The roles of herpes simplex virus in neuroscience

Page created by Edna Schmidt
 
CONTINUE READING
Journal of NeuroVirology (1997) 3, 110 ± 125
                              ã 1997 Journal of NeuroVirology, Inc.
                                        http://www.jneurovirol.com

Review
The roles of herpes simplex virus in neuroscience
Sharon L Turner1 and Frank J Jenkins2
2
Department of Pathology, School of Medicine; 2Department of Infectious Diseases and Microbiology, School of Public
Health; 1,2Division of Behavioral Medicine and Oncology, University of Pittsburgh Cancer Institute; 1,2Center for
Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA

                    Herpes simplex virus (HSV) has been a focus of research in many laboratories
                    during the last 30 ± 35 years, with the majority centered on the virus'
                    replication, molecular biology and pathogenesis. Recently, HSV has begun to
                    receive considerable attention in the field of neuroscience, where scientists
                    have begun to use the virus as a tool or model for several areas of investigation.
                    These areas include the construction and development of HSV-based vectors
                    for gene therapy and the use of HSV as a neuronal tracer, as a model for
                    demyelinating disease and to study interactions between the nervous, immune
                    and endocrine systems. The goal of this paper is to review these different roles
                    for HSV in the broad field of neuroscience.

                    Keywords: herpes simplex virus; neuroscience; viral vectors; latency;
                    demyelination; neuro-immune interactions

Introduction
Herpes simplex virus (HSV) refers to two closely                        HSV infections
related members of the human herpesvirus family;                        In a primary infection, HSV enters the body via
herpes simplex virus type 1 and herpes simplex                          mucosal membrane or abraded skin and establishes
virus type 2. HSV-1 and HSV-2 share a significant                       infection locally in epithelial cells. Active viral
homology at genetic and nucleic acid levels and can                     replication in these cells results in the amplification
produce similar diseases ranging from inapparent                        of virus, the formation of a `fever blister' and the
infections and self-limiting cutaneous lesions to                       stimulation of both cellular and humoral immune
fatal encephalitis (for a review, see Whitley and                       responses (as reviewed in Hill, 1985). HSV spreads
Gnann, 1993). In addition, the virus establishes and                    from infected epithelial cells to axons of sensory
maintains a latent state in sensory neurons from                        neurons innervating the site of local infection where
which recurrent HSV infections arise.                                   it travels by retrograde axonal transport to the
   The properties of HSV that make it a useful tool                     neuronal nuclei (Cook and Stevens, 1973). Studies
for studies in the field of neuroscience include its                    using animal models have indicated that infected
neurotropism, the ability to construct viral mutants                    neurons demonstrate either a limited viral replica-
that express foreign genes and its ability to establish                 tion or establish a latent infection (as discussed in
latent infections in neuronal cells. In this review we                  Cook and Stevens, 1973; Hill, 1985). While the exact
will discuss five different roles for HSV in                            mechanism(s) for the establishment of HSV latency
neuroscience which include (1) a model for                              have yet to be determined, recent studies have
demyelinating disease, (2) a tool for transneuronal                     suggested that the decision between active versus
tracing studies, (3) use as a viral vector for gene                     latent infection is made soon after neuronal
therapy, (4) a model for studying neuro-immune                          infection (Simmons et al, 1992; Speck and Sim-
interactions, and (5) a model for studying health                       mons, 1992).
effects of chronic stress. A complete discussion of                        HSV latency is characterized by the presence of
any of these roles would require its own separate                       viral genomes in the nuclei of infected neurons and
review and is beyond the scope of this review.                          the absence of viral replication or viral protein
Instead, we will briefly discuss each role and, in                      production (for review, see Hill, 1985). The infec-
instances where it is possible, point the reader to                     tion and establishment of latency within neurons
more detailed reviews on each subject.                                  explains why HSV is termed a neurotrophic virus.
                                                                        In a neuron in which the virus is latent, there are no
                                                                        virus-specific proteins produced and thus the host's
Correspondence: FJ Jenkins                                              immune system is unaware of the virus' presence.
Received 31 May 1996; revised 18 November 1996; accepted                As a result, the immune system does not target the
5 December 1996                                                         latently-infected neuron for destruction, and anti-
HSV in neuroscience
                                                          SL Turner and FJ Jenkins
                                                                                                                   111
body levels to HSV antigens are not continuously          its own transcription and the transcription of other
stimulated during viral latency.                          genes containing ICP4 repressor elements (DeLuca
   A latent HSV infection is maintained for the life      and Schaffer, 1988a; Muller, 1985; Watson and
of the host, but the virus can be reactivated             Clements, 1980; Kristie and Roizman, 1986). ICP4
periodically to produce infectious virus and recur-       deletion mutants must be maintained in cell culture
rent disease (as reviewed in Hill, 1985). During          systems which provide the missing ICP4 gene in
reactivation, viral replication occurs within the         trans to the virus. The use of the complementing
reactivated neuron, producing infectious viral            cell lines to propagate ICP4 mutant viruses has
progeny. Mature virions are transported back down         further delineated the functions of ICP4 as well as
the sensory axon where they may establish a               assisted in the development of viral vectors.
recurrent infection in the epithelia of the skin.            Similar to ICP4, another major regulatory pro-
Studies using both animal models and human                tein, ICP0, exhibits more moderate transactivation
subjects have shown that viral reactivation can be        of the b and g class genes (Cai and Schaffer, 1992).
