201
|
Sung JM, Margolis DM. HIV Persistence on Antiretroviral Therapy and Barriers to a Cure. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1075:165-185. [PMID: 30030793 DOI: 10.1007/978-981-13-0484-2_7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
HIV persists within the body despite successful suppression of virus replication with antiretroviral therapy (ART). HIV lurks in latent and active reservoirs, leading to rebound of virus spread if ART is interrupted. The latent HIV reservoir is a natural consequence of the life cycle of HIV, with integration of HIV into the genomes of cells that are or later enter the resting state, resulting in transcriptionally quiescent provirus. Resting CD4 T cells comprise the majority of the latent reservoir, although new evidence points to additional, smaller cellular reservoirs of latent HIV. An alternate, so-called active reservoir of HIV also exists within cells such as those found the B cell follicle of lymph nodes, where expression of HIV RNA can be found, again despite the full suppression of viremia and viral replication. Multiple factors such as the degree of virus exposure, timing of ART, and host factors can influence the size and characteristics of the HIV reservoir. Constructing effective strategies for HIV eradication and measuring their impact will require a sophisticated knowledge of the HIV reservoir.
Collapse
Affiliation(s)
- Julia Marsh Sung
- UNC HIV Cure Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David M Margolis
- UNC HIV Cure Center and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Microbiology & Immunology, and Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
202
|
Mishra A, Nam GH, Gim JA, Lee HE, Jo A, Yoon D, Oh S, Kim S, Kim A, Kim DH, Kim YC, Jeong HD, Cha HJ, Choi YH, Kim HS. Comparative evaluation of MCP gene in worldwide strains of Megalocytivirus (Iridoviridae family) for early diagnostic marker. JOURNAL OF FISH DISEASES 2018; 41:105-116. [PMID: 28914452 DOI: 10.1111/jfd.12685] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/18/2017] [Accepted: 06/19/2017] [Indexed: 06/07/2023]
Abstract
Members of the Iridoviridae family have been considered as aetiological agents of iridovirus diseases, causing fish mortalities and economic losses all over the world. Virus identification based on candidate gene sequencing is faster, more accurate and more reliable than other traditional phenotype methodologies. Iridoviridae viruses are covered by a protein shell (capsid) encoded by the important candidate gene, major capsid protein (MCP). In this study, we investigated the potential of the MCP gene for use in the diagnosis and identification of infections caused Megalocytivirus of the Iridoviridae family. We selected data of 66 Iridoviridae family isolates (53 strains of Megalocytivirus, eight strains of iridoviruses and five strains of Ranavirus) infecting various species of fish distributed all over the world. A total of 53 strains of Megalocytivirus were used for designing the complete primer sets for identifying the most hypervariable region of the MCP gene. Further, our in silico analysis of 102 sequences of related and unrelated viruses reconfirms that primer sets could identify strains more specifically and offers a useful and fast alternative for routine clinical laboratory testing. Our findings suggest that phenotype observation along with diagnosis using universal primer sets can help detect infection or carriers at an early stage.
Collapse
Affiliation(s)
- A Mishra
- Genetic Engineering Institute, Pusan National University, Busan, Korea
| | - G-H Nam
- Genetic Engineering Institute, Pusan National University, Busan, Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| | - J-A Gim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| | - H-E Lee
- Genetic Engineering Institute, Pusan National University, Busan, Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| | - A Jo
- Genetic Engineering Institute, Pusan National University, Busan, Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| | - D Yoon
- Department of Chemistry, College of Natural Sciences, Pusan National University, Busan, Korea
| | - S Oh
- Department of Chemistry, College of Natural Sciences, Pusan National University, Busan, Korea
| | - S Kim
- Department of Chemistry, College of Natural Sciences, Pusan National University, Busan, Korea
| | - A Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, Korea
| | - D-H Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, Korea
| | - Y C Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan, Korea
| | - H D Jeong
- Department of Aquatic Life Medicine, Pukyong National University, Busan, Korea
| | - H-J Cha
- Departments of Parasitology and Genetics, College of Medicine, Kosin University, Busan, Korea
| | - Y H Choi
- Department of Biochemistry, College of Korean Medicine, Dongeui University, Busan, Korea
| | - H-S Kim
- Genetic Engineering Institute, Pusan National University, Busan, Korea
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Korea
| |
Collapse
|
203
|
Lemieux MW, Sonzogni-Desautels K, Ndao M. Lessons Learned from Protective Immune Responses to Optimize Vaccines against Cryptosporidiosis. Pathogens 2017; 7:pathogens7010002. [PMID: 29295550 PMCID: PMC5874728 DOI: 10.3390/pathogens7010002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/16/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023] Open
Abstract
In developing countries, cryptosporidiosis causes moderate-to-severe diarrhea and kills thousands of infants and toddlers annually. Drinking and recreational water contaminated with Cryptosporidium spp. oocysts has led to waterborne outbreaks in developed countries. A competent immune system is necessary to clear this parasitic infection. A better understanding of the immune responses required to prevent or limit infection by this protozoan parasite is the cornerstone of development of an effective vaccine. In this light, lessons learned from previously developed vaccines against Cryptosporidium spp. are at the foundation for development of better next-generation vaccines. In this review, we summarize the immune responses elicited by naturally and experimentally-induced Cryptosporidium spp. infection and by several experimental vaccines in various animal models. Our aim is to increase awareness about the immune responses that underlie protection against cryptosporidiosis and to encourage promotion of these immune responses as a key strategy for vaccine development. Innate and mucosal immunity will be addressed as well as adaptive immunity, with an emphasis on the balance between TH1/TH2 immune responses. Development of more effective vaccines against cryptosporidiosis is needed to prevent Cryptosporidium spp.-related deaths in infants and toddlers in developing countries.
Collapse
Affiliation(s)
- Maxime W Lemieux
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada.
- Department of Medicine, Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| | - Karine Sonzogni-Desautels
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada.
- Faculty of Agricultural and Environmental Sciences, Institute of Parasitology, McGill University, Ste-Anne-de-Bellevue, QC H9X 3V9, Canada.
| | - Momar Ndao
- National Reference Centre for Parasitology, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada.
- Department of Medicine, Division of Infectious Diseases, Faculty of Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
204
|
Chen J, Ren R, Yu F, Wang C, Zhang X, Li W, Tan S, Jiang S, Liu S, Li L. A Degraded Fragment of HIV-1 Gp120 in Rat Hepatocytes Forms Fibrils and Enhances HIV-1 Infection. Biophys J 2017; 113:1425-1439. [PMID: 28978437 DOI: 10.1016/j.bpj.2017.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/03/2017] [Accepted: 08/04/2017] [Indexed: 12/26/2022] Open
Abstract
Identification of the host or viral factors that enhance HIV infection is critical for preventing sexual transmission of HIV. Amyloid fibrils derived from human semen, including semen-derived enhancer of virus infection and semenogelins, enhance HIV-1 infection dramatically in vitro. In this study, we reported that a short-degraded peptide fragment 1 (DPF1) derived from native HIV-1 envelope protein gp120-loaded rat hepatocytes, formed fibrils by self-assembly and thus enhanced HIV-1 infection by promoting the binding of HIV-1 to target cells. Furthermore, DPF1-formed fibrils might be used as a crossing seed to accelerate the formation of semen-derived enhancer of virus infection and semenogelin fibrils. It will be helpful to clarify the viral factors that affect HIV-1 infection. DPF1 as an analog of gp120 containing the critical residues for CD4 binding might be useful for designing of HIV vaccines and developing HIV entry inhibitors.
Collapse
Affiliation(s)
- Jinquan Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China; Jiangsu Protein Drug Engineering Laboratory, Jiangsu Food and Pharmaceutical Science College, Huai'an, China
| | - Ruxia Ren
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Fei Yu
- College of Life Sciences, Agricultural University of Hebei, Baoding, China; Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chunyan Wang
- Center for Clinical Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuanxuan Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Wenjuan Li
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Suiyi Tan
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China; Lindsley F. Kimball Research Institute, New York Blood Center, New York, New York
| | - Shuwen Liu
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Lin Li
- Guangdong Provincial Key Laboratory of New Drug Screening, Guangzhou Key Laboratory of Drug Research for Emerging Virus Prevention and Treatment, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
205
|
McIntosh RC, Ironson G, Antoni M, Lai B, Kumar M, Fletcher MA, Schneiderman N. Psychological Distress Mediates the Effect of Alexithymia on 2-Year Change in HIV Viral Load. Int J Behav Med 2017; 24:294-304. [PMID: 27882489 DOI: 10.1007/s12529-016-9602-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
PURPOSE Individuals with trait alexithymia (AL) display poor cognitive assimilation of thoughts, feelings, and emotions. This may result in the persistence of stress, anxiety, and depressive disorders. The cumulative effect of this psychological distress is also linked clinical markers of human immunodeficiency virus (HIV) disease progression. This study examines the indirect effect of AL on HIV viral load as a function of baseline levels and change in psychological distress. METHODS N = 123 HIV positive adults aged 37.9 ± 9.2 years provided blood samples for HIV-1 viral RNA and CD4 T lymphocytes along with self-reported stress, anxiety, and depression every 6 months for 2 years. A second-order conditional latent growth model was used to represent baseline and 2-year change in cumulative levels of psychological distress and to test the indirect effect of baseline levels of trait AL on change in HIV-1 viral load through this latent measure. RESULTS AL was associated with baseline and latent change in psychological distress. Furthermore, baseline psychological distress predicted 2-year change in HIV-1 viral RNA after controlling for viral load at baseline. Altogether, trait AL had a significant indirect effect on change in viral load (β = 0.16, p = 0.03) as a function of baseline levels of distress. CONCLUSION Identification and communication of thoughts, feelings, and emotions are important for long-term psychological adaptation in HIV. Greater psychological distress, in turn, allows for persistence of peripheral viral replication.
