Next Article in Journal
Comparative Longitudinal Serological Study of Anti-SARS-CoV-2 Antibody Profiles in People with COVID-19
Next Article in Special Issue
Using Cryopreserved Plasmodium falciparum Sporozoites in a Humanized Mouse Model to Study Early Malaria Infection Processes and Test Prophylactic Treatments
Previous Article in Journal
Prevalence and Association of Campylobacter spp., Salmonella spp., and Blastocystis sp. in Poultry
Previous Article in Special Issue
A Comparison of Lymphoid and Myeloid Cells Derived from Human Hematopoietic Stem Cells Xenografted into NOD-Derived Mouse Strains
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Advances in Humanized Mouse Models for Studying NK Cells and HIV Infection

by
Jocelyn T. Kim
1,*,
Gabrielle Bresson-Tan
1 and
Jerome A. Zack
2,3
1
Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
2
Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
3
Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(8), 1984; https://doi.org/10.3390/microorganisms11081984
Submission received: 30 May 2023 / Revised: 28 July 2023 / Accepted: 1 August 2023 / Published: 2 August 2023
(This article belongs to the Special Issue Humanised Mouse Models: Recent Advances in Human Infectious Diseases)

Abstract

:
Human immunodeficiency virus (HIV) has infected millions of people worldwide and continues to be a major global health problem. Scientists required a small animal model to study HIV pathogenesis and immune responses. To this end, humanized mice were created by transplanting human cells and/or tissues into immunodeficient mice to reconstitute a human immune system. Thus, humanized mice have become a critical animal model for HIV researchers, but with some limitations. Current conventional humanized mice are prone to death by graft versus host disease induced by the mouse signal regulatory protein α and CD47 signaling pathway. In addition, commonly used humanized mice generate low levels of human cytokines required for robust myeloid and natural killer cell development and function. Here, we describe recent advances in humanization procedures and transgenic and knock-in immunodeficient mice to address these limitations.

1. Introduction

There are approximately 38 million people currently living with HIV/AIDS worldwide. Anti-retroviral therapy (ART) suppresses HIV replication in people living with HIV/AIDS for years such that viremia is largely undetectable using standard clinical assays [1,2,3]. However, ART is not curative due to the latent viral reservoir. HIV infects and stably integrates a full-length copy of the replication-competent viral genome into the host chromosome of target cells such as CD4+ T cells and macrophages [4]. The most well-defined and prominent ‘latent reservoir’ is within memory CD4+ T cells [5,6]. During HIV infection, a subset of these infected CD4+ T cells avert virus-induced death and transition into memory cells [5,6]. Because memory cells are by nature long-lived and quiescent, these infected cells can persist for decades with their silenced proviral cargo, only to later produce an infectious virus if ART is discontinued [7,8,9]. Due to their longevity, latently infected cells decay very slowly during ART [5]. In addition, recent studies profiling HIV-1 integration sites demonstrated that infected cell clones with identical integration sites could persist for more than 11 years and proliferate over time [10,11]. These breakthrough studies demonstrated that despite effective ART, the latent reservoir can proliferate and increase over time.
In vitro experiments involving cell lines, primary cells, and primary tissues have been critical to understanding HIV biology. Ex vivo organoid models have emerged as a valuable alternative to study HIV neuropathology [12] due to the limited availability of human primary neuronal cells and post-mortem tissue. However, it can be difficult to study HIV pathogenesis and complex immune interactions with only in vitro or ex vivo systems. The most common in vivo models for HIV research utilize humanized mice or non-human primates (NHP). Because NHP are physiologically and immunologically similar to humans, the simian immunodeficient virus (SIV) NHP model [13] has been important for the preclinical evaluation of drugs, vaccines and gene-therapies against HIV, and the study of lentiviral immunity in vivo. However, NHP studies can be costly, lengthy, and vulnerable to monkey shortages [14], making their common use prohibitive. An alternative in vivo model involves the transplantation or introduction of various human immune tissues or cells into immunodeficient mice [15]. Due to the human species tropism of HIV, humanized mice are a widely used small animal model for HIV research. Over the past several decades, different immune-deficient mouse strains have been developed and tested for human immune engraftment and are reviewed elsewhere [15,16,17]. R5 and X4 tropic HIV isolates replicated efficiently in humanized mice (as reviewed previously [18]) and were suppressed by various regimens of ART administered via intraperitoneal injections [19,20,21,22], subcutaneous injection [23,24,25], drinking water [21,26], or animal feed [23,24,27,28,29]. Latent HIV reservoirs were found in various anatomic sites in the infected humanized mice [20,21,26,27,30], with evidence of viral rebound following ART interruption, similar to the rebound viremia seen in people living with HIV if ART is discontinued. This review will outline recent advances in humanized mouse models in the field of HIV research with a particular focus on improving natural killer (NK) cell reconstitution.

2. Commonly Used Humanized Mouse Models

2.1. NSG, NRG, NOG, BRG Mice

The most common approach to creating a humanized mouse model is transferring human immune cells or tissue into immunodeficient mice. This allows for the engraftment of functional human immune cells in the murine context to study various human infections. The most widely used immunodeficient mice are NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG), NOD.Cg-Rag1tm1Mom Il2rgtm1Wjl/SzJ (NRG), NOD.Cg-Prkdcscid Il2rgtm1Sug/JicTac (NOG), and C;129S4-Rag2tm1.1Flv Il2rgtm1.1Flv/J (BRG) as listed in Table 1 and Supplementary Table S1. These mice harbor mutations in either the recombination activating gene (Rag) or protein kinase catalytic polypeptide (Prkdcscid) resulting in the loss of endogenous mouse T cells and B cells. The additional mutation in the IL2RG, which encodes the interleukin-2 receptor subunit γ (IL2rγ), results in deficiencies in multiple cytokines signaling pathways such as IL-2, IL-4, IL-7, IL-9, and IL-15. The loss of murine IL-15 signaling leads to poor murine NK cell development. Thus, the disruption in IL2rγ signaling allows for an increase in tolerance to xenotransplantation. In addition, these mice are on a NOD background, which have inherent deficiencies in mouse macrophage, dendritic cell, NK cell, and the complementary system. The most common approaches to humanization of mice are detailed in this review.

2.2. HuPBL and PDX-NSG Mice

The first model of humanized mice was generated via injection of human peripheral blood lymphocytes (huPBL) into immunodeficient SCID (C.B-17scid) mice over 35 years ago [33]. Human lymphocytes quickly engrafted as soon as two hours after PBL injection and demonstrated an activated phenotype [33]. Given that huPBL-SCID mice were rapidly engrafted with mature human CD4+ T cells, HIV could be administered two hours after PBL injection resulting in robust viral replication and CD4+ T cell decline in vivo. In a later variation of this model, human peripheral lymphocytes from adult donors were injected intraperitoneally into immunodeficient NSG mice (huPBL-NSG), which had an additional IL2RG mutation, leading to improved human immune engraftment [31,32], likely due to the lack of murine NK cells that could inhibit xenogeneic cell engraftment. Human lymphocytes expanded in the huPBL-NSG mouse efficiently and permitted HIV replication in vivo for extended periods of time. However, human myeloid and NK cells lacked proliferative capacity resulting in low reconstitution and contributed to minimal to absent primary immune responses.
Recently, human lymphocytes containing latently infected cells from long-term ART-suppressed patients were used to humanize NSG mice to generate patient-derived xenograft (PDX) mice [52]. These mice developed viremia 2 weeks after PBL injection, indicating the rapid reactivation of proviruses and an in vivo model to study the viral reservoir. However, these humanized mice quickly succumbed to graft versus host disease (GvHD). The rapid development of GvHD in huPBL mice is caused by the xenogeneic reaction of human cells toward mouse tissues, resulting in severe morbidity and mortality, thus limiting their use for long-term in vivo studies.
Interestingly, studies in human patients undergoing allogeneic stem cell transplantation noted that the engraftment of human naïve CD4+ T cells, but not CD4+ memory T cells, were triggering GvHD [53,54,55]. To this end, CD4+ memory T cells from ART suppressed patients were transferred into NSG mice to create patient-derived xenograft (PDX) mice. Importantly, these PDX mice could be followed for approximately eleven weeks without developing GvHD [35]. In addition, approximately eight weeks after naïve CD4+ T cell injection, viremia was detected indicating the reactivation of proviruses from a viral reservoir. In addition, graft-autologous HIV antigen-specific CD8 T cells clones demonstrated viral control, followed by the subsequent generation of viral escape mutations. Thus, the PDX mice transplanted with human naïve CD4+ T cells from ART suppressed patients offers a way for future studies to investigate the viral reservoir after ART interruption and antigen-specific immune responses.

2.3. HuCD34 Mice

Another model of humanized mice is HuCD34 mice, which consists of human CD34+ hematopoietic stem cells (HSCs) injected into immunodeficient mice (e.g., NSG, NRG, NOG, and BRG). These CD34+ HSCs can be processed from human fetal liver or non-fetal human sources such as umbilical cord blood (UCB), adult bone marrow, or granulocyte colony stimulating factor (G-CSF) mobilized peripheral blood stem cells. Varying levels of lymphoid, myeloid, and erythroid lineages may develop depending on the cell source utilized [36]. CD34+ HSCs are most commonly injected intravenously into adult mice or intra-hepatically into newborn mice [37,38]. Prior to transplantation, immunodeficient mice are often conditioned with myeloablative sub-lethal irradiation or chemotherapy (e.g., busulfan) prior to transplantation. Transplanted human CD34+ HSCs have differentiated into functional human T and B cells allowing for efficient HIV replication in vivo [38,39,40,41] via rectal or vaginal mucosal challenges [56]. In addition, HuCD34-NOG mice were capable of anti-HIV IgG responses, but with limited breadth to HIV antigens [42]. Additional advances have improved antigen-specific IgG responses by matching human leukocyte antigen (HLA) class I and class II molecules between human HSCs and human HLA-matched transgenic mice (reviewed in [57]).

