Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Med Primatol ; 52(5): 290-293, 2023 10.
Article in English | MEDLINE | ID: mdl-37658590

ABSTRACT

HIV-2 Group F virus with an origin in NHPs was isolated from only two individuals. Two serial passages in hu-mice showed increased viral loads, CD4+ T cell decline and nonsynonymous genetic changes showing its capacity for further evolution, and spread in the human.


Subject(s)
HIV-2 , Humans , Animals , Mice , HIV-2/genetics , Serial Passage , Viral Load
2.
J Med Primatol ; 52(5): 294-297, 2023 10.
Article in English | MEDLINE | ID: mdl-37658595

ABSTRACT

HIV-1 emerged from SIVcpz evolving in humans. Humanized mice are an effective tool for assessing viral evolution via measuring viral loads, CD4+ T cell decline, and analyzing genetic changes. Four serial passages showed many non-synonymous mutations important for the adaptation and evolution of SIVcpz to human immune cells.


Subject(s)
HIV-1 , Pan troglodytes , Humans , Animals , Mice , HIV-1/genetics , Serial Passage , Viral Load
3.
Front Immunol ; 14: 1060959, 2023.
Article in English | MEDLINE | ID: mdl-36825016

ABSTRACT

Introduction: Immunocompetent and immunocompromised murine models have been instrumental in answering important questions regarding ZIKV pathogenesis and vertical transmission. However, mimicking human congenital zika syndrome (CZS) characteristics in these murine models has been less than optimal and does not address the potential viral effects on the human immune system. Methods: Here, we utilized neonatal humanized Rag2-/-γc-/- mice to model CZS and evaluate the potential viral effects on the differentiation of human hematopoietic stem cells in vivo. Newborn Rag2-/-γc-/- mice were engrafted with ZIKV-infected hematopoietic stem cells (HSC) and monitored for symptoms and lesions. Results: Within 13 days, mice displayed outward clinical symptoms that encompassed stunted growth, hunched posture, ruffled fur, and ocular defects. Striking gross pathologies in the brain and visceral organs were noted. Our results also confirmed that ZIKV actively infected human CD34+ hematopoietic stem cells and restricted the development of terminally differentiated B cells. Histologically, there was multifocal mineralization in several different regions of the brain together with ZIKV antigen co-localization. Diffuse necrosis of pyramidal neurons was seen with collapse of the hippocampal formation. Discussion: Overall, this model recapitulated ZIKV microcephaly and CZS together with viral adverse effects on the human immune cell ontogeny thus providing a unique in vivo model to assess the efficacy of novel therapeutics and immune interventions.


Subject(s)
Microcephaly , Nervous System Malformations , Zika Virus Infection , Animals , Humans , Mice , Cell Differentiation , Microcephaly/virology , Nervous System Malformations/virology , Zika Virus , Zika Virus Infection/complications
4.
J Med Primatol ; 51(5): 284-287, 2022 10.
Article in English | MEDLINE | ID: mdl-36030392

ABSTRACT

Serial passage of SIVmac239 allows for greater understanding of the genetic changes necessary for cross-species transmission of primate lentiviruses into humans. Using humanized mice, we show that adaptive mutations continue to accumulate in SIVmac239 during four serial passages, with persistent CD4+ T cell decline and increases in plasma viral loads.


Subject(s)
Rodent Diseases , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Humans , Macaca mulatta , Mice , Serial Passage , Simian Immunodeficiency Virus/genetics , Viral Load
5.
J Med Primatol ; 51(5): 288-291, 2022 10.
Article in English | MEDLINE | ID: mdl-36030391

ABSTRACT

Critical genetic adaptations needed for SIV chimpanzee to evolve into HIV-1 are not well understood. Using humanized mice, we mimicked the evolution of SIVcpzLB715 into HIV-1 Group M over the course of four generations. Higher initial viral load, increased CD4+ T-cell decline, and nonsynonymous substitutions arose suggesting viral evolution.


