Apr 20, 2024

Public workspaceGeneration of Immunodeficient Mice Bearing Human Immune Systems by the Engraftment of Hematopoietic Stem Cells V.2

Generation of Immunodeficient Mice Bearing Human Immune Systems by the Engraftment of Hematopoietic Stem Cells
  • Suheyla Hasgur1,
  • Ken Edwin Aryee1,
  • Leonard D. Shultz2,
  • Dale L. Greiner1,
  • and Michael A. Brehm1
  • 1Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605;
  • 2The Jackson Laboratory, Bar Harbor, ME 04609
Open access
Protocol CitationSuheyla Hasgur, Ken Edwin Aryee, Leonard D. Shultz, Dale L. Greiner, and Michael A. Brehm 2024. Generation of Immunodeficient Mice Bearing Human Immune Systems by the Engraftment of Hematopoietic Stem Cells. protocols.io https://dx.doi.org/10.17504/protocols.io.36wgq4q43vk5/v2Version created by Sandy Beshir
License: This is an open access protocol distributed under the terms of the Creative Commons Attribution License,  which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited
Protocol status: Working
We use this protocol and it's working
Created: April 20, 2024
Last Modified: April 20, 2024
Protocol Integer ID: 98534
Keywords: Hematopoietic stem cells; SCID; Thymus; HSC; humanized mice
Funders Acknowledgement:
NIH
Grant ID: UC4 DK104218
Disclaimer
UC4 DK104218
Abstract
Immunodeficient mice are being used as recipients of human hematopoietic stem cells (HSC) for in vivo analyses of human immune system development and function. The development of several stocks of immunodeficient Prkdc scid ( scid ), or recombination activating 1 or 2 gene ( Rag1 or Rag2 ) knockout mice bearing a targeted mutation in the gene encoding the IL2 receptor gamma chain ( IL2r γ) have greatly facilitated the engraftment of human HSC and enhanced the development of functional human immune systems. These “humanized” mice are being used to study human hematopoiesis, human-specific immune therapies, human-specific pathogens, and human immune system homeostasis and function. The establishment of these model systems is technically challenging, and levels of human immune system development reported in the literature are variable between laboratories. The use of standard protocols for optimal engraftment of HSC and for monitoring the development of the human immune systems would enable more direct comparisons between humanized mice generated in different laboratories. Here we describe a standard protocol for the engraftment of human HSC into 21-day old NOD-scid IL2r γ ( NSG) mice using an intravenous injection approach. The multi-parameter flow cytometry used to monitor human immune system development and the kinetics of development are described.


ACKNOWLEDGEMENT

This work was supported by National Institutes of Health Grants AI046629, AI112321, DK104218, CA034196, and OD018259 and grants from the Helmsley Charitable Trust and the Juvenile Diabetes Research Foundation. The authors would like to thank to TUBITAK (The Scientific and Technological Research Council of Turkey) for the research support (2214A). MAB and DLR are consultants for the Jackson Laboratory (Bar Harbor, ME).The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official views of the National Institutes of Health.
Guidelines
INTRODUCTION

1.1 Immunodeficient Mice Bearing Mutations Within the IL2ry Gene as Recipients of Human HSC

The generation of immunodeficient mice that support the engraftment of human immune systems has enabled the in vivo study of human immune system development and function(1). Early efforts to engraft human immune systems into mice utilized the C.B-17 strain bearing the Prkdc scid ( scid, severe combined immune deficiency) mutation (2), which permitted low levels of human immune cell engraftment after injection with peripheral blood mononuclear cells (PBMC) or hematopoietic stem cells (HSC) but overall immune system function was limited (1). The development of NOD -scid mice (3) improved engraftment of human immune systems but overall function and levels of take were still suboptimal for the study of human immunobiology (4, 5). The introduction of immunodeficient Prkdc scid ( scid ), or recombination activating 1 or 2 gene ( Rag1 or Rag2 ) knockout mice bearing a mutated IL-2 receptor gamma chain ( IL2r γ null ) facilitated greatly the in vivo engraftment and function of human immune cells (6-9). The IL2rγ-chain is required for high affinity signaling of IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21 cytokines, and IL2r γ null mice have severe defects in NK-cell activity in addition to T and B cell development (10). These new strains of immunodeficient IL2r γ null mice are now being used for studies of human hematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology, and regenerative medicine (11).