2.4. NSG-BLT Mice

A third method for humanization involves the surgical implantation of matching human fetal thymus and liver fragments as well as injection of matched CD34+ HSCs into conditioned immunodeficient adult mice to create bone–liver–thymus (BLT) mice [30,43,44,58,59,60]. In some BLT mice, matching human fetal thymus and fetal liver-derived CD34+ HSCs are used. The BLT-NSG mouse is one of the most commonly used mouse models in HIV research due to the efficient human multi-lineage immune engraftment. The fetal human HSCs engraft and populate the mouse bone marrow, serving as progenitor cells for human lymphoid and myeloid cells. Multi-lineage reconstitution of human T cells, B cells, and myeloid cells was found throughout the mice including the human thymus and murine liver, bone marrow, thymus, spleen, lymph nodes, lungs, female reproductive tract, and gut [30,43,44,60,61,62]. NSG-BLT mice were susceptible to HIV infection via rectal and vaginal HIV challenges [46,47,62,63,64] despite lacking immune aggregates such as gut associated lymphoid tissue [65]. Because the human T cells were educated upon the background of the human thymus tissue, BLT mice demonstrated increased T cell engraftment and anti-HIV specific CD8+ T cell responses [43,45,59]. The BLT mice also demonstrated B cells, albeit mostly immature and minimal memory B cells with limited class-switching [66,67]. HIV-specific human antibodies, particularly human IgM, have been elicited after HIV infection [43,68] or immunization [69,70,71]. However, achieving robust antigen-specific humoral vaccination in BLT mice that recapitulate healthy adult human immunization responses has been difficult. Engraftment of human NK cells has been low [72], but they demonstrate in vivo function. Recently, tissue-specific functional memory NK cells were isolated from the liver of BLT-immunized mice [73]. Despite their limitations, NSG-BLT mice have been used extensively to test anti-HIV drugs, antibodies, and cellular therapies (reviewed in [15,17,74]).

2.5. HuCD34-SRG, HuPBL-SRG, HuCD34-TKO, and TKO-BLT Mice

Humanized mice models utilizing the NSG, NRG, NOG, and BRG mouse strains can be restricted in their experimental timelines due to the development of GvHD [75]. Although there is biological variation between human donors and mouse series, the mice will begin to succumb to GvHD by ~20 weeks transplantation of human CD34+ HSCs or tissues. Signal regulatory protein α (SIRPα) is an immunoglobulin superfamily receptor expressed on myeloid cells and is activated by allogeneic or xenogeneic interactions with non-self CD47 [76,77]. Human CD47 has a low affinity for mouse SIRPα, which results in the phagocytosis of transplanted human cells by mouse macrophages and the induction of GvHD. Interestingly, SIRPα is polymorphic between different strains of mice, and the mouse SIRPα in B6.129S and C57BL/6 mice does not cross-react sufficiently with human CD47. To this end, efficient human immune cell reconstitution and a dramatic reduction in GvHD was achieved in humanized SRG and TKO mice, which altered the CD47/SIRPα anti-phagocytic signaling pathway (reviewed in [78]). By knocking in human SIRPA to generate SIRPAh/m Rag2−/− Il2rg−/− (SRG) mice, huCD34-SRG mice achieved robust humanization and lived for 9 months without succumbing to GvHD [79]. In addition, CD47 was knocked out of mice deficient in Rag1 and IL-2Rγ to generate B6.129S-Rag2tm1FwaCD47tm1FplIl2rgtm1Wjl/J TKO mice. Transplantation of human PBLs led to efficient T cell engraftment and HIV infection in vivo [50]. Importantly, by knocking out CD47, these huPBL mice demonstrated a 24-day delay to the onset of mild GvHD compared to huPBL-NSG mice. In addition, human fetal tissue and autologous CD34+ HSCs were transplanted into TKO mice deficient in Rag2, IL2rg, and CD47 (C57BL/6 Rag2−/−γc−/−CD47−/−) to create TKO-BLT mice. Due to the knockout of mouse CD47 in these TKO mice, excellent and stable human immune cell engraftment was observed with little to no GVHD for up to 45 weeks after transplantation [23,51,72,80]. Indeed, these TKO-BLT mice demonstrated HIV infection and efficient suppression of viremia with ART over extended periods, which was followed by rebound viremia after ART interruption, indicating the establishment of a latent HIV reservoir in vivo. Thus, TKO-BLT mice may be placed on ART for prolonged periods, allowing for reservoir studies, which often require extended timelines [72].

2.6. NeoThy-NSG or NSG(W) Mice

Due to recent increased restrictions on abortion and previous changes on policies in the U.S. related to human fetal tissue research [81], the ability to efficiently source human fetal tissue to generate BLT mice may be affected at some research institutions in the future. Human non-fetal UCB CD34+ HSCs continues to be available to generate humanized mice. However, the disadvantage of using UCB is the variable and low number of CD34+ HSCs collected from a single donor unit. Thus, neonatal human thymic tissue was utilized to humanize NSG or NOD,B6.SCID Il2rγ−/−KitW41/W41 (NSG-W) mice with autologous or allogeneic UCB-derived CD34+ HSCs to generate NeoThy mice [48]. NeoThy mice had equivalent levels of human immune subsets including T cells compared to mice humanized with human fetal thymus and allogeneic UCB CD34+ HSCs. Further investigations are warranted to assess if these mice are efficiently infected with HIV and can establish a stable latent reservoir.

3. Improved Mouse Models for HIV-1 and NK Cell Research

The common and recently used mouse strains in Table 1 do not generate human cytokines and other factors such as chemokines. Although murine cells produce certain mouse cytokines such as IL-7 [82] or IL-12 [83], which can signal to human cytokine receptors, the levels of these murine cytokine levels are minimal or absent in mice with mutations in Rag1, Prkdcscid, and IL2rγ such as NSG mice. Other murine cytokines such as IL-2 [84], IL-3 [85], IL-4 [84], IL-15 [86], granulocyte–macrophage colony-stimulating factor GM-CSF [87], and macrophage colony-stimulating factor (M-CSF) [85] have little to no activity on human cells. Although transplanted human HSCs differentiate into lymphoid cells in humanized mice, reconstitution of innate immune cells is needed to help generate human cytokines and develop a more mature immune environment. Insufficient cross-species reactivity and biological function between mouse factors and their corresponding human receptors and minimal human innate immune engraftment are major factors limiting the development of a fully functional human immune system in humanized mice.
NK cells are innate immune effectors capable of intrinsically recognizing and clearing virally infected cells through multiple mechanisms. Epidemiological and genetic studies have shown NK cell interactions with self-HLA molecules are involved in recognition of HIV-infected cells and may slow disease progression, reduce viral setpoint, or mediate immune pressure [88,89,90,91,92,93,94,95,96]. IL-15 is a critical cytokine required for NK cell and T cell development, maintenance, and function. Myeloid cells can also contribute to NK cell development by providing a richer cytokine environment. However, human NK cell development and function is poor in the humanized mouse models described so far in Table 1. Due to the low level of human cytokine production, particularly IL-15, lack of cross-species IL-15 reactivity, and insufficient myeloid cell engraftment, the frequency of NK cells in HuCD34-NSG [97], NSG-BLT [73], and TKO-BLT [72,73] mice were substantially low (~1% or less of human CD45+ in the blood) compared to the expected frequency of NK cells in human peripheral blood from healthy adult donors (~5–20% of PBMCs). Although there were overall low levels of human NK cells in NSG-BLT mice, liver-specific NK cells, but not spleen or bone marrow-derived NK cells, were capable of mediating vaccination-dependent and antigen-specific killing ex vivo [73]. In addition, an injection of an IL-15 superagonist [98,99] enhanced the human NK cells present in huPBL-mice, thereby inhibiting HIV replication in vivo [100] (Figure 1). Interestingly, administration of the IL-15 superagonist also transiently reduced viral replication in SIV-infected macaques [101].
The low human NK cell engraftment in HuCD34 and BLT mice makes them an ideal model to study the effect of adoptively transferred exogenous NK cells. Although NSG-BLT mice supported transient engraftment of exogenously administered human peripheral NK cells, these NK cells inhibited viral replication in vivo [72]. In addition, a kick and kill combination, comprising a PKC modulator as a latency reversing agent (LRA) and allogenic transfusions of human peripheral NK cells from healthy donors as the killing agent, was tested in TKO-BLT mice, which were suitable for in vivo reservoir studies due to their increased longevity. The LRA and NK cell combination was administered during ART suppression in HIV-infected TKO-BLT mice and delayed viral rebound after ART interruption, reduced number of rebounding viral clones, and eliminated the reservoir in a subset of infected animals [72]. This suggests that the addition of NK cells as a “kick” step in kick and kill approaches to eliminate the HIV reservoir may be efficacious (Figure 1). However, one caveat to this approach is that a subset of NK cells can express CD4, which could make them susceptible to HIV infection [102,103,104].
To increase human NK cell reconstitution in humanized mice, multiple approaches have been explored. Direct injections of recombinant human IL-15 cytokine complexed with the IL-15 receptor α led to efficient human NK cell engraftment in humanized BALB/c Rag2−/−γc−/− mice with CD34+ HSCs [105]. HuCD34-BRG mice administered with exogenous human Flt3 ligand were shown to increase human myeloid and NK cell engraftment and function, suggesting an improvement in human myeloid engraftment may provide a richer human cytokine environment for human NK cell proliferation [106]. However, these exogenous human cytokine injections provided transient levels requiring repeated injections and supra-physiologic, systemic concentrations. Hydrodynamic injection of plasmid DNA encoding human IL-15 [107] or cytokine superagonist [100] was simplified to a single administration, but still led to physiologically high cytokine levels or NK cell stimulation. Other modes of long-term cytokine vector-mediated cytokine expression have been regulated by a strong human cytomegalovirus (CMV) promoter and enhancer sequence [108,109], which was associated with supra-physiologic levels of cytokines. Finally, transgenic or knock-in mice were developed, in which promoter and regulatory sequences were inserted into the mouse genome to induce more physiological expression of human cytokines. An improvement in human NK cell engraftment in humanized mice was achieved in the following transgenic and knock-in strains by expressing human IL-15 and/or improving engraftment of human myeloid cells, which express and trans-present IL-15 thereby promoting human NK cell reconstitution (Table 2 and Supplementary Table S1). Importantly, these recent humanized transgenic mice developed human NK cells that inhibited HIV-1 replication [84] and utilized antibody-dependent cellular toxicity (ADCC) via a broadly neutralizing antibody PGT121 to suppress infection [97] or CD4-induced antibodies and CD4 mimetic to control viral rebound from the reservoir [97,110] (Figure 1). Altogether, these studies demonstrate the importance of enhancing NK cell function in humanized mice.

