. We suggest that broadly reactive B cells may interact with a larger fraction of this repertoire and demonstrate that this would select for bnAbs. Intriguingly, this suggests that broadening the Tfh cell repertoire by vaccination may speed up the evolution of bnAbs.
Collapse
Affiliation(s)
- Rob J De Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, The Netherlands
- Santa Fe Institute, Santa Fe, New Mexico, USA
| | - Alan S Perelson
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
- Santa Fe Institute, Santa Fe, New Mexico, USA
| |
Collapse
|
25
|
Sanche S, Sheehan N, Mesplède T, Wainberg MA, Li J, Nekka F. A Mathematical Model to Predict HIV Virological Failure and Elucidate the Role of Lymph Node Drug Penetration. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:469-476. [PMID: 28556627 PMCID: PMC5706346 DOI: 10.1002/psp4.12200] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/28/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022]
Abstract
Preventing virological failure following HIV treatment remains a difficult task that is further complicated by the emergence of drug resistance. We have developed a mathematical model able to explain and predict HIV virological outcomes for various compounds and patients' drug intake patterns. Compared to current approaches, this model considers, altogether, drug penetration into lymph nodes, a refined adherence representation accounting for the propensity for long drug holidays, population pharmacokinetic and pharmacodynamic variability, drug interaction, and crossresistance. In silico results are consistent with clinical observations for treatment with efavirenz, efavirenz in association with tenofovir DF and emtricitabine, or boosted darunavir. Our findings indicate that limited lymph node drug penetration can account for a large proportion of cases of virological failure and drug resistance. Since a limited amount of information is required by the model, it can be of use in the process of drug discovery and to guide clinical treatment strategies.
Collapse
Affiliation(s)
- S Sanche
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Québec, Canada
| | - N Sheehan
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Québec, Canada.,Chronic Viral Illness Service, McGill University Health Centre, Montréal, Québec, Canada.,McGill University AIDS Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - T Mesplède
- McGill University AIDS Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - M A Wainberg
- McGill University AIDS Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montréal, Québec, Canada
| | - J Li
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Québec, Canada
| | - F Nekka
- Faculté de Pharmacie de l'Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
26
|
Konrad BP, Taylor D, Conway JM, Ogilvie GS, Coombs D. On the duration of the period between exposure to HIV and detectable infection. Epidemics 2017; 20:73-83. [PMID: 28365331 DOI: 10.1016/j.epidem.2017.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/02/2017] [Accepted: 03/08/2017] [Indexed: 11/16/2022] Open
Abstract
HIV infection cannot be detected immediately after exposure because plasma viral loads are too small initially. The duration of this phase of infection (the "eclipse period") is difficult to estimate because precise dates of exposure are rarely known. Therefore, the reliability of clinical HIV testing during the first few weeks of infection is unknown, creating anxiety among HIV-exposed individuals and their physicians. We address this by fitting stochastic models of early HIV infection to detailed viral load records for 78 plasma donors, taken during the period of exposure and infection. We first show that the classic stochastic birth-death model does not satisfactorily describe early infection. We therefore apply a different stochastic model that includes infected cells and virions separately. Since every plasma donor in our data eventually becomes infected, we must condition the model to reflect this bias, before fitting to the data. Applying our best estimates of unknown parameter values, we estimate the mean eclipse period to be 8-10 days. We further estimate the reliability of a negative test t days after potential exposure.
Collapse
Affiliation(s)
- Bernhard P Konrad
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Darlene Taylor
- British Columbia Centre for Disease Control, 655 W 12th Ave., Vancouver, BC V5Z 4R4, Canada
| | - Jessica M Conway
- Department of Mathematics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Gina S Ogilvie
- British Columbia Centre for Disease Control, 655 W 12th Ave., Vancouver, BC V5Z 4R4, Canada
| | - Daniel Coombs
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada.
| |
Collapse
|
27
|
Wagner TA. Combining Cell and Gene Therapy in an Effort to Eradicate HIV. AIDS Patient Care STDS 2016; 30:534-538. [PMID: 27905840 DOI: 10.1089/apc.2016.0226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
More than 30 million people are infected with HIV, and HIV remains the fifth leading cause of disability-adjusted life years worldwide. Antiretroviral therapy (ART) dramatically decreases mortality rate, but there are side effects, long-term toxicities, expenses, stigmas, and inconveniences associated with chronic treatment, and HIV-infected individuals on ART have an increased risk of malignancies, cardiovascular disease, neurologic disease, and shortened life expectancy. Therefore, a cure for HIV remains an important goal. Combining new cell and gene therapy technology is an exciting approach that appears promising in vitro. Animal testing and careful clinical trials will be needed to determine if these strategies are clinically useful.
Collapse
Affiliation(s)
- Thor A. Wagner
- Department of Pediatrics, University of Washington, Seattle, Washington
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington
| |
Collapse
|
28
|
Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature 2016; 540:284-287. [PMID: 27841870 PMCID: PMC5145754 DOI: 10.1038/nature20583] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/01/2016] [Indexed: 02/07/2023]
Abstract
The development of immunologic interventions that can target the viral reservoir in HIV-1-infected individuals is a major goal of the HIV-1 cure field1,2. However, little evidence exists that the viral reservoir can be sufficiently targeted to improve virologic control following discontinuation of antiretroviral therapy (ART). Here we show that Ad26/MVA3,4 therapeutic vaccination with toll-like receptor 7 (TLR7) stimulation improves virologic control and delays viral rebound following ART discontinuation in SIV-infected rhesus monkeys that initiated ART during acute infection. Ad26/MVA therapeutic vaccination resulted in a dramatic increase in the magnitude and breadth of SIV-specific cellular immune responses in virologically suppressed, SIV-infected monkeys. TLR7 agonist administration led to innate immune stimulation and cellular immune activation. The combination of Ad26/MVA vaccination and TLR7 stimulation resulted in decreased levels of viral DNA in lymph nodes and peripheral blood, as well as improved virologic control and delayed viral rebound following ART discontinuation. Cellular immune breadth correlated inversely with setpoint viral loads and correlated directly with time to viral rebound. These data demonstrate the potential of therapeutic vaccination with innate immune stimulation as a strategy aimed at an HIV-1 functional cure.