triggered by a variety of stressful or stress-related     Transient expression assays have also suggested
stimuli including heat, UV light, fever, hormonal         that ICP0 is capable of trans-activating ICP4 and is
changes, menses and physical trauma to the neuron         critical for the replication of HSV following
(e.g. Segal et al, 1974; Spruance, 1985; Carlton and      transfection of viral DNA (Cai et al, 1993a; Cai
Kilbourne, 1952; Pazin et al, 1978). While the virus      and Schaffer, 1989). While the synthesis of ICP0 is
appears to be latent most of the time, HSV infection      not essential for viral replication in cell culture
is probably best characterized by recurrent reactiva-     (Sacks and Schaffer, 1987; Stow and Stow, 1986), it
tions and periods of latency.                             does appear to be involved in viral replication in
                                                          animal models and in viral reactivation triggered
HSV replication                                           by ganglion explanation (Leib et al, 1989a,b). These
The HSV genome is a linear, double-stranded DNA           properties are best demonstrated in a recent study
approximately 150 Kb in length (McGeoch et al,            by Cai et al (1993b) which utilizes nonsense,
1988). There are at least 75 separate open reading        insertion and deletion mutants of ICP0 to delineate
frames encoded within this DNA genome. Molecu-            the effects of mutant ICP0 peptides on viral
lar studies have revealed that many of these genes        replication and reactivation. Plasmid constructs
can be deleted without interfering with the virus'        containing the mutated ICP0 genes (which were
ability to replicate in cell culture lines (as reviewed   used to generate the ICP0 viral mutants) exhibited
in Ward and Roizman, 1994). In addition, it is            unique levels of transactivating activity as mea-
possible to construct site-specific mutations, in-        sured by the ability of the expressed ICP0 peptides
cluding the deletion of viral genes and the insertion     to stimulate transcription of ICP0 promoter-respon-
of foreign genes, into the viral genome (for example,     sive genes in vitro. In a murine model for HSV
Post and Roizman, 1981; Roizman and Jenkins,              replication and latency, using the ICP0 mutants, it
1985; Jenkins and Roizman, 1986).                         was reported that the efficiency of viral replication
   HSV genes are divided into three major temporal        in the eyes and trigeminal ganglia of inoculated
classes (a, b, and g) which are regulated in a            mice, and the frequency of viral reactivation
coordinated, cascade fashion (for review see Roiz-        induced by ganglion explant, correlated with the
man and Sears, 1993). The a or immediate-early (IE)       levels of ICP0 transactivating activity. This data
genes contain the major transcriptional regulatory        suggests a functional role for ICP0 in viral
proteins and their production is required for the         replication and reactivation in vivo. However,
transcription of the b and g gene classes. b proteins     levels of viral DNA as determined by PCR analysis,
are not produced in the absence of a proteins and         did not correlate with the efficiency of viral
their synthesis is required for viral DNA replication.    replication or reactivation, suggesting that other,
The b proteins consist primarily of proteins              as yet undefined factors, may influence the ability
involved in viral nucleic acid metabolism. The g          of the virus to establish neuronal latency.
proteins reach peak rates of synthesis after the onset       HSV latency is defined by the presence of viral
of DNA replication and consist primarily of virus         genomes and the absence of viral protein produc-
structural proteins.                                      tion within infected cells. While this definition
   Two HSV-1 a proteins that are relevant for the         suggests that viral gene transcription is absent, a
discussions in this review are called ICP0 and ICP4       small subset of viral transcripts, termed the latency
(ICP=infected cell protein). ICP4 is the major            associated transcripts (LATs) are transcribed during
regulatory protein of HSV and deletion studies have       active and latent infections (Wechsler et al, 1988;
demonstrated that it is absolutely required for viral     Wagner et al, 1988; Stevens et al, 1988; Krause et al,
replication (DeLuca and Schaffer, 1988b; Dixon and        1990; Rock et al, 1987; Roizman and Sears, 1993).
Schaffer, 1980; Preston, 1979; DeLuca et al, 1984,        The function of the LAT transcripts is unclear. Viral
1985). The ICP4 protein is involved in the                deletion mutants indicate that the LATs are not
transactivation of both b and g genes (as reviewed        required to establish latent infection (Ho and
in Roizman and Sears, 1993), but may also repress         Mocarski, 1989; Steiner et al, 1989). In addition,
HSV in neuroscience
                                            SL Turner and FJ Jenkins
112
      contradicting data indicate variable roles for the               CNS demyelination. Susceptible strains exhibit
      LATs in viral reactivation. Some studies report                  multifocal demyelination, while moderately resis-
      decreased reactivation frequencies in LAT mutants                tant strains exhibit unifocal demyelination and
      whereas others document no difference in reactiva-               resistant strains simply exhibit an increased pre-
      tion frequencies in LAT mutants and wild type                    sence of inflammatory cells at the TREZ (Kastrukoff
      virus (Natarajan et al, 1991; Block et al, 1990; Perng           et al, 1987). This study was followed by a more
      et al, 1996; Maggioncalda et al, 1994). Such                     recent report which described investigations on the
      inconsistencies may be partially explained by the                effects of immunosuppression on TREZ demyelina-
      different mutations in the different LAT mutants                 tion in strains of immunocompetent mice, immune
      which include deletions in LAT promoter se-                      deficient mice, and immunocompetent mice that
      quences and LAT coding sequences. An additional                  had been immunosuppressed with either cyclospor-
      problem arises from the fact that the LAT gene is                in A or cyclophosphamide (Kastrukoff et al, 1993).
      located antisense to the ICP0 gene (Stevens et al,               Animals that were immune deficient or immuno-
      1987). The overlap between the LAT and ICP0 genes                suppressed either lacked evidence of demyelination
      makes it difficult to ascertain possible functions of            or exhibited reduced levels of demyelination when
      LAT since many LAT mutations commonly disrupt                    compared to immunocompetent mice. Taken to-
      ICP0 sequences. As previously discussed, ICP0                    gether, these studies clearly demonstrate that the
      plays an important role in viral replication and                 immune system plays an important role in the
      reactivation in vivo and therefore LAT mutations                 development of demyelination during an acute
      may not always delineate the functions of LAT from               HSV infection and that this response may be
      those of ICP0.                                                   influenced by genetic determinants. In addition, it
                                                                       was demonstrated that infectious virus was required
      A model for virus-induced demyelinating disease                  for the development of demyelinating lesions and
      Animal studies using peripheral inoculations of                  that the presence of virus alone was not sufficient.
