varicella-zoster virus infection of human neural cells in vivo · varicella-zoster virus (vzv) is a...

6
Varicella-zoster virus infection of human neural cells in vivo Armin Baiker* , Klaus Fabel , Antonio Cozzio §¶ , Leigh Zerboni* , Konstanze Fabel , Marvin Sommer* , Nobuko Uchida , Dongping He , Irving Weissman § , and Ann M. Arvin* ** Departments of *Pediatrics, Microbiology and Immunology, Neurosurgery, and § Pathology, Stanford University School of Medicine, Stanford, CA 94305; and StemCells Inc., Palo Alto, CA 94304 Contributed by Irving Weissman, June 8, 2004 Varicella-zoster virus (VZV) establishes latency in sensory ganglia and causes herpes zoster upon reactivation. These investigations in a nonobese diabetic severe combined immunodeficient mouse- human neural cell model showed that VZV infected both neurons and glial cells and spread efficiently from cell to cell in vivo. Neural cell morphology and protein synthesis were preserved, in contrast to destruction of epithelial cells by VZV. Expression of VZV genes in neural cells was characterized by nuclear retention of the major viral transactivating protein and a block in synthesis of the pre- dominant envelope glycoprotein. The attenuated VZV vaccine strain retained infectivity for neurons and glial cells in vivo. VZV gene expression in differentiated human neural cells in vivo differs from neural infection by herpes simplex virus, which is character- ized by latency-associated transcripts, and from lytic VZV replica- tion in skin. The chimeric nonobese diabetic severe combined immunodeficient mouse model may be useful for investigating other neurotropic human viruses. V aricella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has a linear, double-stranded DNA genome encoding at least 70 proteins, including immediate early (IE) regulatory proteins that control viral gene transcription, early genes, such as viral kinases, and late genes, which are predominantly viral glycoproteins that comprise the virion envelope. Like all alphaherpesviruses, VZV establishes latency in neural cells within sensory ganglia. VZV reactivation results in herpes zoster, associated with postherpetic neuralgia in the elderly and dissemination to lungs, liver, and brain in immunocompromised individuals (5). Although most VZV morbidity results from its capacity to infect neural cells, investigating neurotropism has been difficult because the virus is a highly human-specific pathogen (2, 4). Herpes simplex virus 1 produces latency-associated antisense mRNA transcripts (LATs) in neuronal cell nuclei, and viral proteins are not detected in human autopsy specimens or animal models of persistent infection (6). In contrast, VZV has no LAT sequence, and transcription or translation of VZV genes encoding the IE major transactivating protein, IE62, the IE63 coregulatory pro- tein, and ORFs 4, 21, 29, 40, and 66 has been reported in ganglia (2, 4, 7–18). Recently, Uchida et al. (19) described proliferation and dif- ferentiation of human CNS stem cells into neurons and glial cells in mice with nonobese diabetic severe combined immunodefi- ciency (NOD-SCID). Transfer of these cells into the lateral ventricle results in site-appropriate engraftment within the sub- ventricular zone (SVZ), self-renewal, migration, and differenti- ation to human neurons and glia. The expanding populations migrate throughout the brain from the SVZ to the olfactory bulb, express human neuronal and glial cell markers, and develop axonal processes. Skin and thymusliver xenografts in SCID mice permit analysis of VZV tropism for human epidermal and dermal cells and T cells within their intact tissue microenviron- ments in vivo (20–24). Therefore, we used the chimeric NOD- SCID mouse–human neural cell model to investigate VZV infection of differentiated neural cells in vivo. We compared the parent Oka virus (pOka), a low passage clinical isolate, to the vaccine Oka virus (vOka), a pOka derivative used to make live attenuated varicella vaccines, because it is not known whether vOka has reduced infectivity for neural cells (25–27). We also evaluated a VZV deletion mutant, rOka70, that lacks one copy of the gene encoding IE63, because neural cell expression of IE63 has been prominent in autopsy and animal studies (9, 13, 15). This mutant, and two other single-copy ORF6370 variants, exhibits no deficiencies in cell culture and is as virulent as the parent virus in human skin xenografts in SCID mice (24), These experiments in the NOD-SCID mouse–human neural cell model showed that VZV infection with pOka, vOka, and rOka70 was characterized by efficient spread in human neu- rons and glial cells, without disrupting cell morphology or expression of neural cell adhesion molecules (hNCAMs), glial fibrillary acidic protein (hGFAP), or neuron-specific class III -tubulin in vivo. VZV infection of human neural cells exhibited a distinctive intracellular distribution of the IE regulatory pro- teins and a block in synthesis of the major envelope glycoprotein, gE, compared to lytic VZV infection of epidermal and dermal cells in vivo. Materials and Methods NOD-SCID Mice with Neural Cell Xenografts. Processing of fetal brain tissue and the isolation of human CNS stem cells by flow cytometry were carried out as described (19, 28). Fetal brain tissues of 16–20 gestational weeks were used. Sorted CD133CD24lo cells were plated at a density of 10 5 cells per ml in human neurosphere culture media consisting of X-vivo 15 medium (BioWhittaker), N-2 supplement (GIBCO), and 0.2 mgml heparin supplemented with basic fibroblast growth factor (20 ngml), epidermal growth factor (20 ngml), and leukemia inhibitory factor (10 ngml). Sorted cells form neurospheres within 10–14 days in culture. Neurospheres were passaged by enzymatic dissociation into single cells for 5–10 min in collage- nase (0.5 mgml in PBS containing 0.1% human serum albumin). Neurosphere cultures were passaged every 10–14 days. Cryo- anesthetized neonatal mice (24 h) were placed on a stereotaxic device (Stoelting) and injected with 2 l of cells (10 5 cells) into both lateral ventricles (19). Human brain tissue was obtained in accordance with state and federal guidelines. VZV Infection of NOD-SCID Mice. NOD-SCID mice transplanted with human neurosphere cells (StemCells Inc.) were inoculated with VZV-infected human melanoma cells at 4–6 months after transplant. Viruses tested were pOka, live attenuated vOka (Merck), and rOka70, a deletion mutant with only one copy of Abbreviations: VZV, varicella-zoster virus; IE, immediate early; NOD-SCID, nonobese dia- betic severe combined immunodeficiency; pOka, parent Oka virus; vOka, vaccine Oka virus; hNCAM, human neural cell adhesion molecule; hGFAP, human glial fibrillary acidic protein; TBS, Tris-buffered saline; gE, glycoprotein E. Present address: University Hospital of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland. **To whom correspondence should be addressed. E-mail: [email protected]. © 2004 by The National Academy of Sciences of the USA 10792–10797 PNAS July 20, 2004 vol. 101 no. 29 www.pnas.orgcgidoi10.1073pnas.0404016101 Downloaded by guest on November 17, 2020