Collapse
|
29
|
Lythgoe KA, Blanquart F, Pellis L, Fraser C. Large Variations in HIV-1 Viral Load Explained by Shifting-Mosaic Metapopulation Dynamics. PLoS Biol 2016; 14:e1002567. [PMID: 27706164 PMCID: PMC5051940 DOI: 10.1371/journal.pbio.1002567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/08/2016] [Indexed: 12/17/2022] Open
Abstract
The viral population of HIV-1, like many pathogens that cause systemic infection, is structured and differentiated within the body. The dynamics of cellular immune trafficking through the blood and within compartments of the body has also received wide attention. Despite these advances, mathematical models, which are widely used to interpret and predict viral and immune dynamics in infection, typically treat the infected host as a well-mixed homogeneous environment. Here, we present mathematical, analytical, and computational results that demonstrate that consideration of the spatial structure of the viral population within the host radically alters predictions of previous models. We study the dynamics of virus replication and cytotoxic T lymphocytes (CTLs) within a metapopulation of spatially segregated patches, representing T cell areas connected by circulating blood and lymph. The dynamics of the system depend critically on the interaction between CTLs and infected cells at the within-patch level. We show that for a wide range of parameters, the system admits an unexpected outcome called the shifting-mosaic steady state. In this state, the whole body’s viral population is stable over time, but the equilibrium results from an underlying, highly dynamic process of local infection and clearance within T-cell centers. Notably, and in contrast to previous models, this new model can explain the large differences in set-point viral load (SPVL) observed between patients and their distribution, as well as the relatively low proportion of cells infected at any one time, and alters the predicted determinants of viral load variation. A novel metapopulation model of HIV suggests that within-host infections are characterized by a highly dynamic process of localized infection followed by clearance within T cell centers. When a person is infected with HIV, the initial peak level of virus in the blood is usually very high before a lower, relatively stable level is reached and maintained for the duration of the chronic infection. This stable level is known as the set-point viral load (SPVL) and is associated with severity of infection. SPVL is also highly variable among patients, ranging from 100 to a million copies of the virus per mL of blood. The replicative capacity of the infecting virus and the strength of the immune response both influence SPVL. However, standard mathematical models show that variation in these two factors cannot easily reproduce the observed distribution of SPVL among patients. Standard models typically treat infected individuals as well-mixed systems, but in reality viral replication is localised in T-cell centres, or patches, found in secondary lymphoid tissue. To account for this population structure, we developed a carefully parameterised metapopulation model. We find the system can reach a steady state at which the viral load in the blood is relatively stable, representing SPVL, but surprisingly, the patches are highly dynamic, characterised by bursts of infection followed by elimination of virus due to localised host immune responses. Significantly, this model can reproduce the wide distribution of SPVLs found among infected individuals for realistic distributions of viral replicative capacity and strength of immune response. Our model can also be used in the future to understand other aspects of chronic HIV infection.
Collapse
Affiliation(s)
- Katrina A. Lythgoe
- Department of Zoology, Tinbergen Building, University of Oxford, Oxford, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
- * E-mail:
| | - François Blanquart
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Lorenzo Pellis
- Mathematics Institute, Zeeman Building, University of Warwick, Coventry, United Kingdom
| | - Christophe Fraser
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
30
|
Multi-dose Romidepsin Reactivates Replication Competent SIV in Post-antiretroviral Rhesus Macaque Controllers. PLoS Pathog 2016; 12:e1005879. [PMID: 27632364 PMCID: PMC5025140 DOI: 10.1371/journal.ppat.1005879] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/18/2016] [Indexed: 11/19/2022] Open
Abstract
Viruses that persist despite seemingly effective antiretroviral treatment (ART) and can reinitiate infection if treatment is stopped preclude definitive treatment of HIV-1 infected individuals, requiring lifelong ART. Among strategies proposed for targeting these viral reservoirs, the premise of the “shock and kill” strategy is to induce expression of latent proviruses [for example with histone deacetylase inhibitors (HDACis)] resulting in elimination of the affected cells through viral cytolysis or immune clearance mechanisms. Yet, ex vivo studies reported that HDACis have variable efficacy for reactivating latent proviruses, and hinder immune functions. We developed a nonhuman primate model of post-treatment control of SIV through early and prolonged administration of ART and performed in vivo reactivation experiments in controller RMs, evaluating the ability of the HDACi romidepsin (RMD) to reactivate SIV and the impact of RMD treatment on SIV-specific T cell responses. Ten RMs were IV-infected with a SIVsmmFTq transmitted-founder infectious molecular clone. Four RMs received conventional ART for >9 months, starting from 65 days post-infection. SIVsmmFTq plasma viremia was robustly controlled to <10 SIV RNA copies/mL with ART, without viral blips. At ART cessation, initial rebound viremia to ~106 copies/mL was followed by a decline to < 10 copies/mL, suggesting effective immune control. Three post-treatment controller RMs received three doses of RMD every 35–50 days, followed by in vivo experimental depletion of CD8+ cells using monoclonal antibody M-T807R1. RMD was well-tolerated and resulted in a rapid and massive surge in T cell activation, as well as significant virus rebounds (~104 copies/ml) peaking at 5–12 days post-treatment. CD8+ cell depletion resulted in a more robust viral rebound (107 copies/ml) that was controlled upon CD8+ T cell recovery. Our results show that RMD can reactivate SIV in vivo in the setting of post-ART viral control. Comparison of the patterns of virus rebound after RMD administration and CD8+ cell depletion suggested that RMD impact on T cells is only transient and does not irreversibly alter the ability of SIV-specific T cells to control the reactivated virus. Antiretroviral therapy (ART) does not eradicate HIV-1 in infected individuals due to virus persistence in latently infected reservoir cells, despite apparently effective ART. The persistent virus and can rekindle infection when ART is interrupted. The goal of the “shock and kill” viral clearance strategy is to induce expression of latent proviruses and eliminate the infected cells through viral cytolysis or immune clearance mechanisms. Latency reversing agents (LRAs) tested to date have been reported to have variable effects, both on virus reactivation and on immune functions. We performed in vivo reactivation experiments in SIV-infected RMs that controlled viral replication after a period of ART to evaluate the ability of the histone deacetylase inhibitor romidepsin (RMD) to reactivate SIV and its impact on SIV-specific immune responses. Our results suggest that RMD treatment can increase virus expression in this setting, and that it does not markedly or durably impair the ability of SIV-specific T cells to control viral replication.
