<<

The Pigtail Macaque (Macaca nemestrina) in Biomedical Research

Charlotte E. Hotchkiss The Macaca nemestrina (pigtail macaques) is an invaluable non- model that has been utilized to support research in the areas of reproduction and development, immunology, infectious disease, , transplantation and stem cell research.1-30 This summary outlines the natural history of the M. nemestrina and summarizes the important research areas that have benefited from having this unique model available for advancing biomedical research. Natural Biology The southern pigtail macaque (M. nemestrina) is native to southeast , primarily inhabiting islands in Indonesia, but also the mainland in , , and Brunei Darussalam.31 It is considered a separate species from the northern pigtail macaque (M. leonina) based on morphological characteristics,32 but there is range overlap, and interbreeding occurs in Thailand and on the islands of Phuket and Yao Yai.33 Genetically, M. nemestrina is in the silenus group34 which is distinct from the macaques more commonly used in research, namely the rhesus (M. mulatta) and cynomolgus (M. fascicularis) macaques which are in the fascicularis group. The primary habitat of M. nemestrina is the tropical rainforest, with preferred temperatures of 64° – 86° F (18° – 30° C). Unlike some other macaque species, they do not adapt well to cold environments. Macaques that live in temperate climates tend to exhibit seasonality in breeding to ensure that infants are born in the spring. In contrast, M. nemestrina breed and deliver infants year- round. Young infants are at high risk for developing hypothermia; therefore M. nemestrina colonies should not be maintained outdoors in temperate climates. Although the lack of seasonal breeding raises challenges in maintaining M. nemestrina breeding colonies, it increases their utility as a model for biomedical research.

Infectious Disease Models M. nemestrina demonstrate a high susceptibility to human infectious disease pathogens as well as nonhuman primate , such as immunodeficiency (SIV) and simian-human chimeric immunodeficiency virus (SHIV).10,14, 35-45 They are of particular interest in HIV/AIDS research due to the presence of a nonfunctional TRIM5α variant (TRIM-cyp) that eliminates a major barrier for replication of HIV-1 in macaques and permits infection with lentivirus strains closely related to HIV-1.46-50 M. nemestrina is the only macaque species to be successfully infected with HIV-151 and are both more easily infected and more likely to exhibit pathologic changes following infection than other macaque species.52 Comparison of pathogenicity in different NHP species provides valuable information on host factors involved in resistance or susceptibility to infection and disease.4,53-57 Mane-A1*004g Mane-A1*006g Macaca nemestrina are also valuable Mane-A3*13g models for non-AIDS infectious disease Mane-A1*010g research, including study of influenza, Mane-A1*018g Mane-A7*01g Mane-A1*019g 58-61 Mane-A6*01g chlamydia, and tuberculosis. One Mane-A1*031g tool that is useful in studying infectious Mane-A4*14g Mane-A1*032g Mane-A1*038g disease is genetic characterization of Mane-A4*13g individual , including MHC Mane-A4*01g Mane-A1*040g Mane-A1*063g Mane-A1*052g haplotypes which can affect the immune response. MHC Class I major and Mane-A1*082g minor expressed alleles have been Mane-A4*01/14g analyzed for ~600 animals using the Mane-A1*084g 62 next generation sequencing approach. Mane-A1*108g A representation of the most common Mane-A1*131g Mane-A1*114g MHC I lineages within the M. nemestrina Mane-A2*05g population at the Washington National Primate Research Center is shown here.