      HSV on the cornea, snout, footpad or vagina have                 Townsend (1981b) also reported that following a
      demonstrated that the virus spreads from peripheral              peripheral HSV infection, the virus spreads from the
      epithelial cells to both the peripheral nervous                  PNS to the CNS where it initially infects and lyses
      system (PNS) and central nervous system (CNS;                    astrocytes. This infection occurs prior to onset of
      Townsend and Baringer, 1978a,b; Kristensson et al,               demyelination. Following the infection of astro-
      1978, 1979). Cellular damage induced by the virus is             cytes, immune cells appear and demyelination
      markedly different in these two areas. Tissue damage             begins. The results of this study were later confirmed
      in the PNS is generally mild while the damage in the             by Itoyama et al (1991). Thus HSV-induced demye-
      CNS is often extensive within a local foci. A well               lination of the CNS appears to be due to a
      studied example of this concept is seen within the               combination of viral-induced lysis of astrocytes
      trigeminal root entry zone (TREZ) of the brainstem, a            and oligodendrocytes and immuno-pathology
      junction region between the PNS and CNS (Town-                   caused by immune cell infiltrates.
      send and Baringer, 1978a,b; Townsend, 1983).                        A puzzling aspect of HSV-induced demyelination
      Within this region, following an acute HSV infec-                is the development of lesions only within the CNS
      tion, the peripheral myelin is untouched while the               and not in the PNS. One explanation for this has
      CNS side of the trigeminal root develops demyeli-                been a reported inability of Schwann cells to be
      nated lesions. Within the demyelinated lesions,                  productively infected with HSV. Electron micro-
      there is an absence of myelin and the presence of                scopic studies have reported that following a
      both intact axons and a mononuclear cell (MNC)                   peripheral HSV infection (such as corneal inocula-
      infiltrate. The exact mechanism of demyelination is              tion) Schwann cells were found to contain viral
      not known, but results from several studies suggest              capsids, but not enveloped particles, suggesting an
      that it is a combination of cellular infection and host          abortive infection within these cells (Rabin et al,
      immune response. Townsend and Baringer (1978b)                   1968; Hill and Field, 1973). The inability of HSV to
      reported that cyclophosphamide injections reduced                productively infect and lyse Schwann cells would
      the amount of both MNC infiltrate and myelin                     prevent the attraction of immune cells and therefore
      destruction in the CNS following corneal HSV                     demyelination would not occur. This theory has
      infections. Townsend (1981a) later reported that                 been supported in a study by Townsend and Collins
      corneal infections in nude (athymic) mice resulted               (1986) in which infection and lysis of Schwann cells
      in an absence of demyelinating lesions. Kristensson              and subsequent demyelination of the PNS was
      et al (1982) demonstrated that the timing of                     achieved only by the direct inoculation of HSV into
      administration of immunosuppressive agents such                  the sciatic nerve. A productive infection of the
      as cyclophosphamide could either enhance or                      Schwann cells was established along with an
      suppress the immune reaction producing either an                 immune cell infiltrate and subsequent demyelina-
      increase or decrease in demyelination. Studies                   tion. These results suggest that during a peripheral
      comparing genetically distinct strains of mice have              HSV infection, there is some sort of block prevent-
      identified strains which range in their resistance to            ing the virus from productively infecting the
HSV in neuroscience
                                                          SL Turner and FJ Jenkins
                                                                                                                  113
Schwann cells which is overcome by direct inocu-          across synapses resulting in an absence of or
lation of the nerve.                                      relatively weak labeling of linked neurons. In
   Several investigators have also reported that          addition, nonspecific labeling of adjacent neurons
following the development of demyelinating le-            can occur at increased injection concentrations and
sions, some remyelination of the naked axons              extended labeling times. Neurotropic viruses, spe-
occurs. Remyelination was first noted by Kristens-        cifically herpesviruses, possess an advantage to
son et al (1979). Separate reports by Kristensson et      these other contemporary methods in that they are
al (1982) and by Townsend (1983) reported that the        able to replicate within neuron cell bodies provid-
remyelination appeared to be due to the migration         ing signal amplification before infecting second-
of Schwann cells from the PNS to the CNS.                 and third-order neurons. The herpesviruses have
Schwann cell remyelination of CNS axons was later         also been shown to specifically label neuronal
confirmed by Soffer and Martin (1988) using a             connections in both the retrograde and anterograde
vaginal infection with HSV-2. In at least one study,      direction (Ugolini et al, 1987; Norgren Jr, et al,
there was evidence of multiple episodes of demye-         1992).
lination followed by remyelination (Soffer and               The most common viral transneuronal tracers are
Martin, 1988). In instances where HSV establishes         herpes simplex virus 1 and 2 (HSV-1/HSV-2) and
latency and cycles through several rounds of              pseudorabies virus (PRV). All three viruses belong
reactivation, it is reasonable to postulate that cyclic   to the alpha herpesvirinae family and therefore are
episodes of demyelination followed by remyelina-          neurotrophic (Roizman and Sears, 1993). The
tion also occur.                                          ability of these DNA viruses to specifically infect
   As all of the studies just described demonstrate,      neurons contributes to their transneuronal trans-
HSV infection provides a good model for studies           port. The most common method used to detect the
investigating the demyelination of CNS axons              presence of these viruses in neuronal tissue is by
through a combination of cellular damage and              immunohistochemical staining for viral antigen.
immunopathology (ie the cytotoxic effects of an           Other methods that can be used include direct
immune cell infiltrate). Interestingly, HSV and HSV       labeling of virus particles with S35 (by growth of
antibodies have been isolated from patients exhibit-      viral stocks in the presence of S35-labeled amino
ing episodes of multiple sclerosis (MS), supporting       acids) or by incorporation of the b-galactosidase
the relevance of this model to MS-related clinical        gene in their DNA genome (Loewy et al, 1991). In
pathology (Bergstrom et al, 1989). Studies on the         the former method, presence of virus is detected by
mechanisms of CNS demyelination and the long-             photographic emulsion while in the latter method,
term effects of demyelination followed by remyeli-        tissues are incubated with a chromogenic substrate
nation may serve as an excellent model for                (such as X-gal) that detects the presence of the b-
trigeminal neuralgia (Burchiel, 1993). In addition,       galactosidase enzyme.
the HSV model may be helpful in studies investigat-          Viral tracings require certain precautions. In
ing other human demyelinating diseases such as            order to correctly interpret viral tracing studies it
Bell's palsy and multiple sclerosis (Vahlne et al,        is important to understand the mechanisms of viral
1985).                                                    uptake, transport, replication and release as well as
                                                          the generation of specific immune responses by host
A tool for transneuronal tracing studies                  immune cells and their associated neuropathology.