3.1. NSG-Tg (IL-15) Mice

NOD.Cg-Prkdcscid Il2rgtm1Wjl Tg (IL15)1Sz/SzJ (NSG-Tg (IL-15)) mice expressed human IL-15 via the BAC transgenic system, in which the entire human locus including the promoter and flanking regulatory sequences were inserted into the mouse genome leading to more physiological expression. NSG-Tg (IL-15) mice were found to produce physiological serum levels of human IL-15 [111]. The injection of human PBL in these mice resulted in persistent human T, B and NK cell engraftment [112]. In addition, NSG-Tg (IL-15) mice humanized with human CD34+ HSCs supported robust and long-term human NK cell development, survival, and reconstitution in the blood, liver, spleen, bone marrow, and GI tract [119]. Additionally, splenic NK cells from HuCD34-NSG-Tg (IL-15) mice displayed significant and robust cytotoxic ex vivo function against the target K562 cell line compared to splenic NK cells from HuCD34-NSG mice. NK cells from HIV-infected HuCD34-NSG-Tg (IL-15) mice were functional with increased expression of IFN-γ, TNF-α, perforin, Ki67, and CD107a (Lamp-1)) compared to uninfected animals. The HuCD34-NSG-Tg (IL-15) mice did not develop GvHD for 6–9 months post-transplant, thus may be a suitable long-term model to study the effects of NK cells on the HIV reservoir.

3.2. NOG-EXL and NSG-SGM3 Mice

Recently developed transgenic mice have constitutively high levels of human IL-3 and GM-CSF to increase human myeloid and NK cell development. The transgenic NOG-EXL mice (NOD.Cg-Prkdcscid Il2rgtm1Sug Tg (SV40/HTLV-IL3,CSF2)10-7Jic/JicTac) ubiquitously overexpressed human IL3 and CSF2 under the control of the SRα promoter [114]. HuCD34-NOG-EXL mice demonstrated robust engraftment of human CD45+, T, and B cells, and particularly myeloid cells compared to control humanized non-transgenic mice [113,114]. Human NK cell engraftment was modest and <1% of CD45+ cells in HuCD34-NOG-EXL mice [114]. Acute HIV infection led to a reduction of CD4+ T-cells, inversion of the CD4:CD8 ratio, and a reduction of peripheral CD56bright NK cells, which was similar to patterns found during natural human infection [113].
NSG-SGM3 (NOD.Cg-Prkdcscid Il2rgtmWjl Tg (CMV-IL3,CSF2,KITLG)1Eav/MloySzJ) transgenic mice overexpress human IL3, CSF2, and SCF in mice resulting in high levels of IL-3, GM-CSF, and SCF (stem cell factor), respectively [115]. HuCD34-NSG-SGM3 mice had improved myeloid, B, and T regulatory cells compared to HuCD34-NSG mice [115]. BLT-NSG-SGM3 mice showed mature B cells with antigen-specific IgG to dengue virus and Toxoplasma gondii [116,117]. Due to the improved engraftment of myeloid cells, human NK cells engraftment was also improved [117]. In addition, a HIV PDX model was generated by transplanting PBLs from ART-suppressed patients in NSG-SGM3 mice [115]. These mice developed rapid HIV reactivation off ART and significantly more activated circulating T cells compared to NSG-SMG3 mice transplanted with PBLs from uninfected donors. Due to the robust myeloid reconstitution driven by high levels of human IL3, GM-CSF, and SCF, HuCD34-SGM3 mice rapidly succumbed to GvHD-related macrophage activation syndrome, which was characterized by progressive pancytopenia, splenomegaly, and hematophagocytosis [120]. Recently, transgenic overexpression of IL-15 under the human CMV promoter was added to this strain to generate NSG-SGM3-IL15 mice. Further studies to assess NK cells and HIV infection have yet to be investigated in humanized NSG-SGM3-IL15 mice.

3.3. MISTRG Mice

MISTRG mice were constructed by the knock-in expression of human M-CSF, IL-3/GM-CSF, and thrombopoietin (TPO) and BAC-mediated transgenic expression of human SIRPα in Rag2−/− γc−/− mice [118]. HuCD34-MISTRG mice demonstrated improved human CD45+ and myeloid cell engraftment [118]. The human monocytes and macrophages were fully functional, and thus capable of trans-presenting IL-15 for the development of functional human NK cells. HuCD34-MISTRG mice also demonstrated robust T cell engraftment and were efficiently infected by X4 and R5 tropic HIV isolates [80]. However, the humanized MISTRG mice were prone to developing GvHD-related severe anemia after 20 weeks of engraftment likely due to robust macrophage development and subsequent destruction of mouse red blood cells [118].

3.4. SRG-15 Mice

Balb/c x 129 Rag2−/− Il2rg−/− hSIRPA KI hIL15 KI (SRG-15) mice were developed by knock-in replacement of mouse IL15 by its human counterpart in SRG mice [49]. SRG-15 mice humanized with CD34+ HSCs (HuCD34-SRG-15) demonstrated similar levels of human CD45+ cells compared to HuCD34-NSG. However, due to its ability to produce human IL-15, the HuCD34-SRG-15 demonstrated significantly higher levels of peripheral blood and splenic NKp46+ NK cells compared to HuCD34-NSG, HuCD34-SRG, and HuCD34-MISTRG mice [49]. The phenotype and functional profile of NK cells from HuCD34-SRG-15 mice were similar to the human NK cell repertoire by mass cytometry. In addition, the human NK cells were capable of mediating ADCC in vivo using anti-CD20 monoclonal antibody against a xenograft B-cell tumor challenge in HuCD34-SRG-15 mice [49]. HuCD34-SRG-15 were susceptible to HIV infection and established a latent reservoir in vivo [110]. HIV-infected HuCD34-SRG-15 treated with a combination of CD4-induced antibodies and a CD4-mimetic compound, which stabilized the HIV envelope in a conformation conducive to NK cell targeting by ADCC, demonstrated diminished HIV replication, viral reservoir, and viral rebound in vivo [110]. Importantly, these studies suggest that NK cell ADCC function in SRG-15 mice can be harnessed to control and reduce HIV infection and the viral reservoir in vivo.

3.5. MISTRG-6-15 Mice

MISTRG-6-15 mice were created by additionally knocking in human IL-6 and IL-15 into MISTRG mice [97]. HuCD34-MISTRG-6-15 mice demonstrated significantly improved human NK cell repopulation and function in vivo compared with huCD34-NSG mice. Due to human IL-6 expression, myeloid cells engraftment also improved, which likely provided a richer cytokine environment for NK cell development and function. MISTRG-6-15 were not directly compared to MISTRG mice in this study. Human NK cells were longitudinally profiled in HIV-infected HuCD34-MISTRG-6-15 and demonstrated increased activation, proliferation, and functionality during acute infection, followed by immune exhaustion and dysfunction during chronic infection. NK cell engraftment and functionality was only partially restored by ART. These patterns of NK biology were similar to previous clinical reports delineating NK cell function from people living with HIV/AIDS [121,122,123]. Importantly, NK cell depletion by NKp46 antibody resulted in increased HIV-1 RNA levels, indicating NK cells controlled HIV-1 replication in vivo [97]. The effect these NK cells may have on the establishment and dynamics of the HIV reservoir remains to be seen.

4. Other Mouse Strains with Improved NK Reconstitution

The following additional recent mouse strains have been developed to enhance NK cell engraftment, but they have not been studied in the context of HIV infection (Table 3 and Supplementary Table S1).

4.1. NOG-IL15 Mice

BLT (NOG-IL15) mice were generated by microinjecting the human IL-15 cDNA into the mouse zygote, leading to overexpression of human IL15 under the control of a CMV promoter [119]. The supraphysiologic levels of human IL-15 in these mice allowed for the efficient engraftment and in vivo anti-tumor function of adoptively transferred human peripheral NK cells, but ultimately led to NK cell exhaustion and dysfunction [119]. Transplantation with human CD34 HSCs and HIV infection studies have not been pursued with these mice.

4.2. HIL-7xhIL-15 KI Mice

The hIL-7xhIL-15 KI mice were generated by knocking in human IL-7 and IL-15 into mouse counterpart genes of NSG mice [124]. hIL-7xhIL-15 KI NSG mice transplanted with CD34+ HSCs demonstrated abnormally high frequencies of human NK cells in the blood (43.1 ± 5.4%), spleen (28.3 ± 5.3%), and bone marrow (17.7 ± 4.7%) among the gated human CD45+ cells. The high levels of NK cell engraftment were likely due to the supraphysiological levels of human IL-15, which was regulated by a CMV promoter. To note, humanized IL-7 single knock-in mice did not demonstrate elevated frequencies of NK cells. Further studies to assess HIV infection have not been pursued with these mice.

5. Concluding Remarks

There are now a wide range of transgenic mice, human cell, or tissue sources, and humanization procedures to generate humanized mice. Depending on the experimental question, each model continues to have its utility. There have been recent exciting advances in the development of transgenic and knock-in mice with robust human myeloid and/or NK cell reconstitution and function. These newer humanized mice should allow researchers to better analyze the latent reservoir and assess HIV vaccines and curative therapies.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11081984/s1, Table S1: Full name of mouse strains.

Author Contributions

J.T.K. and G.B.-T. were responsible for the literature collection. J.T.K., G.B.-T. and J.A.Z. were responsible for the manuscript writing. J.T.K. and J.A.Z. provided ideas for this review and guided the writing. All authors have read and agreed to the published version of the manuscript.

Funding

The authors’ laboratories have been supported by grants from the National Institutes of Health, including AI155232 to J.T.K., AI161803 to J.Z., AI164568 to J.Z., and AI152501 to J.T.K. from the UCLA-CDU CFAR and UCLA AIDS Institute.

Data Availability Statement

Not applicable.