Upload: others

Post on 14-Aug-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

Varicella-zoster virus infection of human neuralcells in vivoArmin Baiker*†, Klaus Fabel‡, Antonio Cozzio§¶, Leigh Zerboni*†, Konstanze Fabel‡, Marvin Sommer*†, Nobuko Uchida�,Dongping He�, Irving Weissman§, and Ann M. Arvin*†**

Departments of *Pediatrics, †Microbiology and Immunology, ‡Neurosurgery, and §Pathology, Stanford University School of Medicine, Stanford, CA 94305;and �StemCells Inc., Palo Alto, CA 94304

Contributed by Irving Weissman, June 8, 2004

Varicella-zoster virus (VZV) establishes latency in sensory gangliaand causes herpes zoster upon reactivation. These investigations ina nonobese diabetic severe combined immunodeficient mouse-human neural cell model showed that VZV infected both neuronsand glial cells and spread efficiently from cell to cell in vivo. Neuralcell morphology and protein synthesis were preserved, in contrastto destruction of epithelial cells by VZV. Expression of VZV genesin neural cells was characterized by nuclear retention of the majorviral transactivating protein and a block in synthesis of the pre-dominant envelope glycoprotein. The attenuated VZV vaccinestrain retained infectivity for neurons and glial cells in vivo. VZVgene expression in differentiated human neural cells in vivo differsfrom neural infection by herpes simplex virus, which is character-ized by latency-associated transcripts, and from lytic VZV replica-tion in skin. The chimeric nonobese diabetic severe combinedimmunodeficient mouse model may be useful for investigatingother neurotropic human viruses.

Varicella-zoster virus (VZV) is a human alphaherpesvirusthat causes varicella and herpes zoster (1–4). VZV has a

linear, double-stranded DNA genome encoding at least 70proteins, including immediate early (IE) regulatory proteins thatcontrol viral gene transcription, early genes, such as viral kinases,and late genes, which are predominantly viral glycoproteins thatcomprise the virion envelope. Like all alphaherpesviruses, VZVestablishes latency in neural cells within sensory ganglia. VZVreactivation results in herpes zoster, associated with postherpeticneuralgia in the elderly and dissemination to lungs, liver, andbrain in immunocompromised individuals (5). Although mostVZV morbidity results from its capacity to infect neural cells,investigating neurotropism has been difficult because the virus isa highly human-specific pathogen (2, 4). Herpes simplex virus 1produces latency-associated antisense mRNA transcripts(LATs) in neuronal cell nuclei, and viral proteins are notdetected in human autopsy specimens or animal models ofpersistent infection (6). In contrast, VZV has no LAT sequence,and transcription or translation of VZV genes encoding the IEmajor transactivating protein, IE62, the IE63 coregulatory pro-tein, and ORFs 4, 21, 29, 40, and 66 has been reported in ganglia(2, 4, 7–18).

Recently, Uchida et al. (19) described proliferation and dif-ferentiation of human CNS stem cells into neurons and glial cellsin mice with nonobese diabetic severe combined immunodefi-ciency (NOD-SCID). Transfer of these cells into the lateralventricle results in site-appropriate engraftment within the sub-ventricular zone (SVZ), self-renewal, migration, and differenti-ation to human neurons and glia. The expanding populationsmigrate throughout the brain from the SVZ to the olfactory bulb,express human neuronal and glial cell markers, and developaxonal processes. Skin and thymus�liver xenografts in SCIDmice permit analysis of VZV tropism for human epidermal anddermal cells and T cells within their intact tissue microenviron-ments in vivo (20–24). Therefore, we used the chimeric NOD-SCID mouse–human neural cell model to investigate VZVinfection of differentiated neural cells in vivo. We compared the

parent Oka virus (pOka), a low passage clinical isolate, to thevaccine Oka virus (vOka), a pOka derivative used to make liveattenuated varicella vaccines, because it is not known whethervOka has reduced infectivity for neural cells (25–27). We alsoevaluated a VZV deletion mutant, rOka�70, that lacks one copyof the gene encoding IE63, because neural cell expression ofIE63 has been prominent in autopsy and animal studies (9, 13,15). This mutant, and two other single-copy ORF63�70 variants,exhibits no deficiencies in cell culture and is as virulent as theparent virus in human skin xenografts in SCID mice (24),

These experiments in the NOD-SCID mouse–human neuralcell model showed that VZV infection with pOka, vOka, andrOka�70 was characterized by efficient spread in human neu-rons and glial cells, without disrupting cell morphology orexpression of neural cell adhesion molecules (hNCAMs), glialfibrillary acidic protein (hGFAP), or neuron-specific class III�-tubulin in vivo. VZV infection of human neural cells exhibiteda distinctive intracellular distribution of the IE regulatory pro-teins and a block in synthesis of the major envelope glycoprotein,gE, compared to lytic VZV infection of epidermal and dermalcells in vivo.

Materials and MethodsNOD-SCID Mice with Neural Cell Xenografts. Processing of fetalbrain tissue and the isolation of human CNS stem cells by flowcytometry were carried out as described (19, 28). Fetal braintissues of 16 –20 gestational weeks were used. SortedCD133�CD24��lo cells were plated at a density of 105 cells perml in human neurosphere culture media consisting of X-vivo 15medium (BioWhittaker), N-2 supplement (GIBCO), and 0.2mg�ml heparin supplemented with basic fibroblast growth factor(20 ng�ml), epidermal growth factor (20 ng�ml), and leukemiainhibitory factor (10 ng�ml). Sorted cells form neurosphereswithin 10–14 days in culture. Neurospheres were passaged byenzymatic dissociation into single cells for 5–10 min in collage-nase (0.5 mg�ml in PBS containing 0.1% human serum albumin).Neurosphere cultures were passaged every 10–14 days. Cryo-anesthetized neonatal mice (�24 h) were placed on a stereotaxicdevice (Stoelting) and injected with 2 �l of cells (105 cells) intoboth lateral ventricles (19). Human brain tissue was obtained inaccordance with state and federal guidelines.

VZV Infection of NOD-SCID Mice. NOD-SCID mice transplantedwith human neurosphere cells (StemCells Inc.) were inoculatedwith VZV-infected human melanoma cells at 4–6 months aftertransplant. Viruses tested were pOka, live attenuated vOka(Merck), and rOka�70, a deletion mutant with only one copy of

Abbreviations: VZV, varicella-zoster virus; IE, immediate early; NOD-SCID, nonobese dia-betic severe combined immunodeficiency; pOka, parent Oka virus; vOka, vaccine Oka virus;hNCAM, human neural cell adhesion molecule; hGFAP, human glial fibrillary acidic protein;TBS, Tris-buffered saline; gE, glycoprotein E.

¶Present address: University Hospital of Zurich, Gloriastrasse 31, 8091 Zurich, Switzerland.

**To whom correspondence should be addressed. E-mail: [email protected].

© 2004 by The National Academy of Sciences of the USA

10792–10797 � PNAS � July 20, 2004 � vol. 101 � no. 29 www.pnas.org�cgi�doi�10.1073�pnas.0404016101

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 2: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

the gene encoding IE63 (24). Melanoma cells were grown intissue culture medium (DMEM, GIBCO) supplemented withheat-inactivated FCS. VZV-infected melanoma cells weretrypsinized when 75% of cells showed cytopathic changes; cellsfrom one T75 flask were resuspended in 1 ml of PBS. Theinoculum for mouse injection was 10 �l of this virus per cellsuspension, containing �2.0 � 105 cells and �2.0–8.0 � 103

plaque-forming units of infectious virus. Narcotized animalswere placed on a stereotaxic device (Stoelting), and VZV-infected cell preparation (10 �l) was injected into each lateralventricle. Stereotaxic coordinates were 0.4 mm posterior toBregma, 2.2 mm deep, and �1.0 mm lateral (29). Animals werecared for according to Animal Welfare Act PL 94-279, withprotocols approved by the Stanford University AdministrativePanel on Laboratory Animal Care.

Immunohistochemistry. The murine mAb and polyclonal antiseraused to evaluate cellular and viral protein expression included:anti-human nuclei, mouse mAb, human-specific nuclear stain,1:200 (Chemicon) (30); anti-hNCAM (CD56), mouse mAb totwo isoforms, 145 and 185 kDa, of human neural cell adhesionmolecule, 1:200 (Zymed) (31); anti-HMB45, mouse mAb tohuman melanosome antigen in the cytoplasm of melanoma cells,1:500 (Dako) (32); anti-hGFAP, mouse mAb, cytoplasmic stain-ing of human glial cells, 1:2,000 (StemCells Inc.); anti-neuronalclass III �-tubulin, rabbit antiserum to neuron-specific class III�-tubulin in mouse and human neurons, 1:500 (Covance, Prince-ton, NJ) (33); anti-active caspase-3, rabbit antiserum to activehuman or mouse caspase-3, 1:200 (Becton Dickinson-Pharmingen) (34); and anti-nestin, rabbit antiserum to humannestin intermediate filaments expressed in developing CNS cells,1:500 (Chemicon). Primary antibodies to VZV-infected cellproteins were rabbit antisera to IE62 and IE63 (a gift from PaulKinchington, University of Pittsburgh, Pittsburgh), ORF47 ki-nase (20), and gE (35). GK is a high titer human polyclonal VZVantiserum. Secondary antibodies were donkey antisera, conju-gated with FITC (green), Cy3 (red), or Cy5 (blue) and diluted1:500 (Jackson ImmunoResearch).

Confocal Analysis of Mouse Brain Sections. Three weeks after VZVinoculation, mice were perfused with PBS followed by 4%paraformaldehyde (PFA) in PBS. Mouse brains were removedand fixed in 4% PFA�PBS for 2 days and dehydrated in a 30%sucrose�PBS solution for 3 days. Fixed brains were sectionedsagitally at 40-�m thickness with a freezing microtome. Sectionswere stored at �20°C in cryoprotectant solution. Each mousebrain yielded at least eight sections containing human neuralcells. Brain sections were rinsed with Tris-buffered saline (TBS)and blocked for 30 min at room temperature in TBS with 10%donkey serum and 0.3% Triton X-100 (TBS�). Sections wereincubated at 4°C overnight with primary antibodies, diluted inTBS�, washed, and incubated at 4°C overnight with the sec-ondary fluorochrome-conjugated antibodies (Jackson Immu-noResearch) diluted in TBS�. Sections were washed, wet-mounted by using polyvinyl alcohol-1,4 diazabicyclo [2.2.2]octane (36), and evaluated with a Zeiss510 confocal laser-scanning device attached to a Zeiss-Axiovert microscope andLSM510 software. Appropriate gain and black-level settingswere determined on control tissues stained with secondaryantibodies alone. Upper and lower thresholds were set by usingthe range indicator function to minimize data loss caused bysaturation.

Skin Xenografts. As described (22), skin implants were inocu-lated with VZV-infected MRC-5 cells (50 �l) 4–6 weeks afterimplantation and harvested 3 weeks later. Sections wereblocked in TBS (10% goat serum�0.3% Triton X-100) over-night, incubated in TBS (3% goat serum�0.3% Triton X-100)

with primary antisera and washed. Slides were incubated inTBS (3% goat serum�0.3% Triton X-100) with biotin-labeledgoat secondary antibody (1:1,000) (Vector Laboratories),washed with TBS, and incubated in TBS with diluted alkalinephosphatase-conjugated streptavidin (1:400) (Jackson Immu-noResearch). Slides were treated with FastRed (Sigma) andcounterstained with hematoxylin.

VZV-Infected Melanoma Cells. Infected melanoma cells were fixedin 4% paraformaldehyde, washed with PBS, incubated overnightat 4°C in PBS (10% donkey serum�0.3% Triton X-100), andincubated in PBS (3% donkey serum�0.3% Triton X-100) withprimary antisera at 4°C overnight. Slides were washed andincubated in PBS (3% donkey serum�0.3% Triton X-100) withfluorochrome-conjugated secondary antibodies (Jackson Immu-noResearch), washed, and mounted with Vectashield plus 4�,6-diamidino-2-phenylindole.

ResultsInfection of Human Neural Cells by pOka and vOka in Vivo. Extensiveexpression of IE62 and IE63 proteins was detected by 3 weeksafter inoculation of VZV-infected melanoma cells into theventricles of NOD-SCID mice with human neural cell xenografts(Fig. 1). Alternate mouse brain sections containing humanneural cells were stained with antibodies against IE62 or IE63(red), in combination with markers that stained human cellnuclei, hNCAM, or the melanoma cell marker HMB45 (green).IE62 and IE63 proteins were detected whether the infectingvirus was the clinical isolate, pOka (Fig. 1 a–c) or the attenuatedvaricella vaccine strain, vOka (Fig. 1 d and e). VZV-infected cellswere evident within the mouse brain in regions containing cellsthat stained with anti-human nuclei mAb (huNuc) (Fig. 1 a andd) and anti-hNCAM (Fig. 1c). Dual expression of IE63 andhNCAM was detected in many cells (Fig. 1c). Cells expressing IEproteins did not stain with the melanoma marker HMB45 (Fig.1 b and e). Invasion of mouse brain by melanoma cells was notobserved and anti-hNCAM did not react with melanoma cells.IE62 and IE63 expression in engrafted cells expressing thehuman nuclei marker or hNCAM was confirmed in 11 alternatesections examined after pOka inoculation and eight alternatesections after vOka inoculation.

Fig. 1. Infection of human neural cells by VZV pOka and vOka in vivo.Alternate mouse brain sections prepared 3 weeks after infection with pOka(a–c and f ) or vOka (d and e) were stained with anti-human nuclei mAb(green�FITC) in combination with polyclonal rabbit antiserum to IE62 (red�Cy3) (a, d, and f ) the melanoma cell marker anti-HMB45 mAb (green�FITC) incombination with polyclonal rabbit antisera to IE63 (red�Cy3) (b and e), oranti-hNCAM mAb (green�FITC) in combination with polyclonal rabbit antiserato IE63 (red�Cy3) (c). Roman numerals indicate section numbers.

Baiker et al. PNAS � July 20, 2004 � vol. 101 � no. 29 � 10793

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 3: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

Residual melanoma cells were found within ventricles in a fewsections. These cells were identified with anti-human nuclei mAb(Fig. 1f ) and anti-HMB45 (data not shown). A few survivingmelanoma cells expressed IE62 protein (Fig. 1f ). Mouse braintissue beyond the areas of hNCAM-positive or human nuclei-positive cells showed no reactivity with antibodies to humannuclei, HMB45, IE62, or IE63.

Expression of IE62 and IE63 in VZV-Infected Human Neural Cells. TheIE62 and IE63 regulatory proteins showed different patternsof intracellular localization within human neural cells, asillustrated for pOka-infected cells (Fig. 2). IE62 exhibited adistinct nuclear localization (red) and colocalized with thehuman nuclei marker (green) (Fig. 2b). In cells showing dualexpression of IE62 (red) and hNCAM (green), which weresectioned in a plane that included nuclei and membranes, IE62expression was separated from the cell membranes expressinghNCAM by a cytoplasmic zone in which IE62 was not detected(Fig. 2a). In dual positive cells, IE63 staining extended to thehNCAM-expressing cell membranes, with no zone of separa-tion, suggesting that IE63 was present in the cytoplasm as wellas in nuclei (Fig. 2 c and d). In these and other experiments,secondary antibodies did not crossreact with VZV-infectedhuman neural cell xenografts (Fig. 2e).

Expression of IE62 and IE63 in Human Neural Cells Infected withrOka�70. IE63 is an essential VZV protein encoded by identicalgenes, ORF63�70. Our hypothesis was that VZV neurotropismmight require both copies. However, IE62 and IE63 proteinsshowed the same patterns of intracellular localization afterinoculation of the single-copy OR63 mutant, rOka�70, com-pared to pOka or vOka, which have two copies of ORF63. IE62and IE63 (red) were expressed extensively in hNCAM-positivecells (green) within rOka�70-infected xenografts. IE62 localizedto the nuclei, as it did in human neural cells infected with pOkaor vOka (Fig. 2a�). As observed with pOka and vOka, IE63 wasdetected diffusely in the cytoplasm and nuclei (Fig. 2b�). Primaryantibodies did not crossreact with mouse brain (Fig. 2e�). Thesepatterns were confirmed in 13 mouse brain sections examinedafter rOka�70 inoculation.

Expression of Early and Late VZV Proteins in Human Neural Cells. TheVZV ORF47 protein kinase, a viral serine�threonine kinase, isexpressed during the second, or early, phase of the VZVreplication cycle (37). ORF47 protein was detected abundantlywithin the cytoplasm and nuclei of human neural cells in vivo, asillustrated with rOka�70 (Fig. 2c�). VZV glycoprotein E (gE) isan essential late protein that is expressed on cell membranesduring lytic infection and is a predominant component of thevirion envelope (2, 35). VZV gE was detected very rarely inhuman neural cells. Limited gE expression was observed in onlya few cells within regions that contained abundant hNCAM-positive cells (Fig. 2d�) and that showed IE62, IE63, and ORF47protein expression on alternate sections. These observationsindicated that VZV replication was blocked at the third, or late,phase in infected neural cells.

Analysis of Human Neural Cells Infected with VZV. Alternate sectionswere stained for hNCAM (green), neuron-specific class III�-tubulin (blue), which is present in the cytoplasm of human andmouse neurons, and VZV proteins, using the polyclonal humananti-VZV antiserum, GK (red) (Fig. 3 a–c). Mouse neuronsexpressing �-tubulin III were detected in mouse brain tissuesurrounding the human neural cell xenograft (Fig. 3a). Becauseanti-�-tubulin III also stained melanoma cells, the triple stainincluded anti-hNCAM, which did not crossreact with mousebrain or melanoma cells. Two infected cells have been high-lighted that expressed hNCAM (green) on the cell surface,�-tubulin III (blue) in the cytoplasm, and VZV proteins (red)(Fig. 3 b and c). One of these cells had VZV protein expressionvisible along an axonal structure and over the neural cell body.The polyclonal anti-VZV antiserum, GK, did not crossreact withan uninfected human neural cell xenograft that containedhNCAM-positive cells (Fig. 3d).

These sections showed VZV protein expression in cells thatdid not express hNCAM, indicating that other neural cell typeswere infected with VZV in vivo. Nestin is a cytoplasmic markerfor early-stage, undifferentiated human neural cells. Whensections were stained for human nuclei (green), nestin (blue),and VZV proteins (red) (Fig. 3 a� and b�), coexpression of nestinand VZV proteins, resulting in a purple signal, was observed,

Fig. 2. Expression of IE62, IE63, ORF47 kinase, and gE in VZV-infected human neural cells in vivo. Mouse brain sections prepared 3 weeks after infection withpOka (a–e) or rOka�70 (a�–e�) were stained with anti-hNCAM mAb (green�FITC) (a, c, and a�–e�), anti-human nuclei mAb (green�FITC) (b), or the melanoma cellmarker, anti-HMB45 mAb (green�FITC) (d), in combination with polyclonal rabbit antisera to IE62 (red�Cy3) (a, b, and a�), IE63 (red�Cy3) (c, d, and b�), ORF47(red�Cy3) (c�), or gE (red�Cy3) (d�). No nonspecific staining was detected with secondary antibodies within the same region (e) or anti-hNCAM and anti-IE62primary antibodies (e�). Roman numerals indicate section numbers.

10794 � www.pnas.org�cgi�doi�10.1073�pnas.0404016101 Baiker et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 4: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

indicating that VZV could infect immature human neural cellsin vivo (Fig. 3b�).

Neural cell xenografts in the NOD-SCID mice contained cellsthat expressed hGFAP, found in glial cell cytoplasm (Fig. 4).When sections were stained with hGFAP and IE62, cells thatexpressed hGFAP (green) and IE62 (red) were identified withinthe same area at low magnification (Fig. 4a). Higher magnifi-cations showed that many hGFAP-positive cells were infectedwith VZV in vivo (Fig. 4 b and c). As was observed in hNCAM-positive cells, IE62 showed a distinct nuclear localization inhGFAP-positive cells (Fig. 4 b and c). Some IE62-positive cellswere not hGFAP positive, which was because of the presence ofVZV-infected neurons, expressing hNCAM and �-tubulin III,within this region. Primary antibodies to hGFAP and IE62 didnot cause nonspecific staining (Fig. 4d); anti-hGFAP antiserumdid not crossreact with melanoma cells.

Apoptosis within VZV-Infected Human Neural Cell Xenografts. Toanalyze whether infected human neural cells showed evidence ofapoptosis, sections were stained with antibodies against humannuclei (green), caspase 3 active form (blue), and VZV proteins,using polyclonal anti-VZV antiserum, GK (red) (Fig. 4 f and g).Only a few VZV-infected human neural cells expressed caspase3; a few mouse brain cells also stained with anti-caspase 3, whichis not human specific (Fig. 4g). Secondary antibodies did notcrossreact with VZV-infected human neural cells when the samearea was stained on an alternate section (Fig. 4e).

VZV Protein Expression in Infected Skin Xenografts. VZV causes lyticinfection of human dermal and epidermal cells in skin xenograftsin vivo (20–24, 38). To compare VZV protein expression inhuman neural and skin cells in vivo, skin xenografts werecollected 3 weeks after VZV infection, and alternate sectionswere examined for IE62, IE63, ORF47 kinase, or gE synthesis(Fig. 5). In contrast to neural cell xenografts, IE62 was detectedin both the cytoplasm and nuclei of VZV-infected skin cells andwas present in small, dense bodies within the nucleus (Fig. 5a).Intense IE63 expression was prominent in nuclei of VZV-infected cells; cytoplasmic expression was also observed, al-though much less than in neural cells (Fig. 5b). The ORF47protein kinase was abundant and predominantly cytoplasmic(Fig. 5c). In contrast to VZV-infected human neural cells, gEexpression was extensive, with high concentrations along theplasma membranes of VZV-infected skin cells (Fig. 5d). Someskin cells expressed caspase 3 active form, suggesting apoptosis(Fig. 5e).

VZV Protein Expression in Infected Melanoma Cells. IE62, IE63,ORF47 protein, and gE expression was evaluated 4 days afterVZV infection of melanoma cells. During this lytic infection,IE62 was abundant in cytoplasm, in contrast to its nuclearlocalization in VZV-infected neural cells in vivo. IE62 waspresent within distinct small, dense nuclear bodies within nuclei,as in VZV-infected skin cells (Fig. 6a). IE63 showed substantialnuclear localization, as in VZV-infected skin, and in contrast toits extensive cytoplasmic distribution in VZV-infected neural

Fig. 3. Analysis of human neural cells infected with VZV in vivo. (a–c, a�, andb�) Mouse brain sections prepared 3 weeks after infection with rOka�70 weretriple-stained with anti-hNCAM mAb (green�FITC), polyclonal rabbit anti-serum to �-tubulin III (blue�Cy5), and human antiserum against VZV (GK)(red�Cy3). (d) A control uninfected neural cell xenograft section showedstaining with anti-hNCAM and anti-�-tubulin III but not anti-VZV antiserum(GK). (a� and b�) Triple staining with anti-human nuclei (green�FITC), poly-clonal rabbit antiserum to nestin (blue�Cy5), and anti-VZV antiserum GK(red�Cy3) is shown. Roman numerals indicate section numbers.

Fig. 4. Analysis of human neural cells infected with VZV for expression ofGFAP and evidence of apoptosis. (a–d) Mouse brain sections prepared 3 weeksafter infection with rOka�70 were stained with anti-hGFAP mAb (green�FITC)and polyclonal rabbit antiserum to IE62 (red�Cy3). (d) No nonspecific stainingwas observed in an uninfected xenograft. ( f and g) Triple staining was donewith anti-human nuclei mAb (green�FITC), polyclonal rabbit antiserumagainst caspase 3 active form (blue�Cy5), and anti-VZV antiserum, GK (red�Cy3). (e) Secondary antibodies did not stain nonspecifically. Roman numeralsindicate section numbers.

Baiker et al. PNAS � July 20, 2004 � vol. 101 � no. 29 � 10795

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 5: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

cells (Fig. 6b). ORF47 protein was cytoplasmic (Fig. 6c). As inVZV-infected skin cells, abundant gE expression was detectedalong plasma membranes and more diffusely in cytoplasm (Fig.6d). Uninfected melanoma cells were induced to undergo apo-ptosis with UV light, as a positive control for anti-caspase 3antibody (Fig. 6e); melanoma cells stain positive for HMB45(Fig. 6f ); and absence of hNCAM expression by human mela-noma cells was confirmed (Fig. 6g).

DiscussionVZV infection in the chimeric NOD-SCID mouse–human neu-ral cell model provided a unique opportunity to examine theinitial encounter between VZV and human neural cells in vivo.These experiments demonstrated that VZV spreads efficientlybetween neural cells in vivo, without changing cellular morphol-ogy or expression of neural cell proteins, including hNCAM,hGFAP, and �-tubulin III. VZV gene expression in neural cellsdiffered dramatically from VZV infection of epidermal anddermal cells in human skin in vivo, which is associated with celldestruction and release of infectious virions (20–24). IE62, themajor viral transactivating protein and an important componentof the virion tegument (38, 39), was restricted to the nuclei ofneurons and glial cells, whereas IE62 localized to the cytoplasmof infected skin cells. IE63 was abundant in the cytoplasm ofneurons and glial cells, whether expressed by low-passage pOkaor the single-copy rOka�70 recombinant, which differed from itspredominant nuclear expression in VZV-infected skin cells. Incontrast, the essential late glycoprotein, gE, was detected in onlya very few neural cells, which was strikingly different from theextensive expression of gE in skin cells. The block in glycoproteinsynthesis in neural cells occurred despite synthesis of the ORF47protein kinase, which phosphorylates IE62, IE63, and gE, and isrequired for VZV infection of skin and T cells in vivo (20, 37, 40).The patterns of IE62, IE63, ORF47, and gE synthesis anddistribution found with lytic VZV infection of skin were ob-served in cultured melanoma cells, which also produce infectiousVZ virions in vitro. These observations support the concept thatinfection of neural cells by VZV, in which viral gene expressionoccurs, but is limited to IE and early genes in neural cells, differs

fundamentally from herpes simplex virus, in which viral proteinsare not detected in human autopsy specimens (6).

In this analysis of VZV neuropathogenesis, human neural cellswere infected with VZV in vivo and fixed in situ before removalof tissues. The observation that IE proteins were expressed inneural cells in the NOD-SCID mouse–human neural cell modelwhile gE synthesis was reduced serves to validate reports aboutVZV gene transcription and translation in neural cells withinhuman sensory ganglia obtained at autopsy and in animal models(2, 4, 7–18). Conclusions from autopsy specimens have beenquestioned because VZV gene expression might be triggeredduring the time required to obtain tissues, and infection ofnonhuman neural cells in animals may not reproduce the VZVpathogenesis in the human host accurately. VZV gene expres-sion was similar in human neurons, glial cells, and early-stage,less differentiated neural cells in vivo. Thus, VZV appears toregulate viral proteins and abort the full replicative cycle invarious types of cells of neural origin. The conditions forblocking late gene expression were established even thoughVZV gained access to neural cells by transfer from cells with thecapacity to produce infectious virus, rather than by transportalong nerve cell axons. The generalized nature of this geneticprogram is also suggested by VZV infection of guinea pig entericneurons in vitro, in which ORF62 and early genes were tran-scribed, but gE mRNA was not detected (7). Importantly,although VZV-specific T cell immunity is needed to controlVZV reactivation as zoster, these experiments in the NOD-SCID mouse model suggest that the nonlytic interaction of VZV

Fig. 6. VZV protein expression in infected melanoma cells in vitro. (a–d)Melanoma cells infected with rOka were stained with anti-human nuclei(green�FITC) and polyclonal rabbit antiserum against IE62 (a), IE63 (b), ORF47(c), or gE (red�Texas red) (d). (e) Melanoma cells induced to undergo apoptosisby UV light were stained with polyclonal antiserum to caspase 3 active form(red�Texas red); counterstain 4�,6-diamidino-2-phenylindole (DAPI) (blue).Melanoma cells were stained with anti-HMB45 mAb (green�FITC) and coun-terstain DAPI (blue�Cy5) (f ), or anti-hNCAM mAb (green�FITC) and counter-stain DAPI (blue) (g).

Fig. 5. Analysis of VZV protein expression in infected skin xenografts.Alternate sections of human skin prepared 3 weeks after infection with rOkawere stained with polyclonal antiserum against IE62 (a), IE63 (b), ORF47 (c), gE(d), or caspase 3 active form (red�FastRed) (e); counterstain was hematoxylin�eosin. Roman numerals indicate section numbers.

10796 � www.pnas.org�cgi�doi�10.1073�pnas.0404016101 Baiker et al.

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0

Page 6: Varicella-zoster virus infection of human neural cells in vivo · Varicella-zoster virus (VZV) is a human alphaherpesvirus that causes varicella and herpes zoster (1–4). VZV has

with neural cells is established independently of adaptive hostimmunity. It will be of interest to evaluate modulation of viralgene expression by innate neural cell responses in this model.

Expression of the IE63 regulatory protein was extensive andpredominantly cytoplasmic in the NOD-SCID mouse–humanneural cell model, as has been reported in autopsy and animalstudies (9, 13, 15). Cytoplasmic retention of IE63 could interferewith efficient transcription of VZV glycoprotein genes in neuralcells (3, 4, 40). A single copy of ORF63 was sufficient to supportVZV infection of neural cells in vivo. In these experiments, IE62was restricted to the nuclei of neurons and glial cells, rather thanto the cytoplasm, as was found in an autopsy study (13). Thisdifference in IE62 localization could reflect an earlier phase ofVZV infection of neural cells, as opposed to prolonged latencyduring which further modulation of virus–host cell interactionsmay occur. Both patterns contrast with the distribution of IE62to cytoplasm and nuclei in productively infected skin cells in vivo.Either IE62 nuclear exclusion or retention can block VZVreplication, because IE62 is the key transactivating factor forviral gene transcription in the nucleus, and a major componentof the virion tegument, which is assembled in the cytoplasm(2, 39).

Although the genetic basis of vOka attenuation has not beendetermined, vOka and pOka genomes have nucleotide differ-

ences predicted to change amino acid residues in all classes ofviral proteins (2, 41, 42). Although vOka can cause herpes zoster,it seems to reactivate less often than wild-type VZV (25, 26).Herpes zoster may be less common after vaccination becauseinitial access of vOka to neural cells is reduced by limited skinreplication, or vOka reactivation and secondary viral infection ofskin is less efficient, rather than because of an intrinsic attenu-ation of vOka neurotropism. Whereas vOka has a diminishedcapacity to replicate in skin, vOka remains unattenuated in Tcells in vivo (23). Our initial experiments suggest that derivingvOka by passage of pOka in fibroblasts does not appear to havealtered infectivity for neural cells, which can be further exploredin the NOD-SCID mouse–human neural cell model.

In summary, our observations provide evidence for uniquepatterns of VZV gene expression in human neural cells. Theseexperiments support the concept that the chimeric NOD-SCIDmouse–human neural cell model will be valuable for investigat-ing the neurotropism of other human viruses, such as HIV, indifferentiated human neural cells in vivo.

We thank Dr. Brent Harris, Dartmouth Medical School, Hanover, NH,for expert technical assistance. This work was supported by NationalInstitute of Allergy and Infectious Diseases Grants AI-36884 andAI-053846 and Deutsche Forschungsgemeinschaft Fellowship BA 2035�1-1 (to A.B.).

1. Arvin, A. M. (2001) in Field’s Virology, eds. Knipe, D. M. & Howley, P. M.(Lippincott-Williams & Wilkins, Philadelphia), pp. 2731–2767.

2. Cohen, J. & Strauss, S. (2001) in Field’s Virology, eds. Knipe, D. M. & Howley,P. M. (Lippincott-Williams & Wilkins, Philadelphia), pp. 2547–2586.

3. Kinchington, P. R. (1999) Front. Biosci. 4, 200–211.4. Silverstein, S. & Straus, S. E. (2000) in Varicella-Zoster Virus: Virology and

Clinical Management, eds. Arvin, A. M. & Gershon, A. A. (Cambridge Univ.Press, Cambridge, U.K.), pp. 123–141.

5. Gilden, D. H., Kleinschmidt-DeMasters, B. K., LaGuardia, J. J., Mahalingam,R. & Cohrs, R. J. (2000) N. Engl. J. Med. 2, 635–645.

6. Roizman, B. & Knipe, D. M. (2001) in Field’s Virology, eds. Knipe, D. M. &Howley, P. M. (Lippincott-Williams & Wilkins, Philadelphia), pp. 2399–2459.

7. Chen, J. J., Gershon, A. A. Li, Z. S., Lungu, O. & Gershon, M. D. (2003) J. Med.Virol. 70, 71–82.

8. Cohrs, R. J., Gilden, D. H., Kinchington, P. R., Grinfeld, E. & Kennedy,P. G. E. (2003) J. Virol. 77, 6660–6650.

9. Debrus, S., Sadzot-Delvaux, C., Nikkels, A. F., Piette, J. & Rentier, B. (1995)J. Virol. 69, 3240–3245.

10. Kennedy, P. G., Grinfeld, E. & Gow, J. W. (1998) Proc. Natl. Acad. Sci. USA95, 4658–4662.

11. Kennedy, P. G., Grinfeld, E. & Bell, J. E. (2000) J. Virol. 74, 11893–11898.12. Kennedy, P. G., Grinfeld, E., Bontems, S. & Sadzot-Delvaux, C. (2001) Virology

289, 218–223.13. Lungu, O., Panagiotidis, C. A., Annuziato, P. W., Gershon, A. A. & Silverstein,

S. J. (1998) Proc. Natl. Acad. Sci. USA 95, 7080–7085.14. Mahalingam, R., Wellish, M., Wolf, W., Dueland, A. N., Cohrs, R., Vafai, A.

& Gilden, D. (1990) N. Engl. J. Med. 323, 627–631.15. Maghalingam, R., Wellish, M., Cohrs, R., Debrus, S., Piette, J., Renier, B. &

Gilden, D. H. (1996) Proc. Natl. Acad. Sci. USA 93, 2122–2124.16. Meier, J. L., Holman, R. P., Croen, K. D., Smialek, J. E. & Straus, S. E. (1993)

Virology 193, 193–200.17. Pevenstein, S. R., Williams, R. K., McChesney, D., Mont, E. K., Smialek, J. E.

& Straus, S. E. (1999) J. Virol. 73, 10514–10518.18. Sato, H., Pesnicak, L. & Cohen, J. L. (2002) J. Virol. 76, 3575–3578.19. Uchida, N., Buck, D. W., He, D., Reitsma, M. J., Masek, M., Phan, T. V.,

Tsukamoto, A. S., Gage, F. H. & Weissman, I. L. (2000) Proc. Natl. Acad. Sci.USA 97, 14720–14725.

20. Besser, J., Sommer, M. H., Zerboni, L., Bagowski, C., Ito, H., Moffat, J., Ku,C.-C. & Arvin, A. M. (2003) J. Virol. 77, 5964–5974.

21. Ito, H., Sommer, M. H., Zerboni, L. H., He, H., Boucaud, D., Hay, J.,Ruyechan, W. & Arvin, A. M. (2003) J. Virol. 77, 489–498.

22. Moffat, J. F., Stein, M. D., Kaneshima, H. & Arvin, A. M. (1995) J. Virol. 69,5236–5242.

23. Moffat, J., Zerboni, L., Stein, M., Grose, C., Kaneshima, H. & Arvin, A. M.(1998) J. Virol. 72, 965–974.

24. Sommer, M. H., Zagha, E., Serrano, O. K., Ku, C. C., Zerboni. L., Baiker, A.,Santos, R., Spengler, M., Lynch, J., Grose, C., et al. (2001) J. Virol. 75,8224–8239.

25. Annunziato, P. A. & Gershon, A. A. (2000) in Varicella-Zoster Virus: Virologyand Clinical Management, eds. Arvin, A. M. & Gershon, A. A. (CambridgeUniv. Press, New York), pp. 460–476.

26. Gershon, A. A., LaRussa, P., Hardy, I., Steinberg, S. & Silverstein, S. (1992)J. Infect. Dis. S63–S68.

27. Takahashi, M., Otsuka, T., Okuno, Y., Asano, Y. & Yazaki, T. (1974) Lancet2, 1288–1290.

28. Tamaki, S., Eckert, K., He, D., Sutton, R., Dohse, M., Jain, G., Tushinski, R.,Reitsma, M., Harris, B., Tsukamoto, A., et al. (2002) J. Neurosci. Res. 69,976–986.

29. Davidson, B. L., Stein, C. S., Heth, J. A., Martins, I., Koitin, R. M., Derksen,T. A., Zabner, J., Ghodsi, A. & Chiorini, J. A. (2000) Proc. Natl. Acad. Sci. USA97, 3428–3432.

30. Vescovi, A. L., Parati, E. A., Gritti, A., Poulin, P., Ferrario, M., Wanke, E.,Frolichsthal-Schoeller, P., Cova, L., Arcellana-Panlilio, M., Colombo, A., et al.(1999) Exp. Neurol. 156, 71–83.

31. Schol, D. J., Mooi, W. J., van der Gugten, A. A., Wagenaar, S. S. & Hilgers,J. (1988). Int. J. Cancer S2, 33–40.

32. Leong, A. S. & Millos, J. (1989) Surg. Pathol. 2, 137–141.33. Lee, M. K., Tuttle, J. B., Rebhuhn, L. I., Cleveland, D. W. & Frankfurter, A.

(1990) Cell Motil. Cytoskeleton 17, 118–132.34. Patel, T., Gores, G. J. & Kaufmann, S. H. (1996) FASEB J. 10, 587–597.35. Mo, C., Lee, J., Sommer, M., Grose, C. & Arvin, A. M. (2002) Virology 304,

176–186.36. Valnes, K. & Brandtzaeg, P. (1985) J. Histochem. Cytochem. 33, 755–761.37. Kenyon, T. K., Lynch, J., Hay, J., Ruyechan, W. & Grose, C. (2001) J. Virol. 75,

8854–8858.38. Sato, B., Ito, H., Hinchliffe, S., Sommer, M. H., Zerboni, L. & Arvin, A. M.

(2003) J. Virol. 77, 5607–5620.39. Kinchington, P. R., Fite, K. & Turse, S. E. (2000) J. Virol. 74, 2265–2277.40. Lynch, J. M., Kenyon, T. K., Grose, C., Hay, J. & Ruyechan, W. T. (2002)

Virology 302, 71–82.41. Argaw, T., Cohen, J. I., Klutch, M., Lekstrom, K., Yoshikawa, T., Asano, Y. &

Krause, P. R. (2002) J. Infect. Dis. 81, 1153–1157.42. Gomi, Y., Imagawa, T., Takahashi, M. & Yamanishi, K. (2000) J. Med. Virol.

61, 497–503.

Baiker et al. PNAS � July 20, 2004 � vol. 101 � no. 29 � 10797

MIC

ROBI

OLO

GY

Dow

nloa

ded

by g

uest

on

Nov

embe

r 17

, 202

0