Neuroscientists have long desired to map chains of        Careful analyses should identify transneuronal
neurons in order to identify communication path-          connections by comparison of viral tracing results
ways from origin to termination. With the use of          with conventional transneuronal tracers that are
antero- and retrograde tracers such as fast blue and      injected concurrently to viral infection. Other
horseradish peroxidase it is possible to identify         important considerations include the presence of
single neurons along with their axons and termina-        alternate innervation pathways which could con-
tions. However, to identify synaptically linked           tribute to nonspecific labeling; for example, non-
second and third order neurons, transneuronal             specific labeling may result from dissemination of
tracers are required.                                     virus from the site of inoculation to adjacent sites.
   In order for a transneuronal tracer to be effective    Experiments used to obtain transneuronal tracings
it must be specific for synaptically linked connec-       are controlled by the strain of virus used, the host
tions, possess the ability to be transported antero- or   animal, the site of injection, the amount of virus
retrogradely and be sufficiently tagged for efficient     inoculated, the time of post-inoculation analysis,
and sensitive detection. Substances such as cholera       and the extent of neuropathology associated with
toxin, tetanus toxin and wheat germ agglutinin are        the infection. The importance of these parameters is
known to bind specifically at neuronal membranes          apparent from many studies which have reported
and have been used as transneuronal tracers (Cabot        false labeling under conditions of severe gliosis and
et al, 1984; Jansen et al, 1993; Hirakawa et al, 1993).   at late survival times. (Norgren Jr and Lehman,
These methods have limitations however, since             1989; Strack and Loewy, 1990). However, careful
only small amounts of protein are transported             analyses from several transneuronal tracing studies
HSV in neuroscience
                                           SL Turner and FJ Jenkins
114
      which contain proper controls have demonstrated                 of virus-specific transcription and translation, viral
      the use of herpes viruses to identify transneuronal             DNA synthesis, and the assembly of mature
      connections (for example, Strack et al, 1989;                   nucleocapsids. Viral capsid envelopment and
      Spencer et al, 1990; Jansen et al, 1992; Loewy et               egress are complicated pathways which differ
      al, 1991; Rotto-Percelay et al, 1992; Schramm et al,            according to viral strain and the type of cell
      1993; Hedner et al, 1993; Vizzard et al, 1995).                 infected. Regardless of the observed differences,
         Reports from several studies have identified                 two processes seem most important for the produc-
      properties of the herpesviruses that contribute to              tion and passage of mature virions. The first
      the specificity of transneuronal labeling. Vahlne               involves the acquisition of a bilaminar membrane
      and colleagues have studied the attachment and                  or transport vesicle which will permit fusion with
      adsorption of HSV-1 and HSV-2 to glial cells,                   the plasma membrane and viral egress. Secondly,
      neuronal perikarya and synaptosomal fractions of                viral glycoproteins must be processed to their fully
      brain homogenates (Vahlne et al, 1978, 1980). They              functional form by association with the Golgi
      reported that synaptosomal fractions of brain                   apparatus or golgi-derived vesicles which contain
      homogenates exhibited the strongest viral adsorp-               the enzymes required for these modifications.
      tion followed by glial cells and neuronal perikarya.            These processes are necessary to ensure that virions
      In addition, it was observed that HSV-2 bound less              will be released from the cell with the ability to bind
      efficiently than HSV-1 in each respective fraction of           via glycoprotein receptors to infect adjacent cells.
      rat, human or monkey brain preparations. However,                  Our current understanding of the mechanisms by
      HSV-1 and HSV-2 demonstrated equal binding in                   which virions traverse the cell are the result of
      mouse glial cell preparations. Pseudorabies virus               limited analyses offered by electron microscopy. In
      uptake and retrograde transport studies have                    HSV infected neurons, nucleocapsids have been
      reported the presence of limited viral specific                 observed obtaining a primary envelope at the inner
      receptors on axon terminals which mediate viral                 lamina of the nuclear membrane. Virions are
      uptake (Marchand and Schwab, 1986). These                       subsequently released into the endoplasmic reticu-
      findings suggest preferential infectivity at the                lum as naked nucleocapsids by a de-envelopment
      synapse, but also illustrate that host species type,            fusion with the outer nuclear membrane (Marchand
      viral strain and different cell types influence the             and Schwab, 1986; Lycke et al, 1988). HSV
      path of infection.                                              nucleocapsids receive a secondary envelopment
         Electron microscopic studies using HSV and PRV               by the endoplasmic reticulum and follow antero-
      have demonstrated that fusion of the viral envelope             grade transport to the cellular membrane by a
      with the cellular plasma membrane of neuronal                   transport vesicle. Virus observed in the cytoplasm
      extensions is followed by retrograde axonal trans-              of dorsal root ganglion (DRG) neurons appear to be
      port of unenveloped nucleocapsids along axonal                  contained within these transport vesicles. Fusion of
      microtubules (Ugoline et al, 1987). Although this is            vesicles with the plasma membrane at axonal
      the primary mode of viral transport to the neuronal             terminals releases infectious, enveloped virions
      nucleus, it is not exclusive. Other studies have                capable of infecting adjacent cells. Similar replica-
      shown anterograde transport of virus (Norgren Jr, et            tive processes in neurons of the vagal motor nucleus
      al, 1992; Card et al, 1990), and at least one report            have been reported in a study by Card and
      suggests that the direction of transneuronal trans-             colleagues using pseudorabies virus (PRV; Card et
      port may be strain dependent (Zemanick et al,                   al, 1993). In this study, the authors reported that
      1991). By analyzing labeled neurons at progressive              PRV nucleocapsids obtained a single envelope from
      timepoints, it has been determined that retrograde              the inner nuclear membrane and traversed the inner
      transport occurs much faster than anterograde                   lamina to gain access to the endoplasmic reticulum
      transport (Norgren Jr, et al, 1992). Consideration of           (ER). From the ER, virus fused with the outer ER
      the difference in transport rate is important in                membrane, entering the cytoplasm as naked nu-
      tracing analyses and can be useful in determining               cleocapsids which subsequently become enveloped
      connections between groups of neurons. For exam-                in a bilaminar structure by the trans cisternae of the
      ple, in groups of neurons which are highly                      Golgi. Fusion of the outer virion envelope with
      connected by collaterals, one must consider the                 synaptic membranes occurred at adjacent term-
      fact that individual neuron labeling could be due to            inals, releasing single enveloped virions with sur-
      either antero- or retrograde transport, and in such             face glycoproteins in the correct orientation to
      instances, there may be no way to distinguish                   transneuronally fuse.