Acknowledgments

We acknowledge the University of California, Los Angeles-Charles Drew University, Center for AIDS Research, the UCLA AIDS Institute, and the McCarthy Family Foundation for their support. Figure 1 was created at BioRender.com.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Gunthard, H.F.; Frost, S.D.; Leigh-Brown, A.J.; Ignacio, C.C.; Kee, K.; Perelson, A.S.; Spina, C.A.; Havlir, D.V.; Hezareh, M.; Looney, D.J.; et al. Evolution of envelope sequences of human immunodeficiency virus type 1 in cellular reservoirs in the setting of potent antiviral therapy. J. Virol. 1999, 73, 9404–9412. [Google Scholar] [CrossRef]
  2. Persaud, D.; Siberry, G.K.; Ahonkhai, A.; Kajdas, J.; Monie, D.; Hutton, N.; Watson, D.C.; Quinn, T.C.; Ray, S.C.; Siliciano, R.F. Continued production of drug-sensitive human immunodeficiency virus type 1 in children on combination antiretroviral therapy who have undetectable viral loads. J. Virol. 2004, 78, 968–979. [Google Scholar] [CrossRef] [Green Version]
  3. Ruff, C.T.; Ray, S.C.; Kwon, P.; Zinn, R.; Pendleton, A.; Hutton, N.; Ashworth, R.; Gange, S.; Quinn, T.C.; Siliciano, R.F.; et al. Persistence of wild-type virus and lack of temporal structure in the latent reservoir for human immunodeficiency virus type 1 in pediatric patients with extensive antiretroviral exposure. J. Virol. 2002, 76, 9481–9492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Veenhuis, R.T.; Abreu, C.M.; Costa, P.A.G.; Ferreira, E.A.; Ratliff, J.; Pohlenz, L.; Shirk, E.N.; Rubin, L.H.; Blankson, J.N.; Gama, L.; et al. Monocyte-derived macrophages contain persistent latent HIV reservoirs. Nat. Microbiol. 2023, 8, 833–844. [Google Scholar] [CrossRef]
  5. Finzi, D.; Blankson, J.; Siliciano, J.D.; Margolick, J.B.; Chadwick, K.; Pierson, T.; Smith, K.; Lisziewicz, J.; Lori, F.; Flexner, C.; et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat. Med. 1999, 5, 512–517. [Google Scholar] [CrossRef] [PubMed]
  6. Chun, T.W.; Stuyver, L.; Mizell, S.B.; Ehler, L.A.; Mican, J.A.; Baseler, M.; Lloyd, A.L.; Nowak, M.A.; Fauci, A.S. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc. Natl. Acad. Sci. USA 1997, 94, 13193–13197. [Google Scholar] [CrossRef] [PubMed]
  7. Chun, T.W.; Davey, R.T.; Engel, D.; Lane, H.C.; Fauci, A.S. Re-emergence of HIV after stopping therapy. Nature 1999, 401, 874–875. [Google Scholar] [CrossRef] [PubMed]
  8. Davey, R.T.; Bhat, N.; Yoder, C.; Chun, T.W.; Metcalf, J.A.; Dewar, R.; Natarajan, V.; Lempicki, R.A.; Adelsberger, J.W.; Miller, K.D.; et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc. Natl. Acad. Sci. USA 1999, 96, 15109–15114. [Google Scholar] [CrossRef]
  9. Harrigan, P.R.; Whaley, M.; Montaner, J.S. Rate of HIV-1 RNA rebound upon stopping antiretroviral therapy. AIDS 1999, 13, F59–F62. [Google Scholar] [CrossRef]
  10. Maldarelli, F.; Wu, X.; Su, L.; Simonetti, F.R.; Shao, W.; Hill, S.; Spindler, J.; Ferris, A.L.; Mellors, J.W.; Kearney, M.F.; et al. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science 2014, 345, 179–183. [Google Scholar] [CrossRef] [Green Version]
  11. Wagner, T.A.; McLaughlin, S.; Garg, K.; Cheung, C.Y.; Larsen, B.B.; Styrchak, S.; Huang, H.C.; Edlefsen, P.T.; Mullins, J.I.; Frenkel, L.M. HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 2014, 345, 570–573. [Google Scholar] [CrossRef]
  12. Premeaux, T.A.; Mediouni, S.; Leda, A.; Furler, R.L.; Valente, S.T.; Fine, H.A.; Nixon, D.F.; Ndhlovu, L.C. Next-Generation Human Cerebral Organoids as Powerful Tools To Advance NeuroHIV Research. mBio 2021, 12, e0068021. [Google Scholar] [CrossRef] [PubMed]
  13. Nowak, M.A.; Lloyd, A.L.; Vasquez, G.M.; Wiltrout, T.A.; Wahl, L.M.; Bischofberger, N.; Williams, J.; Kinter, A.; Fauci, A.S.; Hirsch, V.M.; et al. Viral dynamics of primary viremia and antiretroviral therapy in simian immunodeficiency virus infection. J. Virol. 1997, 71, 7518–7525. [Google Scholar] [CrossRef]
  14. Grimm, D. Supply of Monkeys for Research Is at a Crisis Point, U.S. Government Report Concludes. 2023. Available online: https://www.science.org/content/article/supply-monkeys-research-crisis-point-u-s-government-report-concludes#:~:text=Monkey%20prices%20have%20also%20skyrocketed,biomedical%20demand%2C%20the%20report%20says (accessed on 27 July 2023).
  15. Marsden, M.D.; Zack, J.A. Humanized Mouse Models for Human Immunodeficiency Virus Infection. Annu. Rev. Virol. 2017, 4, 393–412. [Google Scholar] [CrossRef] [PubMed]
  16. Brunetti, J.E.; Kitsera, M.; Munoz-Fontela, C.; Rodriguez, E. Use of Hu-PBL Mice to Study Pathogenesis of Human-Restricted Viruses. Viruses 2023, 15, 228. [Google Scholar] [CrossRef]
  17. Marsden, M.D.; Zack, J.A. Studies of retroviral infection in humanized mice. Virology 2015, 479–480, 297–309. [Google Scholar] [CrossRef] [Green Version]
  18. Denton, P.W.; Garcia, J.V. Humanized mouse models of HIV infection. AIDS Rev. 2011, 13, 135–148. [Google Scholar] [PubMed]
  19. Choudhary, S.K.; Archin, N.M.; Cheema, M.; Dahl, N.P.; Garcia, J.V.; Margolis, D.M. Latent HIV-1 infection of resting CD4+ T cells in the humanized Rag2−/− γc−/− mouse. J. Virol. 2012, 86, 114–120. [Google Scholar] [CrossRef] [Green Version]
  20. Denton, P.W.; Olesen, R.; Choudhary, S.K.; Archin, N.M.; Wahl, A.; Swanson, M.D.; Chateau, M.; Nochi, T.; Krisko, J.F.; Spagnuolo, R.A.; et al. Generation of HIV latency in humanized BLT mice. J. Virol. 2012, 86, 630–634. [Google Scholar] [CrossRef] [Green Version]
  21. Marsden, M.D.; Kovochich, M.; Suree, N.; Shimizu, S.; Mehta, R.; Cortado, R.; Bristol, G.; An, D.S.; Zack, J.A. HIV latency in the humanized BLT mouse. J. Virol. 2012, 86, 339–347. [Google Scholar] [CrossRef] [Green Version]
  22. Denton, P.W.; Long, J.M.; Wietgrefe, S.W.; Sykes, C.; Spagnuolo, R.A.; Snyder, O.D.; Perkey, K.; Archin, N.M.; Choudhary, S.K.; Yang, K.; et al. Targeted cytotoxic therapy kills persisting HIV infected cells during ART. PLoS Pathog. 2014, 10, e1003872. [Google Scholar] [CrossRef] [Green Version]
  23. Lavender, K.J.; Pace, C.; Sutter, K.; Messer, R.J.; Pouncey, D.L.; Cummins, N.W.; Natesampillai, S.; Zheng, J.; Goldsmith, J.; Widera, M.; et al. An advanced BLT-humanized mouse model for extended HIV-1 cure studies. AIDS 2018, 32, 1–10. [Google Scholar] [CrossRef] [PubMed]
  24. Nischang, M.; Sutmuller, R.; Gers-Huber, G.; Audige, A.; Li, D.; Rochat, M.A.; Baenziger, S.; Hofer, U.; Schlaepfer, E.; Regenass, S.M.; et al. Humanized mice recapitulate key features of HIV-1 infection: A novel concept using long-acting anti-retroviral drugs for treating HIV-1. PLoS ONE 2012, 7, e38853. [Google Scholar] [CrossRef]
  25. Marsden, M.D.; Loy, B.A.; Wu, X.; Ramirez, C.M.; Schrier, A.J.; Murray, D.; Shimizu, A.; Ryckbosch, S.M.; Near, K.E.; Chun, T.W.; et al. In vivo activation of latent HIV with a synthetic bryostatin analog effects both latent cell “kick” and “kill” in strategy for virus eradication. PLoS Pathog. 2017, 13, e1006575. [Google Scholar] [CrossRef] [Green Version]
  26. Satheesan, S.; Li, H.; Burnett, J.C.; Takahashi, M.; Li, S.; Wu, S.X.; Synold, T.W.; Rossi, J.J.; Zhou, J. HIV Replication and Latency in a Humanized NSG Mouse Model during Suppressive Oral Combinational Antiretroviral Therapy. J. Virol. 2018, 92, e02118-17. [Google Scholar] [CrossRef] [Green Version]
  27. Halper-Stromberg, A.; Lu, C.L.; Klein, F.; Horwitz, J.A.; Bournazos, S.; Nogueira, L.; Eisenreich, T.R.; Liu, C.; Gazumyan, A.; Schaefer, U.; et al. Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell 2014, 158, 989–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Marsden, M.D.; Zhang, T.H.; Du, Y.; Dimapasoc, M.; Soliman, M.S.A.; Wu, X.; Kim, J.T.; Shimizu, A.; Schrier, A.; Wender, P.A.; et al. Tracking HIV Rebound following Latency Reversal Using Barcoded HIV. Cell Rep. Med. 2020, 1, 100162. [Google Scholar] [CrossRef]
  29. Zhen, A.; Carrillo, M.A.; Mu, W.; Rezek, V.; Martin, H.; Hamid, P.; Chen, I.S.Y.; Yang, O.O.; Zack, J.A.; Kitchen, S.G. Robust CAR-T memory formation and function via hematopoietic stem cell delivery. PLoS Pathog. 2021, 17, e1009404. [Google Scholar] [CrossRef]