Collapse
Affiliation(s)
- Roger C McIntosh
- Department of Health Psychology, University of Miami, Miami, FL, 33124, USA.
| | - Gail Ironson
- Department of Health Psychology, University of Miami, Miami, FL, 33124, USA
| | - Michael Antoni
- Department of Health Psychology, University of Miami, Miami, FL, 33124, USA
| | - Betty Lai
- School of Public Health, Georgia State University, Atlanta, GA, USA
| | - Mahendra Kumar
- Departments of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mary Ann Fletcher
- Department of Clinical Immunology, Institute of Neuro Immune Medicine, Nova Southeastern University, Davie, FL, USA
| | - Neil Schneiderman
- Department of Health Psychology, University of Miami, Miami, FL, 33124, USA
| |
Collapse
|
206
|
Hadjichrysanthou C, Cauët E, Lawrence E, Vegvari C, de Wolf F, Anderson RM. Understanding the within-host dynamics of influenza A virus: from theory to clinical implications. J R Soc Interface 2017; 13:rsif.2016.0289. [PMID: 27278364 PMCID: PMC4938090 DOI: 10.1098/rsif.2016.0289] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/16/2016] [Indexed: 12/13/2022] Open
Abstract
Mathematical models have provided important insights into acute viral dynamics within individual patients. In this paper, we study the simplest target cell-limited models to investigate the within-host dynamics of influenza A virus infection in humans. Despite the biological simplicity of the models, we show how these can be used to understand the severity of the infection and the key attributes of possible immunotherapy and antiviral drugs for the treatment of infection at different times post infection. Through an analytic approach, we derive and estimate simple summary biological quantities that can provide novel insights into the infection dynamics and the definition of clinical endpoints. We focus on nine quantities, including the area under the viral load curve, peak viral load, the time to peak viral load and the level of cell death due to infection. Using Markov chain Monte Carlo methods, we fitted the models to data collected from 12 untreated volunteers who participated in two clinical studies that tested the antiviral drugs oseltamivir and zanamivir. Based on the results, we also discuss various difficulties in deriving precise estimates of the parameters, even in the very simple models considered, when experimental data are limited to viral load measures and/or there is a limited number of viral load measurements post infection.
Collapse
Affiliation(s)
| | - Emilie Cauët
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Emma Lawrence
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Carolin Vegvari
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Frank de Wolf
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK Janssen Prevention Center, Leiden, The Netherlands
| | - Roy M Anderson
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
| |
Collapse
|
207
|
Reinharz V, Churkin A, Dahari H, Barash D. A Robust and Efficient Numerical Method for RNA-Mediated Viral Dynamics. FRONTIERS IN APPLIED MATHEMATICS AND STATISTICS 2017; 3:20. [PMID: 30854378 PMCID: PMC6404971 DOI: 10.3389/fams.2017.00020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The multiscale model of hepatitis C virus (HCV) dynamics, which includes intracellular viral RNA (vRNA) replication, has been formulated in recent years in order to provide a new conceptual framework for understanding the mechanism of action of a variety of agents for the treatment of HCV. We present a robust and efficient numerical method that belongs to the family of adaptive stepsize methods and is implicit, a Rosenbrock type method that is highly suited to solve this problem. We provide a Graphical User Interface that applies this method and is useful for simulating viral dynamics during treatment with anti-HCV agents that act against HCV on the molecular level.
Collapse
Affiliation(s)
- Vladimir Reinharz
- Department of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Alexander Churkin
- Department of Software Engineering, Sami Shamoon College of Engineering, Beer-Sheva, Israel
| | - Harel Dahari
- Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Loyola University Medical Center, Maywood, IL, United States
| | - Danny Barash
- Department of Computer Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
208
|
Guo J, Yan Y, Zhang J, Ji J, Ge Z, Ge R, Zhang X, Wang H, Chen Z, Luo J. Genetic characterization and antiretroviral resistance mutations among treatment-naive HIV-infected individuals in Jiaxing, China. Oncotarget 2017; 8:18271-18279. [PMID: 28407682 PMCID: PMC5392326 DOI: 10.18632/oncotarget.15382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Accepted: 01/11/2017] [Indexed: 12/23/2022] Open
Abstract
The aim of this study was to characterize HIV-1 genotypes and antiretroviral resistance mutations among treatment-naive HIV-infected individuals in Jiaxing, China. The HIV-1 partial polymerase (pol) genes in 93 of the 99 plasma samples were successfully amplified and analyzed. Phylogenetic analysis revealed the existence of five HIV-1 genotypes, of which the most prevalent genotype was CRF01_AE (38.7%), followed by CRF07_BC (34.4%), CRF08_BC (16.1%), subtype B/B' (5.4%), and CRF55_01B (2.1%). Besides, three types of unique recombination forms (URFs) were also observed, including C/F2/A1, CRF01_AE/B, and CRF08_BC/CRF07_BC. Among 93 amplicons, 46.2% had drug resistance-associated mutations, including 23.7% for protease inhibitors (PIs) mutations, 1.1% for nucleoside reverse transcriptase inhibitors (NRTIs) mutations, and 20.4% for non-nucleoside reverse transcriptase inhibitors (NNRTIs) mutations. Six (6.5%) out of 93 treatment-naive subjects were identified to be resistant to one or more NNRTIs, while resistance to NRTIs or PIs was not observed. Our study showed the genetic diversity of HIV-1 strains circulating in Jiaxing and a relative high proportion of antiretroviral resistance mutations among treatment-naive patients, indicating a serious challenge for HIV prevention and treatment program.
Collapse
Affiliation(s)
- Jinlei Guo
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Yong Yan
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Jiafeng Zhang
- Institute of AIDS Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, PR China
| | - Jimei Ji
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Zhijian Ge
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Rui Ge
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Xiaofei Zhang
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Henghui Wang
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Zhongwen Chen
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| | - Jianyong Luo
- Jiaxing Key Laboratory of Pathogenic Microbiology, Jiaxing Municipal Center for Disease Control and Prevention, Jiaxing 314001, PR China
| |
Collapse
|
209
|
Dorratoltaj N, Nikin-Beers R, Ciupe SM, Eubank SG, Abbas KM. Multi-scale immunoepidemiological modeling of within-host and between-host HIV dynamics: systematic review of mathematical models. PeerJ 2017; 5:e3877. [PMID: 28970973 PMCID: PMC5623312 DOI: 10.7717/peerj.3877] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 09/11/2017] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The objective of this study is to conduct a systematic review of multi-scale HIV immunoepidemiological models to improve our understanding of the synergistic impact between the HIV viral-immune dynamics at the individual level and HIV transmission dynamics at the population level. BACKGROUND While within-host and between-host models of HIV dynamics have been well studied at a single scale, connecting the immunological and epidemiological scales through multi-scale models is an emerging method to infer the synergistic dynamics of HIV at the individual and population levels. METHODS We reviewed nine articles using the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) framework that focused on the synergistic dynamics of HIV immunoepidemiological models at the individual and population levels. RESULTS HIV immunoepidemiological models simulate viral immune dynamics at the within-host scale and the epidemiological transmission dynamics at the between-host scale. They account for longitudinal changes in the immune viral dynamics of HIV+ individuals, and their corresponding impact on the transmission dynamics in the population. They are useful to analyze the dynamics of HIV super-infection, co-infection, drug resistance, evolution, and treatment in HIV+ individuals, and their impact on the epidemic pathways in the population. We illustrate the coupling mechanisms of the within-host and between-host scales, their mathematical implementation, and the clinical and public health problems that are appropriate for analysis using HIV immunoepidemiological models. CONCLUSION HIV immunoepidemiological models connect the within-host immune dynamics at the individual level and the epidemiological transmission dynamics at the population level. While multi-scale models add complexity over a single-scale model, they account for the time varying immune viral response of HIV+ individuals, and the corresponding impact on the time-varying risk of transmission of HIV+ individuals to other susceptibles in the population.
Collapse
Affiliation(s)
| | - Ryan Nikin-Beers
- Department of Mathematics, Virginia Tech, Blacksburg, United States of America
| | - Stanca M. Ciupe
- Department of Mathematics, Virginia Tech, Blacksburg, United States of America
| | - Stephen G. Eubank
- Biocomplexity Institute, Virginia Tech, Blacksburg, United States of America
| | - Kaja M. Abbas
- Department of Population Health Sciences, Virginia Tech, Blacksburg, United States of America
| |
Collapse
|
210
|
Abstract
Advances in technology have made it possible to analyze integration sites in cells from HIV-infected patients. A significant fraction of infected cells in patients on long-term therapy are clonally expanded; in some cases the integrated viral DNA contributes to the clonal expansion of the infected cells. Although the large majority (>95%) of the HIV proviruses in treated patients are defective, expanded clones can carry replication-competent proviruses, and cells from these clones can release infectious virus. As discussed in this Perspective, it is likely that cells that produce virus are strongly selected against in vivo, and cells with replication competent proviruses expand and survive because only a small fraction of the cells produce virus. These findings have implications for strategies that are intended to eliminate the reservoir of infected cells that has made it almost impossible to cure HIV-infected patients.
Collapse
Affiliation(s)
- Stephen H Hughes
- HIV Drug Resistance Program, National Cancer Institute, Frederick, MD 21702, USA.
| | - John M Coffin
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
211
|
Jarne A, Commenges D, Villain L, Prague M, Lévy Y, Thiébaut R. Modeling $\mathrm{CD4}^{+}$ T cells dynamics in HIV-infected patients receiving repeated cycles of exogenous Interleukin 7. Ann Appl Stat 2017. [DOI: 10.1214/17-aoas1047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
212
|
Liao LE, Kowal S, Cardenas DA, Beauchemin CAA. Exploring virus release as a bottleneck for the spread of influenza A virus infection in vitro and the implications for antiviral therapy with neuraminidase inhibitors. PLoS One 2017; 12:e0183621. [PMID: 28837615 PMCID: PMC5570347 DOI: 10.1371/journal.pone.0183621] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/08/2017] [Indexed: 11/24/2022] Open
Abstract
Mathematical models (MMs) have been used to study the kinetics of influenza A virus infections under antiviral therapy, and to characterize the efficacy of antivirals such as neuraminidase inhibitors (NAIs). NAIs prevent viral neuraminidase from cleaving sialic acid receptors that bind virus progeny to the surface of infected cells, thereby inhibiting their release, suppressing infection spread. When used to study treatment with NAIs, MMs represent viral release implicitly as part of viral replication. Consequently, NAIs in such MMs do not act specifically and exclusively on virus release. We compared a MM with an explicit representation of viral release (i.e., distinct from virus production) to a simple MM without explicit release, and investigated whether parameter estimation and the estimation of NAI efficacy were affected by the use of a simple MM. Since the release rate of influenza A virus is not well-known, a broad range of release rates were considered. If the virus release rate is greater than ∼0.1 h−1, the simple MM provides accurate estimates of infection parameters, but underestimates NAI efficacy, which could lead to underdosing and the emergence of NAI resistance. In contrast, when release is slower than ∼0.1 h−1, the simple MM accurately estimates NAI efficacy, but it can significantly overestimate the infectious lifespan (i.e., the time a cell remains infectious and producing free virus), and it will significantly underestimate the total virus yield and thus the likelihood of resistance emergence. We discuss the properties of, and a possible lower bound for, the influenza A virus release rate.