Reproductive and Developmental Models M. nemestrina have a less keratinized vaginal epithelium63 and more appropriate vaginal size than M. mulatta, as well as a year-round menstrual cycle. The vaginal flora of M. nemestrina is similar to that of .64 These factors make them more useful than other macaque species for evaluating vaginal exposure to pathogens, microbicides and the vaginal microbiome.19,20,22,26,27,39,63,65-71 The results from these microbicide research projects can be directly translated to clinical applications. M. nemestrina exhibit a 32-day menstrual cycle similar to humans, and a gestation length averaging 172 days. Menstrual cycles are easy to track in M. nemestrina; not only do they exhibit obvious menstrual bleeding, but they also display marked ovulatory tumescence, as shown here. This ease of tracking is beneficial for studies which require timed-mating as well as experiments where it is necessary to know the stage of the mestrual cycle for treatment or sample collection. Significant contributions to the understanding of premature delivery and infections during pregnancy have been achieved using the M. nemestrina model.72-78 The fact that M. nemestrina breed year round is advantageous for investigators who perform intensive interventions with pregnant animals or neonates, as research subjects are available year round and experiments can be staggered appropriately. There was a quick response and publication to the Zika virus outbreak that was able to take advantage of the readily available pregnant M. nemestrina to evaluate the effects on the developing primate fetus.79 In addition, treatments for neonatal hypoxia which are now in clinical use were developed using the M. nemestrina model.80-87 M. nemestrina adapt well to a jacket and tether system, and with surgical implantation of catheters and monitors it is possible to administer medication, monitor physiological parameters, and collect biological samples without disturbing the animal. Other Models M. nemestrina are suitable animal models for essentially any project where other macaque species are used. They have been used extensively in research involving hematopoietic stem cell transplantation.13,15,88-107 These projects address a wide range of research topics from the basic biology of immunological tolerance and techniques of gene editing to translational topics of improving transplant success in humans and prevention and elimination of HIV infection. M. nemestrina have also been used to demonstrate functional viability of stem cell transplants in a model of myocardial infarction.108-110 In addition, M. nemestrina models have made significant contributions to neuroscience research.111-131

References

1. Beer BE, Brown CR, Whitted S, et al. Immunodeficiency in the absence of high viral load in pig-tailed macaques infected with Simian immunodeficiency virus SIVsun or SIVlhoest. J Virol 2005;79:14044- 14056. 2. Canary LA, Vinton CL, Morcock DR, et al. Rate of AIDS progression is associated with gastrointestinal dysfunction in simian immunodeficiency virus-infected pigtail macaques. J Immunol 2013;190:2959- 2965. 3. Fultz PN, Stallworth J, Porter D, et al. Immunogenicity in pig-tailed macaques of poliovirus replicons expressing HIV-1 and SIV antigens and protection against SHIV-89.6P disease. Virology 2003;315:425- 437. 4. Gooneratne SL, Alinejad-Rokny H, Ebrahimi D, et al. Linking pig-tailed macaque major histocompatibility complex class I haplotypes and cytotoxic T lymphocyte escape mutations in simian immunodeficiency virus infection. J Virol 2014;88:14310-14325. 5. Hirsch VM, G. D, R. G, et al. Phylogeny and natural history of the primate lentiviruses, SIV and HIV. Curr Opin Genet Dev 1995;5:798-806. 6. Joag SV, Li Z, Foresman L, et al. Chimeric simian/human immunodeficiency virus that causes progressive loss of CD4+ T cells and AIDS in pig-tailed macaques. J Virol 1996;70:3189-3197. 