  30. Melkus, M.W.; Estes, J.D.; Padgett-Thomas, A.; Gatlin, J.; Denton, P.W.; Othieno, F.A.; Wege, A.K.; Haase, A.T.; Garcia, J.V. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nat. Med. 2006, 12, 1316–1322. [Google Scholar] [CrossRef] [PubMed]
  31. Spranger, S.; Frankenberger, B.; Schendel, D.J. NOD/scid IL-2Rg(null) mice: A preclinical model system to evaluate human dendritic cell-based vaccine strategies in vivo. J. Transl. Med. 2012, 10, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kim, K.C.; Choi, B.S.; Kim, K.C.; Park, K.H.; Lee, H.J.; Cho, Y.K.; Kim, S.I.; Kim, S.S.; Oh, Y.K.; Kim, Y.B. A Simple Mouse Model for the Study of Human Immunodeficiency Virus. AIDS Res. Hum. Retroviruses 2016, 32, 194–202. [Google Scholar] [CrossRef] [Green Version]
  33. Mosier, D.E.; Gulizia, R.J.; Baird, S.M.; Wilson, D.B. Transfer of a Functional Human Immune-System to Mice with Severe Combined Immunodeficiency. Nature 1988, 335, 256–259. [Google Scholar] [CrossRef]
  34. Rizza, P.; Santini, S.M.; Logozzi, M.; Lapenta, C.; Sestili, P.; Gherardi, G.; Lande, R.; Spada, M.; Parlato, S.; Belardelli, F.; et al. T-cell dysfunctions in hu-PBL-SCID mice infected with human immunodeficiency virus (HIV) shortly after reconstitution: In vivo effects of HIV on highly activated human immune cells. J. Virol. 1996, 70, 7958–7964. [Google Scholar] [CrossRef]
  35. McCann, C.D.; van Dorp, C.H.; Danesh, A.; Ward, A.R.; Dilling, T.R.; Mota, T.M.; Zale, E.; Stevenson, E.M.; Patel, S.; Brumme, C.J.; et al. A participant-derived xenograft model of HIV enables long-term evaluation of autologous immunotherapies. J. Exp. Med. 2021, 218, e20201908. [Google Scholar] [CrossRef]
  36. Holyoake, T.L.; Nicolini, F.E.; Eaves, C.J. Functional differences between transplantable human hematopoietic stem cells from fetal liver, cord blood, and adult marrow. Exp. Hematol. 1999, 27, 1418–1427. [Google Scholar] [CrossRef]
  37. McDermott, S.P.; Eppert, K.; Lechman, E.R.; Doedens, M.; Dick, J.E. Comparison of human cord blood engraftment between immunocompromised mouse strains. Blood 2010, 116, 193–200. [Google Scholar] [CrossRef] [Green Version]
  38. Hiramatsu, H.; Nishikomori, R.; Heike, T.; Ito, M.; Kobayashi, K.; Katamura, K.; Nakahata, T. Complete reconstitution of human lymphocytes from cord blood CD34+ cells using the NOD/SCID/gammacnull mice model. Blood 2003, 102, 873–880. [Google Scholar] [CrossRef] [Green Version]
  39. Watanabe, S.; Terashima, K.; Ohta, S.; Horibata, S.; Yajima, M.; Shiozawa, Y.; Dewan, M.Z.; Yu, Z.; Ito, M.; Morio, T.; et al. Hematopoietic stem cell-engrafted NOD/SCID/IL2Rgamma null mice develop human lymphoid systems and induce long-lasting HIV-1 infection with specific humoral immune responses. Blood 2007, 109, 212–218. [Google Scholar] [CrossRef] [Green Version]
  40. Watanabe, S.; Ohta, S.; Yajima, M.; Terashima, K.; Ito, M.; Mugishima, H.; Fujiwara, S.; Shimizu, K.; Honda, M.; Shimizu, N.; et al. Humanized NOD/SCID/IL2Rgamma(null) mice transplanted with hematopoietic stem cells under nonmyeloablative conditions show prolonged life spans and allow detailed analysis of human immunodeficiency virus type 1 pathogenesis. J. Virol. 2007, 81, 13259–13264. [Google Scholar] [CrossRef] [Green Version]
  41. Nie, C.; Sato, K.; Misawa, N.; Kitayama, H.; Fujino, H.; Hiramatsu, H.; Heike, T.; Nakahata, T.; Tanaka, Y.; Ito, M.; et al. Selective infection of CD4+ effector memory T lymphocytes leads to preferential depletion of memory T lymphocytes in R5 HIV-1-infected humanized NOD/SCID/IL-2Rgammanull mice. Virology 2009, 394, 64–72. [Google Scholar] [CrossRef] [Green Version]
  42. Sato, K.; Nie, C.; Misawa, N.; Tanaka, Y.; Ito, M.; Koyanagi, Y. Dynamics of memory and naive CD8+ T lymphocytes in humanized NOD/SCID/IL-2Rgammanull mice infected with CCR5-tropic HIV-1. Vaccine 2010, 28 (Suppl. 2), B32–B37. [Google Scholar] [CrossRef] [PubMed]
  43. Brainard, D.M.; Seung, E.; Frahm, N.; Cariappa, A.; Bailey, C.C.; Hart, W.K.; Shin, H.S.; Brooks, S.F.; Knight, H.L.; Eichbaum, Q.; et al. Induction of robust cellular and humoral virus-specific adaptive immune responses in human immunodeficiency virus-infected humanized BLT mice. J. Virol. 2009, 83, 7305–7321. [Google Scholar] [CrossRef] [Green Version]
  44. Lan, P.; Tonomura, N.; Shimizu, A.; Wang, S.M.; Yang, Y.G. Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation. Blood 2006, 108, 487–492. [Google Scholar] [CrossRef] [Green Version]
  45. Tonomura, N.; Habiro, K.; Shimizu, A.; Sykes, M.; Yang, Y.G. Antigen-specific human T-cell responses and T cell-dependent production of human antibodies in a humanized mouse model. Blood 2008, 111, 4293–4296. [Google Scholar] [CrossRef] [Green Version]
  46. Sun, Z.; Denton, P.W.; Estes, J.D.; Othieno, F.A.; Wei, B.L.; Wege, A.K.; Melkus, M.W.; Padgett-Thomas, A.; Zupancic, M.; Haase, A.T.; et al. Intrarectal transmission, systemic infection, and CD4+ T cell depletion in humanized mice infected with HIV-1. J. Exp. Med. 2007, 204, 705–714. [Google Scholar] [CrossRef]
  47. Denton, P.W.; Estes, J.D.; Sun, Z.; Othieno, F.A.; Wei, B.L.; Wege, A.K.; Powell, D.A.; Payne, D.; Haase, A.T.; Garcia, J.V. Antiretroviral pre-exposure prophylaxis prevents vaginal transmission of HIV-1 in humanized BLT mice. PLoS Med. 2008, 5, e16. [Google Scholar] [CrossRef] [Green Version]
  48. Brown, M.E.; Zhou, Y.; McIntosh, B.E.; Norman, I.G.; Lou, H.E.; Biermann, M.; Sullivan, J.A.; Kamp, T.J.; Thomson, J.A.; Anagnostopoulos, P.V.; et al. A Humanized Mouse Model Generated Using Surplus Neonatal Tissue. Stem Cell Rep. 2018, 10, 1175–1183. [Google Scholar] [CrossRef] [Green Version]
  49. Herndler-Brandstetter, D.; Shan, L.; Yao, Y.; Stecher, C.; Plajer, V.; Lietzenmayer, M.; Strowig, T.; de Zoete, M.R.; Palm, N.W.; Chen, J.; et al. Humanized mouse model supports development, function, and tissue residency of human natural killer cells. Proc. Natl. Acad. Sci. USA 2017, 114, E9626–E9634. [Google Scholar] [CrossRef] [Green Version]
  50. Holguin, L.; Echavarria, L.; Burnett, J.C. Novel Humanized Peripheral Blood Mononuclear Cell Mouse Model with Delayed Onset of Graft-versus-Host Disease for Preclinical HIV Research. J. Virol. 2022, 96, e0139421. [Google Scholar] [CrossRef]
  51. Lavender, K.J.; Pang, W.W.; Messer, R.J.; Duley, A.K.; Race, B.; Phillips, K.; Scott, D.; Peterson, K.E.; Chan, C.K.; Dittmer, U.; et al. BLT-humanized C57BL/6 Rag2−/−γc−/−CD47−/− mice are resistant to GVHD and develop B- and T-cell immunity to HIV infection. Blood 2013, 122, 4013–4020. [Google Scholar] [CrossRef]
  52. Flerin, N.C.; Bardhi, A.; Zheng, J.H.; Korom, M.; Folkvord, J.; Kovacs, C.; Benko, E.; Truong, R.; Mota, T.; Connick, E.; et al. Establishment of a Novel Humanized Mouse Model To Investigate In Vivo Activation and Depletion of Patient-Derived HIV Latent Reservoirs. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  53. Huang, W.; Chao, N.J. Memory T cells: A helpful guard for allogeneic hematopoietic stem cell transplantation without causing graft-versus-host disease. Hematol. Oncol. Stem Cell Ther. 2017, 10, 211–219. [Google Scholar] [CrossRef] [PubMed]
  54. Anderson, B.E.; McNiff, J.; Yan, J.; Doyle, H.; Mamula, M.; Shlomchik, M.J.; Shlomchik, W.D. Memory CD4+ T cells do not induce graft-versus-host disease. J. Clin. Investig. 2003, 112, 101–108. [Google Scholar] [CrossRef]
  55. Ishikawa, Y.; Usui, T.; Shiomi, A.; Shimizu, M.; Murakami, K.; Mimori, T. Functional engraftment of human peripheral T and B cells and sustained production of autoantibodies in NOD/LtSzscid/IL-2Rγ−/− mice. Eur. J. Immunol. 2014, 44, 3453–3463. [Google Scholar] [CrossRef] [Green Version]
  56. Berges, B.K.; Akkina, S.R.; Folkvord, J.M.; Connick, E.; Akkina, R. Mucosal transmission of R5 and X4 tropic HIV-1 via vaginal and rectal routes in humanized Rag2−/−γc−/− (RAG-hu) mice. Virology 2008, 373, 342–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Gillgrass, A.; Wessels, J.M.; Yang, J.X.; Kaushic, C. Advances in Humanized Mouse Models to Improve Understanding of HIV-1 Pathogenesis and Immune Responses. Front. Immunol. 2020, 11, 617516. [Google Scholar] [CrossRef] [PubMed]