Collapse
Affiliation(s)
- Laura E Liao
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | - Szymon Kowal
- Department of Physics, Ryerson University, Toronto, ON, Canada
| | | | - Catherine A A Beauchemin
- Department of Physics, Ryerson University, Toronto, ON, Canada.,Interdisciplinary Theoretical and Mathematical Sciences (iTHES, iTHEMS) research group at RIKEN, Wako, Japan
| |
Collapse
|
213
|
Santiago DN, Heidbuechel JPW, Kandell WM, Walker R, Djeu J, Engeland CE, Abate-Daga D, Enderling H. Fighting Cancer with Mathematics and Viruses. Viruses 2017; 9:E239. [PMID: 28832539 PMCID: PMC5618005 DOI: 10.3390/v9090239] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 08/18/2017] [Accepted: 08/18/2017] [Indexed: 12/19/2022] Open
Abstract
After decades of research, oncolytic virotherapy has recently advanced to clinical application, and currently a multitude of novel agents and combination treatments are being evaluated for cancer therapy. Oncolytic agents preferentially replicate in tumor cells, inducing tumor cell lysis and complex antitumor effects, such as innate and adaptive immune responses and the destruction of tumor vasculature. With the availability of different vector platforms and the potential of both genetic engineering and combination regimens to enhance particular aspects of safety and efficacy, the identification of optimal treatments for patient subpopulations or even individual patients becomes a top priority. Mathematical modeling can provide support in this arena by making use of experimental and clinical data to generate hypotheses about the mechanisms underlying complex biology and, ultimately, predict optimal treatment protocols. Increasingly complex models can be applied to account for therapeutically relevant parameters such as components of the immune system. In this review, we describe current developments in oncolytic virotherapy and mathematical modeling to discuss the benefit of integrating different modeling approaches into biological and clinical experimentation. Conclusively, we propose a mutual combination of these research fields to increase the value of the preclinical development and the therapeutic efficacy of the resulting treatments.
Collapse
Affiliation(s)
- Daniel N Santiago
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | | | - Wendy M Kandell
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Cancer Biology PhD Program, University of South Florida, Tampa, FL 33612, USA.
| | - Rachel Walker
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Julie Djeu
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
| | - Christine E Engeland
- German Cancer Research Center, Heidelberg University, 69120 Heidelberg, Germany.
- National Center for Tumor Diseases Heidelberg, Department of Translational Oncology, Department of Medical Oncology, 69120 Heidelberg, Germany.
| | - Daniel Abate-Daga
- Department of Immunology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA.
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
214
|
|
215
|
Ren XX, Ma L, Sun WW, Kuang WD, Li TS, Jin X, Wang JH. Dendritic cells maturated by co-culturing with HIV-1 latently infected Jurkat T cells or stimulating with AIDS-associated pathogens secrete TNF-α to reactivate HIV-1 from latency. Virulence 2017; 8:1732-1743. [PMID: 28762863 DOI: 10.1080/21505594.2017.1356535] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Elucidation of mechanisms underlying the establishment, maintenance of and reactivation from HIV-1 latency is essential for the development of therapeutic strategies aimed at eliminating HIV-1 reservoirs. Microbial translocation, as a consequence of HIV-1-induced deterioration of host immune system, is known to result in a systemic immune activation and transient outbursts of HIV-1 viremia in chronic HIV-1 infection. How these microbes cause the robust HIV-1 reactivation remains elusive. Dendritic cells (DCs) have previously been shown to reactivate HIV-1 from latency; however, the precise role of DCs in reactivating HIV-1 from latently infected T-cell remains obscure. In this study, by using HIV-1 latently infected Jurkat T cells, we demonstrated that AIDS-associated pathogens as represented by Mycobacterium bovis (M. bovis) Bacillus Calmette-Guérin (BCG) and bacterial component lipopolysaccharide (LPS) were unable to directly reactivate HIV-1 from Jurkat T cells; instead, they mature DCs to secrete TNF-α to accomplish this goal. Moreover, we found that HIV-1 latently infected Jurkat T cells could also mature DCs and enhance their TNF-α production during co-culture in a CD40-CD40L-signaling-dependent manner. This in turn led to viral reactivation from Jurkat T cells. Our results reveal how DCs help AIDS-associated pathogens to trigger HIV-1 reactivation from latency.
Collapse
Affiliation(s)
- Xiao-Xin Ren
- a Jiangsu Key Laboratory of Infection and Immunity , Institutes of Biology and Medical Sciences, Soochow University , Suzhou , China.,b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China
| | - Li Ma
- a Jiangsu Key Laboratory of Infection and Immunity , Institutes of Biology and Medical Sciences, Soochow University , Suzhou , China.,b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China
| | - Wei-Wei Sun
- b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China.,c University of Chinese Academy of Sciences , Beijing , China
| | - Wen-Dong Kuang
- b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China.,c University of Chinese Academy of Sciences , Beijing , China
| | - Tai-Sheng Li
- d Department of Infectious Diseases , Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College , Beijing , China
| | - Xia Jin
- b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China
| | - Jian-Hua Wang
- b Key Laboratory of Molecular Virology and Immunology , Institut Pasteur of Shanghai, Chinese Academy of Sciences , Shanghai , China.,c University of Chinese Academy of Sciences , Beijing , China
| |
Collapse
|
216
|
Abstract
The bar is high to improve on current combination antiretroviral therapy (ART), now highly effective, safe, and simple. However, antibodies that bind the HIV envelope are able to uniquely target the virus as it seeks to enter new target cells, or as it is expressed from previously infected cells. Furthermore, the use of antibodies against HIV as a therapeutic may offer advantages. Antibodies can have long half-lives, and are being considered as partners for long-acting antiretrovirals for use in therapy or prevention of HIV infection. Early studies in animal models and in clinical trials suggest that such antibodies can have antiviral activity but, as with small-molecule antiretrovirals, the issues of viral escape and resistance will have to be addressed. Most promising, however, are the unique properties of anti-HIV antibodies: the potential ability to opsonize viral particles, to direct antibody-dependent cellular cytotoxicity (ADCC) against actively infected cells, and ultimately the ability to direct the clearance of HIV-infected cells by effector cells of the immune system. These distinctive activities suggest that HIV antibodies and their derivatives may play an important role in the next frontier of HIV therapeutics, the effort to develop treatments that could lead to an HIV cure.
Collapse
Affiliation(s)
- David M Margolis
- UNC HIV Cure Center, Departments of Medicine, Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute and Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
217
|
Vergara TRC, Samer S, Santos-Oliveira JR, Giron LB, Arif MS, Silva-Freitas ML, Cherman LA, Treitsman MS, Chebabo A, Sucupira MCA, Da-Cruz AM, Diaz RS. Thalidomide is Associated With Increased T Cell Activation and Inflammation in Antiretroviral-naive HIV-infected Individuals in a Randomised Clinical Trial of Efficacy and Safety. EBioMedicine 2017; 23:59-67. [PMID: 28822719 PMCID: PMC5605327 DOI: 10.1016/j.ebiom.2017.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/28/2017] [Accepted: 08/07/2017] [Indexed: 01/13/2023] Open
Abstract
Trial Design Open-label, randomised, controlled, pilot proof-of-concept clinical trial. Methods Participants: Antiretroviral naïve adult males with CD4 count ≥ 350 cells/mm3. Interventions: Patients were randomised to receive thalidomide 200 mg QD for 3 weeks (Thalidomide group) or not (Control group) and followed for 48 weeks. Objective: We hypothesized that short-term Thalidomide use would reduce HIV related inflammation and HIV replication among antiretroviral naïve HIV infected individuals. Outcome: Viral loads, CD4/CD8 counts, ultra-sensitive C-reactive protein (US-CRP), cell activation markers, and plasma lipopolysaccharide (LPS) were analyzed. Randomisation: Unrestricted randomisation. Blinding: No blinding was used. Results Numbers randomised: Thirty recruited individuals were randomised to Thalidomide (16 patients) or Control (14 patients) groups. Recruitment: Patients were recruited from April 2011 to January 2013. Outcome: Viral loads remained stable in both groups. During thalidomide treatment, a decrease in CD4/CD8 ratio (p = 0.04), a decrease in CD4 count (p = 0.04), an increase in cell activation calculated by the percentage of CD38 +/HLA-DR+ CD8 cells (p < 0.05) and an increase in US-CRP (p < 0.01) were observed in the Thalidomide group, with all parameters returning to baseline levels after thalidomide interruption. We confirmed that thalidomide increased HIV replication in vitro of 6 of 7 samples from long-term antiretroviral suppressed individuals. Harms: No class 3/4 adverse events occurred. Conclusions Short-term use of thalidomide led to an intense transient increase in T cell activation and inflammation, with a decrease in the CD4+ cell count without changes to the CD8+ cell count. We confirmed that thalidomide acts in vitro as a latency reversal agent and speculate that the in vivo results obtained were due to an increase in HIV replication. Short-term Thalidomide use lead to intense increase in inflammatory markers among HIV+ antiretroviral naïve individuals. Thalidomide use lead to a transitory decrease in CD4+ T cells and CD4/CD8 ratios. In vitro complementary studies revealed that Thalidomide acts as an HIV Latency Reversal Agent.
Thalidomide in general presents a potent anti-inflammatory effect, and is still being used for a number of inflammatory diseases and infection. Therefore, Thalidomide investigated here in a randomised clinical trial aiming to mitigate HIV related inflammation among antiretroviral naïve individuals. Surprisingly, the HIV related inflammation increased during Thalidomide use, and in vitro studies demonstrated that Thalidomide is efficient in interrupting HIV latency, one of the major's obstacles for the cure of HIV chronic infection.
Collapse
Affiliation(s)
- Tânia R C Vergara
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil; Oncohiv, Rio de Janeiro, Brazil
| | - Sadia Samer
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Joanna R Santos-Oliveira
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro (IFRJ), Rio de Janeiro, Brazil
| | - Leila B Giron
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Muhammad Shoaib Arif
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Lia A Cherman
- Secretaria Municipal de Saúde Antônio Ribeiro Neto, Rio de Janeiro, Brazil
| | | | - Alberto Chebabo
- Oncohiv, Rio de Janeiro, Brazil; Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Alda M Da-Cruz
- Laboratório Interdisciplinar de Pesquisas Médicas, Instituto Oswaldo Cruz/FIOCRUZ, Rio de Janeiro, Brazil
| | - Ricardo Sobhie Diaz
- Laboratório de Retrovirologia, Universidade Federal de São Paulo, São Paulo, Brazil.
| | | |
Collapse
|
218
|
Ronsard L, Rai T, Rai D, Ramachandran VG, Banerjea AC. In silico Analyses of Subtype Specific HIV-1 Tat-TAR RNA Interaction Reveals the Structural Determinants for Viral Activity. Front Microbiol 2017; 8:1467. [PMID: 28848502 PMCID: PMC5550727 DOI: 10.3389/fmicb.2017.01467] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/20/2017] [Indexed: 11/24/2022] Open
Abstract
HIV-1 Tat transactivates viral genes through strong interaction with TAR RNA. The stem-loop bulged region of TAR consisting of three nucleotides at the position 23–25 and the loop region consisting of six nucleotides at the position 30–35 are essential for viral transactivation. The arginine motif of Tat (five arginine residues on subtype TatC) is critically important for TAR interaction. Any mutations in this motif could lead to reduce transactivation ability and pathogenesis. Here, we identified structurally important residues (arginine and lysine residues) of Tat in this motif could bind to TAR via hydrogen bond interactions which is critical for transactivation. Natural mutant Ser46Phe in the core motif could likely led to conformational change resulting in more hydrogen bond interactions than the wild type Tat making it highly potent transactivator. Importantly, we report the possible probabilities of number of hydrogen bond interactions in the wild type Tat and the mutants with TAR complexes. This study revealed the differential transactivation of subtype B and C Tat could likely be due to the varying number of hydrogen bonds with TAR. Our data support that the N-terminal and the C-terminal domains of Tat is involved in the TAR interactions through hydrogen bonds which is important for transactivation. This study highlights the evolving pattern of structurally important determinants of Tat in the arginine motif for viral transactivation.