7. Joag SV, Liu ZQ, Stephens EB, et al. Oral immunization of macaques with attenuated vaccine virus induces protection against vaginally transmitted AIDS. J Virol 1998;72:9069-9078. 8. Otten RA, Brown BG, Simon M, et al. Differential replication and pathogenic effect of HIV-1 amd HIV-2 in Macaca nemestrina. AIDS Res Hum Retroviruses 1994;8:297-306. 9. Pullium JK, R. AD, E. J, et al. Pig-tailed macaques infected with human immunodeficiency virus (HIV) type 2GB122 or simian/HIV89.6p express virus in semes during primary infection: new model for genital tract shedding and transmission. J Infect Dis 2001;183:1023-1010-1030. 10. Radzio J, Aung W, Holder A, et al. Prevention of vaginal SHIV transmission in macaques by a coitally- dependent Truvada regimen. PloS one 2012;7:e50632. 11. Radzio J, Spreen W, Yueh YL, et al. The long-acting integrase inhibitor GSK744 protects macaques from repeated intravaginal SHIV challenge. Sci Transl Med 2015;7:270ra275. 12. Wei Q, Stallworth JW, Vance PJ, et al. Simian immunodeficiency virus (SIV)/immunoglobulin G immune complexes in SIV-infected macaques block detection of CD16 but not cytolytic activity of natural killer cells. Clin Vaccine Immunol 2006;13:768-778. 13. Younan PM, Polacino P, Kowalski JP, et al. Positive selection of mC46-expressing CD4+ T cells and maintenance of virus specific immunity in a primate AIDS model. Blood 2013;122:179-187. 14. Parsons MS, Lee WS, Kristensen AB, et al. Fc-dependent functions are redundant to efficacy of anti- HIV antibody PGT121 in macaques. J Clin Invest 2019;129:182-191. 15. Peterson CW, Wang J, Deleage C, et al. Differential impact of transplantation on peripheral and tissue-associated viral reservoirs: Implications for HIV gene therapy. PLoS pathogens 2018;14:e1006956. 16. Mangus LM, Beck SE, Queen SE, et al. Lymphocyte-Dominant Encephalitis and Meningitis in Simian Immunodeficiency Virus-Infected Macaques Receiving Antiretroviral Therapy. Am J Pathol 2018;188:125-134. 17. Beck SE, Queen SE, Metcalf Pate KA, et al. An SIV/macaque model targeted to study HIV-associated neurocognitive disorders. J Neurovirol 2018;24:204-212. 18. Zhen A, Peterson CW, Carrillo MA, et al. Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS pathogens 2017;13:e1006753. 19. Srinivasan P, Zhang J, Dinh CT, et al. Repeated administration of high-dose depot medroxyprogesterone acetate does not alter SHIVSF162p3 viral kinetics and tenofovir pharmacokinetics when delivered via intravaginal rings. J Med Primatol 2017;46:129-136. 20. Smith JM, Moss JA, Srinivasan P, et al. Novel multipurpose pod-intravaginal ring for the prevention of HIV, HSV, and unintended pregnancy: Pharmacokinetic evaluation in a macaque model. PloS one 2017;12:e0185946. 21. Parsons MS, Lloyd SB, Lee WS, et al. Partial efficacy of a broadly neutralizing antibody against cell- associated SHIV infection. Sci Transl Med 2017;9. 22. Makarova N, Henning T, Taylor A, et al. Topical tenofovir protects against vaginal simian HIV infection in macaques coinfected with Chlamydia trachomatis and Trichomonas vaginalis. Aids 2017;31:745-752. 23. Gama L, Abreu CM, Shirk EN, et al. Reactivation of simian immunodeficiency virus reservoirs in the brain of virally suppressed macaques. Aids 2017;31:5-14. 24. Croteau JD, Engle EL, Queen SE, et al. Marked Enteropathy in an Accelerated Macaque Model of AIDS. Am J Pathol 2017;187:589-604. 25. Brocca-Cofano E, Kuhrt D, Siewe B, et al. Pathogenic Correlates of Simian Immunodeficiency Virus- Associated B Cell Dysfunction. J Virol 2017;91. 26. Srinivasan P, Zhang J, Martin A, et al. Safety and Pharmacokinetics of Quick-Dissolving Polymeric Vaginal Films Delivering the Antiretroviral IQP-0528 for Preexposure Prophylaxis. Antimicrob Agents Chemother 2016;60:4140-4150. 