1.2 HSC Engraftment of Immunodeficient NOD-scid IL2rγnull Mice

NOD.Cg-Prkdc scid Il2rg tm1Wjl /Sz (NODscid IL2r γ null or NSG) mice support engraftment of human HSC from a variety of sources, including G-CSF mobilized peripheral blood, bone marrow aspirates, umbilical cord blood, and fetal liver (12-15). In vivo human hematopoietic repopulation through transplantation of human CD34+ HSC in NSG recipients allows high levels of human HSC engraftment, differentiation of human T cells in the murine thymus and human B cells, differentiation of human myeloid subsets and human immune function in vivo (12-14, 16-20). A critical aspect for generating HSC-engrafted immunodeficient mice is the use of standardized protocols that enable consistent and robust human immune system development. For example, age of the mouse recipient has important implications for development of human immune cell subsets. One study has shown that newborn NSG mice support more efficient human T cell development after HSC injection than adult NSG mice (8-12 weeks) (21).

HSC-injection into newborn mice is challenging, as the injection sites (intrahepatic, intracardiac and facial vein) require technical expertise and in some instances survival is problematic (6). Here we describe a standard protocol for the engraftment of human HSC into 21-day old NSG mice using an intravenous injection approach. A description of the multi-parameter flow cytometry used to monitor human immune system development is shown. The kinetics of human immune system development in 21-day old NSG mice were compared to those in HSC-engrafted newborn NSG mice.


REFERENCES
CITATION
1. Shultz LD, et al (2012). Humanized mice for immune system investigation: progress, promise and challenges. Nat Rev Immunol. 2012;12(11):786–98.

CITATION
2. Bosma GC, Custer RP, Bosma MJ (1983). A severe combined immunodeficiency mutation in the mouse. Nature. 1983;301(5900):527–30.

CITATION
3. Shultz LD, et al. Multiple defects in innate and adaptive immunologic function in NOD/LtSz-scid mice. J Immunol. 1995;154(1):180–91..

CITATION
4. Greiner DL, Hesselton RA, Shultz LD. SCID mouse models of human stem cell engraftment. Stem Cells. 1998;16(3):166–77.

CITATION
5. Hesselton RM, et al. High levels of human peripheral blood mononuclear cell engraftment and enhanced susceptibility to human immunodeficiency virus type 1 infection in NOD/LtSz-scid/scid mice. J Infect Dis. 1995;172(4):974–82.

CITATION
6. Pearson T, Greiner DL, Shultz LD. Creation of "humanized" mice to study human immunity. Current protocols in immunology / edited by John E Coligan [et al] 2008 Chapter 15: Unit 15.21.

CITATION
7. Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol. 2007;7(2):118–30..

CITATION
8. Manz MG, Di Santo JP. Renaissance for mouse models of human hematopoiesis and immunobiology. Nat Immunol. 2009;10(10):1039–42.

CITATION
9. Sugamura K, et al. The interleukin-2 receptor gamma chain: its role in the multiple cytokine receptor complexes and T cell development in XSCID. Annu Rev Immunol. 1996;14:179–205.

CITATION
10. Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol. 2009;9(7):480–90.

CITATION
11. Brehm MA, Shultz LD, Greiner DL. Humanized mouse models to study human diseases. Curr Opin Endocrinol Diabetes Obes. 2010;17(2):120–5.

CITATION
12. Shultz LD, et al. Human lymphoid and myeloid cell development in NOD/LtSz-scid IL2R gamma null mice engrafted with mobilized human hemopoietic stem cells. J Immunol. 2005;174(10):6477–89.

CITATION
13. Ito M, et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood. 2002;100(9):3175–82.

CITATION
14. Ishikawa F, et al. Development of functional human blood and immune systems in NOD/SCID/IL2 receptor {gamma} chain(null) mice. Blood. 2005;106(5):1565–73.

CITATION
15. Kalscheuer H, et al. A model for personalized in vivo analysis of human immune responsiveness. Sci Transl Med. 2012;4(125):125ra30.

CITATION
16. Hiramatsu H, et al. Complete reconstitution of human lymphocytes from cord blood CD34+ cells using the NOD/SCID/gammacnull mice model. Blood. 2003;102(3):873–80.

CITATION
17. Tanaka S, et al. Development of mature and functional human myeloid subsets in hematopoietic stem cell-engrafted NOD/SCID/IL2rgammaKO mice. J Immunol. 2012;188(12):6145–55.

CITATION
18. Traggiai E, et al. Development of a human adaptive immune system in cord blood cell-transplanted mice. Science. 2004;304(5667):104–7.

CITATION
19. Legrand N, Weijer K, Spits H. Experimental models to study development and function of the human immune system in vivo. J Immunol. 2006;176(4):2053–8.

CITATION
20. Brehm MA, et al. Parameters for establishing humanized mouse models to study human immunity: analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rgamma(null) mutation. Clin Immunol. 2010;135(1):84–98.

CITATION
21. Brehm MA, et al. Parameters for establishing humanized mouse models to study human immunity: Analysis of human hematopoietic stem cell engraftment in three immunodeficient strains of mice bearing the IL2rγnull mutation. Clinical Immunology. 2010;135(1):84–98.

CITATION
22. Rubinstein P, et al. Processing and cryopreservation of placental/umbilical cord blood for unrelated bone marrow reconstitution. Proc Natl Acad Sci U S A. 1995;92(22):10119–22..

CITATION
23. Feldman DL, Mogelesky TC. Use of Histopaque for isolating mononuclear cells from rabbit blood. J Immunol Methods. 1987;102(2):243–9.

CITATION
24. Rongvaux A, et al. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu Rev Immunol. 2013;31:635–74.

CITATION
25. Lan P, et al. Reconstitution of a functional human immune system in immunodeficient mice through combined human fetal thymus/liver and CD34+ cell transplantation. Blood. 2006;108(2):487–92.

CITATION
26. Covassin L, et al. Human immune system development and survival of NOD-scid IL2rgamma (NSG) mice engrafted with human thymus and autologous hematopoietic stem cells. Clin Exp Immunol. 2013.

CITATION
27. Brehm MA, et al. Overcoming current limitations in humanized mouse research. J Infect Dis. 2013;208(Suppl 2):S125–30.

Materials
1. Human Cord Blood Preparation (See NOTE 1)

1.1 Citrate phosphate dextrose anticoagulant solution, USP (CPD) blood pack unit (Fenwal, Lake Zurich, IL). UCB specimens were provided by the University of Massachusetts Memorial Umbilical Cord Blood Donation Program under Institutional Review Board (IRB) approval.

1.2 Hespan (6% Hetastarch / 0.9% NaCl Solution) (Braun Medical Inc, Bethlehem, PA). Hespan is used for volume reduction of UCB and preserves recovery of HSC (22).

1.3 Histopaque-1077 (Sigma-Aldrich, St Louis, MO, USA). Histopaque is a density medium and is used for separating mononuclear cells from blood (23).




1.7 RPMI 1640 (Gibco, Life technologies, Grand Island, NY USA)

1.8 50mL centrifuge tubes (BD Falcon, Franklin Lakes, NJ, USA)

1.9 Water bath

2. CD3 T cell Depletion (See NOTE 2)

2.1 Human CD3 microbead kit (Miltenyi Biotech, Auburn, CA, USA)

2.2 MidiMACS Separator (Miltenyi Biotech, Auburn, CA, USA)

2.3 MACS multistand (Miltenyi Biotech, Auburn, CA, USA)

2.4 LD columns (Miltenyi Biotech, Auburn, CA, USA)

2.5 MACS Buffer: PBS supplemented with 0.5% fetal bovine serum (FBS, Atlanta Biologicals, Lawrenceville, GA, USA), 2 mM EDTA, and sterilized with a vacuum flask (0.2μm filter).

3. Flow cytometry analysis

3.1 FACS buffer: PBS supplemented with 1% FBS, and 0.1% sodium azide.

3.2 Flow cytometry analysis of T cell depleted UCB

  • ReagentHuman CD3, Clone UCHT1BD BiosciencesCatalog #555333

  • ReagentHuman CD34, Clone 581BD BiosciencesCatalog #555821

  • ReagentHuman CD45, Clone HI30BD BiosciencesCatalog #555485

3.3 Fluorescent antibodies for screening of HSC-engrafted NSG mice.

  • ReagentAnti-Mouse CD45 (mLy5), Clone 30-F11BD BiosciencesCatalog #557235

  • ReagentAnti-Human CD45, Clone HI30BD BiosciencesCatalog #555485

  • ReagentAnti-Human CD3, Clone UCHT1BD BiosciencesCatalog #555333

  • ReagentAnti-Human CD20, Clone 2H7BD BiosciencesCatalog #556632

  • ReagentAnti-Human CD33, Clone WM53BD BiosciencesCatalog #562854

  • ReagentAnti-Human CD14, Clone HCD14BioLegendCatalog #325618

4. HSC injection

The materials listed below are necessary for intravenous injection of HSC into mice that are 21 to 28 days old. The materials used for injecting newborn mice with HSC have been listed in detail previously (6).

4.1 Immunodeficient mice: NOD-PrkdcscidIL2rgtm1Wjl(NSG) mice between 3 to 4 weeks of age. (See NOTE 3)

4.2 137Cs gamma irradiator

4.3 Autoclaved, filtered, ventilated device for holding mice during irradiation.

4.4 Amount1 mL syringe with 27-G needle

4.5 Heating pad or warming lamp

4.6 Tailvein restrainer for mice (Braintree Scientific, BrainTree, MA)




Note
NOTE 1
UCB is a reliable source of functional human HSC. Alternative sources include G-CSF mobilized peripheral blood, bone marrow aspirates and fetal liver (24). Each source will have differences in preparation, in cell yields and engraftment capacity and the characteristics of the developed human immune system.



Note
NOTE 2
The standard protocol for our laboratory is to use CD3 depleted UCB cells for injecting CD34+ HSC into mouse recipients. The depletion of CD3+ T cells is essential to prevent development of acute xenogeneic graft-versus-host disease (GVHD) in the mice. Alternatively, purified CD34+ cells can also be injected into the recipient mice (25). One advantage for the CD3 depletion approach is that accessory cells (CD34-negative) present within the UCB specimen have been shown to enhance engraftment of human HSC in immunodeficient mice (26).


Note
NOTE 3
Immunodeficient mice bearing mutations within the IL2-receptor common gamma chain are the ideal recipients for the engraftment of human HSC. NSG mice were developed at The Jackson Laboratory by back-crossing a complete null mutation at theIL2rglocus onto the NOD.Cg-Prkdcscid(NOD/SCID) strain (12). Alternative mouse strains bearing mutations within theIL2rglocus have been described previously (7, 24).


Before start
The protocols described below involve the manipulation of primary human cells. All work should be done in a standard laminar flow hood and with appropriate personal protective equipment. Waste materials should be disposed of using protocols approved by an Institutional Biosafety Committee. All mouse injections and handling should be done using protocols approved by an Institutional Animal Care and Use Committee.
Methods
Methods
Preparation of umbilical cord blood (UCB)

Allow histopaque and RPMI supplemented with 0.5% BSA to warm to room temperature.
Transfer UBC to Amount50 mL conical tubes (30 mls per tube).

Add hespan to each tube of UBC to a final concentration of 20% per volume and mix gently.
Incubate for 30 minutes at room temperature. The incubation period is to allow red blood cells (RBC) to sediment.


Remove bottom layer of RBC with Amount10 mL pipette, leaving approximately Amount2 mL of the RBC volume.

Add RPMI supplemented with 0.5% BSA to each tube to bring to a total volume of Amount30 mL .

Slowly underlay Amount14 mL of histopaque to each tube containing umbilical cord blood. Ensure a clear interface is maintained between the histopaque and the cell-containing medium.

Spin for 30 min at 300 × g at room temperature with centrifuge brake off.

Remove the top layer of plasma, leaving Amount2 mL volume above buffy coat layer.

Remove buffy coat layer, transfer to a new Amount50 mL conical tube.

Add Amount30 mL of RPMI supplemented with 0.5% BSA and centrifuge for 5 min 400 × g and Temperature4 °C .

Discard supernatant

Pool pellets from each Amount50 mL conical tube in a total volume of 10 mls of RPMI supplemented with 0.5% BSA. (See NOTE 4)


Note
NOTE 4
DNAse (15 ul 10,000 U/ml DNAse/30 ml) can be added if pellet is viscous after resuspension.

Perform viability count.
Reserve 100,000 cells for flow cytometric analysis.
Centrifuge cells for 5 min at 400 × g and Temperature4 °C and proceed to step 2.

Depletion of CD3+ T cells

Our laboratory uses reagents from Miltenyi Biotech to deplete human CD3+ T cells from UCB samples. The manufacture’s protocol was followed for depletion of human T cells (See NOTE 2)
Resuspend CD3-depleted UCB cells in 1 ml RPMI supplemented with 0.5% BSA and perform viability counts.
To validate depletion of CD3+ T cells, stain recovered cells with antibodies specific for human CD3 and human CD45. T cell levels (cells staining double positive for CD3 and human CD45) should be below 0.5% of total cells. Cells saved from step 1.15 should be used as a positive staining control.


To determine the CD34+ HSC levels, stain the recovered cells with antibodies specific for human CD34 and human CD45. HSC are identified as CD45 dim and CD34 positive.
Resuspend cells in RPMI supplemented with 0.5% BSA. Keep cells on ice until injection.
HSC injection

The protocol for injecting newborn mice with HSC has been described in detail previously (6). Here we will focus on intravenous injection of HSC into 21 to 28 day old mice.
Irradiate recipient mice by whole body gamma irradiation. For young (21 to 28 days old) NSG mice a conditioning dose of 100 cGy is normally well tolerated and enables efficient engraftment. For engraftment of newborn NSG mice, 100 cGy is also used. HSC injection should be performed between four and 24 hours after irradiation.
Warm recipient mice using warming lamp.
Inject CD3-depleted UCB cells to obtain a total of 1×105CD34+ cells per injection into the lateral tail vein. For newborn mice show in here, 5×104CD34+ cells were injected by intracardiac route into 24 to 72 hour old NSG pups.
Ensure that bleeding from injection site has stopped and return recipient mouse to cage.
Evaluation of Human Cell Chimerism Levels by Flow Cytometry