Subject(s)
HIV-1 , Rodent Diseases , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Animals , Disease Models, Animal , Evolution, Molecular , HIV-1/genetics , Mice , Pan troglodytes/genetics , Simian Immunodeficiency Virus/genetics , Viral Load
6.
Front Virol ; 12021.
Article in English | MEDLINE | ID: mdl-37168442

ABSTRACT

Simian immunodeficiency virus native to sooty mangabeys (SIVsm) is believed to have given rise to HIV-2 through cross-species transmission and evolution in the human. SIVmac239 and SIVB670, pathogenic to macaques, and SIVhu, isolated from an accidental human infection, also have origins in SIVsm. With their common ancestral lineage as that of HIV-2 from the progenitor SIVsm, but with different passage history in different hosts, they provide a unique opportunity to evaluate cross-species transmission to a new host and their adaptation/evolution both in terms of potential genetic and phenotypic changes. Using humanized mice with a transplanted human system, we evaluated in vivo replication kinetics, CD4+ T cell dynamics and genetic adaptive changes during serial passage with a goal to understand their evolution under human selective immune pressure. All the three viruses readily infected hu-mice causing chronic viremia. While SIVmac and SIVB670 caused CD4+ T cell depletion during sequential passaging, SIVhu with a deletion in nef gene was found to be less pathogenic. Deep sequencing of the genomes of these viruses isolated at different times revealed numerous adaptive mutations of significance that increased in frequency during sequential passages. The ability of these viruses to infect and replicate in humanized mice provides a new small animal model to study SIVs in vivo in addition to more expensive macaques. Since SIVmac and related viruses have been indispensable in many areas of HIV pathogenesis, therapeutics and cure research, availability of this small animal hu-mouse model that is susceptible to both SIV and HIV viruses is likely to open novel avenues of investigation for comparative studies using the same host.

7.
Front Microbiol ; 11: 1889, 2020.
Article in English | MEDLINE | ID: mdl-32849468

ABSTRACT

The genetic evolution of HIV-1 from its progenitor virus SIV following cross-species transmission is not well understood. Here we simulated the SIVcpz initial transmission to humans using humanized mice and followed the viral evolution during serial passages lasting more than a year. All three SIVcpz progenitor viruses used, namely LB715 and MB897 (group M) as well as EK505 (group N) readily infected hu-mice resulting in chronic viremia. Viral loads increased progressively to higher set-points and the CD4+ T cell decline became more pronounced by the end of the second serial passage indicating viral adaptation and increased pathogenicity. Viral genomes sequenced at different time points revealed many non-synonymous variants not previously reported that occurred throughout the viral genome, including the gag, pol, env, and nef genes. These results shed light on the potential changes that the SIVcpz genome had undergone during the initial stages of human infection and subsequent spread.

8.
J Med Primatol ; 49(5): 280-283, 2020 10.
Article in English | MEDLINE | ID: mdl-32777101

ABSTRACT

Through the accumulation of adaptive mutations, HIV-2 originated from SIVsm. To identify these evolutionary changes, a humanized mouse model recapitulated the process that likely enabled this cross-species transmission event. Various adaptive mutations arose, as well as increased virulence and CD4+ T-cell decline as the virus was passaged in humanized mice.


Subject(s)
CD4 Lymphocyte Count , Evolution, Molecular , HIV-2/genetics , HIV-2/pathogenicity , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Animals , Cercocebus atys , Disease Models, Animal , Mice , Mice, Transgenic , Monkey Diseases , Mutation , Virulence
9.
Antimicrob Agents Chemother ; 64(10)2020 09 21.
Article in English | MEDLINE | ID: mdl-32661005