Collapse
|
31
|
Notwithstanding Circumstantial Alibis, Cytotoxic T Cells Can Be Major Killers of HIV-1-Infected Cells. J Virol 2016; 90:7066-7083. [PMID: 27226367 PMCID: PMC4984658 DOI: 10.1128/jvi.00306-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Several experiments suggest that in the chronic phase of human immunodeficiency virus type 1 (HIV-1) infection, CD8+ cytotoxic T lymphocytes (CTL) contribute very little to the death of productively infected cells. First, the expected life span of productively infected cells is fairly long, i.e., about 1 day. Second, this life span is hardly affected by the depletion of CD8+ T cells. Third, the rate at which mutants escaping a CTL response take over the viral population tends to be slow. Our main result is that all these observations are perfectly compatible with killing rates that are much faster than one per day once we invoke the fact that infected cells proceed through an eclipse phase of about 1 day before they start producing virus. Assuming that the major protective effect of CTL is cytolytic, we demonstrate that mathematical models with an eclipse phase account for the data when the killing is fast and when it varies over the life cycle of infected cells. Considering the steady state corresponding to the chronic phase of the infection, we find that the rate of immune escape and the rate at which the viral load increases following CD8+ T cell depletion should reflect the viral replication rate, ρ. A meta-analysis of previous data shows that viral replication rates during chronic infection vary between 0.5 ≤ ρ ≤ 1 day−1. Balancing such fast viral replication requires killing rates that are several times larger than ρ, implying that most productively infected cells would die by cytolytic effects. IMPORTANCE Most current data suggest that cytotoxic T cells (CTL) mediate their control of human immunodeficiency virus type 1 (HIV-1) infection by nonlytic mechanisms; i.e., the data suggest that CTL hardly kill. This interpretation of these data has been based upon the general mathematical model for HIV infection. Because this model ignores the eclipse phase between the infection of a target cell and the start of viral production by that cell, we reanalyze the same data sets with novel models that do account for the eclipse phase. We find that the data are perfectly consistent with lytic control by CTL and predict that most productively infected cells are killed by CTL. Because the killing rate should balance the viral replication rate, we estimate both parameters from a large set of published experiments in which CD8+ T cells were depleted in simian immunodeficiency virus (SIV)-infected monkeys. This confirms that the killing rate can be much faster than is currently appreciated.
Collapse
|
32
|
Lu CL, Murakowski DK, Bournazos S, Schoofs T, Sarkar D, Halper-Stromberg A, Horwitz JA, Nogueira L, Golijanin J, Gazumyan A, Ravetch JV, Caskey M, Chakraborty AK, Nussenzweig MC. Enhanced clearance of HIV-1-infected cells by broadly neutralizing antibodies against HIV-1 in vivo. Science 2016; 352:1001-4. [PMID: 27199430 DOI: 10.1126/science.aaf1279] [Citation(s) in RCA: 276] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 04/11/2016] [Indexed: 01/09/2023]
Abstract
Antiretroviral drugs and antibodies limit HIV-1 infection by interfering with the viral life cycle. In addition, antibodies also have the potential to guide host immune effector cells to kill HIV-1-infected cells. Examination of the kinetics of HIV-1 suppression in infected individuals by passively administered 3BNC117, a broadly neutralizing antibody, suggested that the effects of the antibody are not limited to free viral clearance and blocking new infection but also include acceleration of infected cell clearance. Consistent with these observations, we find that broadly neutralizing antibodies can target CD4(+) T cells infected with patient viruses and can decrease their in vivo half-lives by a mechanism that requires Fcγ receptor engagement in a humanized mouse model. The results indicate that passive immunotherapy can accelerate elimination of HIV-1-infected cells.
Collapse
Affiliation(s)
- Ching-Lan Lu
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA. Weill Cornell Medical College, New York, NY 10065, USA
| | - Dariusz K Murakowski
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stylianos Bournazos
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Till Schoofs
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Debolina Sarkar
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Joshua A Horwitz
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Lilian Nogueira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jovana Golijanin
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Anna Gazumyan
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Jeffrey V Ravetch
- Laboratory of Molecular Genetics and Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Arup K Chakraborty
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA. Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA. Howard Hughes Medical Institute.
| |
Collapse
|
33
|
Kumberger P, Frey F, Schwarz US, Graw F. Multiscale modeling of virus replication and spread. FEBS Lett 2016; 590:1972-86. [PMID: 26878104 DOI: 10.1002/1873-3468.12095] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 01/21/2016] [Accepted: 02/07/2016] [Indexed: 01/16/2023]
Abstract
Replication and spread of human viruses is based on the simultaneous exploitation of many different host functions, bridging multiple scales in space and time. Mathematical modeling is essential to obtain a systems-level understanding of how human viruses manage to proceed through their life cycles. Here, we review corresponding advances for viral systems of large medical relevance, such as human immunodeficiency virus-1 (HIV-1) and hepatitis C virus (HCV). We will outline how the combination of mathematical models and experimental data has advanced our quantitative knowledge about various processes of these pathogens, and how novel quantitative approaches promise to fill remaining gaps.