HSC-engrafted NSG mice are normally screened for human cell chimerism levels in peripheral blood between 12 and 15 weeks post-injection. This evaluation is easily done by flow cytometric analysis of peripheral blood cells to validate human immune system development. For analysis, all cells positive for mouse CD45 are excluded from the gating strategy. Human immune cells are identified as staining positive for human CD45 and subpopulations are defined from those cells. The immune cell subsets that develop will be dependent on age of the recipient mouse and the specific time point evaluated. The results shown below compare human immune system development between HSC-engrafted newborn NSG mice and HSC-engrafted young (21-day old) NSG mice.
Levels of human CD45+ cells. Newborn and 21-day old NSG mice were irradiated and injected IV by intracardiac route (newborn) or via the tail vein (3 weeks old) with T cell depleted UCB containing 1×105CD34+cells as described above. Between 6 to 18 weeks after HSC injection, the percentages of human CD45+ cells in the blood were determined by flow cytometry (Figure 1). Each shape represents an individual animal and mice injected with the same HSC are identified by symbols of similar shape and color. Higher proportions of human CD45+ cells were detected in the blood of 21-day old, HSC-engrafted NSG mice at 6, 9,12 and 18 weeks as compared to newborn mice (Figure 1). In addition higher levels of human CD45+ cells were detected in the spleen and bone marrow of the 21-day old NSG mice compared with levels of circulating cells (Figure 2). These results show that 21-day old NSG mice and newborn NSG mice support HSC engraftment and that 21-day old mice show faster kinetics of human immune system development.

Figure 1. Human cell chimerism levels in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD45 positive cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 1. Human cell chimerism levels in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD45 positive cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 2. Human cell chimerism levels in spleen and bone marrow of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. 18 weeks after HSC injection, the percentages and number of human CD45+ cells in the spleen and percentages in the bone marrow were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 2. Human cell chimerism levels in spleen and bone marrow of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. 18 weeks after HSC injection, the percentages and number of human CD45+ cells in the spleen and percentages in the bone marrow were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).

Human T cell development. The kinetics of human CD3+ T cell development were compared between newborn and 21-day old NSG mice that had been injected with human HSC as described above (Figure 3.) The levels of human T cells detected in the peripheral blood were low at the 6 and 9 week time points for both groups of HSC-engrafted NSG mice and began to increase by week 12. At weeks 15 and 18 the levels of T cells were significantly higher in newborn NSG mice as compared to 21-day old NSG mice. These data suggest that both newborn and 21-day old NSG mice support human T cells development, but the kinetics are accelerated in mice injected as newborns (See NOTE 5)


Note
NOTE 5
Immunodeficient mice expressing HLA class I and II have been used support HLA-rerstricted human T cell development and are commercially available (27).

Figure 3. Human CD3+ T cell development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD3 positive T cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 3. Human CD3+ T cell development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD3 positive T cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).

B cell development. The kinetics of human CD20+ B cell development were compared between newborn and 21-day old NSG mice that had been injected with human HSC as described above (Figure 4). Both newborn and 21-day old injected NSG mice supported B cell development and significantly higher levels were detected in 21-day old mice at 6, 15 and 18 weeks. Overall a significant proportion of the CD45+ cells in NSG mice were human B cells at all time points.

Figure 4. Human CD20+ B cell development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD20 positive T cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 4. Human CD20+ B cell development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 18 weeks after HSC injection, the percentages of human CD20 positive T cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).

Monocyte/Macrophage development. The kinetics of human CD14+/CD33+ monocyte/macrophage development were compared between newborn and 21-day old NSG mice that had been injected with human HSC as described above (Figure 5). Both newborn and 21-day old NSG mice supported low levels of human monocyte/macrophage development with slightly higher levels detected in 21-day old mice at the 15 week time point. At all time points tested CD14+/CD33+ cells were detectable in the peripheral blood.

Figure 5. Human monocyte/macrophage development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 15 weeks after HSC injection, the percentages of human CD14/CD33+ positive myeloid cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).
Figure 5. Human monocyte/macrophage development in peripheral blood of NSG mice engrafted with human HSC

Newborn and 21-day old NSG mice were irradiated and injected IV via intracardiac route (newborn) or the tail vein (21-day old) with T cell depleted UCB containing CD34+cells. At 6– 15 weeks after HSC injection, the percentages of human CD14/CD33+ positive myeloid cells were determined by flow cytometry. Each shape represents an individual animal, and points of the same color and shape are from independent experiments (N=3).