  58. Long, B.R.; Stoddart, C.A. Alpha interferon and HIV infection cause activation of human T cells in NSG-BLT mice. J. Virol. 2012, 86, 3327–3336. [Google Scholar] [CrossRef] [Green Version]
  59. Dudek, T.E.; No, D.C.; Seung, E.; Vrbanac, V.D.; Fadda, L.; Bhoumik, P.; Boutwell, C.L.; Power, K.A.; Gladden, A.D.; Battis, L.; et al. Rapid evolution of HIV-1 to functional CD8+ T cell responses in humanized BLT mice. Sci. Transl. Med. 2012, 4, 143ra98. [Google Scholar] [CrossRef] [Green Version]
  60. Denton, P.W.; Nochi, T.; Lim, A.; Krisko, J.F.; Martinez-Torres, F.; Choudhary, S.K.; Wahl, A.; Olesen, R.; Zou, W.; Di Santo, J.P.; et al. IL-2 receptor gamma-chain molecule is critical for intestinal T-cell reconstitution in humanized mice. Mucosal Immunol. 2012, 5, 555–566. [Google Scholar] [CrossRef] [Green Version]
  61. Kitchen, S.G.; Levin, B.R.; Bristol, G.; Rezek, V.; Kim, S.; Aguilera-Sandoval, C.; Balamurugan, A.; Yang, O.O.; Zack, J.A. In vivo suppression of HIV by antigen specific T cells derived from engineered hematopoietic stem cells. PLoS Pathog. 2012, 8, e1002649. [Google Scholar] [CrossRef]
  62. Deruaz, M.; Luster, A.D. BLT Humanized Mice as Model to Study HIV Vaginal Transmission. J. Infect. Dis. 2013, 208, S131–S136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Denton, P.W.; Krisko, J.F.; Powell, D.A.; Mathias, M.; Kwak, Y.T.; Martinez-Torres, F.; Zou, W.; Payne, D.A.; Estes, J.D.; Garcia, J.V. Systemic Administration of Antiretrovirals Prior to Exposure Prevents Rectal and Intravenous HIV-1 Transmission in Humanized BLT Mice. PLoS ONE 2010, 5, e8829. [Google Scholar] [CrossRef] [PubMed]
  64. Stoddart, C.A.; Maidji, E.; Galkina, S.A.; Kosikova, G.; Rivera, J.M.; Moreno, M.E.; Sloan, B.; Joshi, P.; Long, B.R. Superior human leukocyte reconstitution and susceptibility to vaginal HIV transmission in humanized NOD-scid IL-2R γ−/− (NSG) BLT mice. Virology 2011, 417, 154–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Nochi, T.; Denton, P.W.; Wahl, A.; Garcia, J.V. Cryptopatches are essential for the development of human GALT. Cell Rep. 2013, 3, 1874–1884. [Google Scholar] [CrossRef] [Green Version]
  66. Martinez-Torres, F.; Nochi, T.; Wahl, A.; Garcia, J.V.; Denton, P.W. Hypogammaglobulinemia in BLT humanized mice—An animal model of primary antibody deficiency. PLoS ONE 2014, 9, e108663. [Google Scholar] [CrossRef]
  67. Chang, H.; Biswas, S.; Tallarico, A.S.; Sarkis, P.T.N.; Geng, S.; Panditrao, M.M.; Zhu, Q.; Marasco, W.A. Human B-cell ontogeny in humanized NOD/SCID gamma c(null) mice generates a diverse yet auto/poly- and HIV-1-reactive antibody repertoire. Genes Immun. 2012, 13, 399–410. [Google Scholar] [CrossRef] [Green Version]
  68. Biswas, S.; Chang, H.; Sarkis, P.T.N.; Fikrig, E.; Zhu, Q.; Marasco, W.A. Humoral immune responses in humanized BLT mice immunized with West Nile virus and HIV-1 envelope proteins are largely mediated via human CD5+ B cells. Immunology 2011, 134, 419–433. [Google Scholar] [CrossRef]
  69. Claiborne, D.T.; Dudek, T.E.; Maldini, C.R.; Power, K.A.; Ghebremichael, M.; Seung, E.; Mellors, E.F.; Vrbanac, V.D.; Krupp, K.; Bisesi, A.; et al. Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  70. Norton, T.D.; Zhen, A.; Tada, T.; Kim, J.; Kitchen, S.; Landau, N.R. Lentiviral Vector-Based Dendritic Cell Vaccine Suppresses HIV Replication in Humanized Mice. Mol. Ther. 2019, 27, 960–973. [Google Scholar] [CrossRef] [Green Version]
  71. Cheng, L.; Wang, Q.; Li, G.; Banga, R.; Ma, J.; Yu, H.; Yasui, F.; Zhang, Z.; Pantaleo, G.; Perreau, M.; et al. TLR3 agonist and CD40-targeting vaccination induces immune responses and reduces HIV-1 reservoirs. J. Clin. Investig. 2018, 128, 4387–4396. [Google Scholar] [CrossRef] [Green Version]
  72. Kim, J.T.; Zhang, T.H.; Carmona, C.; Lee, B.; Seet, C.S.; Kostelny, M.; Shah, N.; Chen, H.; Farrell, K.; Soliman, M.S.A.; et al. Latency reversal plus natural killer cells diminish HIV reservoir in vivo. Nat. Commun. 2022, 13, 121. [Google Scholar] [CrossRef]
  73. Nikzad, R.; Angelo, L.S.; Aviles-Padilla, K.; Le, D.T.; Singh, V.K.; Bimler, L.; Vukmanovic-Stejic, M.; Vendrame, E.; Ranganath, T.; Simpson, L.; et al. Human natural killer cells mediate adaptive immunity to viral antigens. Sci. Immunol. 2019, 4, eaat8116. [Google Scholar] [CrossRef]
  74. Carrillo, M.A.; Zhen, A.; Zack, J.A.; Kitchen, S.G. New approaches for the enhancement of chimeric antigen receptors for the treatment of HIV. Transl. Res. 2017, 187, 83–92. [Google Scholar] [CrossRef]
  75. Greenblatt, M.B.; Vrbanac, V.; Tivey, T.; Tsang, K.; Tager, A.M.; Aliprantis, A.O. Graft versus host disease in the bone marrow, liver and thymus humanized mouse model. PLoS ONE 2012, 7, e44664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Oberbarnscheidt, M.H.; Zeng, Q.; Li, Q.; Dai, H.H.; Williams, A.L.; Shlomchik, W.D.; Rothstein, D.M.; Lakkis, F.G. Non-self recognition by monocytes initiates allograft rejection. J. Clin. Investig. 2014, 124, 3579–3589. [Google Scholar] [CrossRef] [PubMed]
  77. Pengam, S.; Durand, J.; Usal, C.; Gauttier, V.; Dilek, N.; Martinet, B.; Daguin, V.; Mary, C.; Thepenier, V.; Teppaz, G.; et al. SIRP alpha/CD47 axis controls the maintenance of transplant tolerance sustained by myeloid-derived suppressor cells. Am. J. Transpl. 2019, 19, 3263–3275. [Google Scholar] [CrossRef] [PubMed]
  78. Martinov, T.; McKenna, K.M.; Tan, W.H.; Collins, E.J.; Kehret, A.R.; Linton, J.D.; Olsen, T.M.; Shobaki, N.; Rongvaux, A. Building the Next Generation of Humanized Hemato-Lymphoid System Mice. Front. Immunol. 2021, 12, 643852. [Google Scholar] [CrossRef]
  79. Strowig, T.; Rongvaux, A.; Rathinam, C.; Takizawa, H.; Borsotti, C.; Philbrick, W.; Eynon, E.E.; Manz, M.G.; Flavell, R.A. Transgenic expression of human signal regulatory protein alpha in Rag2−/−γc−/− mice improves engraftment of human hematopoietic cells in humanized mice. Proc. Natl. Acad. Sci. USA 2011, 108, 13218–13223. [Google Scholar] [CrossRef]
  80. Ivic, S.; Rochat, M.; Li, D.; Audigé, A.; Schlaepfer, E.; Münz, C.; Manz, M.G.; Spec, R.F. Differential Dynamics of HIV Infection in Humanized MISTRG versus MITRG Mice. Immunohorizons 2017, 1, 162–175. [Google Scholar] [CrossRef] [Green Version]
  81. Langin, K. For Scientists, Roe’s End Raises Concerns About Personal Safety And Professional Choices. Available online: https://www.science.org/content/article/scientists-roe-s-end-raises-concerns-about-personal-safety-and-professional-choices (accessed on 1 July 2023).
  82. Barata, J.T.; Silva, A.; Abecasis, M.; Carlesso, N.; Cumano, A.; Cardoso, A.A. Molecular and functional evidence for activity of murine IL-7 on human lymphocytes. Exp. Hematol. 2006, 34, 1133–1142. [Google Scholar] [CrossRef]
  83. Zou, J.J.; Schoenhaut, D.S.; Carvajal, D.M.; Warrier, R.R.; Presky, D.H.; Gately, M.K.; Gubler, U. Structure-function analysis of the p35 subunit of mouse interleukin 12. J. Biol. Chem. 1995, 270, 5864–5871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Mosmann, T.R.; Yokota, T.; Kastelein, R.; Zurawski, S.M.; Arai, N.; Takebe, Y. Species-specificity of T cell stimulating activities of IL 2 and BSF-1 (IL 4): Comparison of normal and recombinant, mouse and human IL 2 and BSF-1 (IL 4). J. Immunol. 1987, 138, 1813–1816. [Google Scholar] [CrossRef]
  85. Manz, M.G. Human-hemato-lymphoid-system mice: Opportunities and challenges. Immunity 2007, 26, 537–541. [Google Scholar] [CrossRef] [Green Version]
  86. Eisenman, J.; Ahdieh, M.; Beers, C.; Brasel, K.; Kennedy, M.K.; Le, T.; Bonnert, T.P.; Paxton, R.J.; Park, L.S. Interleukin-15 interactions with interleukin-15 receptor complexes: Characterization and species specificity. Cytokine 2002, 20, 121–129. [Google Scholar] [CrossRef] [PubMed]
  87. Kitamura, T.; Hayashida, K.; Sakamaki, K.; Yokota, T.; Arai, K.; Miyajima, A. Reconstitution of functional receptors for human granulocyte/macrophage colony-stimulating factor (GM-CSF): Evidence that the protein encoded by the AIC2B cDNA is a subunit of the murine GM-CSF receptor. Proc. Natl. Acad. Sci. USA 1991, 88, 5082–5086. [Google Scholar] [CrossRef] [PubMed]