27. Srinivasan P, Moss JA, Gunawardana M, et al. Topical Delivery of Tenofovir Disoproxil Fumarate and Emtricitabine from Pod-Intravaginal Rings Protects Macaques from Multiple SHIV Exposures. PloS one 2016;11:e0157061. 28. Peterson CW, Haworth KG, Burke BP, et al. Multilineage polyclonal engraftment of Cal-1 gene- modified cells and in vivo selection after SHIV infection in a nonhuman primate model of AIDS. Molecular therapy Methods & clinical development 2016;3:16007. 29. Pandrea I, Xu C, Stock JL, et al. Antibiotic and Antiinflammatory Therapy Transiently Reduces Inflammation and Hypercoagulation in Acutely SIV-Infected Pigtailed Macaques. PLoS pathogens 2016;12:e1005384. 30. Ortiz AM, Klase ZA, DiNapoli SR, et al. IL-21 and probiotic therapy improve Th17 frequencies, microbial translocation, and microbiome in ARV-treated, SIV-infected macaques. Mucosal Immunol 2016;9:458-467. 31. Richardson M, Mittermeier RA, Rylands AB, et al. Macaca nemestrina . The IUCN Red List of Threatened Species 2008: e.T12555A3356892. 2008; https://dx.doi.org/10.2305/IUCN.UK.2008.RLTS.T12555A3356892.en. . 32. Malaivijitnond S, Arsaithamkul V, Tanaka H, et al. Boundary zone between northern and southern pig-tailed macaques and their morphological differences. 2012;53:377-389. 33. Groves C. Primate : Smithsonian, 2001. 34. Li J, Han K, Xing J, et al. Phylogeny of the macaques (Cercopithecidae: Macaca) based on Alu elements. Gene 2009;448:242-249. 35. Baas T, Baskin CR, Diamond DL, et al. Integrated molecular signature of disease: analysis of influenza virus-infected macaques through functional genomics and proteomics. J Virol 2006;80:10813-10828. 36. Baskin CR, Garcia-Sastre A, Tumpey TM, et al. Integration of clinical data, pathology, and cDNA microarrays in influenza virus-infected pigtailed macaques (Macaca nemestrina). J Virol 2004;78:10420- 10432. 37. Fornek JL, Korth MJ, Katze MG. Use of functional genomics to understand influenza-host interactions. Advances in virus research 2007;70:81-100. 38. Haigwood NL, Montefiori DC, Sutton WF, et al. Passive immunotherapy in simian immunodeficiency virus-infected macaques accelerates the development of neutralizing antibodies. J Virol 2004;78:5983- 5995. 39. Henning T, Fakile Y, Phillips C, et al. Development of a pigtail macaque model of sexually transmitted infection/HIV coinfection using Chlamydia trachomatis, Trichomonas vaginalis, and SHIV(SF162P3). J Med Primatol 2011;40:214-223. 40. Jegaskanda S, Reece JC, De Rose R, et al. Comparison of influenza and SIV specific CD8 T cell responses in macaques. PloS one 2012;7:e32431. 41. Patton DL, Cosgrove Sweeney YT, Paul KJ. A summary of preclinical topical microbicide vaginal safety and chlamydial efficacy evaluations in a pigtailed macaque model. Sex Transm Dis 2008;35:889-897. 42. Patton DL, Sweeney YT, Agnew KJ, et al. Development of a nonhuman primate model for Trichomonas vaginalis infection. Sex Transm Dis 2006;33:743-746. 43. Patton DL, Sweeney YT, Paul KJ. A summary of preclinical topical microbicide rectal safety and efficacy evaluations in a pigtailed macaque model. Sex Transm Dis 2009;36:350-356. 44. Reece JC, Alcantara S, Gooneratne S, et al. Trivalent live attenuated influenza-simian immunodeficiency virus vaccines: efficacy and evolution of cytotoxic T lymphocyte escape in macaques. J Virol 2013;87:4146-4160. 45. Sexton A, De Rose R, Reece JC, et al. Evaluation of recombinant influenza virus-simian immunodeficiency virus vaccines in macaques. J Virol 2009;83:7619-7628. 46. Brennan G, Kozyrev Y, Kodama T, et al. Novel TRIM5 isoforms expressed by Macaca nemestrina. J Virol 2007;81:12210-12217. 47. Liao CH, Kuang YQ, Liu HL, et al. A novel fusion gene, TRIM5-Cyclophilin A in the pig-tailed macaque determines its susceptibility to HIV-1 infection. AIDS 2007;21 Suppl 8:S19-26. 