Collapse
Affiliation(s)
- Peter Kumberger
- BioQuant-Center, Heidelberg University, Germany.,Center for Modeling and Simulation in the Biosciences (BIOMS), Heidelberg University, Germany
| | - Felix Frey
- BioQuant-Center, Heidelberg University, Germany.,Institute for Theoretical Physics, Heidelberg University, Germany
| | - Ulrich S Schwarz
- BioQuant-Center, Heidelberg University, Germany.,Institute for Theoretical Physics, Heidelberg University, Germany
| | - Frederik Graw
- BioQuant-Center, Heidelberg University, Germany.,Center for Modeling and Simulation in the Biosciences (BIOMS), Heidelberg University, Germany
| |
Collapse
|
34
|
Butler TC, Barton JP, Kardar M, Chakraborty AK. Identification of drug resistance mutations in HIV from constraints on natural evolution. Phys Rev E 2016; 93:022412. [PMID: 26986367 DOI: 10.1103/physreve.93.022412] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Indexed: 11/07/2022]
Abstract
Human immunodeficiency virus (HIV) evolves with extraordinary rapidity. However, its evolution is constrained by interactions between mutations in its fitness landscape. Here we show that an Ising model describing these interactions, inferred from sequence data obtained prior to the use of antiretroviral drugs, can be used to identify clinically significant sites of resistance mutations. Successful predictions of the resistance sites indicate progress in the development of successful models of real viral evolution at the single residue level and suggest that our approach may be applied to help design new therapies that are less prone to failure even where resistance data are not yet available.
Collapse
Affiliation(s)
- Thomas C Butler
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - John P Barton
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02139, USA
| | - Mehran Kardar
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Arup K Chakraborty
- Department of Physics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts 02139, USA.,Departments of Chemistry and Biological Engineering, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
35
|
Conway JM, Perelson AS. Residual Viremia in Treated HIV+ Individuals. PLoS Comput Biol 2016; 12:e1004677. [PMID: 26735135 PMCID: PMC4703306 DOI: 10.1371/journal.pcbi.1004677] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 11/26/2015] [Indexed: 12/20/2022] Open
Abstract
Antiretroviral therapy (ART) effectively controls HIV infection, suppressing HIV viral loads. However, some residual virus remains, below the level of detection, in HIV-infected patients on ART. The source of this viremia is an area of debate: does it derive primarily from activation of infected cells in the latent reservoir, or from ongoing viral replication? Observations seem to be contradictory: there is evidence of short term evolution, implying that there must be ongoing viral replication, and viral strains should thus evolve. However, phylogenetic analyses, and rare emergent drug resistance, suggest no long-term viral evolution, implying that virus derived from activated latent cells must dominate. We use simple deterministic and stochastic models to gain insight into residual viremia dynamics in HIV-infected patients. Our modeling relies on two underlying assumptions for patients on suppressive ART: that latent cell activation drives viral dynamics and that the reproductive ratio of treated infection is less than 1. Nonetheless, the contribution of viral replication to residual viremia in patients on ART may be non-negligible. However, even if the portion of viremia attributable to viral replication is significant, our model predicts (1) that latent reservoir re-seeding remains negligible, and (2) some short-term viral evolution is permitted, but long-term evolution can still be limited: stochastic analysis of our model shows that de novo emergence of drug resistance is rare. Thus, our simple models reconcile the seemingly contradictory observations on residual viremia and, with relatively few parameters, recapitulates HIV viral dynamics observed in patients on suppressive therapy. In HIV+ individuals, antiretroviral therapy (ART) effectively controls HIV viral loads to below levels detectable by routine tests. However, more sensitive tests can detect some residual viremia. The source of this virus is a matter of debate: does it derive from ongoing viral replication, or from viral production following activation of latently infected cells? Experimental observations support both sides of the argument: in patients on therapy, HIV shows no long-term evolution, and emergence of drug-resistant mutants is rare, implying no ongoing viral replication, but there remains short-term evolution, implying the opposite. To reconcile these observations, we propose a mathematical model of latently and productively infected cells and virus. Using our models we predict that, in patients on suppressive ART, the contribution of viral replication to residual virus, while small, yields short term-evolution. But even if the contribution is large, for example if adherence to therapy is poor, long-term evolution can still be limited, and de novo emergence of drug resistance is rare. Thus, our simple models reconcile the seemingly contradictory observations on residual viremia.
Collapse
Affiliation(s)
- Jessica M. Conway
- Department of Mathematics and Center for Infectious Disease Dynamics (CIDD), The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| | - Alan S. Perelson
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| |
Collapse
|
36
|
Pandit A, de Boer RJ. HIV-1 CCR5 gene therapy will fail unless it is combined with a suicide gene. Sci Rep 2015; 5:18088. [PMID: 26674113 PMCID: PMC4682191 DOI: 10.1038/srep18088] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022] Open
Abstract
Highly active antiretroviral therapy (ART) has successfully turned Human immunodeficiency virus type 1 (HIV-1) from a deadly pathogen into a manageable chronic infection. ART is a lifelong therapy which is both expensive and toxic, and HIV can become resistant to it. An alternative to lifelong ART is gene therapy that targets the CCR5 co-receptor and creates a population of genetically modified host cells that are less susceptible to viral infection. With generic mathematical models we show that gene therapy that only targets the CCR5 co-receptor fails to suppress HIV-1 (which is in agreement with current data). We predict that the same gene therapy can be markedly improved if it is combined with a suicide gene that is only expressed upon HIV-1 infection.
Collapse
Affiliation(s)
- Aridaman Pandit
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, 3584CH, The Netherlands
| | - Rob J de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, 3584CH, The Netherlands
| |
Collapse
|
37
|
Maziane M, Lotfi EM, Hattaf K, Yousfi N. Dynamics of a Class of HIV Infection Models with Cure of Infected Cells in Eclipse Stage. Acta Biotheor 2015; 63:363-80. [PMID: 26082312 DOI: 10.1007/s10441-015-9263-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 06/09/2015] [Indexed: 02/07/2023]
Abstract
In this paper, we propose two HIV infection models with specific nonlinear incidence rate by including a class of infected cells in the eclipse phase. The first model is described by ordinary differential equations (ODEs) and generalizes a set of previously existing models and their results. The second model extends our ODE model by taking into account the diffusion of virus. Furthermore, the global stability of both models is investigated by constructing suitable Lyapunov functionals. Finally, we check our theoretical results with numerical simulations.