  88. Flores-Villanueva, P.O.; Yunis, E.J.; Delgado, J.C.; Vittinghoff, E.; Buchbinder, S.; Leung, J.Y.; Uglialoro, A.M.; Clavijo, O.P.; Rosenberg, E.S.; Kalams, S.A.; et al. Control of HIV-1 viremia and protection from AIDS are associated with HLA-Bw4 homozygosity. Proc. Natl. Acad. Sci. USA 2001, 98, 5140–5145. [Google Scholar] [CrossRef]
  89. Gondois-Rey, F.; Cheret, A.; Granjeaud, S.; Mallet, F.; Bidaut, G.; Lecuroux, C.; Ploquin, M.; Muller-Trutwin, M.; Rouzioux, C.; Avettand-Fenoel, V.; et al. NKG2C+ memory-like NK cells contribute to the control of HIV viremia during primary infection: Optiprim-ANRS 147. Clin. Transl. Immunol. 2017, 6, e150. [Google Scholar] [CrossRef]
  90. Ma, M.; Wang, Z.; Chen, X.; Tao, A.; He, L.; Fu, S.; Zhang, Z.; Fu, Y.; Guo, C.; Liu, J.; et al. NKG2C+NKG2A Natural Killer Cells are Associated with a Lower Viral Set Point and may Predict Disease Progression in Individuals with Primary HIV Infection. Front. Immunol. 2017, 8, 1176. [Google Scholar] [CrossRef] [Green Version]
  91. Martin, M.P.; Gao, X.; Lee, J.H.; Nelson, G.W.; Detels, R.; Goedert, J.J.; Buchbinder, S.; Hoots, K.; Vlahov, D.; Trowsdale, J.; et al. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat. Genet. 2002, 31, 429–434. [Google Scholar] [CrossRef]
  92. Holzemer, A.; Thobakgale, C.F.; Jimenez Cruz, C.A.; Garcia-Beltran, W.F.; Carlson, J.M.; van Teijlingen, N.H.; Mann, J.K.; Jaggernath, M.; Kang, S.G.; Korner, C.; et al. Selection of an HLA-C*03:04-Restricted HIV-1 p24 Gag Sequence Variant Is Associated with Viral Escape from KIR2DL3+ Natural Killer Cells: Data from an Observational Cohort in South Africa. PLoS Med. 2015, 12, e1001900. [Google Scholar] [CrossRef] [Green Version]
  93. Alter, G.; Martin, M.P.; Teigen, N.; Carr, W.H.; Suscovich, T.J.; Schneidewind, A.; Streeck, H.; Waring, M.; Meier, A.; Brander, C.; et al. Differential natural killer cell-mediated inhibition of HIV-1 replication based on distinct KIR/HLA subtypes. J. Exp. Med. 2007, 204, 3027–3036. [Google Scholar] [CrossRef]
  94. Kiani, Z.; Bruneau, J.; Geraghty, D.E.; Bernard, N.F. HLA-F on Autologous HIV-Infected Cells Activates Primary NK Cells Expressing the Activating Killer Immunoglobulin-Like Receptor KIR3DS1. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Norman, J.M.; Mashiba, M.; McNamara, L.A.; Onafuwa-Nuga, A.; Chiari-Fort, E.; Shen, W.; Collins, K.L. The antiviral factor APOBEC3G enhances the recognition of HIV-infected primary T cells by natural killer cells. Nat. Immunol. 2011, 12, 975–983. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Martin, M.P.; Qi, Y.; Gao, X.; Yamada, E.; Martin, J.N.; Pereyra, F.; Colombo, S.; Brown, E.E.; Shupert, W.L.; Phair, J.; et al. Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat. Genet. 2007, 39, 733–740. [Google Scholar] [CrossRef]
  97. Sungur, C.M.; Wang, Q.; Ozanturk, A.N.; Gao, H.; Schmitz, A.J.; Cella, M.; Yokoyama, W.M.; Shan, L. Human natural killer cells confer protection against HIV-1 infection in humanized mice. J. Clin. Investig. 2022. [Google Scholar] [CrossRef]
  98. Han, K.P.; Zhu, X.; Liu, B.; Jeng, E.; Kong, L.; Yovandich, J.L.; Vyas, V.V.; Marcus, W.D.; Chavaillaz, P.A.; Romero, C.A.; et al. IL-15:IL-15 receptor alpha superagonist complex: High-level co-expression in recombinant mammalian cells, purification and characterization. Cytokine 2011, 56, 804–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Rhode, P.R.; Egan, J.O.; Xu, W.; Hong, H.; Webb, G.M.; Chen, X.; Liu, B.; Zhu, X.; Wen, J.; You, L.; et al. Comparison of the Superagonist Complex, ALT-803, to IL15 as Cancer Immunotherapeutics in Animal Models. Cancer Immunol. Res. 2016, 4, 49–60. [Google Scholar] [CrossRef] [PubMed]
  100. Seay, K.; Church, C.; Zheng, J.H.; Deneroff, K.; Ochsenbauer, C.; Kappes, J.C.; Liu, B.; Jeng, E.K.; Wong, H.C.; Goldstein, H. In Vivo Activation of Human NK Cells by Treatment with an Interleukin-15 Superagonist Potently Inhibits Acute In Vivo HIV-1 Infection in Humanized Mice. J. Virol. 2015, 89, 6264–6274. [Google Scholar] [CrossRef] [Green Version]
  101. Ellis-Connell, A.L.; Balgeman, A.J.; Zarbock, K.R.; Barry, G.; Weiler, A.; Egan, J.O.; Jeng, E.K.; Friedrich, T.; Miller, J.S.; Haase, A.T.; et al. ALT-803 Transiently Reduces Simian Immunodeficiency Virus Replication in the Absence of Antiretroviral Treatment. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [Green Version]
  102. Bernstein, H.B.; Plasterer, M.C.; Schiff, S.E.; Kitchen, C.M.; Kitchen, S.; Zack, J.A. CD4 expression on activated NK cells: Ligation of CD4 induces cytokine expression and cell migration. J. Immunol. 2006, 177, 3669–3676. [Google Scholar] [CrossRef] [Green Version]
  103. Bernstein, H.B.; Wang, G.; Plasterer, M.C.; Zack, J.A.; Ramasastry, P.; Mumenthaler, S.M.; Kitchen, C.M. CD4+ NK cells can be productively infected with HIV.; leading to downregulation of CD4 expression and changes in function. Virology 2009, 387, 59–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Valentin, A.; Rosati, M.; Patenaude, D.J.; Hatzakis, A.; Kostrikis, L.G.; Lazanas, M.; Wyvill, K.M.; Yarchoan, R.; Pavlakis, G.N. Persistent HIV-1 infection of natural killer cells in patients receiving highly active antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2002, 99, 7015–7020. [Google Scholar] [CrossRef] [PubMed]
  105. Huntington, N.D.; Legrand, N.; Alves, N.L.; Jaron, B.; Weijer, K.; Plet, A.; Corcuff, E.; Mortier, E.; Jacques, Y.; Spits, H.; et al. IL-15 trans-presentation promotes human NK cell development and differentiation in vivo. J. Exp. Med. 2009, 206, 25–34. [Google Scholar] [CrossRef] [PubMed]
  106. Lopez-Lastra, S.; Masse-Ranson, G.; Fiquet, O.; Darche, S.; Serafini, N.; Li, Y.; Dusseaux, M.; Strick-Marchand, H.; Di Santo, J.P. A functional DC cross talk promotes human ILC homeostasis in humanized mice. Blood Adv. 2017, 1, 601–614. [Google Scholar] [CrossRef] [Green Version]
  107. Chen, Q.; Khoury, M.; Chen, J. Expression of human cytokines dramatically improves reconstitution of specific human-blood lineage cells in humanized mice. Proc. Natl. Acad. Sci. USA 2009, 106, 21783–21788. [Google Scholar] [CrossRef]
  108. Douam, F.; Ziegler, C.G.K.; Hrebikova, G.; Fant, B.; Leach, R.; Parsons, L.; Wang, W.; Gaska, J.M.; Winer, B.Y.; Heller, B.; et al. Selective expansion of myeloid and NK cells in humanized mice yields human-like vaccine responses. Nat. Commun. 2018, 9, 5031. [Google Scholar] [CrossRef] [Green Version]
  109. O’Connell, R.M.; Balazs, A.B.; Rao, D.S.; Kivork, C.; Yang, L.; Baltimore, D. Lentiviral vector delivery of human interleukin-7 (hIL-7) to human immune system (HIS) mice expands T lymphocyte populations. PLoS ONE 2010, 5, e12009. [Google Scholar] [CrossRef]
  110. Rajashekar, J.K.; Richard, J.; Beloor, J.; Prevost, J.; Anand, S.P.; Beaudoin-Bussieres, G.; Shan, L.; Herndler-Brandstetter, D.; Gendron-Lepage, G.; Medjahed, H.; et al. Modulating HIV-1 envelope glycoprotein conformation to decrease the HIV-1 reservoir. Cell Host Microbe 2021, 29, 904–916. e6. [Google Scholar] [CrossRef]
  111. Abeynaike, S.A.; Huynh, T.R.; Mehmood, A.; Kim, T.; Frank, K.; Gao, K.; Zalfa, C.; Gandarilla, A.; Shultz, L.; Paust, S. Human Hematopoietic Stem Cell Engrafted IL-15 Transgenic NSG Mice Support Robust NK Cell Responses and Sustained HIV-1 Infection. Viruses 2023, 15, 365. [Google Scholar] [CrossRef]
  112. Aryee, K.E.; Burzenski, L.M.; Yao, L.C.; Keck, J.G.; Greiner, D.L.; Shultz, L.D.; Brehm, M.A. Enhanced development of functional human NK cells in NOD-scid-IL2rg(null) mice expressing human IL15. FASEB J. 2022, 36, e22476. [Google Scholar] [CrossRef]
  113. Perdomo-Celis, F.; Medina-Moreno, S.; Davis, H.; Bryant, J.; Zapata, J.C. HIV Replication in Humanized IL-3/GM-CSF-Transgenic NOG Mice. Pathogens 2019, 8, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ito, R.; Takahashi, T.; Katano, I.; Kawai, K.; Kamisako, T.; Ogura, T.; Ida-Tanaka, M.; Suemizu, H.; Nunomura, S.; Ra, C.; et al. Establishment of a human allergy model using human IL-3/GM-CSF-transgenic NOG mice. J. Immunol. 2013, 191, 2890–2899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Billerbeck, E.; Barry, W.T.; Mu, K.; Dorner, M.; Rice, C.M.; Ploss, A. Development of human CD4+FoxP3+ regulatory T cells in human stem cell factor-, granulocyte-macrophage colony-stimulating factor-, and interleukin-3-expressing NOD-SCID IL2Rgamma(null) humanized mice. Blood 2011, 117, 3076–3086. [Google Scholar] [CrossRef] [Green Version]