Collapse
Affiliation(s)
- Larance Ronsard
- Laboratory of Virology, National Institute of ImmunologyNew Delhi, India.,Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur HospitalNew Delhi, India
| | - Tripti Rai
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical SciencesNew Delhi, India
| | - Devesh Rai
- Department of Microbiology, All India Institute of Medical SciencesNew Delhi, India
| | - Vishnampettai G Ramachandran
- Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur HospitalNew Delhi, India
| | - Akhil C Banerjea
- Laboratory of Virology, National Institute of ImmunologyNew Delhi, India
| |
Collapse
|
219
|
Murray AJ, Kwon KJ, Farber DL, Siliciano RF. The Latent Reservoir for HIV-1: How Immunologic Memory and Clonal Expansion Contribute to HIV-1 Persistence. THE JOURNAL OF IMMUNOLOGY 2017; 197:407-17. [PMID: 27382129 DOI: 10.4049/jimmunol.1600343] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/12/2016] [Indexed: 12/15/2022]
Abstract
Combination antiretroviral therapy (ART) for HIV-1 infection reduces plasma virus levels to below the limit of detection of clinical assays. However, even with prolonged suppression of viral replication with ART, viremia rebounds rapidly after treatment interruption. Thus, ART is not curative. The principal barrier to cure is a remarkably stable reservoir of latent HIV-1 in resting memory CD4(+) T cells. In this review, we consider explanations for the remarkable stability of the latent reservoir. Stability does not appear to reflect replenishment from new infection events but rather normal physiologic processes that provide for immunologic memory. Of particular importance are proliferative processes that drive clonal expansion of infected cells. Recent evidence suggests that in some infected cells, proliferation is a consequence of proviral integration into host genes associated with cell growth. Efforts to cure HIV-1 infection by targeting the latent reservoir may need to consider the potential of latently infected cells to proliferate.
Collapse
Affiliation(s)
- Alexandra J Murray
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kyungyoon J Kwon
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032; Department of Surgery, Columbia University Medical Center, New York, NY 10032; and
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; Howard Hughes Medical Institute, Baltimore MD 21250
| |
Collapse
|
220
|
Abstract
Human immunodeficiency virus (HIV) remains a significant source of morbidity and mortality worldwide. No effective vaccine is available to prevent HIV transmission, and although antiretroviral therapy can prevent disease progression, it does not cure HIV infection. Substantial effort is therefore currently directed toward basic research on HIV pathogenesis and persistence and developing methods to stop the spread of the HIV epidemic and cure those individuals already infected with HIV. Humanized mice are versatile tools for the study of HIV and its interaction with the human immune system. These models generally consist of immunodeficient mice transplanted with human cells or reconstituted with a near-complete human immune system. Here, we describe the major humanized mouse models currently in use, and some recent advances that have been made in HIV research/therapeutics using these models.
Collapse
Affiliation(s)
- Matthew D Marsden
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, California 90095;
| | - Jerome A Zack
- Department of Medicine, Division of Hematology and Oncology, University of California, Los Angeles, California 90095; .,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California 90095;
| |
Collapse
|
221
|
Bolton DL, McGinnis K, Finak G, Chattopadhyay P, Gottardo R, Roederer M. Combined single-cell quantitation of host and SIV genes and proteins ex vivo reveals host-pathogen interactions in individual cells. PLoS Pathog 2017; 13:e1006445. [PMID: 28654687 PMCID: PMC5507340 DOI: 10.1371/journal.ppat.1006445] [Citation(s) in RCA: 22] [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: 04/11/2017] [Revised: 07/10/2017] [Accepted: 06/04/2017] [Indexed: 12/27/2022] Open
Abstract
CD4 T cells harboring HIV-1/SIV represent a formidable hurdle to eradicating infection, and yet their detailed phenotype remains unknown. Here we integrate two single-cell technologies, flow cytometry and highly multiplexed quantitative RT-PCR, to characterize SIV-infected CD4 T cells directly ex vivo. Within individual cells, we correlate the cellular phenotype, in terms of host protein and RNA expression, with stages of the viral life cycle defined by combinatorial expression of viral RNAs. Spliced RNA+ infected cells display multiple memory and activation phenotypes, indicating virus production by diverse CD4 T cell subsets. In most (but not all) cells, progressive infection accompanies post-transcriptional downregulation of CD4 protein, while surface MHC class I is largely retained. Interferon-stimulated genes were also commonly upregulated. Thus, we demonstrate that combined quantitation of transcriptional and post-transcriptional regulation at the single-cell level informs in vivo mechanisms of viral replication and immune evasion. HIV-1, and its simian counterpart, SIV, infect and kill CD4 T cells, resulting in their massive depletion that ultimately leads to AIDS in the absence of antiretroviral therapy. With effective therapy, these cells are largely preserved, but a subset harbors latent virus that can persist for decades and reemerge upon therapy interruption, preventing HIV-1 cure. To prevent or eliminate productive cellular infection, there is tremendous demand to identify host factors expressed by these cells in vivo, which may serve as unique biomarkers or drug targets. Here we provide the first detailed combined transcriptomic and protein expression profile of SIV-infected cells directly ex vivo using novel single-cell technologies. Our survey of activation markers, interferon-stimulated genes, and viral restriction factors identified multiple host genes differentially expressed by SIV-infected cells and will inform future therapeutic strategies.
Collapse
Affiliation(s)
- Diane L. Bolton
- US Military HIV Research Program, Henry M. Jackson Foundation, Walter Reed Army Institute of Research, Silver Spring, Maryland, United States of America
- * E-mail:
| | - Kathleen McGinnis
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Greg Finak
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Pratip Chattopadhyay
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Raphael Gottardo
- Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Mario Roederer
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, Maryland, United States of America
| |
Collapse
|
222
|
Agrawal P, Nawadkar R, Ojha H, Kumar J, Sahu A. Complement Evasion Strategies of Viruses: An Overview. Front Microbiol 2017; 8:1117. [PMID: 28670306 PMCID: PMC5472698 DOI: 10.3389/fmicb.2017.01117] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
Being a major first line of immune defense, the complement system keeps a constant vigil against viruses. Its ability to recognize large panoply of viruses and virus-infected cells, and trigger the effector pathways, results in neutralization of viruses and killing of the infected cells. This selection pressure exerted by complement on viruses has made them evolve a multitude of countermeasures. These include targeting the recognition molecules for the avoidance of detection, targeting key enzymes and complexes of the complement pathways like C3 convertases and C5b-9 formation - either by encoding complement regulators or by recruiting membrane-bound and soluble host complement regulators, cleaving complement proteins by encoding protease, and inhibiting the synthesis of complement proteins. Additionally, viruses also exploit the complement system for their own benefit. For example, they use complement receptors as well as membrane regulators for cellular entry as well as their spread. Here, we provide an overview on the complement subversion mechanisms adopted by the members of various viral families including Poxviridae, Herpesviridae, Adenoviridae, Flaviviridae, Retroviridae, Picornaviridae, Astroviridae, Togaviridae, Orthomyxoviridae and Paramyxoviridae.
Collapse
Affiliation(s)
- Palak Agrawal
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Renuka Nawadkar
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Hina Ojha
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Jitendra Kumar
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| | - Arvind Sahu
- Complement Biology Laboratory, National Centre for Cell Science, Savitribai Phule Pune UniversityPune, India
| |
Collapse
|
223
|
Dagne GA. Bayesian two-part bent-cable Tobit models with skew distributions: Application to AIDS studies. Stat Methods Med Res 2017; 27:3696-3708. [PMID: 28560896 DOI: 10.1177/0962280217710679] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This paper presents a new development of a bent-cable two-part Tobit model to identify both phasic patterns and mixture of advancing (to AIDS) and non-advancing patients of HIV. In identification of such phasic patterns, estimation of a transition period for the development of drug resistance to antiretroviral (ARV) drug or therapy is carried out using longitudinal data that have a gradual change from a declining phase to an increasing phase. In addition to phasic changes, there are also problems of skewness and left-censoring in the response variable because of a lower limit of detection. A relatively large percentage of data below limit of detection are recorded more than expected under an assumed skew-distribution. To properly accommodate these features, we present an extension of the random effects bent-cable Tobit model that incorporates a mixture of true undetectable observations and those values from a skew-normal distribution for a response with left-censoring, skewness and phasic patterns. The proposed methods are illustrated using real data from an AIDS clinical study.
Collapse
Affiliation(s)
- Getachew A Dagne
- Department of Epidemiology and Biostatistics, University of South Florida, Tampa, FL, USA
| |
Collapse
|
224
|
Fajardo-Ortiz D, Lopez-Cervantes M, Duran L, Dumontier M, Lara M, Ochoa H, Castano VM. The emergence and evolution of the research fronts in HIV/AIDS research. PLoS One 2017; 12:e0178293. [PMID: 28542584 PMCID: PMC5444800 DOI: 10.1371/journal.pone.0178293] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/10/2017] [Indexed: 11/19/2022] Open
Abstract
In this paper, we have identified and analyzed the emergence, structure and dynamics of the paradigmatic research fronts that established the fundamentals of the biomedical knowledge on HIV/AIDS. A search of papers with the identifiers "HIV/AIDS", "Human Immunodeficiency Virus", "HIV-1" and "Acquired Immunodeficiency Syndrome" in the Web of Science (Thomson Reuters), was carried out. A citation network of those papers was constructed. Then, a sub-network of the papers with the highest number of inter-citations (with a minimal in-degree of 28) was selected to perform a combination of network clustering and text mining to identify the paradigmatic research fronts and analyze their dynamics. Thirteen research fronts were identified in this sub-network. The biggest and oldest front is related to the clinical knowledge on the disease in the patient. Nine of the fronts are related to the study of specific molecular structures and mechanisms and two of these fronts are related to the development of drugs. The rest of the fronts are related to the study of the disease at the cellular level. Interestingly, the emergence of these fronts occurred in successive "waves" over the time which suggest a transition in the paradigmatic focus. The emergence and evolution of the biomedical fronts in HIV/AIDS research is explained not just by the partition of the problem in elements and interactions leading to increasingly specialized communities, but also by changes in the technological context of this health problem and the dramatic changes in the epidemiological reality of HIV/AIDS that occurred between 1993 and 1995.