ABSTRACT

Adequate antiretroviral (ARV) concentrations in lymphoid tissues are critical for optimal antiretroviral therapy (ART). While the spleen contains 25% of the body's lymphocytes, there are minimal data on ARV penetration in this organ. This study quantified total and protein-unbound splenic ARV concentrations and determined whether drug transporters, sex, or infection status were modifiers of these concentrations in animal models and humans. Two humanized mice models (hu-HSC-Rag [n = 36; 18 HIV-positive (HIV+) and 18 HIV-negative (HIV-)] and bone marrow-liver-thymus [n = 13; 7 HIV+ and 6 HIV-]) and one nonhuman primate (NHP) model (rhesus macaque [n = 18; 10 SHIV+ and 8 SHIV-]) were dosed to steady state with ARV combinations. HIV+ human spleens (n = 14) from the National NeuroAIDS Tissue Consortium were analyzed postmortem (up to 24 h postdose). ARV concentrations were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), drug transporter concentrations were measured with LC-MS proteomics, and protein binding in NHP spleens was determined by rapid equilibrium dialysis. Mice generally had the lowest splenic concentrations of the three species. Protein binding in splenic tissue was 6 to 96%, compared to 76 to 99% in blood plasma. NHPs had quantifiable Mrp4, Bcrp, and Ent1 concentrations, and humans had quantifiable ENT1 concentrations. None significantly correlated with tissue ARV concentrations. There was also no observable influence of infection status or sex. With these dosing strategies, NHP splenic penetration most closely resembled that of humans. These data can inform tissue pharmacokinetic scaling to humans to target HIV reservoirs by identifying important species-related differences.


Subject(s)
Anti-HIV Agents , HIV Infections , Pharmaceutical Preparations , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Anti-HIV Agents/therapeutic use , Chromatography, Liquid , HIV Infections/drug therapy , Humans , Macaca mulatta , Mice , Models, Animal , Neoplasm Proteins , Spleen , Tandem Mass Spectrometry
11.
J Med Primatol ; 49(5): 284-287, 2020 10.
Article in English | MEDLINE | ID: mdl-33460210

ABSTRACT

HIV-1 evolved from SIV during cross-species transmission events, though viral genetic changes are not well understood. Here, we studied the evolution of SIVcpzLB715 into HIV-1 Group M using humanized mice. High viral loads, rapid CD4+ T-cell decline, and non-synonymous substitutions were identified throughout the viral genome suggesting viral adaptation.


Subject(s)
Ape Diseases/virology , HIV-1/genetics , Mutation , Pan troglodytes , Simian Immunodeficiency Virus/genetics , Animals , Disease Models, Animal , Evolution, Molecular
12.
Curr HIV Res ; 18(1): 19-28, 2020.
Article in English | MEDLINE | ID: mdl-31870268

ABSTRACT

The main advantage of animal models of infectious diseases over in vitro studies is the gain in the understanding of the complex dynamics between the immune system and the pathogen. While small animal models have practical advantages over large animal models, it is crucial to be aware of their limitations. Although the small animal model at least needs to be susceptible to the pathogen under study to obtain meaningful data, key elements of pathogenesis should also be reflected when compared to humans. Well-designed small animal models for HIV, hepatitis viruses and tuberculosis require, additionally, a thorough understanding of the similarities and differences in the immune responses between humans and small animals and should incorporate that knowledge into the goals of the study. To discuss these considerations, the NIAID hosted a workshop on 'Small Animal Models for HIV, Hepatitis B, and Tuberculosis' on May 30, 2019. Highlights of the workshop are outlined below.


Subject(s)
Disease Models, Animal , HIV Infections/pathology , HIV-1/immunology , Hepatitis B virus/immunology , Hepatitis B/pathology , Mycobacterium tuberculosis/immunology , Tuberculosis/pathology , Animals , Coinfection/microbiology , Guinea Pigs , HIV Infections/immunology , Hepatitis B/immunology , Humans , Macaca mulatta , Marmota , Mice , National Institute of Allergy and Infectious Diseases (U.S.) , Rabbits , Tuberculosis/immunology , United States
13.
J Med Primatol ; 49(1): 40-43, 2020 02.
Article in English | MEDLINE | ID: mdl-31576587

ABSTRACT

HIV-1 evolved from its progenitor SIV strains, but details are lacking on its adaptation to the human host. We followed the evolution of SIVcpz in humanized mice to mimic cross-species transmission. Increasing viral loads, CD4+ T-cell decline, and non-synonymous mutations were seen in the entire genome reflecting viral adaptation.