48. Hatziioannou T, Ambrose Z, Chung NP, et al. A macaque model of HIV-1 infection. Proc Natl Acad Sci U S A 2009;106:4425-4429. 49. Hatziioannou T, Del Prete GQ, Keele BF, et al. HIV-1-induced AIDS in monkeys. Science 2014;344:1401-1405. 50. Igarashi T, Iyengar R, Byrum RA, et al. Human immunodeficiency virus type 1 derivative with 7% simian immunodeficiency virus genetic content is able to establish infections in pig-tailed macaques. J Virol 2007;81:11549-11552. 51. Agy MB, Frumkin LR, Corey L, et al. Infection of Macaca nemestrina by human immunodeficiency virus type-1. Science 1992;257:103-106. 52. Garcia-Tellez T, Huot N, Ploquin MJ, et al. Non-human primates in HIV research: Achievements, limits and alternatives. Infect Genet Evol 2016;46:324-332. 53. Kimata JT, L. K, D. A, et al. Emerging cytopathic and antigenic SIV variants influce AIDS progression. Nature Medicine 1999;5:535-541. 54. Kinman LM, Worlein JM, Leigh J, et al. HIV in central nervous system and behavioral development: an HIV-2287 macaque model of AIDS. AIDS 2004;18:1363-1370. 55. Kuller L, Benveniste RE, Tsai CC, et al. Intrarectal inoculation of macaques by the simian immunodeficiency virus, SIVmne E11S: CD4+ depletion and AIDS. J Med Primatol 1994;23:397-409. 56. Kuller L, Benveniste RE, Watanabe R, et al. Transmission of SIVMne from female to male Macaca nemestrina. J Med Primatol 1992;21:299-307. 57. McClure J, Schmidt AM, Rey-Cuille MA, et al. Derivation and characterization of a highly pathogenic isolate of human immunodeficiency virus type 2 that causes rapid CD4+ cell depletion in Macaca nemestrina. J Med Primatol 2000;29:114-126. 58. Gardner MB, Luciw PA. Macaque models of human infectious disease. ILAR J 2008;49:220-255. 59. Jegaskanda S, Amarasena TH, Laurie KL, et al. Standard trivalent influenza virus protein vaccination does not prime antibody-dependent cellular cytotoxicity in macaques. J Virol 2013;87:13706-13718. 60. Patton DL, Teng A, Randall A, et al. Whole genome identification of C. trachomatis immunodominant antigens after genital tract infections and effect of antibiotic treatment of pigtailed macaques. Journal of proteomics 2014;108:99-109. 61. Shen Y, Shen L, Sehgal P, et al. Clinical latency and reactivation of AIDS-related mycobacterial infections. J Virol 2004;78:14023-14032. 62. Wiseman RW, Karl JA, Bimber BN, et al. Major histocompatibility complex genotyping with massively parallel pyrosequencing. Nat Med 2009;15:1322-1326. 63. Hadzic SV, Wang X, Dufour J, et al. Comparison of the vaginal environment of Macaca mulatta and Macaca nemestrina throughout the menstrual cycle. American journal of reproductive immunology 2014;71:322-329. 64. Patton DL, Sweeney YC, Rabe LK, et al. The vaginal microflora of pig-tailed macaques and the effects of chlorhexidine and benzalkonium on this ecosystem. Sex Transm Dis 1996;23:489-493. 65. Butler K, Ritter JM, Ellis S, et al. A Depot Medroxyprogesterone Acetate Dose That Models Human Use and Its Effect on Vaginal SHIV Acquisition Risk. J Acquir Immune Defic Syndr 2016;72:363-371. 66. Dietz Ostergaard S, Butler K, Ritter JM, et al. A combined oral contraceptive affects mucosal SHIV susceptibility factors in a pigtail macaque (Macaca nemestrina) model. J Med Primatol 2015;44:97-107. 67. Engel RM, Morris M, Henning T, et al. Evaluation of pigtail macaques as a model for the effects of copper intrauterine devices on HIV infection. J Med Primatol 2014;43:349-359. 68. Henning TR, Butler K, Hanson D, et al. Increased susceptibility to vaginal simian/human immunodeficiency virus transmission in pig-tailed macaques coinfected with Chlamydia trachomatis and Trichomonas vaginalis. J Infect Dis 2014;210:1239-1247. 69. Kersh EN, Henning T, Vishwanathan SA, et al. SHIV susceptibility changes during the menstrual cycle of pigtail macaques. J Med Primatol 2014;43:310-316. 