Collapse
Affiliation(s)
- David Fajardo-Ortiz
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Luis Duran
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Michel Dumontier
- Institute of Data Science, Maastricht University, Maastricht, The Netherlands
| | - Miguel Lara
- Instituto de Biología, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Victor M. Castano
- Centro de Física Aplicada y Tecnología Avanzada, Universidad Nacional Autónoma de México, Queretaro, Mexico
| |
Collapse
|
225
|
Vidya Vijayan KK, Karthigeyan KP, Tripathi SP, Hanna LE. Pathophysiology of CD4+ T-Cell Depletion in HIV-1 and HIV-2 Infections. Front Immunol 2017; 8:580. [PMID: 28588579 PMCID: PMC5440548 DOI: 10.3389/fimmu.2017.00580] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/01/2017] [Indexed: 12/20/2022] Open
Abstract
The hall mark of human immunodeficiency virus (HIV) infection is a gradual loss of CD4+ T-cells and imbalance in CD4+ T-cell homeostasis, with progressive impairment of immunity that leads ultimately to death. HIV infection in humans is caused by two related yet distinct viruses: HIV-1 and HIV-2. HIV-2 is typically less virulent than HIV-1 and permits the host to mount a more effective and sustained T-cell immunity. Although both infections manifest the same clinical spectrum, the much lower rate of CD4+ T-cell decline and slower progression of disease in HIV-2 infected individuals have grabbed the attention of several researchers. Here, we review the most recent findings on the differential rate of decline of CD4+ T-cell in HIV-1 and HIV-2 infections and provide plausible reasons for the observed differences between the two groups.
Collapse
Affiliation(s)
- K K Vidya Vijayan
- Division of HIV/AIDS, Department of Clinical Research, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | | | - Srikanth P Tripathi
- Division of HIV/AIDS, Department of Clinical Research, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Luke Elizabeth Hanna
- Division of HIV/AIDS, Department of Clinical Research, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| |
Collapse
|
226
|
Abstract
In this article, we show how to estimate a transition period for the evolvement of drug resistance to antiretroviral (ARV) drug or other related treatments in the framework of developing a Bayesian method for jointly analyzing time-to-event and longitudinal data. For HIV/AIDS longitudinal data, developmental trajectories of viral loads tend to show a gradual change from a declining trend after initiation of treatment to an increasing trend without an abrupt change. Such characteristics of trajectories are also associated with a time-to-event process. To assess these clinically important features, we develop a joint bent-cable Tobit model for the time-to-event and left-censored response variable with skewness and phasic developments. Random effects are used to determine the stochastic dependence between the time-to-event process and response process. The proposed method is illustrated using real data from an AIDS clinical study.
Collapse
Affiliation(s)
- Getachew A Dagne
- a Department of Epidemiology & Biostatistics , College of Public Health, University of South Florida , Tampa , Florida , USA
| |
Collapse
|
227
|
Ciupe SM, Heffernan JM. In-host modeling. Infect Dis Model 2017; 2:188-202. [PMID: 29928736 PMCID: PMC6001971 DOI: 10.1016/j.idm.2017.04.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 01/14/2023] Open
Abstract
Understanding the mechanisms governing host-pathogen kinetics is important and can guide human interventions. In-host mathematical models, together with biological data, have been used in this endeavor. In this review, we present basic models used to describe acute and chronic pathogenic infections. We highlight the power of model predictions, the role of drug therapy, and advantage of considering the dynamics of immune responses. We also present the limitations of these models due in part to the trade-off between the complexity of the model and their predictive power, and the challenges a modeler faces in determining the appropriate formulation for a given problem.
Collapse
Affiliation(s)
- Stanca M. Ciupe
- Department of Mathematics, Virginia Tech, Blacksburg, VA, USA
| | - Jane M. Heffernan
- Centre for Disease Modelling, Department of Mathematics & Statistics, York University, Toronto, ON, Canada
| |
Collapse
|
228
|
Ronsard L, Ganguli N, Singh VK, Mohankumar K, Rai T, Sridharan S, Pajaniradje S, Kumar B, Rai D, Chaudhuri S, Coumar MS, Ramachandran VG, Banerjea AC. Impact of Genetic Variations in HIV-1 Tat on LTR-Mediated Transcription via TAR RNA Interaction. Front Microbiol 2017; 8:706. [PMID: 28484443 PMCID: PMC5399533 DOI: 10.3389/fmicb.2017.00706] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/05/2017] [Indexed: 01/10/2023] Open
Abstract
HIV-1 evades host defense through mutations and recombination events, generating numerous variants in an infected patient. These variants with an undiminished virulence can multiply rapidly in order to progress to AIDS. One of the targets to intervene in HIV-1 replication is the trans-activator of transcription (Tat), a major regulatory protein that transactivates the long terminal repeat promoter through its interaction with trans-activation response (TAR) RNA. In this study, HIV-1 infected patients (n = 120) from North India revealed Ser46Phe (20%) and Ser61Arg (2%) mutations in the Tat variants with a strong interaction toward TAR leading to enhanced transactivation activities. Molecular dynamics simulation data verified that the variants with this mutation had a higher binding affinity for TAR than both the wild-type Tat and other variants that lacked Ser46Phe and Ser61Arg. Other mutations in Tat conferred varying affinities for TAR interaction leading to differential transactivation abilities. This is the first report from North India with a clinical validation of CD4 counts to demonstrate the influence of Tat genetic variations affecting the stability of Tat and its interaction with TAR. This study highlights the co-evolution pattern of Tat and predominant nucleotides for Tat activity, facilitating the identification of genetic determinants for the attenuation of viral gene expression.
Collapse
Affiliation(s)
- Larance Ronsard
- Laboratory of Virology, National Institute of ImmunologyDelhi, India.,Department of Microbiology, University College of Medical Sciences and Guru Teg Bahadur HospitalDelhi, India
| | - Nilanjana Ganguli
- Laboratory of Virology, National Institute of ImmunologyDelhi, India
| | - Vivek K Singh
- Centre for Bioinformatics, School of Life Sciences, Pondicherry UniversityPondicherry, India
| | - Kumaravel Mohankumar
- Department of Biochemistry and Molecular Biology, Pondicherry UniversityPondicherry, India.,Department of Veterinary Physiology and Pharmacology, Texas A&M University, College StationTX, USA
| | - Tripti Rai
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical SciencesDelhi, India
| | - Subhashree Sridharan
- Department of Biochemistry and Molecular Biology, Pondicherry UniversityPondicherry, India.,Department of Symptom Research, The University of Texas MD Anderson Cancer Center, HoustonTX, USA
| | - Sankar Pajaniradje
- Department of Biochemistry and Molecular Biology, Pondicherry UniversityPondicherry, India
| | - Binod Kumar
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, ChicagoIL, USA
| | - Devesh Rai
- Department of Microbiology, All India Institute of Medical SciencesDelhi, India
| | - Suhnrita Chaudhuri
- Department of Neurological Surgery, Northwestern University, ChicagoIL, USA
| | - Mohane S Coumar
- Centre for Bioinformatics, School of Life Sciences, Pondicherry UniversityPondicherry, India
| | | | - Akhil C Banerjea
- Laboratory of Virology, National Institute of ImmunologyDelhi, India
| |
Collapse
|
229
|
HIV persistence in tissue macrophages of humanized myeloid-only mice during antiretroviral therapy. Nat Med 2017; 23:638-643. [PMID: 28414330 PMCID: PMC5419854 DOI: 10.1038/nm.4319] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/13/2017] [Indexed: 12/23/2022]
Abstract
Despite years of fully suppressive antiretroviral therapy (ART), HIV persists in the host and is never eradicated. One major barrier to eradication is that multiple different cell types are infected that may individually contribute to HIV persistence. Tissue macrophages are critical contributors to HIV disease (1–3); however, their specific role in HIV persistence during long-term suppressive ART has not been established (4–6). Using humanized myeloid-only mice (MoM), we demonstrate that HIV infection of tissue macrophages is rapidly suppressed by ART, as determined by a rapid drop in plasma viral load and a dramatic drop in the levels of cell-associated viral RNA and DNA. No virus rebound was observed in the plasma of 67% of the ART treated animals at seven weeks post-ART interruption, and no replication competent virus was rescued from the tissue macrophages obtained from these animals. In contrast, in a subset of animals (~33%), a significantly delayed viral rebound was observed that is consistent with the establishment of persistent infection in tissue macrophages. These observations represent the first direct evidence of HIV persistence in tissue macrophages in vivo.
Collapse
|
230
|
Hosmane NN, Kwon KJ, Bruner KM, Capoferri AA, Beg S, Rosenbloom DIS, Keele BF, Ho YC, Siliciano JD, Siliciano RF. Proliferation of latently infected CD4 + T cells carrying replication-competent HIV-1: Potential role in latent reservoir dynamics. J Exp Med 2017; 214:959-972. [PMID: 28341641 PMCID: PMC5379987 DOI: 10.1084/jem.20170193] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
A latent reservoir for HIV-1 in resting CD4+ T lymphocytes precludes cure. Mechanisms underlying reservoir stability are unclear. Recent studies suggest an unexpected degree of infected cell proliferation in vivo. T cell activation drives proliferation but also reverses latency, resulting in productive infection that generally leads to cell death. In this study, we show that latently infected cells can proliferate in response to mitogens without producing virus, generating progeny cells that can release infectious virus. Thus, assays relying on one round of activation underestimate reservoir size. Sequencing of independent clonal isolates of replication-competent virus revealed that 57% had env sequences identical to other isolates from the same patient. Identity was confirmed by full-genome sequencing and was not attributable to limited viral diversity. Phylogenetic and statistical analysis suggested that identical sequences arose from in vivo proliferation of infected cells, rather than infection of multiple cells by a dominant viral species. The possibility that much of the reservoir arises by cell proliferation presents challenges to cure.