  116. Jangalwe, S.; Shultz, L.D.; Mathew, A.; Brehm, M.A. Improved B cell development in humanized NOD-scid IL2Rgamma(null) mice transgenically expressing human stem cell factor, granulocyte-macrophage colony-stimulating factor and interleukin-3. Immun. Inflamm. Dis. 2016, 4, 427–440. [Google Scholar] [CrossRef]
  117. Wunderlich, M.; Chou, F.S.; Sexton, C.; Presicce, P.; Chougnet, C.A.; Aliberti, J.; Mulloy, J.C. Improved multilineage human hematopoietic reconstitution and function in NSGS mice. PLoS ONE 2018, 13, e0209034. [Google Scholar] [CrossRef] [Green Version]
  118. Rongvaux, A.; Willinger, T.; Martinek, J.; Strowig, T.; Gearty, S.V.; Teichmann, L.L.; Saito, Y.; Marches, F.; Halene, S.; Palucka, A.K.; et al. Development and function of human innate immune cells in a humanized mouse model. Nat. Biotechnol. 2014, 32, 364–372. [Google Scholar] [CrossRef]
  119. Katano, I.; Nishime, C.; Ito, R.; Kamisako, T.; Mizusawa, T.; Ka, Y.; Ogura, T.; Suemizu, H.; Kawakami, Y.; Ito, M.; et al. Long-term maintenance of peripheral blood derived human NK cells in a novel human IL-15- transgenic NOG mouse. Sci. Rep. 2017, 7, 17230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Wunderlich, M.; Stockman, C.; Devarajan, M.; Ravishankar, N.; Sexton, C.; Kumar, A.R.; Mizukawa, B.; Mulloy, J.C. A xenograft model of macrophage activation syndrome amenable to anti-CD33 and anti-IL-6R treatment. J. Clin. Investig. 2016, 1, e88181. [Google Scholar] [CrossRef] [Green Version]
  121. Mavilio, D.; Lombardo, G.; Benjamin, J.; Kim, D.; Follman, D.; Marcenaro, E.; O’Shea, M.A.; Kinter, A.; Kovacs, C.; Moretta, A.; et al. Characterization of CD56-/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc. Natl. Acad. Sci. USA 2005, 102, 2886–2891. [Google Scholar] [CrossRef]
  122. Giuliani, E.; Vassena, L.; Di Cesare, S.; Malagnino, V.; Desimio, M.G.; Andreoni, M.; Barnaba, V.; Doria, M. NK cells of HIV-1-infected patients with poor CD4+ T-cell reconstitution despite suppressive HAART show reduced IFN-gamma production and high frequency of autoreactive CD56(bright) cells. Immunol. Lett. 2017, 190, 185–193. [Google Scholar] [CrossRef]
  123. Goodier, M.R.; Imami, N.; Moyle, G.; Gazzard, B.; Gotch, F. Loss of the CD56hiCD16- NK cell subset and NK cell interferon-gamma production during antiretroviral therapy for HIV-1: Partial recovery by human growth hormone. Clin. Exp. Immunol. 2003, 134, 470–476. [Google Scholar] [CrossRef] [PubMed]
  124. Matsuda, M.; Ono, R.; Iyoda, T.; Endo, T.; Iwasaki, M.; Tomizawa-Murasawa, M.; Saito, Y.; Kaneko, A.; Shimizu, K.; Yamada, D.; et al. Human NK cell development in hIL-7 and hIL-15 knockin NOD/SCID/IL2rgKO mice. Life Sci. Alliance 2019, 2, e201800195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Harnessing NK cells to fight HIV infection.
Figure 1. Harnessing NK cells to fight HIV infection.
Microorganisms 11 01984 g001
Table 1. Commonly used humanized mouse strains and models.
Table 1. Commonly used humanized mouse strains and models.
Mouse NameMouse ModelXenograftSummary of Key Findings
NSG
NRG
NOG
BRG
HuPBL [16,31,32,33,34]
HuPDX [16,35]
HuCD34 [36,37,38,39,40,41,42]
BLT [30,43,44,45,46,47]
NeoThy [48]
Human PBL (HuPBL, HuPDX), CD34+ HSC (HuCD34), or human thymus (NeoThy) ± human liver and CD34+ HSCs (BLT)
  • Minimal to low NK cell engraftment in these humanized mouse models, but appropriate model to test exogenous NK cell adoptive therapies.
  • Prone to GvHD.
SRGHuCD34 [49]Human CD34+ HSC
  • Minimal to low NK cell engraftment.
  • Less susceptible to GvHD due to human SIRPA knock-in.
TKOHuPBL [50]
BLT [23,51]
Human PBL or
human thymus, liver and CD34+ HSCs (BLT)
  • Minimal NK cell engraftment in humanized TKO mice, but appropriate model to test exogenous NK cell adoptive therapies.
  • Less susceptible to GvHD due to knockout of CD47.
  • Mice remain healthy for 45 weeks post-humanization.
  • Mice can be placed on ART for prolonged periods.
Table 2. Recently advanced humanized mouse models to study NK cells and HIV infection.
Table 2. Recently advanced humanized mouse models to study NK cells and HIV infection.
Mouse NameHuman
Cytokines
Expressed
Mouse ModelXenograftsSummary of Key Findings
NSG-Tg(IL-15)IL-15HuCD34 [111,112]Human PBL (HuPBL, HuPDX), CD34+ HSC (HuCD34), or human thymus (NeoThy) ± human liver and CD34+ HSCs (BLT)
  • Robust and long-term engraftment of human NK cells with increased expression of IFN-γ, TNF-α, perforin, Ki67, and CD107a after HIV infection in vivo.
  • Less susceptible to GvHD.
  • May be a suitable long-term model to study the effects of NK cells on the HIV reservoir.
NOG-EXLGM-CSF
IL-3
HuCD34 [113,114]Human CD34+ HSC
  • Robust myeloid cell engraftment, but low NK cell reconstitution.
  • Acute HIV infection decreased levels of CD56bright NK cells.
NSG-SMG3GM-CSF
IL-3
SCF
HuPBL
HuCD34 [115]
BLT [116,117]
Human PBL (HuPBL),
CD34+ HSC (HuCD34), or human thymus, liver and CD34+ HSCs (BLT)
  • Robust myeloid cell engraftment, and thereby NK cell development.
  • Early death due to GvHD-related macrophage activation syndrome.
MISTRGGM-CSF
IL-3
M-CSF
TPO
HuCD34 [80,118]Human CD34+ HSC
  • Robust engraftment of myeloid cells, which aid in engraftment of human NK cells.
  • Robust T cell engraftment with efficiently infection by X4 and R5 tropic HIV isolates.
  • Prone to GvHD-related severe anemia 20 weeks post-transplantation.
SRG-15IL-15HuCD34 [49,110]Human CD34+ HSC
  • Significantly improved engraftment of human NK cells compared to HuCD34-SRG or HuCD34-MISTRG mice.
  • Appropriate long-term pre-clinical model to test curative therapies against the HIV reservoir.
MISTRG-GM-CSF
IL-3
IL-6
IL-10
M-CSF
TPO
HuCD34 [97]Human CD34+ HSC
  • Significantly improved myeloid and NK cell engraftment and function compared to HuCD34-NSG mice.
  • Human NK cells during acute infection, ART suppression, and rebound infection show functional profile patterns similar to clinical studies.
  • Importantly, human NK cells were shown to directly control HIV-1 replication in vivo.
Table 3. Other recent humanized mouse models to study NK cells.
Table 3. Other recent humanized mouse models to study NK cells.
Mouse
Strain
Human
Cytokines
Expressed
Mouse ModelXenograftsSummary of Key Findings
NOG-IL15IL-15PBL [119]Human PBL
  • Robust engraftment of human NK cells with subsequent exhaustion and dysfunction, likely due to supraphysiologic level of human IL-15.
  • Prone to GvHD.
  • In vivo HIV infection has not been studied yet.
hIL-7xhIL-15 KI NSGIL-7
IL-15
HuCD34 [124]Human CD34+ HSC
  • Abnormally high frequencies of human NK cells, likely due to supraphysiologic level of human IL-15.
  • In vivo HIV infection has not been studied yet.
  • Likely prone to GvHD.
NSG-SMG3-IL15GM-CSF
IL-3
IL-15
SCF
None
  • Transgenic expression of IL-15 in NSG-SGM3 mice.
  • Further studies to assess NK cells and HIV infection in vivo have yet to be performed.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kim, J.T.; Bresson-Tan, G.; Zack, J.A. Current Advances in Humanized Mouse Models for Studying NK Cells and HIV Infection. Microorganisms 2023, 11, 1984. https://doi.org/10.3390/microorganisms11081984

AMA Style

Kim JT, Bresson-Tan G, Zack JA. Current Advances in Humanized Mouse Models for Studying NK Cells and HIV Infection. Microorganisms. 2023; 11(8):1984. https://doi.org/10.3390/microorganisms11081984

Chicago/Turabian Style

Kim, Jocelyn T., Gabrielle Bresson-Tan, and Jerome A. Zack. 2023. "Current Advances in Humanized Mouse Models for Studying NK Cells and HIV Infection" Microorganisms 11, no. 8: 1984. https://doi.org/10.3390/microorganisms11081984

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop