Generic placeholder image

Current HIV Research

Editor-in-Chief

ISSN (Print): 1570-162X
ISSN (Online): 1873-4251

Conference Proceedings

Small Animal Models for Human Immunodeficiency Virus (HIV), Hepatitis B, and Tuberculosis: Proceedings of an NIAID Workshop

Author(s): Ramesh Akkina, Daniel L. Barber, Moses T. Bility, Karl-Dimiter Bissig, Benjamin J. Burwitz, Katrin Eichelberg, Janice J. Endsley, J. Victor Garcia, Richard Hafner, Petros C. Karakousis, Brent E. Korba, Rajen Koshy, Chris Lambros, Stephan Menne, Eric L. Nuermberger, Alexander Ploss, Brendan K. Podell, Larisa Y. Poluektova, Brigitte E. Sanders-Beer*, Selvakumar Subbian and Angela Wahl

Volume 18, Issue 1, 2020

Page: [19 - 28] Pages: 10

DOI: 10.2174/1570162X18666191223114019

open access plus

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.

Keywords: HIV, AIDS, co-infections, HBV, tuberculosis, animal models.

Graphical Abstract
[1]
Wahl A, De C, Abad Fernandez M, et al. Precision mouse models with expanded tropism for human pathogens. Nat Biotechnol 2019; 37(10): 1163-73.
[http://dx.doi.org/10.1038/s41587-019-0225-9] [PMID: 31451733]
[2]
Honeycutt JB, Garcia JV. Humanized mice: models for evaluating NeuroHIV and cure strategies. J Neurovirol 2018; 24(2): 185-91.
[http://dx.doi.org/10.1007/s13365-017-0567-3] [PMID: 28831774]
[3]
Kovarova M, Benhabbour SR, Massud I, et al. Ultra-long-acting removable drug delivery system for HIV treatment and prevention. Nat Commun 2018; 9(1): 4156.
[http://dx.doi.org/10.1038/s41467-018-06490-w] [PMID: 30297889]
[4]
Denton PW, Garcia JV. Mucosal HIV-1 transmission and prevention strategies in BLT humanized mice. Trends Microbiol 2012; 20(6): 268-74.
[http://dx.doi.org/10.1016/j.tim.2012.03.007] [PMID: 22503637]
[5]
Wahl A, Swanson MD, Nochi T, et al. Human breast milk and antiretrovirals dramatically reduce oral HIV-1 transmission in BLT humanized mice. PLoS Pathog 2012; 8(6):e1002732
[http://dx.doi.org/10.1371/journal.ppat.1002732] [PMID: 22737068]
[6]
Charlins P, Schmitt K, Remling-Mulder L, et al. A humanized mouse-based HIV-1 viral outgrowth assay with higher sensitivity than in vitro qVOA in detecting latently infected cells from individuals on ART with undetectable viral loads. Virology 2017; 507: 135-9.
[http://dx.doi.org/10.1016/j.virol.2017.04.011] [PMID: 28432928]
[7]
Schmitt K, Akkina R. Ultra-sensitive HIV-1 latency viral outgrowth assays using humanized mice. Front Immunol 2018; 9: 344.
[http://dx.doi.org/10.3389/fimmu.2018.00344] [PMID: 29556230]
[8]
Thompson CG, Rosen EP, Prince HMA, et al. Heterogeneous antiretroviral drug distribution and HIV/SHIV detection in the gut of three species. Sci Transl Med 2019; 11(499)eaap8758
[http://dx.doi.org/10.1126/scitranslmed.aap8758] [PMID: 31270274]
[9]
Hu S, Neff CP, Kumar DM, et al. A humanized mouse model for HIV-2 infection and efficacy testing of a single-pill triple-drug combination anti-retroviral therapy. Virology 2017; 501: 115-8.
[http://dx.doi.org/10.1016/j.virol.2016.11.013] [PMID: 27912079]
[10]
Schmitt K, Charlins P, Veselinovic M, et al. Zika viral infection and neutralizing human antibody response in a BLT humanized mouse model. Virology 2018; 515: 235-42.
[http://dx.doi.org/10.1016/j.virol.2017.12.026] [PMID: 29310105]
[11]
Gendelman HE, McMillan J, Bade AN, Edagwa B, Kevadiya BD. The promise of long-acting antiretroviral therapies: from need to manufacture. Trends Microbiol 2019; 27(7): 593-606.
[http://dx.doi.org/10.1016/j.tim.2019.02.009] [PMID: 30981593]
[12]
Edagwa B, McMillan J, Sillman B, Gendelman HE. Long-acting slow effective release antiretroviral therapy. Expert Opin Drug Deliv 2017; 14(11): 1281-91.
[http://dx.doi.org/10.1080/17425247.2017.1288212] [PMID: 28128004]
[13]
Dagur RS, Wang W, Makarov E, Sun Y, Poluektova LY. Establishment of the dual humanized TK-NOG mouse model for HIVassociated liver pathogenesis. J Vis Exp 2019; (151):
[14]
Bility MT, Cheng L, Zhang Z, et al. Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: Induction of human-specific liver fibrosis and M2-like macrophages. PLoS Pathog 2014; 10(3)e1004032
[http://dx.doi.org/10.1371/journal.ppat.1004032] [PMID: 24651854]
[15]
Cheng L, Li F, Bility MT, Murphy CM, Su L. Modeling hepatitis B virus infection, immunopathology and therapy in mice. Antiviral Res 2015; 121: 1-8.
[http://dx.doi.org/10.1016/j.antiviral.2015.06.012] [PMID: 26099683]
[16]
Samal J, Kelly S, Na-Shatal A, et al. Human immunodeficiency virus infection induces lymphoid fibrosis in the BM-liver-thymus-spleen humanized mouse model. JCI Insight 2018; 3(18)120430
[http://dx.doi.org/10.1172/jci.insight.120430] [PMID: 30232273]
[17]
Bility MT, Nio K, Li F, et al. Chronic hepatitis C infection-induced liver fibrogenesis is associated with M2 macrophage activation. Sci Rep 2016; 6: 39520.
[http://dx.doi.org/10.1038/srep39520] [PMID: 287000758]
[18]
Winer BY, Ding Q, Gaska JM, Ploss A. In vivo models of hepatitis B and C virus infection. FEBS Lett 2016; 590(13): 1987-99.
[http://dx.doi.org/10.1002/1873-3468.12157] [PMID: 27009462]
[19]
Douam F, Ploss A. The use of humanized mice for studies of viral pathogenesis and immunity. Curr Opin Virol 2018; 29: 62-71.
[http://dx.doi.org/10.1016/j.coviro.2018.03.003] [PMID: 29604551]
[20]
de Jong YP, Rice CM, Ploss A. New horizons for studying human hepatotropic infections. J Clin Invest 2010; 120(3): 650-3.
[http://dx.doi.org/10.1172/JCI42338] [PMID: 20179350]
[21]
Bissig KD, Le TT, Woods NB, Verma IM. Repopulation of adult and neonatal mice with human hepatocytes: A chimeric animal model. Proc Natl Acad Sci USA 2007; 104(51): 20507-11.
[http://dx.doi.org/10.1073/pnas.0710528105] [PMID: 18077355]
[22]
Bissig KD, Wieland SF, Tran P, et al. Human liver chimeric mice provide a model for hepatitis B and C virus infection and treatment. J Clin Invest 2010; 120(3): 924-30.
[http://dx.doi.org/10.1172/JCI40094] [PMID: 20179355]
[23]
Dandri M, Burda MR, Török E, et al. Repopulation of mouse liver with human hepatocytes and in vivo infection with hepatitis B virus. Hepatology 2001; 33(4): 981-8.
[http://dx.doi.org/10.1053/jhep.2001.23314] [PMID: 11283864]
[24]
Barzi M, Pankowicz FP, Zorman B, et al. A novel humanized mouse lacking murine P450 oxidoreductase for studying human drug metabolism. Nat Commun 2017; 8(1): 39.
[http://dx.doi.org/10.1038/s41467-017-00049-x] [PMID: 28659616]
[25]
Bissig-Choisat B, Wang L, Legras X, et al. Development and rescue of human familial hypercholesterolaemia in a xenograft mouse model. Nat Commun 2015; 6: 7339.
[http://dx.doi.org/10.1038/ncomms8339] [PMID: 26081744]
[26]
Menne S, Cote PJ. The woodchuck as an animal model for pathogenesis and therapy of chronic hepatitis B virus infection. World J Gastroenterol 2007; 13(1): 104-24.
[http://dx.doi.org/10.3748/wjg.v13.i1.104] [PMID: 17206759]
[27]
Alioto TS, Cruz F, Gomez-Garrido J, et al. The genome sequence of the Eastern woodchuck (Marmota monax): A preclinical animal model for chronic hepatitis B. G3 (Bethesda) 2019; 9(12): 3943-52.
[28]
Suslov A, Wieland S, Menne S. Modulators of innate immunity as novel therapeutics for treatment of chronic hepatitis B. Curr Opin Virol 2018; 30: 9-17.
[http://dx.doi.org/10.1016/j.coviro.2018.01.008] [PMID: 29444493]
[29]
