8
Viral pathogenesis in hu-PBL-SCID mice Donald E. Mosier The transplantation of human cells into immunodeficient mice has provided new models for human immune function, infection by pathogenic viruses that grow in lymphocytes or other hematopoietic cells, and development of hematopoietic lineages. SCID mice reconstituted with adult peripheral blood mononuclear cells (hu-PBL-SCID mice) maintain some established immune responses, but do not produce a full spectrum of primary cellular or humoral responses. Nonetheless, hu-PBL-SCID mice are a valuable tool for studying primary infection with human immunodeficiency virus (HIV) and reactivation of latent Epstein-Barr virus (EBV) infection. This review will summarize findings from such studies. Key words: animal models / EBV / HIV / viral pathogenesis ©1996 Academic Press Ltd THERE WAS GREAT EXCITEMENT in the fall of 1988 when three different laboratories reported the successful transplantation of adult or fetal human cells to immunodeficient SCID or beige.nude.xid mice. 1-3 Two of the models, ours 1 and McCune’s, 2 were motivated by the desire to create a small animal model to study HIV infection. 4-10 The study by Kamel-Reid and Dick 3 had as its goal the development of human hematopoietic precursors in immunodeficient mouse bone marrow. 11-13 The transfer of mature peripheral blood mononuclear cells to SCID mice gave hope of establishing an adoptively transferred human immune response that would reproduce the full spectrum of human immunity, while the transplanta- tion of hematopoietic progenitors and fetal thymus might allow the development of lymphoid and other hematopoietic lineages and generate a stable human– mouse xenochimera. Now, almost a decade later, we see that the early expectations have been partially fulfilled. As is detailed in the rest of this issue, a fully functional human immune response has yet to be documented, but adoptive transfer of human immu- nity has been demonstrated. 14-17 Sustained, multi- lineage engraftment of human hematopoietic cells has been achieved. 13,18-20 As if in answer to the original motivation for their creation, xenotransplant models have proven to be remarkably useful for studying HIV-1 infection. Moreover, mice trans- planted with adult PBL develop EBV-related B-cell lymphoproliferative disease 21-24 that is remarkably similar to a subset of AIDS-associated lymphomas. 25,26 The primary infection of hu-PBL-SCID mice with variants of HIV-1 has given insight into pathogenesis and immune suppression of infection. Likewise, the reactivation of EBV and its in-vivo transformation of human B lymphocytes has provided new insights into the pathogenesis of B-cell lymphomas. Persistence of human cells in the hu-PBL-SCID model The intraperitoneal injection of human peripheral blood mononuclear cells into SCID mice leads to the selective survival and expansion of human CD3 + T cells, with smaller numbers of human B cells, mono- cytes and NK cells surviving. 27,28 CD3 + T cells rapidly show signs of activation and selective expansion of CD45RO + memory T cells in both the CD4 + and CD8 + subsets. We have recently observed that anti- bodies to CD40 ligand 29 block T-cell activation and substantially reduce engraftment of human cells in SCID mice (M. Eckert, I. Atencio, D. Mosier, unpub- lished observations), suggesting that T-cell activation is essential for the success of the human graft. This may occur because of recognition of mouse xenoanti- gens 30,31 on human antigen presenting cells (B cells or monocytes), although it is not clear that xenoanti- gens are the sole source of T-cell stimulation. We and others have observed fatal graft-versus-host disease when adult PBL are transplanted to neonatal SCID mice. 32,33 Conversely, human cord blood cells are more easily engrafted in newborn SCID mice than in adults. 32 These observations suggest that antigenic priming has occurred in adult donors leading to a From the Department of Immunology, IMM7, The Scripps Research Institute, 10666 North Torrey Pines Road, La Jolla, CA 92037, USA seminars in IMMUNOLOGY, Vol 8, 1996: pp 255–262 ©1996 Academic Press Ltd 1044-5323/96/040255 + 08$18.00/0 255

Viral pathogenesis in hu-PBL-SCID mice

Embed Size (px)

Citation preview

Page 1: Viral pathogenesis in hu-PBL-SCID mice

Viral pathogenesis in hu-PBL-SCID miceDonald E. Mosier

The transplantation of human cells into immunodeficientmice has provided new models for human immune function,infection by pathogenic viruses that grow in lymphocytes orother hematopoietic cells, and development of hematopoieticlineages. SCID mice reconstituted with adult peripheral bloodmononuclear cells (hu-PBL-SCID mice) maintain someestablished immune responses, but do not produce a fullspectrum of primary cellular or humoral responses.Nonetheless, hu-PBL-SCID mice are a valuable tool forstudying primary infection with human immunodeficiencyvirus (HIV) and reactivation of latent Epstein-Barr virus(EBV) infection. This review will summarize findings fromsuch studies.

Key words: animal models / EBV / HIV / viralpathogenesis

©1996 Academic Press Ltd

THERE WAS GREAT EXCITEMENT in the fall of 1988 whenthree different laboratories reported the successfultransplantation of adult or fetal human cells toimmunodeficient SCID or beige.nude.xid mice.1-3

Two of the models, ours1 and McCune’s,2 weremotivated by the desire to create a small animal modelto study HIV infection.4-10 The study by Kamel-Reidand Dick3 had as its goal the development of humanhematopoietic precursors in immunodeficient mousebone marrow.11-13 The transfer of mature peripheralblood mononuclear cells to SCID mice gave hope ofestablishing an adoptively transferred humanimmune response that would reproduce the fullspectrum of human immunity, while the transplanta-tion of hematopoietic progenitors and fetal thymusmight allow the development of lymphoid and otherhematopoietic lineages and generate a stable human–mouse xenochimera. Now, almost a decade later, wesee that the early expectations have been partiallyfulfilled. As is detailed in the rest of this issue, a fullyfunctional human immune response has yet to be

documented, but adoptive transfer of human immu-nity has been demonstrated.14-17 Sustained, multi-lineage engraftment of human hematopoietic cellshas been achieved.13,18-20 As if in answer to theoriginal motivation for their creation, xenotransplantmodels have proven to be remarkably useful forstudying HIV-1 infection. Moreover, mice trans-planted with adult PBL develop EBV-related B-celllymphoproliferative disease21-24 that is remarkablysimilar to a subset of AIDS-associated lymphomas.25,26

The primary infection of hu-PBL-SCID mice withvariants of HIV-1 has given insight into pathogenesisand immune suppression of infection. Likewise, thereactivation of EBV and its in-vivo transformation ofhuman B lymphocytes has provided new insights intothe pathogenesis of B-cell lymphomas.

Persistence of human cells in the hu-PBL-SCIDmodel

The intraperitoneal injection of human peripheralblood mononuclear cells into SCID mice leads to theselective survival and expansion of human CD3+ Tcells, with smaller numbers of human B cells, mono-cytes and NK cells surviving.27,28 CD3+ T cells rapidlyshow signs of activation and selective expansion ofCD45RO+ memory T cells in both the CD4+ andCD8+ subsets. We have recently observed that anti-bodies to CD40 ligand29 block T-cell activation andsubstantially reduce engraftment of human cells inSCID mice (M. Eckert, I. Atencio, D. Mosier, unpub-lished observations), suggesting that T-cell activationis essential for the success of the human graft. Thismay occur because of recognition of mouse xenoanti-gens30,31 on human antigen presenting cells (B cellsor monocytes), although it is not clear that xenoanti-gens are the sole source of T-cell stimulation. We andothers have observed fatal graft-versus-host diseasewhen adult PBL are transplanted to neonatal SCIDmice.32,33 Conversely, human cord blood cells aremore easily engrafted in newborn SCID mice than inadults.32 These observations suggest that antigenicpriming has occurred in adult donors leading to a

From the Department of Immunology, IMM7, The ScrippsResearch Institute, 10666 North Torrey Pines Road, La Jolla, CA92037, USA

seminars in IMMUNOLOGY, Vol 8, 1996: pp 255–262

©1996 Academic Press Ltd1044-5323/96/040255 + 08$18.00/0

255

Page 2: Viral pathogenesis in hu-PBL-SCID mice

Rep

lica

tion

rat

e

Extent of CD4 T-cell depletionMacrophage tropic,non-cytopathic

T-cell tropic,low cytopathicity

T-cell tropic,high cytopathicity

JR-C

SF

BaL

SF

128A

UC

1

SF

162

SF

2∆N

EF

MN

SF

2

NL

4-3

∆ N

EF

SF

13

NL

4-3

LA

I

SF

33

89.6

∆NE

F

WE

AU

89.6

, 89.

6∆V

PU

, 89.

6∆V

PR

cross-reactive T-cell response to xenoantigens.34-36

This response leads to lethal graft-versus-host diseasein newborn SCID recipients, but it is required toestablish a successful graft in adult SCID mice. Cordblood T cells, which have few xenoreactive T cells, canonly survive in the more permissive environment ofthe newborn SCID recipient. Although one groupfinds all human T cells to be ‘anergic’ in hu-PBL-SCIDmice30,31 at later time points after engraftment, wefind that human T cells recovered from hu-PBL-SCIDmice at 4–6 weeks after reconstitution are stillresponsive to antigen or anti-CD3 stimulation [(ref27) and B. Torbett, D. Mosier, unpublishedobservations].

Human B cells survive in hu-PBL-SCID mice mainlyas differentiated plasma cells localized in local lymphnodes or cell adhesions to the peritoneal cavity.37

Human immunoglobulin production can occur forup to a year after PBL transplantation, and we haveobserved EBV-related B-cell lymphomas occurring aslate at 9 months after engraftment in some experi-ments. Although small numbers of human monocyte/macrophages persist in local lymphoid tissue, theymay be critical for establishment of HIV-1 infection(see later). There is no evidence for engraftment ofcirculating human stem cells following PBLinjection.

Human cells can be recovered not only from theperitoneal site of injection into hu-PBL-SCID mice,but also from lymph nodes that drain the peritonealcavity, the spleen, and bone marrow. Low numbers ofT cells are present in the peripheral blood and otherlymph nodes. We have not detected human cells inthe thymus, but they are often found in perithymiclymph nodes adherent to the thymic capsule. CD4+ Tcells usually exceed CD8+ T cells in lymph nodes,whereas the reverse is true in cells recovered byperitoneal lavage.

The extensive T-cell activation in the hu-PBL-SCIDmodel provides a ready target for HIV-1 infection, andmimics the extensive lymphocyte activation seen inchronic HIV-1 infection.38,39 Injection of HIV-1 into ahu-PBL-SCID mouse is thus not similar to a needlestick injury in a normal individual, but it is a relevantand important model for HIV-1 research as long asthe underlying biology is understood.

Consequences of HIV-1 infection in thehu-PBL-SCID model

HIV-1 infection of hu-PBL-SCID mice leads to theprimary consequence of HIV-1 infection of humans,

loss of CD4+ T cells.8,9 All patient and laboratoryisolates of HIV-1 and HIV-2 tested to date have beeninfectious in the hu-PBL-SCID model; this includesthree HIV-2 isolates, and HIV-1 clade B, D and Eisolates. Individual HIV-1 isolates differ in the effi-ciency of primary infection, the rate of viral replica-tion, and the rate of CD4+ T-cell depletion.9,40,41 Acomparison of many of the viruses we have studied ispresented in Figure 1, which shows rate of CD4+ T-celldepletion plotted versus replication rate and cyto-pathic effects (rapid cell killing, syncytial induction).The rate of CD4+ T-cell loss varies from very rapid(e.g. isolate 89.6 which causes total loss of CD4+ Tcells within 2 weeks of infection) to very slow (lessthan 25% reduction in CD4+ T cells at 4 weeks afterinfection). Few viruses establish infection withoutcausing CD4+ T-cell depletion. Two HIV-2 isolates, aswell as HIV-1SF2∆nef (nef accessory gene depletion)and the Mu1 envelope mutation42,43 fall into thiscategory. Although the HIV-2 isolates replicate well inthe hu-PBL-SCID model, the two mutated HIV-1isolates show low infectivity and replication.

Several findings are evident in the data presented inFigure 1. First, there is considerable variation between

Figure 1. The relationship between extent of CD4+ T-celldepletion caused by HIV infection of hu-PBL-SCID mice (Xaxis), the in-vivo replication rate of the virus isolate (Y axis),and the cell tropism and in-vitro cytopathic effects of thevirus (Z axis). Each of the virus isolates (designated on thecolumn) was employed in at least two replicate experimentswith five mice per infected group, and some of the viruses(SF162, SF33, NL4-3, 89.6) have been used in 5–10experiments. Overlapping columns indicate no differencesbetween the three viruses studied. There is a tendency forviruses with high replication and high cytopathicity to causerapid CD4+ T-cell depletion, but some notable exceptionsto this trend exist (e.g. HIV1 SF33). Deletion of the nefgene seems to have a more deleterious effect on a lesspathogenic isolate like SF2 than on a highly pathogenic onelike 89.6.

D. E. Mosier

256

Page 3: Viral pathogenesis in hu-PBL-SCID mice

different viral isolates in the hu-PBL-SCID model,which reflects the biologic heterogeneity of HIV-1 ingeneral.44 Second, there is a correlation betweenreplication rate and extent of CD4+ T-cell depletion,although several of the nef deletion mutants and theHIV-1SF33 isolate45 are exceptions to this generaltrend. Deletion of the nef gene slows CD4+ T-celldepletion, but does not prevent it except in the case ofthe poorly infectious SF2 mutant. These results aresimilar to those seen with nef deletion mutants ofSIV46 and those reported with nef deletion mutants inthe SCID-hu model.47 Thus, use of the hu-PBL-SCIDmodel yields a clear phenotype for nef deletionmutants as do other animal models. In contrast, thein-vitro phenotype of nef deletion mutants is oftensubtle, and has led to conflicting interpretations48-50

of nef function (hence its misnomer, negative effectfactor). Third, several non-cytopathic, macrophage-tropic HIV-1 and HIV-2 (UC1) isolates cause rapid lossof CD4+ T cells, so there is no consistent relationshipbetween in-vitro cytopathic effects and in-vivo patho-genicity.9 Patient isolates from rapid progressors[WEAU, 89.651,52] do cause very rapid loss of CD4+ Tcells in hu-PBL-SCID mice, however. We are thus inagreement with Kaneshima et al 53 that highly cyto-pathic isolates from rapid progressors often (but notalways; e.g. SF33) show a more severe phenotype inSCID-hu or hu-PBL-SCID mice. The other findingpresented in Figure 1 is that deletion of the viralaccessory genes vpr or vpu in the highly pathogenic89.6 HIV-1 isolate54 has no obvious impact on virusinfection and rate of CD4+ T-cell depletion in the hu-PBL-SCID model. This result is in agreement withrecently published findings of Aldrovandi et al 55 inthe SCID-hu thy/liver model.

Macrophage-tropism and HIV-1 transmissionefficiency

We have further investigated the basis of the surpris-ing rapid CD4+ T-cell depletion caused by infectionwith macrophage-tropic HIV-1 isolates. Macrophage-tropism refers to the ability of virus isolates to grow inprimary human macrophage/monocytes as well asprimary T cells but not in established T-cell lines ortumors. Cell tropism is determined by the gp120envelope gene.56-58 Analysis of recombinant virusesgenerated from HIV-1SF162 and HIV-1SF2

42,43,59 havemapped the rapid CD4+ T-cell depletion phenotypeto envelope, although mutations in the V3 loop hadvariable effects on viral replication and CD4+ T-cellloss in the hu-PBL-SCID model.41 Quantitative analy-

sis of viral load by proviral DNA copy number orplasma viral RNA content at different times afterinfection of hu-PBL-SCID mice with HIV-1SF162 andHIV-1SF33 has shown that mice infected with SF162show higher viral loads at days 7–9 after infection, butequal or lower virus burden at days 14–28 afterinfection [ref 9, and unpublished observations]. Themacrophage-tropic SF162 isolate thus appears to havean advantage in initial infection efficiency rather thanreplication rate when compared to SF33. Severaldifferent types of experiments suggest that macro-phage-tropic virus has a substantial advantage in theinitial cycle of virus replication in the hu-PBL-SCIDmodel. The experiments compared the macrophage-tropic SF162 isolate and the T-cell line-tropic SF33isolate (see Figure 1). First, the rate of CD4+ T-celldepletion caused by SF162 and SF33 differed sub-stantially if cell-free virus was used to infect mice, butthat difference disappeared if virus-infected cells wereused to transmit infection.40 Infected cells are knownto be highly efficient at transmitting virus.60 Second,cell-free virus stocks of SF33 prepared in mitogen-activated PBL or macrophage-depleted PBL wereindistinguishable in terms of infectious titer andability to cause CD4+ T-cell depletion in hu-PBL-SCIDmice. In contrast, SF162 prepared in the absence ofprimary macrophages was both less infectious thanvirus grown in unseparated PBL, and caused muchless CD4+ T-cell depletion in hu-PBL-SCID mice if thePBL graft had been macrophage-depleted prior toreconstitution of the SCID mice. These experimentssuggest that macrophage-tropic virus derived frominfected macrophages is much more easily trans-mitted than the same virus derived from infectedCD4+ T cells. However, virus derived from CD4+ Tcells can be pathogenic in the hu-PBL-SCID model ifit quickly finds a human macrophage/monocyte toinfect. These findings strongly suggest that macro-phage-derived HIV-1 is different than T-cell-derivedvirus. The structure of envelope,61,62 the level ofglycosylation,63-65 the state of oligomerization,66,67

and the acquisition of host cell membrane compo-nents68 could all contribute to these differences. Amodel highlighting these differences is presented inFigure 2.