      between originating and target cells. It is particu-               Although the release of herpes viruses occurs at
      larly important to consider this limitation when                neuronal terminals, sites of virion egress do not
      interpreting the specificity of viral tracing studies.          always occur directly into synaptic clefts. Herpes-
         Once delivered to the neuronal cell body, herpes             containing vesicles have been reported to fuse at
      virus nucleocapsids are observed at the pores of                presynaptic terminals, releasing enveloped virus
      nuclear membranes where they release their DNA                  which fuses to postsynaptic membranes adjacent to
      into the nucleus. The viral replicative cycle consists          presynaptic terminals. This results in the entry of
HSV in neuroscience
                                                          SL Turner and FJ Jenkins
                                                                                                                    115
naked nucleocapsids into the neuron. Astrocytes           expression of the target gene within the neuronal
are also susceptible to PRV and HSV infection, but        cells. Herpesviruses, particularly herpes simplex
infected cells are only observed subsequent to an         virus type 1, have unique characteristics of infec-
adjacent neuronal infection. Ultrastructural ana-         tion, replication and pathogenesis which make
lyses of PRV-infected astrocytes have revealed a          them potentially ideal candidates for the develop-
defect in the cytoplasmic envelopment of viral            ment of viral vectors capable of altering endogenous
nucleocapsids. This defect renders nucleocapsids          gene expression or delivery of foreign genes both in
incapable of plasma membrane fusion, resulting in         vivo and in vitro. The reader is directed to several
an absence of viral egress and an accumulation of         reviews on these subjects (Glorioso et al, 1994,
virion particles within the cellular cytoplasm (Card      1995; Neuwelt et al, 1995; Kwong and Frenkel,
et al, 1993). The resulting abortive infection            1995).
effectively prevents astrocytic PRV virions from             Herpes viruses have several advantages which
contributing to nonspecific extracellular spread. At      lend to their ability to act as neuronal vectors. HSV
present, no such mechanisms are known for HSV. In         exists as a large approximately 150 Kb double
fact, HSV has been reported to be quite capable of        stranded DNA virus which has been entirely
establishing a productive infection in astrocytes         sequenced and extensively studied (McGeoch et
(Itoyama et al, 1991). The inability of PRV to            al, 1988). As a result of many years of intense
establish a productive infection in astrocytes            research, a general knowledge exists of which genes
provides a great advantage to PRV in ensuring             and DNA sequences may be deleted and at which
specific transneuronal transport and is a major           sites foreign DNA may be inserted into the DNA
reason why PRV is considered by many to be the            genome (as reviewed in Ward and Roizman, 1994).
virus of choice for transneuronal tracing studies.        Research has also defined the minimal require-
   Additional host mechanisms restricting virus           ments for viral replication and packaging (as
spread to non-neuronal cells are provided by the          discussed in Kwong and Frenkel, 1995). HSV-based
host's immune response to both PRV and HSV                vector strategies rely on the ability of HSV to infect
infections. Resident microglia, monocytes and             neuronal cells and establish a latent infection.
macrophages are activated in the nervous system              The two main strategies for HSV-based vectors
during viral infection and may effectively phagocy-       are genetically-engineered viruses and plasmid
tose virus and degenerating cellular debris (Rina-        derived `amplicon' vectors. The first strategy
man et al, 1993). The importance of these                 involves the construction of recombinant viruses
mechanisms is apparent considering the large viral        containing deletions in one or more viral genes
load which may be released from necrotic cells to         whose expression is essential for viral replication
the extracellular space. T-lymphocytes may also           (for reviews, see Glorioso et al, 1994, 1995). The
play a role in the delineation of viral spread (Zhao et   deletion of genes whose expression is essential for
al, 1995). Factors regulating these mechanisms have       virus replication (for example, ICP4), results in the
yet to be elucidated, but most likely involve             production of a virus that cannot replicate in
immune-mediated cytokine production and the               normal cells. Replication of these defective viruses
induction of major histocompatibility antigen ex-         requires a cell line that provides the missing gene
pression within the nervous system.                       (or genes) in trans (termed a complementing cell
                                                          line). Foreign genes can be inserted into these
A viral vector for gene therapy                           mutated viral genomes with the goal of producing
The advances of modern molecular biology and in           a virus vector that will infect the target cell (ie.,
vivo gene therapy have challenged neuroscientists         neurons), and express the foreign gene without
with the potential prospect of gene manipulation in       killing the cell. The second strategy involves the use
postmitotic neurons. The ability to alter gene            of plasmid derived vectors containing HSV-1
expression in these cells would open the door             origins of DNA replication and DNA packaging
towards potential therapies for several disorders         signals which enable multiple copies of the vector
such as Parkinson's disease, Huntington's disease         genomes to be packaged into helper virus virions
and amyotrophic lateral sclerosis. Gene therapy           (for reviews see Neuwelt et al, 1995; Kwong and
using viral-based vectors has received considerable       Frenkel, 1995). Helper viruses can be either
attention and represents a major focus of ongoing         recombinant viruses containing a deletion within
research in many laboratories. Viral vectors using        an essential viral gene or viruses containing
several different human viruses such as adeno-            temperature-sensitive mutations that prevent repli-
viruses, retroviruses and herpes viruses are cur-         cation at 378C (normal body temperature). In the
rently being developed. Gene therapy directed             case of the former, the replication of the helper virus
towards neuronal cells however, presents unique           and packaging of the amplicon vector DNA must
problems. These problems include the genetic              occur in a cell line capable of complementing the
manipulation of post-mitotic (ie., non-dividing)          mutations in the helper virus. Plasmid-derived
cells, the ability to specifically infect neurons,        vectors (amplicons) are advantageous because the
long-term maintenance of the vector DNA and               DNA constructs can be easily manipulated to test
HSV in neuroscience
                                           SL Turner and FJ Jenkins
116
      endogenous, foreign, antisense or promoter gene                 Originally, strong promoter systems such as the
      expression in the target cell. Although the effi-               human cytomegalovirus IE promoter and the Rous
      ciency of delivery of these multiple copy vectors is            sarcoma virus long terminal repeat (RSV LTR) along
      high, the primary disadvantage of this system is the            with several HSV promoters were used to drive gene
      fluctuating helper virus to amplicon ratio with                 expression. Although such promoter systems were
      passage. These fluctuations must be monitored to                capable of expression they were only active
      ensure high amplicon delivery and experimental                  transiently in the CNS and did not result in long-
      reproducibility in the absence of wild-type recom-              term gene expression (as reviewed in Glorioso et al,
      binants (Kwong and Frenkel, 1995).                              1995). Neuronal specific promoters (such as the
         Regardless of the vector system used, two                    neurofilament and neuronal-specific enolase pro-
      primary goals must be achieved to enable long-term              moters) which are believed to be constitutively
      gene expression in neuronal cells. The first goal               active in neurons, also produced only transient
      involves the construct of mutant vectors which                  expression in the CNS (Andersen et al, 1993).
      themselves are non-cytotoxic to cells. Several                     A second approach to achieving long term
      studies have noted active expression of foreign                 expression in the CNS has been to use the LAT
      genes by HSV vector constructs which subsequently               promoters. As mentioned earlier, during HSV
      become inactivated (for examples see Kwong and                  latency the only viral transcripts consistently
      Frenkel, 1995; Johnson et al, 1992, 1994). Reasons              detected are the latency associated transcripts
      for the inactivation of the HSV construct are not               (LATs). The possibility that the LATs are constitu-
      apparent, but evidence suggests that it is a result of          tively expressed in latently infected neurons has
      cytotoxic effects induced by the vector. This                   made them strong candidates for long-term gene
      problem has been shown in both plasmid derived                  expression in neuronal systems. Two latency active
      and recombinant virus vectors alike. Several lines              promoter (LAP) regions upstream of the LAT
      of evidence suggest that viral immediate early (IE)             transcripts have been identified, termed LAP1 and
      gene expression may induce CPE (Johnson et al,                  LAP2 (Dobson et al, 1989; Goins et al, 1994).
      1992). Two approaches to circumvent this include                Transient expression assays and construction of
      HSV vectors with multiple IE gene deletions and                 specific deletion mutants have suggested that the
      vectors with deletions in both IE genes and the HSV             LAP1 promoter is the major latency promoter.
      VP16 gene. VP16 is a structural HSV protein                     Deletion of this promoter results in the production
      responsible for transactivating the HSV IE genes                of a virus that can establish latency but does not
      (as reviewed in Roizman and Sears, 1993). It has                express LAT transcripts (Dobson et al, 1989; Nicosia
      been reported that a HSV mutant containing                      et al, 1993). The role of LAP2 is less clear, but
      deletions in two of the five HSV IE genes, namely               appears to be required for full expression of the LAT
      ICP4 and ICP27, fails to express early and late viral           transcripts (Glorioso et al, 1995). The use of LAT
      proteins and is less cytotoxic than the parent virus            promoters for gene expression has met with mixed
      (Glorioso et al, 1994, 1995). Also, it has been                 success. LAP1 constructs have failed to produce
      reported that a virus lacking VP16 and ICP4 exhibits            long term expression in the CNS (Margolis et al,
      reduced CPE when compared to an ICP4 deletion                   1993; Lokensgard et al, 1994), but have been
      mutant (Glorioso et al, 1995). HSV expresses a                  reported to demonstrate reasonable expression in
      virion host shut-off (VHS) protein which is respon-             the PNS (Dobson et al, 1989; Wolfe et al, 1992).
      sible for degrading host cell RNA during an active              Glorioso and coworkers have reported that a HSV
      viral replication cycle (Read and Frenkel, 1983).               construct containing a lac Z gene under the control
      This protein may also be involved in cytopathic                 of the LAP2 promoter is expressed in the brain for at
      effects although a recent study reported no reduc-              least 41 h following inoculation (Glorioso et al,
      tion in CPE using vectors containing mutations in               1995). Thus, while short term expression in either
      the VHS gene (Johnson et al, 1994). Other packaging             the CNS or PNS appears achievable, long term
      alternatives may also be explored as ways to deliver            expression in the CNS is still not a definite reality
      HSV vectors that are incapable of establishing a                and more research is required to produce an
      lytic infection. One such method involves the use of            acceptable HSV vector.
      L particles, which are defective HSV virions lacking
      viral DNA, yet are capable of entering cells without            A model for studying interactions among the
      causing host cell damage (Glorioso et al, 1994;                 nervous, immune and endocrine systems
      Rixon et al, 1992).                                             In immunocompetent hosts, active herpes simplex
         The second goal in the development of HSV                    virus infections rapidly stimulate immune re-
      vectors capable of long-term gene expression                    sponses which function to restrict viral replication
      involves designing stable, active promoters capable             and the spread of virus. In addition, the host's
      of expressing appropriate levels of the foreign                 immune response is most likely involved in the
      protein. The specific promoter involved in indivi-              establishment of latent infections. In fact, the
      dual therapies may change according to the type,                establishment of latency could be viewed as a
      status, and activity of the neuronal cell of interest.          double-edged sword. It provides the host with a
HSV in neuroscience
                                                            SL Turner and FJ Jenkins
                                                                                                                      117
mechanism to limit viral spread and cellular                by a cellular protein termed Oct-1 (Gerster and
damage which, at the same time, ensures the                 Roeder, 1988; Kristie and Roizman, 1987; O'Hare
persistence and survival of the virus. In HSV-              and Goding, 1988). The Oct-1 DNA complex is then
infected neonates and immunocompromised hosts,              bound by a viral protein called the alpha trans-
virus replicates to high titers, is often widely            inducing factor (aTIF) or VP16 (Gerster and Roeder,
disseminated and the resulting viral pathology              1988; McKnight et al, 1987; O'Hare et al, 1988;
generally extensive. While the mechanisms in-               Preston et al, 1988). The aTIF protein is contained
volved in controlling HSV replication and establish-        within infectious HSV viral particles and is there-
ment of latency are not completely understood, it           fore introduced into the cell upon viral infection.