Collapse
Affiliation(s)
- Nina N Hosmane
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Kyungyoon J Kwon
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Katherine M Bruner
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Adam A Capoferri
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Subul Beg
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Daniel I S Rosenbloom
- Department of Biomedical Informatics, Columbia University Medical Center, New York, NY 10032
| | - Brandon F Keele
- AIDS and Cancer Virus Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, Frederick, MD 21702
| | - Ya-Chi Ho
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Janet D Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Robert F Siliciano
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205
- Howard Hughes Medical Institute, Baltimore, MD 21205
| |
Collapse
|
231
|
Kariuki SM, Selhorst P, Ariën KK, Dorfman JR. The HIV-1 transmission bottleneck. Retrovirology 2017; 14:22. [PMID: 28335782 PMCID: PMC5364581 DOI: 10.1186/s12977-017-0343-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 03/05/2017] [Indexed: 02/07/2023] Open
Abstract
It is well established that most new systemic infections of HIV-1 can be traced back to one or a limited number of founder viruses. Usually, these founders are more closely related to minor HIV-1 populations in the blood of the presumed donor than to more abundant lineages. This has led to the widely accepted idea that transmission selects for viral characteristics that facilitate crossing the mucosal barrier of the recipient’s genital tract, although the specific selective forces or advantages are not completely defined. However, there are other steps along the way to becoming a founder virus at which selection may occur. These steps include the transition from the donor’s general circulation to the genital tract compartment, survival within the transmission fluid, and establishment of a nascent stable local infection in the recipient’s genital tract. Finally, there is the possibility that important narrowing events may also occur during establishment of systemic infection. This is suggested by the surprising observation that the number of founder viruses detected after transmission in intravenous drug users is also limited. Although some of these steps may be heavily selective, others may result mostly in a stochastic narrowing of the available founder pool. Collectively, they shape the initial infection in each recipient.
Collapse
Affiliation(s)
- Samuel Mundia Kariuki
- Division of Immunology, Department of Pathology, Falmouth 3.25, University of Cape Town, Anzio Rd, Observatory, Cape Town, 7925, South Africa.,International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa.,Department of Biological Sciences, University of Eldoret, Eldoret, Kenya
| | - Philippe Selhorst
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jeffrey R Dorfman
- Division of Immunology, Department of Pathology, Falmouth 3.25, University of Cape Town, Anzio Rd, Observatory, Cape Town, 7925, South Africa.
| |
Collapse
|
232
|
Abstract
During the evolution of human immunodeficiency virus (HIV), transmissions between humans and primates resulted in multiple HIV lineages in humans. This evolution has been rapid, giving rise to a complex classification and allowing for worldwide spread and intermixing of subtypes, which has consequently led to dozens of circulating recombinant forms. In the Republic of Korea, 12,522 cases of HIV infection have been reported between 1985, when AIDS was first identified, and 2015. This review focuses on the evolution of HIV infection worldwide and the molecular epidemiologic characteristics of HIV in Korea.
Collapse
Affiliation(s)
- Bum Sik Chin
- Center for Infectious Diseases, National Medical Center, Seoul, Korea.
| |
Collapse
|
233
|
Huang Y, Yan C. Piecewise Mixture Modeling for Longitudinal Virologic Data With Heterogeneity, Nonnormality, and Missingness. Stat Biopharm Res 2017. [DOI: 10.1080/19466315.2016.1215347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Yangxin Huang
- Department of Epidemiology and Biostatistics, College of Public Health, University of South Florida, Tampa, FL
| | - Chunning Yan
- School of Management, Shanghai University, Shanghai, China
| |
Collapse
|
234
|
Zhang J, Crumpacker CS. Toward a Cure: Does Host Immunity Play a Role? mSphere 2017; 2:e00138-17. [PMID: 28497113 PMCID: PMC5422033 DOI: 10.1128/msphere.00138-17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/01/2017] [Indexed: 01/18/2023] Open
Abstract
Three decades of research on human immunodeficiency virus (HIV) and AIDS reveal that the human body has developed through evolution a genome immune system embodying epigenetic regulation against pathogenic nucleic acid invasion. In HIV infection, this epigenetic regulation plays a cardinal role in HIV RNA production that silences HIV transcription at a molecular (RNA) level, controls viral load at a cellular (biological) level, and governs the viremic stage of AIDS at the clinical (patient) level. Even though the human genome is largely similar among humans and HIV is a single viral species, human hosts show significant differences in viral RNA levels, ranging from cell to organ to individual and expressed as elite controllers, posttreatment controllers, and patients with AIDS. These are signature biomarkers of typical epigenetic regulation whose importance has been shunted aside by interpreting all of AIDS pathogenesis by the known properties of innate and adaptive immunity. We propose that harnessing the host genome immune system, defined as epigenetic immunity, against HIV infection will lead toward a cure.
Collapse
Affiliation(s)
- Jielin Zhang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Clyde S Crumpacker
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
235
|
Abstract
This is an exciting time for immunology because the future promises to be replete with exciting new discoveries that can be translated to improve health and treat disease in novel ways. Immunologists are attempting to answer increasingly complex questions concerning phenomena that range from the genetic, molecular, and cellular scales to that of organs, whole animals or humans, and populations of humans and pathogens. An important goal is to understand how the many different components involved interact with each other within and across these scales for immune responses to emerge, and how aberrant regulation of these processes causes disease. To aid this quest, large amounts of data can be collected using high-throughput instrumentation. The nonlinear, cooperative, and stochastic character of the interactions between components of the immune system as well as the overwhelming amounts of data can make it difficult to intuit patterns in the data or a mechanistic understanding of the phenomena being studied. Computational models are increasingly important in confronting and overcoming these challenges. I first describe an iterative paradigm of research that integrates laboratory experiments, clinical data, computational inference, and mechanistic computational models. I then illustrate this paradigm with a few examples from the recent literature that make vivid the power of bringing together diverse types of computational models with experimental and clinical studies to fruitfully interrogate the immune system.
Collapse
Affiliation(s)
- Arup K Chakraborty
- Institute for Medical Engineering and Science, Departments of Chemical Engineering, Physics, Chemistry, and Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139; .,Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139
| |
Collapse
|
236
|
Histidine 375 Modulates CD4 Binding in HIV-1 CRF01_AE Envelope Glycoproteins. J Virol 2017; 91:JVI.02151-16. [PMID: 27928014 DOI: 10.1128/jvi.02151-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 11/29/2016] [Indexed: 01/11/2023] Open
Abstract
The envelope glycoproteins (Envs) from human immunodeficiency virus type 1 (HIV-1) mediate viral entry. The binding of the HIV-1 gp120 glycoprotein to CD4 triggers conformational changes in gp120 that allow high-affinity binding to its coreceptors. In contrast to all other Envs from the same phylogenetic group, M, which possess a serine (S) at position 375, those from CRF01_AE strains possess a histidine (H) at this location. This residue is part of the Phe43 cavity, where residue 43 of CD4 (a phenylalanine) engages with gp120. Here we evaluated the functional consequences of replacing this residue in two CRF01_AE Envs (CM244 and 92TH023) by a serine. We observed that reversion of amino acid 375 to a serine (H375S) resulted in a loss of functionality of both CRF01_AE Envs as measured by a dramatic loss in infectivity and ability to mediate cell-to-cell fusion. While no effects on processing or trimer stability of these variants were observed, decreased functionality could be linked to a major defect in CD4 binding induced by the replacement of H375 by a serine. Importantly, mutations of residues 61 (layer 1), 105 and 108 (layer 2), and 474 to 476 (layer 3) of the CRF01_AE gp120 inner domain layers to the consensus residues present in group M restored CD4 binding and wild-type levels of infectivity and cell-to-cell fusion. These results suggest a functional coevolution between the Phe43 cavity and the gp120 inner domain layers. Altogether, our observations describe the functional importance of amino acid 375H in CRF01_AE envelopes. IMPORTANCE A highly conserved serine located at position 375 in group M is replaced by a histidine in CRF01_AE Envs. Here we show that H375 is required for efficient CRF01_AE Env binding to CD4. Moreover, this work suggests that specific residues of the gp120 inner domain layers have coevolved with H375 in order to maintain its ability to mediate viral entry.
Collapse
|
237
|
Rodriguez-Brenes IA, Hofacre A, Fan H, Wodarz D. Complex Dynamics of Virus Spread from Low Infection Multiplicities: Implications for the Spread of Oncolytic Viruses. PLoS Comput Biol 2017; 13:e1005241. [PMID: 28107341 PMCID: PMC5249046 DOI: 10.1371/journal.pcbi.1005241] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/08/2016] [Indexed: 12/22/2022] Open
Abstract
While virus growth dynamics have been well-characterized in several infections, data are typically collected once the virus population becomes easily detectable. Earlier dynamics, however, remain less understood. We recently reported unusual early dynamics in an experimental system using adenovirus infection of human embryonic kidney (293) cells. Under identical experimental conditions, inoculation at low infection multiplicities resulted in either robust spread, or in limited spread that eventually stalled, with both outcomes occurring with approximately equal frequencies. The reasons underlying these observations have not been understood. Here, we present further experimental data showing that inhibition of interferon-induced antiviral states in cells results in a significant increase in the percentage of robust infections that are observed, implicating a race between virus replication and the spread of the anti-viral state as a central mechanism. Analysis of a variety of computational models, however, reveals that this alone cannot explain the simultaneous occurrence of both viral growth outcomes under identical conditions, and that additional biological mechanisms have to be invoked to explain the data. One such mechanism is the ability of the virus to overcome the antiviral state through multiple infection of cells. If this is included in the model, two outcomes of viral spread are found to be simultaneously stable, depending on initial conditions. In stochastic versions of such models, the system can go by chance to either state from identical initial conditions, with the relative frequency of the outcomes depending on the strength of the interferon-based anti-viral response, consistent with the experiments. This demonstrates considerable complexity during the early phase of the infection that can influence the ability of a virus to become successfully established. Implications for the initial dynamics of oncolytic virus spread through tumors are discussed. We investigate in vitro adenovirus spread starting from the lowest infection multiplicities. This phase of virus dynamics remains poorly understood and is likely critical for ensuring that engineered oncolytic viruses successfully spread and destroy tumors. We find unexpectedly complex dynamics, which are analyzed with a combination of experiments and mathematical models. The experiments indicate that the induction of an interferon-based anti-viral state is a crucial underlying mechanism. The mathematical models demonstrate that this mechanism alone cannot explain the experiments, and that additional mechanisms must be invoked to account for the data. The models suggest that the ability of the virus to overcome the anti-viral state through multiple infection of cells might be one such mechanism.
Collapse
Affiliation(s)
- Ignacio A. Rodriguez-Brenes
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, California, United States of America
| | - Andrew Hofacre
- Department of Molecular Biology and Biochemistry, Cancer Research Institute, University of California, Irvine, Irvine, California, United States of America
| | - Hung Fan
- Department of Molecular Biology and Biochemistry, Cancer Research Institute, University of California, Irvine, Irvine, California, United States of America
| | - Dominik Wodarz
- Department of Mathematics, University of California, Irvine, Irvine, California, United States of America
- Department of Ecology and Evolutionary Biology, University of California, Irvine, Irvine, California, United States of America
- * E-mail:
| |
Collapse
|
238
|
Xiao Y, Sun X, Tang S, Zhou Y, Peng Z, Wu J, Wang N. Personalized life expectancy and treatment benefit index of antiretroviral therapy. Theor Biol Med Model 2017; 14:1. [PMID: 28100241 PMCID: PMC5242026 DOI: 10.1186/s12976-016-0047-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/29/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The progression of Human Immunodeficiency Virus (HIV) within host includes typical stages and the Antiretroviral Therapy (ART) is shown to be effective in slowing down this progression. There are great challenges in describing the entire HIV disease progression and evaluating comprehensive effects of ART on life expectancy for HIV infected individuals on ART. METHODS We develop a novel summative treatment benefit index (TBI), based on an HIV viral dynamics model and linking the infection and viral production rates to the Weibull function. This index summarizes the integrated effect of ART on the life expectancy (LE) of a patient, and more importantly, can be reconstructed from the individual clinic data. RESULTS The proposed model, faithfully mimicking the entire HIV disease progression, enables us to predict life expectancy and trace back the timing of infection. We fit the model to the longitudinal data in a cohort study in China to reconstruct the treatment benefit index, and we describe the dependence of individual life expectancy on key ART treatment specifics including the timing of ART initiation, timing of emergence of drug resistant virus variants and ART adherence. CONCLUSIONS We show that combining model predictions with monitored CD4 counts and viral loads can provide critical information about the disease progression, to assist the design of ART regimen for maximizing the treatment benefits.
Collapse
Affiliation(s)
- Yanni Xiao
- Department of Applied Mathematics, Xi'an Jiaotong University, Xianning West Road, Xi'an, 710049, China
| | - Xiaodan Sun
- Department of Applied Mathematics, Xi'an Jiaotong University, Xianning West Road, Xi'an, 710049, China.
| | - Sanyi Tang
- College of Mathematics and Information Science, Shaanxi Normal University, West Chang'an Avenue, Xi'an, 710119, China
| | - Yicang Zhou
- Department of Applied Mathematics, Xi'an Jiaotong University, Xianning West Road, Xi'an, 710049, China
| | - Zhihang Peng
- School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Jianhong Wu
- Laboratory for Industrial and Applied Mathematics, Centre for Disease Modelling, York Institute for Health Research, York University, Toronto, M3J 1P3, Canada
| | - Ning Wang
- National Center for AIDS/STD Prevention and Control, Chinese Center for Disease Control and Prevention, 155 Changbai Road, Beijing, 102206, China
| |
Collapse
|
239
|
Abstract
Viral latency is a major barrier to curing HIV infection with antiretroviral therapy, and consequently, for eliminating the disease globally. The establishment, maintenance, and potential clearance of latent infection are complex dynamic processes and can be best understood and described with the help of mathematical models. Here we review the use of viral dynamics models for HIV, with a focus on applications to the latent reservoir. Such models have been used to explain the multiphasic decay of viral load during antiretroviral therapy, the early seeding of the latent reservoir during acute infection and the limited inflow during treatment, the dynamics of viral blips, and the phenomenon of posttreatment control. In addition, mathematical models have been used to predict the efficacy of potential HIV cure strategies, such as latency-reversing agents, early treatment initiation, or gene therapies, and to provide guidance for designing trials of these novel interventions.
Collapse
|
240
|
Siliciano JD, Siliciano RF. Assays to Measure Latency, Reservoirs, and Reactivation. Curr Top Microbiol Immunol 2017; 417:23-41. [PMID: 29071475 DOI: 10.1007/82_2017_75] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
HIV-1 persists even in patients who are successfully treated with combination antiretroviral therapy. The major barrier to cure is a small pool of latently infected resting CD4+ T cells carrying an integrated copy of the viral genome that is not expressed while the cells remain in a resting state. Targeting this latent reservoir is a major focus of HIV-1 cure research, and the development of a rapid and scalable assay for the reservoir is a rate-limiting step in the search for a cure. The most commonly used assays are standard PCR assays targeting conserved regions of the HIV-1 genome. However, because the vast majority of HIV-1 proviruses are defective, such assays may not accurately capture changes in the minor subset of proviruses that are replication-competent and that pose a barrier to cure. On the other hand, the viral outgrowth assay that was used to initially define the latent reservoir may underestimate reservoir size because not all replication-competent proviruses are induced by a single round of T cell activation in this assay. Therefore, this assay is best regarded as a definitive minimal estimate of reservoir size. The best approach may be to measure all of the proviruses with the potential to cause viral rebound. A variety of novel assays have recently been described. Ultimately, the assay that best predicts time to viral rebound will be the most useful to the cure effort.
Collapse
Affiliation(s)
- Janet D Siliciano
- Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert F Siliciano
- Johns Hopkins University School of Medicine, Howard Hughes Medical Institute, Baltimore, MD, 21205, USA.
| |
Collapse
|
241
|
Scagnolari C, Turriziani O, Monteleone K, Pierangeli A, Antonelli G. Consolidation of molecular testing in clinical virology. Expert Rev Anti Infect Ther 2016; 15:387-400. [PMID: 28002969 DOI: 10.1080/14787210.2017.1271711] [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: 02/08/2023]
Abstract
INTRODUCTION The development of quantitative methods for the detection of viral nucleic acids have significantly improved our ability to manage disease progression and to assess the efficacy of antiviral treatment. Moreover, major advances in molecular technologies during the last decade have allowed the identification of new host genetic markers associated with antiviral drug response but have also strongly revolutionized the way we see and perform virus diagnostics in the coming years. Areas covered: In this review, we describe the history and development of virology diagnostic methods, dedicating particular emphasis on the gradual evolution and recent advances toward the introduction of multiparametric platforms for the syndromic diagnosis. In parallel, we outline the consolidation of viral genome quantification practice in different clinical settings. Expert commentary: More rapid, accurate and affordable molecular technology can be predictable with particular emphasis on emerging techniques (next generation sequencing, digital PCR, point of care testing and syndromic diagnosis) to simplify viral diagnosis in the next future.
Collapse
Affiliation(s)
- Carolina Scagnolari
- a Laboratory of Virology, Department of Molecular Medicine, and Istituto Pasteur Italia-Cenci Bolognetti Foundation , 'Sapienza' University of Rome , Rome , Italy
| | - Ombretta Turriziani
- a Laboratory of Virology, Department of Molecular Medicine, and Istituto Pasteur Italia-Cenci Bolognetti Foundation , 'Sapienza' University of Rome , Rome , Italy
| | - Katia Monteleone
- a Laboratory of Virology, Department of Molecular Medicine, and Istituto Pasteur Italia-Cenci Bolognetti Foundation , 'Sapienza' University of Rome , Rome , Italy
| | - Alessandra Pierangeli
- a Laboratory of Virology, Department of Molecular Medicine, and Istituto Pasteur Italia-Cenci Bolognetti Foundation , 'Sapienza' University of Rome , Rome , Italy
| | - Guido Antonelli
- a Laboratory of Virology, Department of Molecular Medicine, and Istituto Pasteur Italia-Cenci Bolognetti Foundation , 'Sapienza' University of Rome , Rome , Italy
| |
Collapse
|
242
|
Bayesian analysis of piecewise growth mixture models with skew-t distributions: Application to AIDS studies. J Biopharm Stat 2016; 27:691-704. [PMID: 28010168 DOI: 10.1080/10543406.2016.1269782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A major problem in HIV/AIDS studies is the development of drug resistance to antiretroviral (ARV) drug or therapy. Estimating the time at which such drug resistance would develop is usually sought. The goal of this article is to perform this estimation by developing growth mixture models with change-points and skew-t distributions based on longitudinal data. For such data, following ARV treatment, the profile of each subject's viral load tends to follow a 'broken stick' like growth trajectory, indicating multiple phases of decline and increase in viral loads. These multiple phases with multiple change-points are captured by subject-specific random parameters of growth curve models. To account for heterogeneity of drug resistance among subjects, the change-points are also allowed to differ by subgroups (subpopulations) of patients classified into latent classes on the basis of trajectories of observed viral loads. The proposed methods are illustrated using real data from an AIDS clinical study.
Collapse
|
243
|
Abstract
Emergent responses of the immune system result from the integration of molecular and cellular networks over time and across multiple organs. High-content and high-throughput analysis technologies, concomitantly with data-driven and mechanistic modeling, hold promise for the systematic interrogation of these complex pathways. However, connecting genetic variation and molecular mechanisms to individual phenotypes and health outcomes has proven elusive. Gaps remain in data, and disagreements persist about the value of mechanistic modeling for immunology. Here, we present the perspectives that emerged from the National Institute of Allergy and Infectious Disease (NIAID) workshop 'Complex Systems Science, Modeling and Immunity' and subsequent discussions regarding the potential synergy of high-throughput data acquisition, data-driven modeling, and mechanistic modeling to define new mechanisms of immunological disease and to accelerate the translation of these insights into therapies.
Collapse
|
244
|
Schiffer JT, Swan DA, Magaret A, Corey L, Wald A, Ossig J, Ruebsamen-Schaeff H, Stoelben S, Timmler B, Zimmermann H, Melhem MR, Van Wart SA, Rubino CM, Birkmann A. Mathematical modeling of herpes simplex virus-2 suppression with pritelivir predicts trial outcomes. Sci Transl Med 2016; 8:324ra15. [PMID: 26843190 DOI: 10.1126/scitranslmed.aad6654] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pharmacokinetic and pharmacodynamic models estimate the potency of antiviral agents but do not capture viral and immunologic factors that drive the natural dynamics of infection. We designed a mathematical model that synthesizes pharmacokinetics, pharmacodynamics, and viral pathogenesis concepts to simulate the activity of pritelivir, a DNA helicase-primase inhibitor that targets herpes simplex virus. Our simulations recapitulate detailed viral kinetic shedding features in five dosage arms of a phase 2 clinical trial. We identify that in vitro estimates of median effective concentration (EC50) are lower than in vivo values for the drug. Nevertheless, pritelivir potently decreases shedding at appropriate doses based on its mode of action and long half-life. Although pritelivir directly inhibits replication in epithelial cells, our model indicates that pritelivir also indirectly limits downstream viral spread from neurons to genital keratinocytes, within genital ulcers, and from ulcer to new mucosal sites of infection. We validate our model based on its ability to predict outcomes in a subsequent trial with a higher dose. The model can therefore be used to optimize dose selection in clinical practice.
Collapse
Affiliation(s)
- Joshua T Schiffer
- Department of Medicine, University of Washington, Seattle, WA 98105, USA. Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - David A Swan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Amalia Magaret
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. Department of Laboratory Medicine, University of Washington, Seattle, WA 98105, USA
| | - Lawrence Corey
- Department of Medicine, University of Washington, Seattle, WA 98105, USA. Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. Department of Laboratory Medicine, University of Washington, Seattle, WA 98105, USA
| | - Anna Wald
- Department of Medicine, University of Washington, Seattle, WA 98105, USA. Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. Department of Laboratory Medicine, University of Washington, Seattle, WA 98105, USA. Department of Epidemiology, University of Washington, Seattle, WA 98105, USA
| | | | | | | | | | | | - Murad R Melhem
- Institute for Clinical Pharmacodynamics, Latham, NY 12307, USA
| | - Scott A Van Wart
- Institute for Clinical Pharmacodynamics, Latham, NY 12307, USA. School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Christopher M Rubino
- Institute for Clinical Pharmacodynamics, Latham, NY 12307, USA. School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
245
|
Achieving HIV-1 Control through RNA-Directed Gene Regulation. Genes (Basel) 2016; 7:genes7120119. [PMID: 27941595 PMCID: PMC5192495 DOI: 10.3390/genes7120119] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 12/13/2022] Open
Abstract
HIV-1 infection has been transformed by combined anti-retroviral therapy (ART), changing a universally fatal infection into a controllable infection. However, major obstacles for an HIV-1 cure exist. The HIV latent reservoir, which exists in resting CD4+ T cells, is not impacted by ART, and can reactivate when ART is interrupted or ceased. Additionally, multi-drug resistance can arise. One alternate approach to conventional HIV-1 drug treatment that is being explored involves gene therapies utilizing RNA-directed gene regulation. Commonly known as RNA interference (RNAi), short interfering RNA (siRNA) induce gene silencing in conserved biological pathways, which require a high degree of sequence specificity. This review will provide an overview of the silencing pathways, the current RNAi technologies being developed for HIV-1 gene therapy, current clinical trials, and the challenges faced in progressing these treatments into clinical trials.
Collapse
|
246
|
Fuchs SP, Desrosiers RC. Promise and problems associated with the use of recombinant AAV for the delivery of anti-HIV antibodies. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16068. [PMID: 28197421 PMCID: PMC5289440 DOI: 10.1038/mtm.2016.68] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/11/2016] [Indexed: 02/07/2023]
Abstract
Attempts to elicit antibodies with potent neutralizing activity against a broad range of human immunodeficiency virus (HIV) isolates have so far proven unsuccessful. Long-term delivery of monoclonal antibodies (mAbs) with such activity is a creative alternative that circumvents the need for an immune response and has the potential for creating a long-lasting sterilizing barrier against HIV. This approach is made possible by an incredible array of potent broadly neutralizing antibodies (bnAbs) that have been identified over the last several years. Recombinant adeno-associated virus (rAAV) vectors are ideally suited for long-term delivery for a variety of reasons. The only products made from rAAV are derived from the transgenes that are put into it; as long as those products are not viewed as foreign, expression from muscle tissue may continue for decades. Thus, use of rAAV to achieve long-term delivery of anti-HIV mAbs with potent neutralizing activity against a broad range of HIV-1 isolates is emerging as a promising concept for the prevention or treatment of HIV-1 infection in humans. Experiments in mice and monkeys that have demonstrated protective efficacy against AIDS virus infection have raised hopes for the promise of this approach. However, all published experiments in monkeys have encountered unwanted immune responses to the AAV-delivered antibody, and these immune responses appear to limit the levels of delivered antibody that can be achieved. In this review, we highlight the promise of rAAV-mediated antibody delivery for the prevention or treatment of HIV infection in humans, but we also discuss the obstacles that will need to be understood and solved in order for the promise of this approach to be realized.
Collapse
Affiliation(s)
- Sebastian P Fuchs
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, Florida, USA; Institut für Klinische und Molekulare Virologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ronald C Desrosiers
- Department of Pathology, Miller School of Medicine, University of Miami , Miami, Florida, USA
| |
Collapse
|
247
|
2,4,5-Trisubstituted thiazole derivatives as HIV-1 NNRTIs effective on both wild-type and mutant HIV-1 reverse transcriptase: Optimization of the substitution of positions 4 and 5. Eur J Med Chem 2016; 123:309-316. [DOI: 10.1016/j.ejmech.2016.07.047] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 11/18/2022]
|
248
|
Shet A, Nagaraja P, Dixit NM. Viral Decay Dynamics and Mathematical Modeling of Treatment Response: Evidence of Lower in vivo Fitness of HIV-1 Subtype C. J Acquir Immune Defic Syndr 2016; 73:245-251. [PMID: 27273158 PMCID: PMC5172519 DOI: 10.1097/qai.0000000000001101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 05/16/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Despite the high prevalence of HIV-1 subtype C (HIV-1C) worldwide, information on HIV-1C viral dynamics and response to antiretroviral therapy (ART) is limited. We sought to measure viral load decay dynamics during treatment and estimate the within-host basic reproductive ratio, R0, and the critical efficacy, εc, for successful treatment of HIV-1C infection. METHODS Individuals initiated on first-line ART in India and monitored for 6 months of treatment were considered. Viral load, CD4 count, and adherence data were collected at baseline, 4, 12, 16 and 24 weeks after ART initiation. Drug resistance genotyping was performed at baseline. R0 and εc were estimated using a mathematical model. RESULTS Among 257 patients with complete data, mean baseline viral load was 5.7 log10 copies per milliliter and median CD4 count was 165 cells per cubic millimeter. Primary drug resistance was present in 3.1% at baseline. At 6 months, 87.5% had undetectable viral load, indicating excellent response to ART despite high baseline viremia. After excluding those with transmitted resistance, suboptimal adherence and viral rebound, data from 112 patients were analyzed using a mathematical model. We estimated the median R0 to be 5.3. The corresponding εc was ∼0.8. CONCLUSIONS These estimates of R0 and εc are smaller than current estimates for HIV-1B, suggesting that HIV-1C exhibits lower in vivo fitness compared with HIV-1B, which allows successful treatment despite high baseline viral loads. The lower fitness, and potentially lower virulence, together with high viral loads may underlie the heightened transmission potential of HIV-1C and its growing global spread.
Collapse
Affiliation(s)
- Anita Shet
- Department of Pediatrics, Division of Infectious Diseases, St. John's Medical College Hospital, Bangalore, India
| | - Pradeep Nagaraja
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India; and
| | - Narendra M. Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India; and
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
249
|
Deconstructing the Antiviral Neutralizing-Antibody Response: Implications for Vaccine Development and Immunity. Microbiol Mol Biol Rev 2016; 80:989-1010. [PMID: 27784796 DOI: 10.1128/mmbr.00024-15] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The antibody response plays a key role in protection against viral infections. While antiviral antibodies may reduce the viral burden via several mechanisms, the ability to directly inhibit (neutralize) infection of cells has been extensively studied. Eliciting a neutralizing-antibody response is a goal of many vaccine development programs and commonly correlates with protection from disease. Considerable insights into the mechanisms of neutralization have been gained from studies of monoclonal antibodies, yet the individual contributions and dynamics of the repertoire of circulating antibody specificities elicited by infection and vaccination are poorly understood on the functional and molecular levels. Neutralizing antibodies with the most protective functionalities may be a rare component of a polyclonal, pathogen-specific antibody response, further complicating efforts to identify the elements of a protective immune response. This review discusses advances in deconstructing polyclonal antibody responses to flavivirus infection or vaccination. Our discussions draw comparisons to HIV-1, a virus with a distinct structure and replication cycle for which the antibody response has been extensively investigated. Progress toward deconstructing and understanding the components of polyclonal antibody responses identifies new targets and challenges for vaccination strategies.
Collapse
|
250
|
Gelpi M, Hartling HJ, Thorsteinsson K, Gerstoft J, Ullum H, Nielsen SD. Immune recovery in acute and chronic HIV infection and the impact of thymic stromal lymphopoietin. BMC Infect Dis 2016; 16:591. [PMID: 27769179 PMCID: PMC5073883 DOI: 10.1186/s12879-016-1930-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 10/13/2016] [Indexed: 02/06/2023] Open
Abstract
Background Symptomatic primary HIV infection is associated with an adverse prognosis, and immediate initiation of combination antiretroviral therapy (cART) is recommended. However, little is known about immunological predictors of immune recovery. Thymic Stromal Lymphopoietin (TSLP) is a cytokine that promotes CD4+ T cells homeostatic polyclonal proliferation and regulates Th17/regulatory T-cell balance, immunological functions known to be affected during primary HIV infection. The aim of this study was to describe immune recovery in primary and chronic HIV infection and possible impact of TSLP. Methods Prospective study including 100 HIV-infected individuals (primary HIV infection (N = 14), early presenters (>350 CD4+ T cells/μL, N = 42), late presenters without advanced disease (200–350 CD4+ T cells/μL, N = 24) and with advanced disease (<200 CD4+ T cells/μL, N = 20) and). Immune recovery was defined as increase in CD4+ T cells count from baseline to a given time of follow-up. Plasma TSLP was determined using ELISA and CD4+ T cell subpopulations (recent thymic emigrants, naïve and memory cells) were measured using flow cytometry at baseline and after 6, 12 and 24 months of cART. Results Immune recovery was comparable in all groups, and no differences in immune homeostasis were found between primary HIV infection and early presenters, whereas differences in absolute counts and proportions of CD4+ T cell subpopulations were found between primary HIV infection and late presenters. TSLP was elevated in primary HIV infection at baseline and after 24 months of cART. Interestingly, TSLP was negatively associated with proportion of recent thymic emigrants (correlation coefficient −0.60, p = 0.030). TSLP was not associated with immune recovery in primary HIV infection. Conclusions Immune recovery was comparable in primary and chronic HIV infection whereas differences in absolute counts and proportions of CD4+ T cell subpopulations were found between primary HIV infection and late presenters supporting early initiation of cART. Higher plasma TSLP was found in primary HIV infection, and TSLP was associated with lower thymic output, but not with immune recovery. These findings indicate a possible role of TSLP in immune homeostasis in HIV infection but do not support TSLP to affect immune recovery in primary HIV infection.
Collapse
Affiliation(s)
- Marco Gelpi
- Department of Infectious Diseases, Viro-Immunology Research Unit, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Hans J Hartling
- Department of Infectious Diseases, Viro-Immunology Research Unit, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Kristina Thorsteinsson
- Department of Infectious Disease, University Hospital of Copenhagen Hvidovre, Copenhagen, Denmark
| | - Jan Gerstoft
- Department of Infectious Diseases, Viro-Immunology Research Unit, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark
| | - Susanne D Nielsen
- Department of Infectious Diseases, Viro-Immunology Research Unit, University Hospital of Copenhagen Rigshospitalet, Copenhagen, Denmark.
| |
Collapse
|