Subject(s)
CD4 Lymphocyte Count , Evolution, Molecular , Genome, Viral , HIV-1/physiology , Simian Immunodeficiency Virus/physiology , Viral Load , Animals , Biological Evolution , HIV Infections/veterinary , HIV Infections/virology , HIV-1/genetics , Mice , Mice, Transgenic , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics
14.
Antiviral Res ; 172: 104645, 2019 12.
Article in English | MEDLINE | ID: mdl-31697957

ABSTRACT

The Global Virus Network (GVN) was established in 2011 to strengthen research and responses to emerging viral causes of human disease and to prepare against new viral pandemics. There are now 52 GVN Centers of Excellence and 9 Affiliate laboratories in 32 countries. The 11th International GVN meeting was held from June 9-11, 2019 in Barcelona, Spain and was jointly organized with the Spanish Society of Virology. A common theme throughout the meeting was globalization and climate change. This report highlights the recent accomplishments of GVN researchers in several important areas of medical virology, including severe virus epidemics, anticipation and preparedness for changing disease dynamics, host-pathogen interactions, zoonotic virus infections, ethical preparedness for epidemics and pandemics, one health and antivirals.


Subject(s)
Communicable Diseases, Emerging , Global Health , One Health/trends , Virus Diseases , Animals , Antiviral Agents , Arboviruses/drug effects , Arboviruses/genetics , Arboviruses/metabolism , Climate Change , Communicable Diseases, Emerging/drug therapy , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/transmission , Coronavirus/drug effects , Coronavirus/genetics , Coronavirus/metabolism , Ebolavirus/drug effects , Ebolavirus/genetics , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/prevention & control , Hepatitis B/drug therapy , Hepatitis B/epidemiology , Hepatitis B/prevention & control , Host-Pathogen Interactions , Humans , Influenza, Human/drug therapy , Influenza, Human/epidemiology , Influenza, Human/prevention & control , Internationality , Pandemics , Viral Vaccines , Virus Diseases/drug therapy , Virus Diseases/epidemiology , Virus Diseases/transmission , Viruses/drug effects , Viruses/genetics , Viruses/metabolism , Zoonoses/drug therapy , Zoonoses/epidemiology , Zoonoses/transmission
15.
Article in English | MEDLINE | ID: mdl-31611355

ABSTRACT

For HIV cure strategies like "kick and kill" to succeed, antiretroviral (ARV) drugs must reach effective concentrations in putative viral reservoirs. We characterize penetration of six ARVs in three preclinical animal models and humans. We found that standard dosing strategies in preclinical species closely mimicked tissue concentrations in humans for some, but not all, ARVs. These results have implications for interpreting HIV treatment, prevention, or cure interventions between preclinical and clinical models.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/drug therapy , Animals , Anti-HIV Agents/therapeutic use , Atazanavir Sulfate/therapeutic use , Emtricitabine/therapeutic use , Female , Humans , In Vitro Techniques , Maraviroc/therapeutic use , Mice , Raltegravir Potassium/therapeutic use , Tenofovir/therapeutic use
16.
Sci Transl Med ; 11(499)2019 07 03.
Article in English | MEDLINE | ID: mdl-31270274