Collapse
Affiliation(s)
- Mehdi Maziane
- Department of Mathematics and Computer Science, Faculty of Sciences Ben M'sik, Hassan II University, P.O. Box 7955, Sidi Othman, Casablanca, Morocco
| | - El Mehdi Lotfi
- Department of Mathematics and Computer Science, Faculty of Sciences Ben M'sik, Hassan II University, P.O. Box 7955, Sidi Othman, Casablanca, Morocco
| | - Khalid Hattaf
- Department of Mathematics and Computer Science, Faculty of Sciences Ben M'sik, Hassan II University, P.O. Box 7955, Sidi Othman, Casablanca, Morocco.
- Centre Régional des Métiers de l'Education et de la Formation (CRMEF), 20340, Derb Ghalef, Casablanca, Morocco.
| | - Noura Yousfi
- Department of Mathematics and Computer Science, Faculty of Sciences Ben M'sik, Hassan II University, P.O. Box 7955, Sidi Othman, Casablanca, Morocco
| |
Collapse
|
38
|
Nakaoka S, Iwami S, Sato K. Dynamics of HIV infection in lymphoid tissue network. J Math Biol 2015; 72:909-938. [PMID: 26507442 DOI: 10.1007/s00285-015-0940-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 09/28/2015] [Indexed: 12/12/2022]
Abstract
Human immunodeficiency virus (HIV) is a fast replicating ribonucleic acid virus, which can easily mutate in order to escape the effects of drug administration. Hence, understanding the basic mechanisms underlying HIV persistence in the body is essential in the development of new therapies that could eradicate HIV infection. Lymphoid tissues are the primary sites of HIV infection. Despite the recent progress in real-time monitoring technology, HIV infection dynamics in a whole body is unknown. Mathematical modeling and simulations provide speculations on global behavior of HIV infection in the lymphatic system. We propose a new mathematical model that describes the spread of HIV infection throughout the lymphoid tissue network. In order to represent the volume difference between lymphoid tissues, we propose the proportionality of several kinetic parameters to the lymphoid tissues' volume distribution. Under this assumption, we perform extensive numerical computations in order to simulate the spread of HIV infection in the lymphoid tissue network. Numerical computations simulate single drug treatments of an HIV infection. One of the important biological speculations derived from this study is a drug saturation effect generated by lymphoid network connection. This implies that a portion of reservoir lymphoid tissues to which drug is not sufficiently delivered would inhibit HIV eradication despite of extensive drug injection.
Collapse
Affiliation(s)
- Shinji Nakaoka
- Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan.
| | - Shingo Iwami
- Department of Biology, Kyushu University, Fukuoka, 812-8581, Japan
| | - Kei Sato
- Institute for Virus Research, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
39
|
Sherrill-Mix S, Ocwieja KE, Bushman FD. Gene activity in primary T cells infected with HIV89.6: intron retention and induction of genomic repeats. Retrovirology 2015; 12:79. [PMID: 26377088 PMCID: PMC4574318 DOI: 10.1186/s12977-015-0205-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 08/28/2015] [Indexed: 02/07/2023] Open
Abstract
Background HIV infection has been reported to alter cellular gene activity, but published studies have commonly assayed transformed cell lines and lab-adapted HIV strains, yielding inconsistent results. Here we carried out a deep RNA-Seq analysis of primary human T cells infected with the low passage HIV isolate HIV89.6. Results Seventeen percent of cellular genes showed altered activity 48 h after infection. In a meta-analysis including four other studies, our data differed from studies of HIV infection in cell lines but showed more parallels with infections of primary cells. We found a global trend toward retention of introns after infection, suggestive of a novel cellular response to infection. HIV89.6 infection was also associated with activation of several human endogenous retroviruses (HERVs) and retrotransposons, of interest as possible novel antigens that could serve as vaccine targets. The most highly activated group of HERVs was a subset of the ERV-9. Analysis showed that activation was associated with a particular variant of ERV-9 long terminal repeats that contains an indel near the U3-R border. These data also allowed quantification of >70 splice forms of the HIV89.6 RNA and specified the main types of chimeric HIV89.6-host RNAs. Comparison to over 100,000 integration site sequences from the same infected cell populations allowed quantification of authentic versus artifactual chimeric reads, showing that 5′ read-in, splicing out of HIV89.6 from the D4 donor and 3′ read-through were the most common HIV89.6-host cell chimeric RNA forms. Conclusions Analysis of RNA abundance after infection of primary T cells with the low passage HIV89.6 isolate disclosed multiple novel features of HIV-host interactions, notably intron retention and induction of transcription of retrotransposons and endogenous retroviruses. Electronic supplementary material The online version of this article (doi:10.1186/s12977-015-0205-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Scott Sherrill-Mix
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, 425 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA.