70. McNicholl JM, Henning TC, Vishwanathan SA, et al. Non-human primate models of hormonal contraception and HIV. American journal of reproductive immunology 2014;71:513-522. 71. Radzio J, Hanley K, Mitchell J, et al. Physiologic doses of depot-medroxyprogesterone acetate do not increase acute plasma simian HIV viremia or mucosal virus shedding in pigtail macaques. Aids 2014;28:1431-1439. 72. Mitchell T, MacDonald JW, Srinouanpranchanh S, et al. Evidence of cardiac involvement in the fetal inflammatory response syndrome: disruption of gene networks programming cardiac development in nonhuman primates. Am J Obstet Gynecol 2018;218:438 e431-438 e416. 73. Adams Waldorf KM, Nelson BR, Stencel-Baerenwald JE, et al. Congenital Zika virus infection as a silent pathology with loss of neurogenic output in the fetal brain. Nat Med 2018;24:368-374. 74. McAdams RM, Bierle CJ, Boldenow E, et al. Choriodecidual Group B Streptococcal Infection Induces miR-155-5p in the Fetal Lung in Macaca nemestrina. Infect Immun 2015;83:3909-3917. 75. Adams Waldorf KM, Singh N, Mohan AR, et al. Uterine overdistention induces preterm labor mediated by inflammation: observations in pregnant women and nonhuman primates. Am J Obstet Gynecol 2015;213:830 e831-830 e819. 76. Vanderhoeven JP, Bierle CJ, Kapur RP, et al. Group B streptococcal infection of the choriodecidua induces dysfunction of the cytokeratin network in amniotic epithelium: a pathway to membrane weakening. PLoS pathogens 2014;10:e1003920. 77. Adams Waldorf KM, McAdams RM. Influence of infection during pregnancy on fetal development. Reproduction 2013;146:R151-162. 78. Adams Waldorf KM, Gammill HS, Lucas J, et al. Dynamic changes in fetal microchimerism in maternal peripheral blood mononuclear cells, CD4+ and CD8+ cells in normal pregnancy. Placenta 2010;31:589- 594. 79. Adams Waldorf KM, Stencel-Baerenwald JE, Kapur RP, et al. Fetal brain lesions after subcutaneous inoculation of Zika virus in a pregnant nonhuman primate. Nat Med 2016;22:1256-1259. 80. Peeples ES, Ezeokeke CK, Juul SE, et al. Evaluating a Targeted Bedside Measure of Cerebral Perfusion in a Nonhuman Primate Model of Neonatal Hypoxic-Ischemic Encephalopathy. Journal of ultrasound in medicine : official journal of the American Institute of Ultrasound in Medicine 2018;37:913-920. 81. McAdams RM, McPherson RJ, Kapur RP, et al. Focal Brain Injury Associated with a Model of Severe Hypoxic-Ischemic Encephalopathy in Nonhuman Primates. Developmental neuroscience 2017;39:107- 123. 82. McAdams RM, Fleiss B, Traudt C, et al. Long-Term Neuropathological Changes Associated with Cerebral Palsy in a Nonhuman Primate Model of Hypoxic-Ischemic Encephalopathy. Developmental neuroscience 2017;39:124-140. 83. Juul SE, Mayock DE, Comstock BA, et al. Neuroprotective potential of erythropoietin in neonates; design of a randomized trial. Matern Health Neonatol Perinatol 2015;1:27. 84. Chun PT, McPherson RJ, Marney LC, et al. Serial plasma metabolites following hypoxic-ischemic encephalopathy in a nonhuman primate model. Developmental neuroscience 2015;37:161-171. 85. Traudt CM, McPherson RJ, Bauer LA, et al. Concurrent erythropoietin and hypothermia treatment improve outcomes in a term nonhuman primate model of perinatal asphyxia. Developmental neuroscience 2013;35:491-503. 86. Jacobson Misbe EN, Richards TL, McPherson RJ, et al. Perinatal asphyxia in a nonhuman primate model. Developmental neuroscience 2011;33:210-221. 87. Juul SE, Aylward E, Richards T, et al. Prenatal cord clamping in newborn Macaca nemestrina: a model of perinatal asphyxia. Developmental neuroscience 2007;29:311-320. 88. Radtke S, Perez AM, Venkataraman R, et al. Preparation and Gene Modification of Nonhuman Primate Hematopoietic Stem and Progenitor Cells. J Vis Exp 2019. 