ABSTRACT

HIV replication within tissues may increase in response to a reduced exposure to antiretroviral drugs. Traditional approaches to measuring drug concentrations in tissues are unable to characterize a heterogeneous drug distribution. Here, we used mass spectrometry imaging (MSI) to visualize the distribution of six HIV antiretroviral drugs in gut tissue sections from three species (two strains of humanized mice, macaques, and humans). We measured drug concentrations in proximity to CD3+ T cells that are targeted by HIV, as well as expression of HIV or SHIV RNA and expression of the MDR1 drug efflux transporter in gut tissue from HIV-infected humanized mice, SHIV-infected macaques, and HIV-infected humans treated with combination antiretroviral drug therapy. Serial 10-µm sections of snap-frozen ileal and rectal tissue were analyzed by MSI for CD3+ T cells and MDR1 efflux transporter expression by immunofluorescence and immunohistochemistry, respectively. The tissue slices were analyzed for HIV/SHIV RNA expression by in situ hybridization and for antiretroviral drug concentrations by liquid chromatography-mass spectrometry. The gastrointestinal tissue distribution of the six drugs was heterogeneous. Fifty percent to 60% of CD3+ T cells did not colocalize with detectable drug concentrations in the gut tissue. In all three species, up to 90% of HIV/SHIV RNA was found to be expressed in gut tissue with no exposure to drug. These data suggest that there may be gut regions with little to no exposure to antiretroviral drugs, which may result in low-level HIV replication contributing to HIV persistence.


Subject(s)
Anti-Retroviral Agents/pharmacology , Gastrointestinal Tract/virology , HIV/drug effects , Simian Immunodeficiency Virus/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adolescent , Adult , Aged , Animals , CD3 Complex/metabolism , Female , Gastrointestinal Tract/drug effects , Gene Expression Regulation, Viral/drug effects , HIV/genetics , Humans , Macaca mulatta , Male , Mice , Middle Aged , RNA, Viral/genetics , RNA, Viral/metabolism , Simian Immunodeficiency Virus/genetics , Species Specificity , T-Lymphocytes/drug effects , Young Adult
17.
J Pharmacol Exp Ther ; 370(3): 360-368, 2019 09.
Article in English | MEDLINE | ID: mdl-31235531

ABSTRACT

In a "kick and kill" strategy for human immunodeficiency virus (HIV) eradication, protective concentrations of antiretrovirals (ARVs) in the lymph node are important to prevent vulnerable cells from further HIV infection. However, the factors responsible for drug distribution and concentration into these tissues are largely unknown. Although humanized mice and nonhuman primates (NHPs) are crucial to HIV research, ARV tissue pharmacology has not been well characterized across species. This study investigated the influence of drug transporter expression, viral infection, and sex on ARV penetration within lymph nodes of animal models and humans. Six ARVs were dosed for 10 days in humanized mice and NHPs. Plasma and lymph nodes were collected at necropsy, 24 hours after the last dose. Human lymph node tissue and plasma from deceased patients were collected from tissue banks. ARV, active metabolite, and endogenous nucleotide concentrations were measured by liquid chromatography-tandem mass spectrometry, and drug transporter expression was measured using quantitative polymerase chain reaction and quantitative targeted absolute proteomics. In NHPs and humans, lymph node ARV concentrations were greater than or equal to plasma, and tenofovir diphosphate/deoxyadenosine triphosphate concentration ratios achieved efficacy targets in lymph nodes from all three species. There was no effect of infection or sex on ARV concentrations. Low drug transporter expression existed in lymph nodes from all species, and no predictive relationships were found between transporter gene/protein expression and ARV penetration. Overall, common preclinical models of HIV infection were well suited to predict human ARV exposure in lymph nodes, and low transporter expression suggests primarily passive drug distribution in these tissues. SIGNIFICANCE STATEMENT: During human immunodeficiency virus (HIV) eradication strategies, protective concentrations of antiretrovirals (ARVs) in the lymph node prevent vulnerable cells from further HIV infection. However, ARV tissue pharmacology has not been well characterized across preclinical species used for HIV eradication research, and the influence of drug transporters, HIV infection, and sex on ARV distribution and concentration into the lymph node is largely unknown. Here we show that two animal models of HIV infection (humanized mice and nonhuman primates) were well suited to predict human ARV exposure in lymph nodes. Additionally, we found that drug transporter expression was minimal and-along with viral infection and sex-did not affect ARV penetration into lymph nodes from any species.