| | - Karen E Ocwieja
- Children's Hospital of Philadelphia, 3401 Civic Center Blvd., Philadelphia, PA, 19104, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine at the University of Pennsylvania, 425 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
40
|
Simple mathematical models do not accurately predict early SIV dynamics. Viruses 2015; 7:1189-217. [PMID: 25781919 PMCID: PMC4379566 DOI: 10.3390/v7031189] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/27/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023] Open
Abstract
Upon infection of a new host, human immunodeficiency virus (HIV) replicates in the mucosal tissues and is generally undetectable in circulation for 1–2 weeks post-infection. Several interventions against HIV including vaccines and antiretroviral prophylaxis target virus replication at this earliest stage of infection. Mathematical models have been used to understand how HIV spreads from mucosal tissues systemically and what impact vaccination and/or antiretroviral prophylaxis has on viral eradication. Because predictions of such models have been rarely compared to experimental data, it remains unclear which processes included in these models are critical for predicting early HIV dynamics. Here we modified the “standard” mathematical model of HIV infection to include two populations of infected cells: cells that are actively producing the virus and cells that are transitioning into virus production mode. We evaluated the effects of several poorly known parameters on infection outcomes in this model and compared model predictions to experimental data on infection of non-human primates with variable doses of simian immunodifficiency virus (SIV). First, we found that the mode of virus production by infected cells (budding vs. bursting) has a minimal impact on the early virus dynamics for a wide range of model parameters, as long as the parameters are constrained to provide the observed rate of SIV load increase in the blood of infected animals. Interestingly and in contrast with previous results, we found that the bursting mode of virus production generally results in a higher probability of viral extinction than the budding mode of virus production. Second, this mathematical model was not able to accurately describe the change in experimentally determined probability of host infection with increasing viral doses. Third and finally, the model was also unable to accurately explain the decline in the time to virus detection with increasing viral dose. These results suggest that, in order to appropriately model early HIV/SIV dynamics, additional factors must be considered in the model development. These may include variability in monkey susceptibility to infection, within-host competition between different viruses for target cells at the initial site of virus replication in the mucosa, innate immune response, and possibly the inclusion of several different tissue compartments. The sobering news is that while an increase in model complexity is needed to explain the available experimental data, testing and rejection of more complex models may require more quantitative data than is currently available.
Collapse
|
41
|
Garcia V, Regoes RR. The Effect of Interference on the CD8(+) T Cell Escape Rates in HIV. Front Immunol 2015; 5:661. [PMID: 25628620 PMCID: PMC4292734 DOI: 10.3389/fimmu.2014.00661] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/09/2014] [Indexed: 12/15/2022] Open
Abstract
In early human immunodeficiency virus (HIV) infection, the virus population escapes from multiple CD8+ cell responses. The later an escape mutation emerges, the slower it outgrows its competition, i.e., the escape rate is lower. This pattern could indicate that the strength of the CD8+ cell responses is waning, or that later viral escape mutants carry a larger fitness cost. In this paper, we investigate whether the pattern of decreasing escape rates could also be caused by genetic interference among different escape strains. To this end, we developed a mathematical multi-epitope model of HIV dynamics, which incorporates stochastic effects, recombination, and mutation. We used cumulative linkage disequilibrium measures to quantify the amount of interference. We found that nearly synchronous, similarly strong immune responses in two-locus systems enhance the generation of genetic interference. This effect, combined with a scheme of densely spaced sampling times at the beginning of infection and sparse sampling times later, leads to decreasing successive escape rate estimates, even when there were no selection differences among alleles. These predictions are supported by empirical data from one HIV-infected patient. Thus, interference could explain why later escapes are slower. Considering escape mutations in isolation, neglecting their genetic linkage, conceals the underlying haplotype dynamics and can affect the estimation of the selective pressure exerted by CD8+ cells. In systems in which multiple escape mutations appear, the occurrence of interference dynamics should be assessed by measuring the linkage between different escape mutations.
Collapse
Affiliation(s)
- Victor Garcia
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH Zürich , Zurich , Switzerland
| | - Roland Robert Regoes
- Institute of Integrative Biology, Department of Environmental Systems Science, ETH Zürich , Zurich , Switzerland
| |
Collapse
|
42
|
Althaus CL, Joos B, Perelson AS, Günthard HF. Quantifying the turnover of transcriptional subclasses of HIV-1-infected cells. PLoS Comput Biol 2014; 10:e1003871. [PMID: 25340797 PMCID: PMC4207463 DOI: 10.1371/journal.pcbi.1003871] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 07/14/2014] [Indexed: 11/18/2022] Open
Abstract
HIV-1-infected cells in peripheral blood can be grouped into different transcriptional subclasses. Quantifying the turnover of these cellular subclasses can provide important insights into the viral life cycle and the generation and maintenance of latently infected cells. We used previously published data from five patients chronically infected with HIV-1 that initiated combination antiretroviral therapy (cART). Patient-matched PCR for unspliced and multiply spliced viral RNAs combined with limiting dilution analysis provided measurements of transcriptional profiles at the single cell level. Furthermore, measurement of intracellular transcripts and extracellular virion-enclosed HIV-1 RNA allowed us to distinguish productive from non-productive cells. We developed a mathematical model describing the dynamics of plasma virus and the transcriptional subclasses of HIV-1-infected cells. Fitting the model to the data allowed us to better understand the phenotype of different transcriptional subclasses and their contribution to the overall turnover of HIV-1 before and during cART. The average number of virus-producing cells in peripheral blood is small during chronic infection. We find that a substantial fraction of cells can become defectively infected. Assuming that the infection is homogenous throughout the body, we estimate an average in vivo viral burst size on the order of 104 virions per cell. Our study provides novel quantitative insights into the turnover and development of different subclasses of HIV-1-infected cells, and indicates that cells containing solely unspliced viral RNA are a good marker for viral latency. The model illustrates how the pool of latently infected cells becomes rapidly established during the first months of acute infection and continues to increase slowly during the first years of chronic infection. Having a detailed understanding of this process will be useful for the evaluation of viral eradication strategies that aim to deplete the latent reservoir of HIV-1. Gaining a quantitative understanding of the development and turnover of different HIV-1-infected subpopulations of cells is crucial to improve the outcome of patients on combination antiretroviral therapy (cART). The population of latently infected cells is of particular interest as they represent the major barrier to a cure of HIV-1 infection. We developed a mathematical model that describes the dynamics of different transcriptionally active subclasses of HIV-1-infected cells and the viral load in peripheral blood. The model was fitted to previously published data from five chronically HIV-1-infected patients starting cART. This allowed us to estimate critical parameters of the within-host dynamics of HIV-1, such as the the number of virions produced by a single infected cell. The model further allowed investigation of HIV-1 dynamics during the acute phase. Computer simulations illustrate that latently infected cells become rapidly established during the first months of acute infection and continue to increase slowly during the first years of chronic infection. This illustrates the opportunity for strategies that aim to eradicate the virus during early cART as the pool of HIV-1 infected cells is substantially smaller during acute infection than during chronic infection.