Korolowizc KE, Li B, Huang X, et al. Liver-targeted toll-like receptor 7 agonist combined with entecavir promotes a functional cure in the woodchuck model of hepatitis B virus. Hepatol Commun 2019; 3(10): 1296-310.
[http://dx.doi.org/10.1002/hep4.1397] [PMID: 31592075]
[30]
Burwitz BJ, Wettengel JM, Mück-Häusl MA, et al. Hepatocytic expression of human sodium-taurocholate cotransporting polypeptide enables hepatitis B virus infection of macaques. Nat Commun 2017; 8(1): 2146.
[http://dx.doi.org/10.1038/s41467-017-01953-y] [PMID: 29247188]
[31]
Calderon VE, Valbuena G, Goez Y, et al. A humanized mouse model of tuberculosis. PLoS One 2013; 8(5)e63331
[http://dx.doi.org/10.1371/journal.pone.0063331] [PMID: 23691024]
[32]
Nusbaum RJ, Calderon VE, Huante MB, et al. Pulmonary tuberculosis in humanized mice infected with HIV-1. Sci Rep 2016; 6: 21522.
[http://dx.doi.org/10.1038/srep21522] [PMID: 26908312]
[33]
Huante M, Nusbaum R, Endsley J. Co-infection with TB and HIV: converging epidemics, clinical challenges, and microbial synergy 2019; 123-53.
[34]
Jondle CN, Sharma A, Simonson TJ, Larson B, Mishra BB, Sharma J. Macrophage galactose-type lectin-1 deficiency is associated with increased neutrophilia and hyperinflammation in gram-negative pneumonia 2016; 196(7): 3088-96.
[http://dx.doi.org/10.4049/jimmunol.1501790]
[35]
Sakai S, Kauffman KD, Schenkel JM, McBerry CC, Mayer-Barber KD, Masopust D, et al. Cutting edge: control of Mycobacterium tuberculosis infection by a subset of lung parenchyma-homing CD4 T cells. Journal of Immunology (Baltimore, Md : 1950) 2014; 192(7): 2965-9.
[36]
Sakai S, Mayer-Barber KD, Barber DL. Defining features of protective CD4 T cell responses to Mycobacterium tuberculosis. Curr Opin Immunol 2014; 29: 137-42.
[http://dx.doi.org/10.1016/j.coi.2014.06.003] [PMID: 25000593]
[37]
Kauffman KD, Sallin MA, Sakai S, et al. Defective positioning in granulomas but not lung-homing limits CD4 T-cell interactions with Mycobacterium tuberculosis-infected macrophages in rhesus macaques. Mucosal Immunol 2018; 11(2): 462-73.
[http://dx.doi.org/10.1038/mi.2017.60] [PMID: 28745326]
[38]
Sallin MA, Kauffman KD, Riou C, et al. Host resistance to pulmonary Mycobacterium tuberculosis infection requires CD153 expression. Nat Microbiol 2018; 3(11): 1198-205.
[http://dx.doi.org/10.1038/s41564-018-0231-6] [PMID: 30202016]
[39]
Kolloli A, Subbian S. Host-directed therapeutic strategies for tuberculosis. Front Med (Lausanne) 2017; 4: 171.
[http://dx.doi.org/10.3389/fmed.2017.00171] [PMID: 29094039]
[40]
Frank DJ, Horne DJ, Dutta NK, et al. Remembering the host in tuberculosis drug development. J Infect Dis 2019; 219(10): 1518-24.
[http://dx.doi.org/10.1093/infdis/jiy712] [PMID: 30590592]
[41]
Singhal A, Jie L, Kumar P, et al. Metformin as adjunct antituberculosis therapy. Sci Transl Med 2014; 6(263)263ra159
[http://dx.doi.org/10.1126/scitranslmed.3009885] [PMID: 25411472]
[42]
Dutta NK, Bruiners N, Pinn ML, et al. Statin adjunctive therapy shortens the duration of TB treatment in mice. J Antimicrob Chemother 2016; 71(6): 1570-7.
[http://dx.doi.org/10.1093/jac/dkw014] [PMID: 26903278]
[43]
Dutta NK, Bruiners N, Zimmerman MD, et al. Adjunctive host-directed therapy with statins improves tuberculosis-related outcomes in mice. J Infect Dis 2019. pii: jiz517
[http://dx.doi.org/10.1093/infdis/jiz517] [PMID: 31605489]
[44]
Maiga M, Ahidjo BA, Maiga MC, et al. Efficacy of adjunctive tofacitinib therapy in mouse models of tuberculosis. EBioMedicine 2015; 2(8): 868-73.
[http://dx.doi.org/10.1016/j.ebiom.2015.07.014] [PMID: 26425693]
[45]