Subject(s)
Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Gene Expression Regulation/drug effects , HIV/physiology , Lymph Nodes/metabolism , Membrane Transport Proteins/metabolism , Sex Characteristics , Animals , Anti-HIV Agents/blood , Female , HIV/drug effects , Humans , Lymph Nodes/drug effects , Macaca mulatta , Male , Mice , Species Specificity
18.
Methods Mol Biol ; 1937: 125-134, 2019.
Article in English | MEDLINE | ID: mdl-30706393

ABSTRACT

Lentiviral vectors (LVs) are widely used in gene transfer protocols due to many advantages that include stable gene expression, higher transgene payloads, and, importantly, the ability to pseudotype the vectors with a diverse number of heterologous viral envelopes with broad or restricted cell tropism depending on the need. The pseudotyping process also allows for incorporation of specific antibodies/ligands to engineer LVs. These features greatly facilitate customization of lentiviral vectors for cell/tissue specific gene delivery. The VSV-G protein containing envelope remains the most widely used among the viral glycoproteins used for LV pseudotyping due to its versatile host range and stability. However, many other viral envelopes are being identified for special applications of LVs. Here we describe the methodology to generate pseudotyped LVs using a four-plasmid transient transfection system focusing on aspects to generate high-titer vector stocks.


Subject(s)
Lentivirus/physiology , Plasmids/genetics , Viral Envelope Proteins/metabolism , Virus Cultivation/methods , Gene Transfer Techniques , Genetic Vectors , HEK293 Cells , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Viral Envelope Proteins/genetics , Viral Load , Viral Tropism , Virus Assembly
20.
Xenobiotica ; 49(10): 1192-1201, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30346892

ABSTRACT

1. Antiretroviral concentrations in cerebrospinal fluid (CSF) are used as surrogate for brain tissue, although sparse data support this. We quantified antiretrovirals in brain tissue across preclinical models, compared them to CSF, and calculated 90% inhibitory quotients (IQ90) for nonhuman primate (NHP) brain tissue. Spatial distribution of efavirenz was performed by mass-spectrometry imaging (MSI). 2. HIV or RT-SHIV-infected and uninfected animals from two humanized mouse models (hemopoietic-stem cell/RAG2-, n = 36; bone marrow-liver-thymus/BLT, n =13) and an NHP model (rhesus macaque, n =18) were dosed with six antiretrovirals. Brain tissue, CSF (NHPs), and plasma were collected at necropsy. Drug concentrations were measured by LC-MS/MS. Rapid equilibrium dialysis determined protein binding in NHP brain. 3. Brain tissue penetration of most antiretrovirals were >10-fold lower (p < 0.02) in humanized mice than NHPs. NHP CSF concentrations were >13-fold lower (p <0.02) than brain tissue with poor agreement except for efavirenz (r = 0.91, p = 0.001). Despite 97% brain tissue protein binding, efavirenz achieved IQ90>1 in all animals and 2-fold greater white versus gray matter concentration. 4. Brain tissue penetration varied across animal models for all antiretrovirals except raltegravir, and extrapolating brain tissue concentrations between models should be avoided. With the exception of efavirenz, CSF is not a surrogate for brain tissue concentrations.


Subject(s)
Anti-HIV Agents , Benzoxazines , Brain , HIV Infections , HIV-1 , Alkynes , Animals , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/pharmacology , Benzoxazines/pharmacokinetics , Benzoxazines/pharmacology , Brain/metabolism , Brain/pathology , Brain/virology , Cyclopropanes , Drug Evaluation, Preclinical , Female , HIV Infections/cerebrospinal fluid , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Macaca mulatta , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...