Immunity to HIV-1 infection

One of the unique advantages of the hu-PBL-SCIDmodel is that the PBL graft can be derived fromrecently immunized donors, which permits the adop-tive transfer of an ongoing human immune response

HIV and EBV in hu-PBL-SCID mice

257

Page 4: Viral pathogenesis in hu-PBL-SCID mice

to the SCID mouse. This approach was used toevaluate resistance to HIV-1 infection in hu-PBL-SCIDmice derived from volunteers participating in HIV-1vaccine trials.69 Only a subset of donors had PBL thatwould adoptively transfer HIV-1 immunity to hu-PBL-SCID mice. These donors were initially immunizedwith recombinant gp160 envelope in a vacciniavector70 and were later reimmunized with recombi-nant gp160 alone.71 If PBL were collected within sixmonths of the last immunization, they were able toprotect hu-PBL-SCID mice against challenge with thevaccine strain of virus.69 Interestingly, the protectiveeffect correlated better with T-cell immune status thanwith neutralizing antibody in the donor. These resultsstimulated two studies of passive transfer of immunityto HIV-1 with cytotoxic T-lymphocyte (CTL) clones15

or neutralizing antibody.16 Transfer of high numbersof CTL was protective against HIV-1 challenge if CTLinjection preceded virus challenge. However, theprotective effect appeared to have two components:an HLA-restricted, cytotoxic effect and an antiviralactivity mediated by any activated CD8+ T-cell clone.

This finding suggests that the CD8+ T-cell antiviraleffect that is prominent in vitro72-74 can also bedetected in the hu-PBL-SCID animal model, and maybe an important component of cellular resistance toviral spread in humans. In a second study,16 therecombinant b12-IgG1 neutralizing antibody75,76 wasinjected into hu-PBL-SCID mice prior to challengewith the SF2 isolate of HIV-1. The antibody showedcomplete protection of the mice at doses between 3–7mg/kg. Substantial levels of neutralizing antibodythus appear to be required for sterilizing immunity,although lower amounts might slow virus spread.Koup and colleagues17,77 have performed similarstudies which are reviewed elsewhere in this volume.

Epstein-Barr virus and human B-celllymphoproliferative disease in hu-PBL-SCIDmice

The introduction of PBL from donors infected withEpstein-Barr virus (EBV) into SCID mice leads to ahigh incidence of EBV-associated lymphoproliferative

Figure 2. HIV produced by human macrophages is different from the same virus produced by CD4T cells. These differences are primarily in the envelope gp120 molecule, and perhaps acquired cellmembrane components. The CD4+ T-cell-derived virion is shown in the standard cartoon withsingle gp120 monomers displayed on the membrane [adapted from Greene, WC, ref 87 withAuthor’s permission], whereas the macrophage-derived virion has trimers of gp120 that have beendifferentially glycosylated and have different antibody-exposed epitopes. The small squiggly lineson the T-cell-derived virion represent host components, while the extensive globular surfacemolecules adjacent to the gp120 trimers represent much more extensive representation ofmacrophage host components. The most easily detected host components are MHC molecules, butother important surface components could be passively acquired and lead to altered infectivity(e.g. LFA-1, ICAM-1, other integrins, CD14, etc.).

D. E. Mosier

258

Page 5: Viral pathogenesis in hu-PBL-SCID mice

disease similar to that seen in immunosuppressedorgan transplant recipients78,79 or in late stageAIDS25,26 The use of the hu-PBL-SCID model hasgiven considerable insight into how this process mighttake place. A previously described23 model for thepathogenesis of EBV-driven lymphoproliferative dis-ease is presented in Figure 3. Activation of CD4+

T-cell lymphocytes is important in initiating theprocess. CD4 T cells are required for lymphoprolifer-ative disease (ref 80, our unpublished observations)and administration of anti-CD40 ligand (CD40L)antibodies blocks appearance of tumors (M. Eckert, I.Atencio, D. Mosier, unpublished observations). How-ever, both of these manipulations substantiallydecrease the success of the PBL reconstitution, andonly a few B lymphocytes survive two weeks afterinjection. Nonetheless, anti-CD40L treatment doesnot totally block tumor induction. We have observed areduction to 33% tumor incidence in hu-PBL-SCIDmice treated with anti-CD40L29 for the first six weeksafter PBL reconstitution. The latent period untiltumor detection increased from 35 ± 3 days to 56–274days. Some small number of human B cells mustsurvive without activated T-cell help for at least sixweeks, and these cells can eventually give rise to EBV-driven B-cell lymphoproliferative disease. A secondline of evidence that T-cell-dependent B-cell differ-entiation is a critical event in tumor formation is therestriction of lytic cycle EBV replication to plasmacy-

toid cells expressing high levels of CD38 and lowerlevels of CD23.24,81 The lymphoblastoid componentof hu-PBL-SCID tumors harbors EBV that is tightlylatent.24 The production of infectious EBV by plasma-cytoid cells may lead to the infection and transforma-tion of new B cells, thus establishing a self-renewingcycle of transformation.

There is heterogeneity in the ability of PBL fromEBV-positive donors to give rise to tumors.22,82 Severalfactors seem to contribute to the variable outcome.One is likely to be the strength of the CD8+ T-cellresponse to EBV and how well it is maintained inSCID mice. Rapid loss of control of EBV-infected cellsmay set up a situation analogous to immune suppres-sion associated with post-transplant lymphomas. Sec-ondary genetic events in individual B cells may add totheir tumorogenic potential.82,83 Variations in EBVfrom different patients may lead to different trans-forming potential in the hu-PBL-SCID model.84 Cer-tain EBV-positive donors whose PBL rarely give rise totumors clearly have EBV with low transforming activity(G. Picchio, R. Rochford, D. Mosier; unpublishedobservations). B cells infected with EBV with deletionsin the critical EBNA-2 gene are attenuated for tumorformation in hu-PBL-SCID mice.85 Finally, the naturalkiller cells present in SCID mice may vary in theirability to control early tumors. Tumor latency isshortened in hu-PBL-SCID beige mice,86 where thebeige mutation introduces impaired NK cell lysis. As

Figure 3. Hypothetical scheme of B-cell transformation caused by EBV activation in hu-PBL-SCIDmice23. CD4+ human T lymphocytes are activated in the SCID environment. They provide cognatehelp (see review by Ifversen and Borrebaeck, this issue) to resting B lymphocytes, a small numberof which harbor latent EBV episomes. This T-dependent B-cell activation leads to thedifferentiation of the B cell to a plasma cell, at which time cell division stops and the EBV lytic cycleis activated. The plasma cell then produces infectious EBV which can infect and transform otherB cells, with the virus remaining latent until another round of plasma cell differentiation takesplace.

HIV and EBV in hu-PBL-SCID mice

259

Page 6: Viral pathogenesis in hu-PBL-SCID mice

detailed by Amadori et al elsewhere in this issue, thehuman cytokine profile may impact the incidence oflymphoproliferative disease. We have noted that PBLfrom atopic individuals are more likely to causeantibody-mediated graft-versus-host disease (anti-platelet and anti-mouse RBC antibodies) in hu-PBL-SCID mice, so the fate of the B-cell graft may be quitedifferent depending on the human donor. Finally, wehave noted differences in IL-10 and IL-6 levels intumors derived from different donors (R. Rochford,G. Picchio, D. Mosier; unpublished observations),suggesting that intrinsic cytokine production could berelated to tumor progression. Despite these variables,the study of the interaction of EBV and differentiatinghuman B lymphocytes in the hu-PBL-SCID modelremains a fertile area of investigation.

Acknowledgements

The work cited in this review is the result of experimentsperformed by several individuals, including R. Gulizia, G.Picchio, R. Rochford, B. Torbett, R. Van Kuyk, P. Parren, J.Glynn, Y. Ling, M. Eckert and I. Atencio. Many collaboratorshave supplied virus isolates and provided helpful discussion.These include J. Levy, D. Trono, R. Collman, G. Shaw and H.Kestler. This work was partially supported by NIH grantsAI29182 and CA65391, the Leukemia Society (R.R.), andthe University of California AIDS Research Program (J.G.).This is publication number 10085-IMM from The ScrippsResearch Institute.

References

1. Mosier DE, Gulizia RJ, Baird SM, Wilson DB (1988) Transfer ofa functional human immune system to mice with severecombined immunodeficiency. Nature (London) 335:256-259

2. McCune JM, Namikawa R, Kaneshima H, Shultz LD, LiebermanM, Weissman IL (1988) The SCID-hu mouse: murine model forthe analysis of human hematolymphoid differentiation andfunction. Science 241:1632-1639

3. Kamel-Reid S, Dick JE (1988) Engraftment of immune-deficient mice with human hematopoietic stem cells. Science242:1706-1709

4. Namikawa R, Kaneshima H, Lieberman M, Weissman IL,McCune JM (1988) Infection of the SCID-hu mouse by HIV-1.Science 242:1684-1686

5. McCune JM, Namikawa R, Lieberman M, Weissman I, Kane-shima H (1990) The SCID-hu mouse as a model system for HIVinfection in Human Retroviruses, pp 347-359. Alan R. Liss, Inc.,New York, NY

6. Bonyadi M, Rabin L, Salimi S, Brown D, Kosek J, McCune J,Kaneshima H (1993) HIV induces thymus depletion in vivo.Nature 363:728-732

7. Su L, Kaneshima H, Bonyhadi M, Salimi S, Kraft D, Rabin L,McCune J (1995) HIV-1-induced thymocyte depletion is asso-ciated with indirect cytopathogenicity and infection of progeni-tor cells in vivo. Immunity 2:25-36

8. Mosier DE, Gulizia RJ, Baird SM, Wilson DB, Spector DH,Spector SA (1991) Human immunodeficiency virus infection ofhuman-PBL-SCID mice. Science 251:791-794

9. Mosier D, Gulizia R, MacIsaac P, Torbett B, Levy J (1993) Rapidloss of CD4 + T cells in human-PBL-SCID mice by non-cytopathic HIV isolates. Science 260:689-692

10. Mosier DE, Gulizia RJ, Baird SM, Spector S, Spector D, KippsTJ, Fox RI, Carson DA, Cooper N, Richman DD, Wilson DB(1989) Studies of HIV infection and the development ofEpstein-Barr virus-related B cell lymphomas following transferof human lymphocytes to mice with severe combined immuno-deficiency, in Current Topics in Microbiology and Immunologypp 195-199. Springer, Berlin-Heidelberg

11. Dick JE, Plfumio F, Lapidot T (1991) Mouse models for humanhematopoiesis. Semi-Immunol 3:367-378

12. Kamel-Reid S, Dick JE, Greaves A, Murdoch B, Doedens M,Grunberger T, Thorner P, Freedman MH, Phillips RA, LetarteM (1992) Differential kinetics of engraftment and induction ofCD10 on human pre-B leukemia cell lines in immune deficientscid mice. Leukemia 6:8-17

13. Lapidot T, Pflumio F, Doedens M, Murdoch B, Williams DE,Dick JE (1992) Cytokine stimulation of multilineage hemato-poiesis from immature human cells engrafted in SCID mice.Science 255:1137-1141

14. Mosier DE, Gulizia RJ, MacIsaac P, Mathieson BJ, Smith G, HuSL, Corey L, Greenberg P (1992) Evaluation of gp160 vaccineesin the hu-PBL-SCID mouse model. AIDS Res Hum Retroviruses8:1387

15. van Kuyk R, Torbett BE, Gulizia RJ, Leath S, Mosier DE, KoenigS (1994) Cloned human CD8 + cytotoxic T lymphocytesprotect human peripheral blood leukocyte-severe combinedimmunodeficient mice from HIV-1 infection by an HLA-unrestricted mechanism. J Immunol 153:4826-4833

16. Parren P, Ditzel H, Gulizia R, Binley J, Barbas CI, Burton D,Mosier D (1995) Protection against HIV-1 infection in hu-PBL-SCID mice by passive immunization with a neutralizing humanmonoclonal antibody against the gp120 CD4-binding site. AIDS9:1-6

17. Safrit JT, Fung MS, Andrews CA, Braun DG, Sun WN, ChangTW, Koup RA (1993) hu-PBL-SCID mice can be protected fromHIV-1 infection by passive transfer of monoclonal antibody tothe principal neutralizing determinant of envelope gp120. Aids7:15-21

18. Kyoizumi S, Baum CM, Kaneshima H, McCune JM, Yee EJ,Namikawa R (1992) Implantation and maintenance of func-tional human bone marrow in SCID-hu mice. Blood79:1704-1711

19. Chen BP, Galy A, Kyoizumi S, Namikawa R, Scarborough J,Webb S, Ford B, Cen DZ, Chen SC (1994) Engraftment ofhuman hematopoietic precursor cells with secondary transferpotential in SCID-hu mice. Blood 84:2497-2505

20. Fraser CC, Kaneshima H, Hansteen G, Kilpatrick M, HoffmanR, Chen BP (1995) Human allogeneic stem cell maintenanceand differentiation in a long-term multilineage SCID-hu graft.Blood 86:1680-1693

21. Mosier DE, Picchio GR, Baird SM, Kobayashi R, Kipps TJ(1992) Epstein-Barr virus-induced human B-cell lymphomas inSCID mice reconstituted with human peripheral blood leuko-cytes. Cancer Res 52:5552s-5553s

22. Picchio GR, Kobayashi R, Kirven M, Baird SM, Kipps TJ, MosierDE (1992) Heterogeneity among Epstein-Barr virus-seroposi-tive donors in the generation of immunoblastic B-cell lympho-mas in SCID mice receiving human peripheral blood leukocytegrafts. Cancer Res 52:2468-2477

D. E. Mosier

260

Page 7: Viral pathogenesis in hu-PBL-SCID mice

23. Rochford R, Mosier D (1994) Immunobiology of Epstein-Barrvirus-associated lymphomas. Clin Immunol Immunopath71:256-259

24. Rochford R, Mosier DE (1995) Differential Epstein-Barr virusgene expression in B-cell subsets recovered from lymphomas inSCID mice after transplantation of human peripheral bloodlymphocytes. J Virol 69:150-155

25. Neri A, Barriga F, Inghirami G, Knowles DM, Neequaye J,Magrath IT, Dalla-Favera R (1991) Epstein-Barr virus infectionprecedes clonal expansion in Burkitt’s and acquired immuno-deficiency syndrome-associated lymphoma. Blood77:1092-1095

26. McGrath MS, Shiramize B, Meeker TC, Kaplan LD, Herndier B(1991) AIDS-Associated Polyclonal Lymphoma: Identificationof a New HIV-Associated Disease Process. J AIDS 4:408-415

27. Torbett BE, Picchio G, Mosier DE (1991) hu-PBL-SCID mice: Amodel for human immune function, AIDS, and lymphomage-nesis. Immunol Rev 124:139-164

28. Carlsson R, Martensson C, Kalliomaki S, Ohlin M, BorrebaeckCAK (1992) Human peripheral blood lymphocytes trans-planted into SCID mice constitute an in vivo culture systemexhibiting several parameters found in a normal humoralimmune response and are a source of immunocytes for theproduction of human monoclonal antibodies. J Immunol148:1065-1071

29. Lederman S, Yellin MJ, Inghirami G, Lee JJ, Knowles DM, ChessL (1992) Molecular interactions mediating T-B lymphocytecollaboration in human lymphoid follicles. Roles of T cell-B-cell-activating molecule (5c8 antigen) and CD40 in contact-dependent help. J Immunol 149:3817-3838

30. Tary-Lehmann M, Saxon A (1992) Human mature T cells thatare anergic in vivo prevail in SCID mice reconstituted withhuman peripheral blood. J Exp Med 175:503-516

31. Tary-Lehmann M, Lehmann PV, Schols D, Roncarolo MG,Saxon A (1994) Anti-SCID mouse reactivity shapes the humanCD4 + T cell repertoire in hu-PBL-SCID chimeras. J Exp Med180:1817-1827

32. Reinhardt B, Torbett BE, Gulizia RJ, Reinhardt PP, Spector SA,Mosier DE (1994) Human immunodeficiency virus type 1infection of neonatal severe combined immunodeficient micexenografted with human cord blood cells. AIDS Res HumRetroviruses 10:131-141

33. Pflumio F, Lapidot T, Murdoch B, Patterson B, Dick JE (1993)Engraftment of human lymphoid cells into newborn SCID miceleads to graft-versus-host disease. Int Immunol 5:1509-1522

34. Alter BJ, Bach FH (1990) Cellular basis of the proliferativeresponse of human T cells to mouse xenoantigens. J Exp Med171:333-338

35. Lindahl KF, Bach FH (1976) Genetic and cellular aspects ofxenogeneic mixed leukocyte culture reaction. J Exp Med144:305-318

36. Swain SL, Dutton RW, Schwab R, Yamamoto J (1983) Xenoge-neic human anti-mouse T cell responses are due to the activityof the same functional T cell subsets responsible for allospecificand major histocompatiblity complex-restricted responses. JExp Med 157:720-729

37. Hoffmann-Fezer G, Kranz B, Gall C, Thierfelder S (1992)Peritoneal sanctuary for human lymphopoiesis in SCID miceinjected with human peripheral blood lymphocytes fromEpstein-Barr virus-negative donors. Eur J Immunol22:3161-3166

38. Gougeon M-L, Lecoeur H, Dulioust A, Enouf M-G, CrouvoisierM, Goujard C, Debord T, Montagnier L (1996) Programmedcell death in peripheral lymphocytes from HIV-infectedpatients. J Immunol 156:3509-3520

39. Giorgi JV, Detels R (1989) T-cell subset alterations in HIV-infected homosexual men: NIAID multicenter AIDS cohortstudy. Clin Immunol Immunopathol 52:10-18

40. Mosier D, Sieburg H (1994) Macrophage-tropic HIV: criticalfor AIDS pathogenesis? Immunol Today 15:332-339

41. Gulizia R, Levy J, Mosier D (1996) The envelope gp120 gene ofhuman immunodeficiency virus type 1 determines the rate ofCD4-positive T cell depletion in SCID mice engrafted withhuman peripheral blood leukocytes. J Virol 70:4184-4187

42. Shioda T, Levy JA, Cheng-Mayer C (1991) Macrophage and Tcell-line tropisms of HIV-1 are determined by specific regions ofthe envelope gp120 gene. Nature 349:167-169

43. Shioda T, Levy JA, Cheng-Mayer C (1992) Small amino acidchanges in the V3 hypervariable region of gp120 can affect theT-cell-line and macrophage tropism of human immunodefi-ciency virus type 1. Proc Natl Acad Sci USA 89:9434-9438

44. Levy JA (1993) Pathogenesis of human immunodeficiency virusinfection. Microbiol Rev 57:183-289

45. York-Higgins D, Cheng-Mayer C, Bauer D, Levy J, Dina D(1990) Human immunodeficiency virus type 1 cellular hostrange, replication and cytopathicity are linked to the enveloperegion of the viral genome. J Virol 64:4016-4020

46. Kestler H, Ringler D, Mori K, Panicali D, Sehgal P, Daniel M,Desrosiers R (1991) Importance of the nef gene for main-tenance of high virus loads and for development of AIDS. Cell65:651-662

47. Jamieson B, Aldrovandi G, Planelles V, Jowett J, Gao L, Bloch L,Chen I, Zack J (1994) Requirement of human immunodefi-ciency virus type 1 nef for in vivo replication and pathogenicity.J Virol 68:3478-3485

48. Zazopoulos E, Haseltine WA (1993) Effect of nef alleles onreplication of human immunodeficiency virus type 1. Virology194:20-27

49. Bandres JC, Ratner L (1994) Human immunodeficiency virustype 1 Nef protein down-regulates transcription factors NF-kappa B and AP-1 in human T cells in vitro after T-cell receptorstimulation. J Virol 68:3243-3249

50. Miller MD, Warmerdam MT, Gaston I, Greene WC, FeinbergMB (1994) The human immunodeficiency virus-1 nef geneproduct: a positive factor for viral infection and replication inprimary lymphocytes and macrophages. J Exp Med179:101-113

51. Li Y, Kappes J, Conway J, Price R, Shaw G, Hahn B (1991)Molecular characterization of human immunodeficiency virustype 1 cloned directly from uncultured human brain tissue:identification of replication-competent and -defective gen-omes. J Virol 65:3973-3985

52. Collman R, Balliet JW, Gregory SA, Friedman H, Kolson DL,Nathanson N, Srinivasan A (1992) An infectious molecularclone of an unusual macrophage-tropic and highly cytopathicstrain of human immunodeficiency virus type 1. J Virol66:7517-7521

53. Kaneshima H, Su L, Bonyhadi ML, Connor RI, Ho DD,McCune JM (1994) Rapid-high, syncytium-inducing isolates ofhuman immunodeficiency virus type 1 induce cytopathicity inthe human thymus of the SCID-hu mouse. J Virol68:8188-8192

54. Balliet JW, Kolson DL, Eiger G, Kim FM, McGann KA,Srinivasan A, Collman R (1994) Distinct effects in primarymacrophages and lymphocytes of the human immunodefi-ciency virus type 1 accessory genes vpr, vpu, and nef: mutationalanalysis of a primary HIV-1 isolate. Virology 200:623-631

55. Aldrovandi G, Zack J (1996) Replication and pathogenicity ofhuman immunodeficiency virus type 1 accessory gene mutantsin SCID-hu mice. J Virol 70:1505-1511

56. Hwang SS, Boyle TJ, Lyerly HK, Cullen B (1991) Identificationof the envelope V3 loop as the primary determinant of celltropism in HIV-1. Science 253:71-74

57. Westervelt P, Trowbridge D, Epstein L, Blumberg B, Li Y, HahnB, Shaw G, Price R, Ratner L (1992) Macrophage tropismdeterminants of human immunodeficiency virus type 1 in vivo.J Virol 66:2577-2582

HIV and EBV in hu-PBL-SCID mice

261

Page 8: Viral pathogenesis in hu-PBL-SCID mice

58. Chesebro B, Wehrly K, Nishio J, Perryman S (1992) Macro-phage-tropic human immunodeficiency virus isolates fromdifferent patients exhibit unusual V3 envelope sequencehomogeneity in comparison to T cell-tropic isolates: definitionof critical amino acids involved in cell tropism. J Virol66:6547-6554

59. Cheng-Mayer C, Shioda T, Levy JA (1991) Host range,replicative, and cytopathic properties of human immunoefi-ciency virus type 1 are determined by very few amino acidchanges in tat and gp120. J Virol 65:6931-6941

60. Dimitrov DS, Willey RL, Sato H, Chang LJ, Blumenthal RMartin MA (1993) Quantitation of human immunodeficiencyvirus type 1 infection kinetics. J Virol 67:2182-2190

61. Sullivan N, Thali M, Furman C, Ho DD, Sodroski J (1993)Effect of amino acid changes in the V1/V2 region of the humanimmunodeficiency virus type 1 gp120 glycoprotein on subunitassociation, syncytium formation, and recognition by a neutral-izing antibody. J Virol 67:3674-3679

62. Sullivan N, Sun Y, Li J, Hofmann W, Sodroski J (1995)Replicative function and neutralization sensitivity of envelopeglycoproteins from primary and T-cell line-passaged humanimmunodeficiency virus type 1 isolates. J Virol 69:4413-4422

63. Cheng-Mayer C, Seto D, Levy JA (1991) Altered host range ofHIV-1 after passage through various human cell types. Virology181:288-294

64. Wu Z, Kayman SC, Honnen W, Revesz K, Chen H, Vijh-WarrierS, Tilley SA, McKeating J, Shotton C, Pinter A (1995)Characterization of neutralization epitopes in the V2 region ofhuman immunodeficiency virus type 1 gp120: role of glycosyla-tion in the correct folding of the V1/V2 domain. J Virol69:2271-2278

65. Fenouillet E, Jones IM (1995) The glycosylation of humanimmunodeficiency virus type 1 transmembrane glycoprotein(gp41) is important for the efficient intracellular transport ofthe envelope precursor gp160. J Gen Virol 76:1509-1514

66. Moore JP, McKeating JA, Huang YX, Ashkenazi A, Ho DD(1992) Virions of primary human immunodeficiency virus type1 isolates resistant to soluble CD4 (sCD4) neutralization differin sCD4 binding and glycoprotein gp120 retention from sCD4-sensitive isolates. J Virol 66:235-243

67. Moore JP, Cao Y, Qing L, Sattentau QJ, Pyati J, Koduri R,Robinson J, Barbas III CF, Burton DR, Ho DD (1995) Primaryisolates of human immunodeficiency virus type 1 are relativelyresistant to neutralization by monoclonal antibodies to gp120,and their neutralization is not predicted by studies withmonomeric gp120. J Virol 69:101-109

68. Arthur L, Bess J, Jr, Sowder R, II, Benveniste R, Mann D,Chermann J-C, Henderson L (1992) Cellular proteins bound toimmunodeficiency viruses: implications for pathogenesis andvaccines. Science 258:1935-1938

69. Mosier D, Gulizia R, MacIsaac P, Corey L, Greenberg P (1993)Resistance to human immunodeficiency virus 1 infection ofSCID mice reconstituted with peripheral blood leukocytes fordonors vaccinated with vaccinia gp160 and recombinant gp160.Proc Natl Acad Sci USA 90:2443-2447

70. Hu S-L, Kosowshi SG, Dalrymple JM (1986) Expression of AIDSvirus envelope gene in recombinant vaccinia viruses. Nature320:537-540

71. Redfield RR, Birx DL, Ketter N, Tramont E, Polonis V, Davis C,Brundage JF, Smith G, Johnson S, Fowler A, Wierzba T,Shafferman A, Volvovitz F, Oster C, Burke DS (1991) A phase Ievaluation of the safety and immunogenicity of vaccination withrecombinant gp160 in patients with early human immunodefi-ciency virus infection. N Engl J Med 324:1677-1684

72. Walker CM, Moody DJ, Stites DP, Levy JA (1989) CD8 Tlymphocyte control of HIV replication in cultured CD4 cellsvaries among infected individuals. Cell Immunol 119:470-475

73. Levy JA (1991) Viral and cellular factors influencing HIVtropism. Adv Exp Med Biol 300:1-15

74. Mackewicz CE, Ortega HW, Levy JA (1991) CD8 + cell anti-HIVactivity correlates with the clinical state of the infectedindividual. J Clin Invest 87:1462-1466

75. Burton DR, Pyati J, Koduri R, Sharp SJ, Thornton GB, ParrenPW, Sawyer LS, Hendry RM, Dunlop N, Nara PL, Lamacchia M,Garratty E, Stiehm ER, Bryson YJ, Cao Y, Moore JP, Ho DD,Barbas CF (1994) Efficient neutralization of primary isolates ofHIV-1 by a recombinant human monoclonal antibody. Science266:1024-1027

76. Roben P, Moore JP, Thali M, Sodroski J, Barbas 3rd CF, BurtonDR (1994) Recognition properties of a panel of humanrecombinant Fab fragments to the CD4 binding site of gp120that show differing abilities to neutralize human immunodefi-ciency virus type 1. J Virol 68:4821-4828

77. Gauduin MC, Safrit JT, Weir R, Fung MS, Koup RA (1995) Pre-and postexposure protection against human immunodefi-ciency virus type 1 infection mediated by a monoclonalantibody. J Infect Dis 171:1203-1209

78. Nalesnik MA, Jaffe R, Starzl TE, Demetris AJ, Porter K,Burnham JA, Makowka L, Ho M, Locker J (1988) Thepathology of post-transplant lymphoproliferative disordersoccurring in the setting of cyclosporine A-prednisone immuno-suppression. Am J Pathol 133:173-192

79. Locker J, Nalesnik M (1989) Molecular genetic analysis oflymphoid tumors arising after organ transplantation. Am JPathol 135:977-987

80. Veronese ML, Veronesi A, D’Andrea E, Del Mistro A, Indrac-colo S, Mazza MR, Mion M, Zamarchi R, Menin C, Panozzo M,Amadori A, Chieco-Bianchi L (1992) Lymphoproliferativedisease in human peripheral blood mononuclear cell-injectedSCID mice. I. T lymphocyte requirement for B cell tumorgeneration. J Exp Med 176:1763-1767

81. Rochford R, Hobbs MV, Garnier J-L, Cooper NR, Cannon MJ(1993) Plasmacytoid differentiation of Epstein-Barr virus-trans-formed B cells in vivo is associated with reduced expression ofviral latent genes. Proc Natl Acad Sci, USA 90:352-356

82. Picchio GR, Cohen JI, Wyatt ER, Mosier DE (1993) Enhancedtumorigenicity of an Epstein-Barr virus-transformed lympho-blastoid cell line is associated with a unique 1:18 chromosomaltranslocation and decreased expression of lymphocyte functionassociated antigen-1a (CD11a). Am J Pathol 143:342-349

83. Thangavelu M, Snyder L, Anastasi J, Le Beau MM, Kirven M,Picchio G, Mosier DE, Rowley JD (1992) Cytogenetic character-ization of B-cell lymphomas from severe combined immunode-ficiency disease mice given injections of lymphocytes fromEpstein-Barr virus-positive donors. Cancer Res 52:4678-4681

84. Menin C, Ometto L, Veronesi A, Montagna M, Coppola V,Veronese ML, Indraccolo S, Bruni L, Corneo B, Amadori A, DeRossi A, Chieco-Bianchi L, D’Andrea E (1995) Dominance of asingle Epstein-Barr virus strain in SCID-mouse tumors inducedby injection of peripheral blood mononuclear cells fromhealthy human donors. Virus Res 36:215-231

85. Cohen JI, Picchio GR, Mosier DE (1992) Epstein-Barr virusnuclear protein 2 is a critical determinant for tumor growth inSCID mice and transformation in vitro. J Virol 66:7555-7559

86. Mosier DE, Stell KL, Gulizia RJ, Torbett BE, Gilmore GL (1993)Homozygous scid/scid;beige/beige mice have low levels ofspontaneous or neonatal T cell-induced B cell generation. JExp Med 177:191-194

87. Greene WC (1991) The molecular biology of human immuno-deficiency virus type 1 infection. N Engl J Med 324:308-317

D. E. Mosier

262