Collapse
Affiliation(s)
- Christian L Althaus
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Beda Joos
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Alan S Perelson
- Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| |
Collapse
|
43
|
Modelling Hepatotoxicity of Antiretroviral Therapy in the Liver during HIV Monoinfection. ACTA ACUST UNITED AC 2014. [DOI: 10.1155/2014/659675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Liver related complications are currently the leading cause of morbidity and mortality among human immunodeficiency virus (HIV) infected individuals. In HIV monoinfected individuals on therapy, liver injury has been associated with the use of antiretroviral agents as most of them exhibit some degree of toxicity. In this study we proposed a mathematical model with the aim of investigating hepatotoxicity of combinational therapy of antiretroviral drugs. Therapy efficacy and toxicity were incorporated in the model as dose-response functions. With the parameter values used in the study, protease inhibitors-based regimens were found to be more toxic than nonnucleoside reverse transcriptase inhibitors-based regimens. In both regimens, the combination of stavudine and zidovudine was the most toxic baseline nucleoside reverse transcriptase inhibitors followed by didanosine with stavudine. However, the least toxic combinations were zidovudine and lamivudine followed by didanosine and lamivudine. The study proposed that, under the same second line regimens, the most toxic first line combination gives the highest viral load and vice versa.
Collapse
|
44
|
Abstract
This review outlines how mathematical models have been helpful, and continue to be so, for obtaining insights into the in vivo dynamics of HIV infection. The review starts with a discussion of a basic mathematical model that has been frequently used to study HIV dynamics. Some crucial results are described, including the estimation of key parameters that characterize the infection, and the generation of influential theories which argued that in vivo virus evolution is a key player in HIV pathogenesis. Subsequently, more recent concepts are reviewed that have relevance for disease progression, including the multiple infection of cells and the direct cell-to-cell transmission of the virus through the formation of virological synapses. These are important mechanisms that can influence the rate at which HIV spreads through its target cell population, which is tightly linked to the rate at which the disease progresses towards AIDS.
Collapse
Affiliation(s)
- Dominik Wodarz
- Department of Ecology and Evolutionary Biology, University of California, 321 Steinhaus Hall, Irvine, CA, 926967, USA,
| |
Collapse
|
45
|
Miron RE, Smith RJ. Resistance to protease inhibitors in a model of HIV-1 infection with impulsive drug effects. Bull Math Biol 2013; 76:59-97. [PMID: 24194434 DOI: 10.1007/s11538-013-9903-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 09/02/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND The emergence of drug resistance is one of the most prevalent reasons for treatment failure in HIV therapy. This has severe implications for the cost of treatment, survival and quality of life. METHODS We use mathematical modelling to describe the interaction between T cells, HIV-1 and protease inhibitors. We use impulsive differential equations to examine the effects of different levels of protease inhibitors in a T cell. We classify three different regimes according to whether the drug efficacy is low, intermediate or high. The model includes two strains: the wild-type strain, which initially dominates in the absence of drugs, and the mutant strain, which is the less efficient competitor, but has more resistance to the drugs. RESULTS Drug regimes may take trajectories through one, two or all three regimes, depending on the dosage and the dosing schedule. Stability analysis shows that resistance does not emerge at low drug levels. At intermediate drug levels, drug resistance is guaranteed to emerge. At high drug levels, either the drug-resistant strain will dominate or, in the absence of longer-lived reservoirs of infected cells, a region exists where viral elimination could theoretically occur. We provide estimates of a range of dosages and dosing schedules where the trajectories lie either solely within a region or cross multiple regions. CONCLUSION Under specific circumstances, if the drug level is physiologically tolerable, elimination of free virus is theoretically possible. This forms the basis for theoretical control using combination therapy and for understanding the effects of partial adherence.
Collapse
Affiliation(s)
- Rachelle E Miron
- Department of Mathematics, The University of Ottawa, 585 King Edward Ave, Ottawa, ON, K1N 6N5, Canada
| | | |
Collapse
|
46
|
Meditz AL, Folkvord JM, Lyle NH, Searls K, Lie YS, Coakley EP, McCarter M, Mawhinney S, Connick E. CCR5 expression is reduced in lymph nodes of HIV type 1-infected women, compared with men, but does not mediate sex-based differences in viral loads. J Infect Dis 2013; 209:922-30. [PMID: 24179109 DOI: 10.1093/infdis/jit575] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Human immunodeficiency virus type 1 (HIV-1)-infected women have lower viral loads than men but similar rates of disease progression. We hypothesized that sex-based differences in CCR5 expression mediate viral load differences. METHODS CCR5 was analyzed by flow cytometry in disaggregated lymph node cells from untreated HIV-1-infected women (n = 28) and men (n = 27). The frequencies of HIV-1 RNA-producing cells in the lymph node were determined by in situ hybridization. Linear and generalized linear regression models were used. RESULTS The percentage of CCR5(+)CD3(+)CD4(+) cells was lower in women (mean, 12%) than men (mean, 16%; P = .034). Neither the percentage of CCR5(+)CD3(+)CD4(+) cells nor the CCR5 density predicted viral load or HIV-1 RNA-producing lymph node cells (P ≥ .24), after adjusting for CD4(+) T-cell count, race, and age. Women had marginally fewer HIV-1 RNA-producing cells (mean, 0.21 cells/mm(2)) than men (mean, 0.44 cells/mm(2); P = .046). After adjusting for the frequency of HIV-1 RNA-producing cells and potential confounders, the viral load in women were 0.46 log10 copies/mL lower than that in men (P = .018). CONCLUSIONS Reduced lymph node CCR5 expression in women did not account for the viral load difference between sexes. CCR5 expression did not predict viral load or frequencies of HIV-1 RNA-producing cells, indicating that physiologic levels of CCR5 do not limit HIV-1 replication in lymph node. Less plasma virus was associated with each HIV-1 RNA-producing cell in women as compared to men, suggesting that women may either produce fewer virions per productively infected cell or more effectively clear extracellular virus.
Collapse
Affiliation(s)
- Amie L Meditz
- Division of Infectious Diseases, Department of Medicine
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
van Deutekom HWM, Wijnker G, de Boer RJ. The rate of immune escape vanishes when multiple immune responses control an HIV infection. THE JOURNAL OF IMMUNOLOGY 2013; 191:3277-86. [PMID: 23940274 DOI: 10.4049/jimmunol.1300962] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During the first months of HIV infection, the virus typically evolves several immune escape mutations. These mutations are found in epitopes in viral proteins and reduce the impact of the CD8⁺ T cells specific for these epitopes. Recent data show that only a subset of the epitopes escapes, that most of these escapes evolve early, and that the rate of immune escape slows down considerably. To investigate why the evolution of immune escape slows down over the time of infection, we have extended a consensus mathematical model to allow several immune responses to control the virus together. In the extended model, most escapes also occur early, and the immune escape rate becomes small later, and typically only a minority of the epitopes escape. We show that escaping one of the many immune responses provides little advantage after viral setpoint has been approached because the total killing rate hardly depends on the breadth of the immune response. If the breadth of the immune response slowly wanes during disease progression, the model predicts an increase in the rate of immune escape at late stages of infection. Overall, the most striking prediction of the model is that HIV evolves a small number of immune escapes, in both relative and absolute terms, when the CTL immune response is broad.
Collapse
|
48
|
Giorgi EE, Korber BT, Perelson AS, Bhattacharya T. Modeling sequence evolution in HIV-1 infection with recombination. J Theor Biol 2013; 329:82-93. [PMID: 23567647 PMCID: PMC3667750 DOI: 10.1016/j.jtbi.2013.03.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 02/12/2013] [Accepted: 03/27/2013] [Indexed: 12/20/2022]
Abstract
Previously we proposed two simplified models of early HIV-1 evolution. Both showed that under a model of neutral evolution and exponential growth, the mean Hamming distance (HD) between genetic sequences grows linearly with time. In this paper we describe a more realistic continuous-time, age-dependent mathematical model of infection and viral replication, and show through simulations that even in this more complex description, the mean Hamming distance grows linearly with time. This remains unchanged when we introduce recombination, though the confidence intervals of the mean HD obtained ignoring recombination are overly conservative.
Collapse
Affiliation(s)
- Elena E Giorgi
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | | | | | |
Collapse
|
49
|
Zhang J, Perelson AS. Contribution of follicular dendritic cells to persistent HIV viremia. J Virol 2013; 87:7893-901. [PMID: 23658450 PMCID: PMC3700220 DOI: 10.1128/jvi.00556-13] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/01/2013] [Indexed: 11/20/2022] Open
Abstract
HIV-1 infections cannot be completely eradicated by drug therapy, as the virus persists in reservoirs. Low-level plasma viremia has been detected in patients treated for over 7 years, but the cellular compartments that support this low-level viremia have not been identified. The decay of HIV-1 during treatment appears to occur in four phases, with the 3rd and 4th phases occurring when the virus is below the limit of detection of conventional assays. Here, we focus on the 3rd phase of decay, which has been estimated to have a half-life of 39 months. We show that follicular dendritic cells (FDC), which have been identified as an HIV reservoir, can be the main source of the low-level viremia detected during the 3rd phase of decay and contribute to viremia at even longer times. Our calculations show that the kinetics of leakage of virus from FDC is consistent with three types of available clinical data.
Collapse
Affiliation(s)
- Jingshan Zhang
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | | |
Collapse
|
50
|
Könnyű B, Sadiq SK, Turányi T, Hírmondó R, Müller B, Kräusslich HG, Coveney PV, Müller V. Gag-Pol processing during HIV-1 virion maturation: a systems biology approach. PLoS Comput Biol 2013; 9:e1003103. [PMID: 23754941 PMCID: PMC3675044 DOI: 10.1371/journal.pcbi.1003103] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Accepted: 05/01/2013] [Indexed: 12/19/2022] Open
Abstract
Proteolytic processing of Gag and Gag-Pol polyproteins by the viral protease (PR) is crucial for the production of infectious HIV-1, and inhibitors of the viral PR are an integral part of current antiretroviral therapy. The process has several layers of complexity (multiple cleavage sites and substrates; multiple enzyme forms; PR auto-processing), which calls for a systems level approach to identify key vulnerabilities and optimal treatment strategies. Here we present the first full reaction kinetics model of proteolytic processing by HIV-1 PR, taking into account all canonical cleavage sites within Gag and Gag-Pol, intermediate products and enzyme forms, enzyme dimerization, the initial auto-cleavage of full-length Gag-Pol as well as self-cleavage of PR. The model allows us to identify the rate limiting step of virion maturation and the parameters with the strongest effect on maturation kinetics. Using the modelling framework, we predict interactions and compensatory potential between individual cleavage rates and drugs, characterize the time course of the process, explain the steep dose response curves associated with PR inhibitors and gain new insights into drug action. While the results of the model are subject to limitations arising from the simplifying assumptions used and from the uncertainties in the parameter estimates, the developed framework provides an extendable open-access platform to incorporate new data and hypotheses in the future.
Collapse
Affiliation(s)
- Balázs Könnyű
- Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - S. Kashif Sadiq
- Computational Biophysics Laboratory (GRIB-IMIM), Universitat Pompeu Fabra, Barcelona, Spain
| | - Tamás Turányi
- Institute of Chemistry, Eötvös Loránd University, Budapest, Hungary
| | - Rita Hírmondó
- Institute of Biology, Eötvös Loránd University, Budapest, Hungary
- Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | - Barbara Müller
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, University of Heidelberg, Heidelberg, Germany
| | - Peter V. Coveney
- Centre for Computational Science, Christopher Ingold Laboratories, University College London, London, United Kingdom
| | - Viktor Müller
- Institute of Biology, Eötvös Loránd University, Budapest, Hungary
- Research Group of Theoretical Biology and Evolutionary Ecology, Eötvös Loránd University and the Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail:
| |
Collapse
|