89. Humbert O, Peterson CW, Norgaard ZK, et al. A Nonhuman Primate Transplantation Model to Evaluate Hematopoietic Stem Cell Gene Editing Strategies for beta-Hemoglobinopathies. Molecular therapy Methods & clinical development 2018;8:75-86. 90. Radtke S, Adair JE, Giese MA, et al. A distinct hematopoietic stem cell population for rapid multilineage engraftment in nonhuman primates. Sci Transl Med 2017;9. 91. Gori JL, Butler JM, Kunar B, et al. Endothelial Cells Promote Expansion of Long-Term Engrafting Marrow Hematopoietic Stem and Progenitor Cells in Primates. Stem Cells Transl Med 2017;6:864-876. 92. Chiarelli PA, Revia RA, Stephen ZR, et al. Nanoparticle Biokinetics in Mice and Nonhuman Primates. ACS Nano 2017;11:9514-9524. 93. Watts KL, Beard BC, Wood BL, et al. No evidence of clonal dominance after transplant of HOXB4- expanded cord blood cells in a nonhuman primate model. Experimental hematology 2014;42:497-504. 94. Chandrasekaran D, Nakamoto B, Watts KL, et al. Modeling promising nonmyeloablative conditioning regimens in nonhuman primates. Human gene therapy 2014;25:1013-1022. 95. Peterson CW, Younan P, Polacino PS, et al. Robust suppression of env-SHIV viremia in Macaca nemestrina by 3-drug ART is independent of timing of initiation during chronic infection. J Med Primatol 2013;42:237-246. 96. Watts KL, Nelson V, Wood BL, et al. Hematopoietic stem cell expansion facilitates multilineage engraftment in a nonhuman primate cord blood transplantation model. Experimental hematology 2012;40:187-196. 97. Gori JL, Chandrasekaran D, Kowalski JP, et al. Efficient generation, purification, and expansion of CD34(+) hematopoietic progenitor cells from nonhuman primate-induced pluripotent stem cells. Blood 2012;120:e35-44. 98. Zhong B, Trobridge GD, Zhang X, et al. Efficient generation of nonhuman primate induced pluripotent stem cells. Stem cells and development 2011;20:795-807. 99. Trobridge GD, Kiem HP. Large animal models of hematopoietic stem cell gene therapy. Gene therapy 2010;17:939-948. 100. Trobridge GD, Wu RA, Beard BC, et al. Protection of stem cell-derived lymphocytes in a primate AIDS gene therapy model after in vivo selection. PloS one 2009;4:e7693. 101. Munoz NM, Trobridge GD, Kiem HP. Ex vivo expansion and lentiviral transduction of Macaca nemestrina CD4+ T cells. J Med Primatol 2009;38:438-443. 102. Trobridge GD, Beard BC, Gooch C, et al. Efficient transduction of pigtailed macaque hematopoietic repopulating cells with HIV-based lentiviral vectors. Blood 2008;111:5537-5543. 103. Shepherd BE, Kiem HP, Lansdorp PM, et al. Hematopoietic stem-cell behavior in nonhuman primates. Blood 2007;110:1806-1813. 104. Trobridge G, Beard BC, Kiem HP. Hematopoietic stem cell transduction and amplification in large animal models. Human gene therapy 2005;16:1355-1366. 105. Schmidt M, Glimm H, Wissler M, et al. Efficient characterization of retro-, lenti-, and foamyvector- transduced cell populations by high-accuracy insertion site sequencing. Annals of the New York Academy of Sciences 2003;996:112-121. 106. Schmidt M, Zickler P, Hoffmann G, et al. Polyclonal long-term repopulating stem cell clones in a primate model. Blood 2002;100:2737-2743. 107. Berger C, Huang ML, Gough M, et al. Nonmyeloablative immunosuppressive regimen prolongs In vivo persistence of gene-modified autologous T cells in a nonhuman primate model. J Virol 2001;75:799- 808. 108. Liu YW, Chen B, Yang X, et al. Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat Biotechnol 2018;36:597-605. 109. Chong JJ, Yang X, Don CW, et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 2014;510:273-277. 110. Chong JJ, Murry CE. Cardiac regeneration using pluripotent stem cells--progression to large animal models. Stem Cell Res 2014;13:654-665. 111. Zanos S, Rembado I, Chen D, et al. Phase-Locked Stimulation during Cortical Beta Oscillations Produces Bidirectional Synaptic Plasticity in Awake Monkeys. Curr Biol 2018;28:2515-2526 e2514. 