Vilaplana C, Marzo E, Tapia G, Diaz J, Garcia V, Cardona PJ. Ibuprofen therapy resulted in significantly decreased tissue bacillary loads and increased survival in a new murine experimental model of active tuberculosis. J Infect Dis 2013; 208(2): 199-202.
[http://dx.doi.org/10.1093/infdis/jit152] [PMID: 23564636]
[46]
Apt A, Kramnik I. Man and mouse TB: contradictions and solutions. Tuberculosis (Edinb) 2009; 89(3): 195-8.
[http://dx.doi.org/10.1016/j.tube.2009.02.002] [PMID: 19345146]
[47]
Nuermberger EL. Preclinical efficacy testing of new drug candidates. Microbiol Spectr 2017; 5(3)
[http://dx.doi.org/10.1128/microbiolspec.TBTB2-0034-2017] [PMID: 28643624]
[48]
Zhang T, Li SY, Williams KN, Andries K, Nuermberger EL. Short-course chemotherapy with TMC207 and rifapentine in a murine model of latent tuberculosis infection. Am J Respir Crit Care Med 2011; 184(6): 732-7.
[http://dx.doi.org/10.1164/rccm.201103-0397OC] [PMID: 21659613]
[49]
Dartois V. The path of anti-tuberculosis drugs: From blood to lesions to mycobacterial cells. Nat Rev Microbiol 2014; 12(3): 159-67.
[http://dx.doi.org/10.1038/nrmicro3200] [PMID: 24487820]
[50]
Lanoix JP, Ioerger T, Ormond A, et al. Selective inactivity of pyrazinamide against tuberculosis in C3HeB/FeJ mice is best explained by neutral pH of caseum. Antimicrob Agents Chemother 2015; 60(2): 735-43.
[http://dx.doi.org/10.1128/AAC.01370-15] [PMID: 26574016]
[51]
Bartelink IH, Zhang N, Keizer RJ, et al. New paradigm for translational modeling to predict long-term tuberculosis treatment response. Clin Transl Sci 2017; 10(5): 366-79.
[http://dx.doi.org/10.1111/cts.12472] [PMID: 28561946]
[52]
Turner OC, Basaraba RJ, Orme IM. Immunopathogenesis of pulmonary granulomas in the guinea pig after infection with Mycobacterium tuberculosis. Infect Immun 2003; 71(2): 864-71.
[http://dx.doi.org/10.1128/IAI.71.2.864-871.2003] [PMID: 12540568]
[53]
Ly LH, Russell MI, McMurray DN. Cytokine profiles in primary and secondary pulmonary granulomas of Guinea pigs with tuberculosis. Am J Respir Cell Mol Biol 2008; 38(4): 455-62.
[http://dx.doi.org/10.1165/rcmb.2007-0326OC] [PMID: 18032570]
[54]
Kiran D, Podell BK, Chambers M, Basaraba RJ. Host-directed therapy targeting the Mycobacterium tuberculosis granuloma: a review. Semin Immunopathol 2016; 38(2): 167-83.
[http://dx.doi.org/10.1007/s00281-015-0537-x] [PMID: 26510950]
[55]
Fernandez ML. Guinea pigs as models for cholesterol and lipoprotein metabolism. J Nutr 2001; 131(1): 10-20.
[http://dx.doi.org/10.1093/jn/131.1.10] [PMID: 11208932]
[56]
Ernst JD. The immunological life cycle of tuberculosis. Nat Rev Immunol 2012; 12(8): 581-91.
[http://dx.doi.org/10.1038/nri3259] [PMID: 22790178]
[57]
Subbian S, Tsenova L, Yang G, et al. Chronic pulmonary cavitary tuberculosis in rabbits: A failed host immune response. Open Biol 2011; 1(4)110016
[http://dx.doi.org/10.1098/rsob.110016] [PMID: 22645653]
[58]
Esteves PJ, Abrantes J, Baldauf HM, et al. The wide utility of rabbits as models of human diseases. Exp Mol Med 2018; 50(5): 66.
[http://dx.doi.org/10.1038/s12276-018-0094-1] [PMID: 29789565]
[59]
Allen TM, Brehm MA, Bridges S, et al. Humanized immune system mouse models: Progress, challenges and opportunities. Nat Immunol 2019; 20(7): 770-4.
[http://dx.doi.org/10.1038/s41590-019-0416-z] [PMID: 31160798]
[60]
Akkina R, Allam A, Balazs AB, et al. Improvements and limitations of humanized mouse models for HIV research: NIH/NIAID “Meet the experts” 2015 workshop summary. AIDS Res Hum Retroviruses 2016; 32(2): 109-19.
[http://dx.doi.org/10.1089/aid.2015.0258] [PMID: 26670361]

© 2024 Bentham Science Publishers | Privacy Policy