112. Seeman SC, Mogen BJ, Fetz EE, et al. Paired Stimulation for Spike-Timing-Dependent Plasticity in Primate Sensorimotor Cortex. The Journal of neuroscience : the official journal of the Society for Neuroscience 2017;37:1935-1949. 113. Rembado I, Zanos S, Fetz EE. Cycle-Triggered Cortical Stimulation during Slow Wave Sleep Facilitates Learning a BMI Task: A Case Report in a Non-Human Primate. Front Behav Neurosci 2017;11:59. 114. Libey T, Fetz EE. Open-Source, Low Cost, Free-Behavior Monitoring, and Reward System for Neuroscience Research in Non-human Primates. Front Neurosci 2017;11:265. 115. Lajoie G, Krouchev NI, Kalaska JF, et al. Correlation-based model of artificially induced plasticity in motor cortex by a bidirectional brain-computer interface. PLoS computational biology 2017;13:e1005343. 116. Eaton RW, Libey T, Fetz EE. Operant conditioning of neural activity in freely behaving monkeys with intracranial reinforcement. Journal of neurophysiology 2017;117:1112-1125. 117. Casimo K, Levinson LH, Zanos S, et al. An interspecies comparative study of invasive electrophysiological functional connectivity. Brain Behav 2017;7:e00863. 118. Wander JD, Sarma D, Johnson LA, et al. Cortico-Cortical Interactions during Acquisition and Use of a Neuroprosthetic Skill. PLoS computational biology 2016;12:e1004931. 119. Zaidi AD, Munk MH, Schmidt A, et al. Simultaneous epidural functional near-infrared spectroscopy and cortical electrophysiology as a tool for studying local neurovascular coupling in primates. NeuroImage 2015;120:394-399. 120. Milovanovic I, Robinson R, Fetz EE, et al. Simultaneous and independent control of a brain- computer interface and contralateral limb movement. Brain Comput Interfaces (Abingdon) 2015;2:174- 185. 121. Fetz EE. Restoring motor function with bidirectional neural interfaces. Prog Brain Res 2015;218:241-252. 122. Wander JD, Blakely T, Miller KJ, et al. Distributed cortical during learning of a brain- computer interface task. Proc Natl Acad Sci U S A 2013;110:10818-10823. 123. Nishimura Y, Perlmutter SI, Fetz EE. Restoration of upper limb movement via artificial corticospinal and musculospinal connections in a monkey with spinal cord injury. Front Neural Circuits 2013;7:57. 124. Nishimura Y, Perlmutter SI, Eaton RW, et al. Spike-timing-dependent plasticity in primate corticospinal connections induced during free behavior. Neuron 2013;80:1301-1309. 125. Lucas TH, Fetz EE. Myo-cortical crossed feedback reorganizes primate motor cortex output. The Journal of neuroscience : the official journal of the Society for Neuroscience 2013;33:5261-5274. 126. Zanos S, Richardson AG, Shupe L, et al. The Neurochip-2: an autonomous head-fixed computer for recording and stimulating in freely behaving monkeys. IEEE transactions on neural systems and rehabilitation engineering : a publication of the IEEE Engineering in Medicine and Biology Society 2011;19:427-435. 127. Moritz CT, Fetz EE. Volitional control of single cortical neurons in a brain-machine interface. Journal of neural engineering 2011;8:025017. 128. Pallus A, Walton MMG, Mustari M. Activity of near-response cells during disconjugate saccades in strabismic monkeys. Journal of neurophysiology 2018;120:2282-2295. 129. Walton MMG, Pallus A, Fleuriet J, et al. Neural mechanisms of oculomotor abnormalities in the infantile strabismus syndrome. Journal of neurophysiology 2017;118:280-299. 130. Mustari MJ. Nonhuman Primate Studies to Advance Vision Science and Prevent Blindness. ILAR J 2017;58:216-225. 131. Willoughby CL, Christiansen SP, Mustari MJ, et al. Effects of the sustained release of IGF-1 on extraocular muscle of the infant non-human primate: at the effector organ level. Invest Ophthalmol Vis Sci 2012;53:68-75.