has become increasingly evident that they involve           The binding of aTIF to the Oct-1/DNA complex
interactions among nervous, immune and endo-                results in a transactivation of the HSV alpha gene
crine systems. The strongest evidence for these             promoters and is responsible for the rapid induction
interactions is seen in the establishment of HSV            of the HSV alpha genes immediately following
latency.                                                    infection. Since the complex between aTIF and
   As mentioned earlier, several studies have sug-          Oct-1 is required for the transcription of the a genes,
gested that within neurons, the decision to establish       the Oct-1 protein represents a positive cellular
either latency or active viral replication occurs very      transcriptional factor used in the regulation of
soon after infection (Simmons et al, 1992; Speck and        HSV genes. The cellular Oct-1 protein belongs to a
Simmons, 1992). This is indicated by the identifica-        family of proteins that are capable of binding to
tion of at least three distinct populations of neurons in   DNA sequences containing octomer motifs such as
infected ganglia. These populations include neurons         TAATGARAT.
that express HSV antigens but not LAT, neurons that            Several laboratories have reported that HSV
express LAT but not HSV antigens, and a small               replication is restricted in the mouse neuroblastoma
proportion of neurons that coexpress LAT and HSV            cell line C1300 (Vahlne and Lycke, 1978; Kemp and
antigens (Simmons et al, 1992; Speck and Simmons,           Latchman, 1989) and in neuronal cell lines derived
1992). It is reasoned that neurons which express HSV        from dorsal root ganglia (Wheatley et al, 1990). The
antigens and no LAT represent active infections             restriction of viral replication within these cells
while those that express LAT and no antigen                 occurs at the level of HSV a gene expression and
represent latent infections. Early divergence (to           appears to be due to the presence of a cellular
active replication or latency) is further supported by      repressor that binds to the TAATGARAT motif
the realization that lytic replication commits the cell     (Kemp and Latchman, 1989; Werstuck et al, 1990).
to viral control and cell death as discussed below.         Lillycrop et al (1991) defined the cellular repressor
   The infection of cells by HSV rapidly results in         as a neuronal homolog to the Oct-2 transcriptional
cell death. Consequences of HSV infection include           factor. Oct-2 is related to Oct-1 and has been shown
the disruption of polyribosomes, inhibition and             to be capable of binding to the TAATGARAT
shut-off of host macromolecular synthesis and               sequence (Gerster and Roeder, 1988). Presumably
margination of nuclear chromatin (for review see            Oct-2 repression occurs as a result of binding
Roizman and Furlong, 1974). The establishment of a          (without the aTIF protein) to the TAATGARAT
latent HSV infection requires that the infected             sequence in a promoters. They also demonstrated
neuron must survive infection. In order to prevent          that this Oct-2 protein was expressed in dorsal root
cell death by HSV, viral replication must be stopped        ganglion neurons. Increasing the levels of Oct-2 in
relatively early in its replication cycle. Once the         the neuron cultures by transfecting a plasmid
infected cell is committed to viral DNA replication,        containing the Oct-2 gene decreased the amount of
it is targeted for death. Therefore, to establish a         HSV a gene transcription in these cells. It is
latent infection, the HSV replicative cycle must be         important to note that Oct-2 was originally defined
stopped prior to b protein synthesis. Inhibition at         as a transcriptional activator of genes in B cells
the level of a gene transcription would achieve this        (Singh et al, 1986; Staudt et al, 1986) and had not
goal. The establishment and maintenance of HSV              been attributed with any repressor activity. More
latency therefore, may be due to either the absence         recent results have demonstrated that differential
of specific cellular transcriptional regulatory pro-        splicing results in the formation of different iso-
teins that are involved in the positive induction of        forms of the Oct-2 protein (Wirth et al, 1991), and
HSV a genes or to the presence of cellular proteins         the predominant form in lymphocytes is distinct
that repress the specific transcription of the a genes.     from the predominant form found in neurons
   At the molecular level, the HSV a genes are              (Lillycrop and Latchman, 1992). In transient ex-
unique with regard to the other HSV genes in that           pression assays the lymphocyte form was reported
their promoter DNA sequences contain one or more            to act as a positive transactivator of HSV IE
copies of the octomer DNA sequence motif TAAT-              promoters while the neuronal form was found to
GARAT (R=Adenine or Guanine; Mackem and                     be a repressor of IE transcription (Lillycrop and
Roizman (1982); Kristie and Roizman (1984). In a            Latchman, 1992). In addition, Wood and co-workers
lytic viral replicative cycle, this DNA motif is bound      (Wood et al, 1992) reported that NGF could
HSV in neuroscience
                                           SL Turner and FJ Jenkins
118
      upregulate (three- to fourfold) the level of Oct-2 in           ment of HSV latency which is dependent on
      neuronal cultures from dorsal root ganglia. NGF had             interactions between immune cells, the endocrine
      no effect on the levels of Oct-1 or a neuronal-                 system and cells of the PNS (Jenkins and Baum,
      specific transcriptional factor Brn-3. These results            1995, Figure 1). During an active HSV infection
      suggested that NGF could mediate the transcrip-                 virus replicates peripherally and is transported to
      tional effects of Oct-2 in neurons.                             the nuclei of the sensory ganglia innervating the site
         Studies using nerve transection or nerve organ               of infection. Viral replication occurs both at the
      cultures have found that the NGF levels in non-                 peripheral site and in some neurons, resulting in an
      neuronal cells increase following nerve injury                  immune response characterized by the attraction of
      (Heumann, 1987; Heumann et al, 1987a,b). This                   activated macrophages, at both the peripheral site
      increase in NGF was due, at least in part, to the               and within the sensory ganglia. The activated
      presence of activated macrophages that were                     macrophages produce a local increase in IL-1 and
      responding to the site of injury, and this macro-               TNF which in turn results in an increase of NGF.
      phage-induced increase in NGF was subsequently                  The additional NGF is taken up by the neurons
      found to be due to the secretion of the cytokine IL-            creating an increase in the Oct-2 transcriptional
      1 (Lindholm et al, 1987). Using cultured explants               factor. This increase acts to suppress HSV a gene
      of sciatic nerves, the addition of cultured medium              transcription, leading to the establishment of
      from activated macrophages or of IL-1 resulted in               latency. Levels of Oct-2 would increase in neighbor-
      a 14-fold increase in NGF mRNA (Lindholm et al,                 ing neurons as well, including both infected and un-
      1987). This increase was transient and in order to              infected cells. Neurons in which viral replication
      maintain the higher levels of NGF mRNA, the IL-1                had proceeded beyond viral DNA replication, and
      had to be replenished. This increase was not                    which express viral proteins, would most likely be
      totally IL-1 specific since the addition of tumor               targeted for destruction either through viral-in-
      necrosis factor (TNF) or platelet-derived growth                duced lysis or immune-mediated cytolysis. Under
      factor resulted in a doubling of the NGF mRNA                   these conditions, latency would occur in those
      levels. However, the addition of prostaglandin E2               neurons which contained viral DNA but had yet to
      or fibroblast growth factor did not result in any               initiate viral replication (ie expressing LAT but not
      mRNA induction. In a subsequent study, Lind-                    HSV antigens). Thus the IL-1/NGF/Oct-2 mediated
      holm and co-workers reported that the mechanism                 resistance to HSV transcription is somewhat analo-
      of the IL-1-induced increase in NGF was an                      gous to viral resistance mediated by the interferons.
      increase in the stability of the NGF mRNA                       That is to say that peripheral replication of HSV
      (Lindholm et al, 1988).                                         may induce a non-permissive environment for viral
         Other evidence further supports a role for NGF in            replication in neurons innervating the site of
      the maintenance of neuronal latency. Wilcox et al               replication. Animal models using corneal infec-
      (1990); Wilcox and Johnson (1988); Wilcox and                   tions, have demonstrated that infectious HSV is not
      Johnson Jr (1987), describe an in vitro model system            detected within trigeminal ganglia until 48 ± 72 h
      in which latency is established in cultured sympa-              post-infection (Coen et al, 1989; Jenkins and Martin,
      thetic and dorsal root ganglion neurons. This                   1990; Tenser and Edris, 1987). This delay would
      system relies upon the addition of NGF in addition              allow for the establishment of a IL-1/NGF/Oct-2-
      to antiviral drugs to establish latency. In this                mediated non-permissive environment. This model
      system, removal of NGF disrupts latency, lytic gene             is based on studies performed in vitro using either
      transcription resumes and viral reactivation occurs.            established cell culture lines or primary cell lines. It
      This model suggests that NGF is required for the                will be important to determine if this model is
      maintenance of neuronal latency since its removal               correct for the establishment of HSV latency in vivo
      results in viral reactivation. Later studies have               using well-established animal models.
      shown that the mechanism of NGF dependent
      latency involves binding of NGF to the NGF                      A model for studying health consequences of
      receptor (TrkA) and the stimulation of second                   psychological stress
      messenger pathways (Smith et al, 1992). It has also             In addition to the establishment and maintence of
      been found that interruption of RNA transcription               a latent infection, herpesvirus infections are also
      or protein synthesis in these latently-infected cells           characterized by recurrent infections that are
      results in viral reactivation with similar kinetics to          caused by stress or stress-related reactivations
      NGF withdrawal. These data suggest the presence of              from the latent state. Scientists in the fields of
      a neuronal factor, regulated by NGF receptor                    neuroscience and behavioral medicine have long
      binding, which contributes to the maintenance of                desired to understand the mechanisms by which
      the latent state. The similarities between this in              psychological processes affect biological function.
      vitro system and the in vivo condition have yet to be           The reactivation of herpesviruses provides an
      determined.                                                     excellent model for studying the direct relation-
         Based on the studies described in this section, we           ship between biobehavioral processes and dis-
      have postulated a molecular model for the establish-            ease.
HSV in neuroscience
                                                                       SL Turner and FJ Jenkins
                                                                                                                                             119

Figure 1 Model for the establishment of HSV latency in sensory neurons. Sensory neurons are depicted as large triangular cells with
a circular nucleus. Surrounding the neurons are smaller satellite cells shown as small circles. A primary HSV infection occurs at a
peripheral site producing a `fever blister' (A). The virus is transported to the nuclei of sensory ganglia by retrograde transport along
axonal fibers (B). (1) and (2) represent un-infected and infected neurons respectively, surrounded by satellite cells. Virus replication
occurs in some of the neurons (3) resulting in the insertion of viral proteins on the surface of the neuron (4). Macrophages are attracted
to these neurons (5) where they become activated and begin producing IL-1 (6). The IL-1 causes an increase in the levels of NGF
produced by the satellite cells which is taken up by the neurons (7). Increased levels of NGF in the neuron result in an increase in the
levels of Oct-2 (8) which results in an inhibition of viral transcription resulting in the establishment of latency (9). Reprinted with
permission from Jenkins and Baum, 1995.

   Studies in behavioral medicine have demon-                          strated an increase in HSV antibody levels in
strated the ability of psychological stress to induce                  chronically-stressed individuals. McKinnon et al
viral reactivation. Several studies from the Glaser's                  (1989) reported that individuals living within a
laboratories at Ohio State University have demon-                      three mile radius of the Three Mile Island nuclear
strated that antibody titers to latent herpesviruses,                  plant in Middletown, Pennsylvania demonstrated
particularly Epstein-Barr virus (EBV) are elevated in                  higher antibody titers to HSV than did control
subjects who have been chronically stressed as                         subjects living in Fredrick, Maryland. This increase
compared to non-stressed control groups (Kiecolt-                      is specific for HSV and not due to a nonspecific
Glaser and Glaser, 1986; Kiecolt-Glaser et al, 1991;                   polyclonal B cell activation since antibody titers to
Glaser et al, 1985; Kiecolt-Glaser et al, 1987). In one                Rubella virus did not differ between groups. In
example, stress associated with exam-taking in first                   addition, increases in HSV antibody titers were not
year medical students resulted in significant in-                      correlated to a concurrent increase in the presence
creases in EBV antibody titers (Glaser et al, 1994). In                of HSV lesions (A Baum, personal communication)
addition, the modulation of EBV antibody levels has                    suggesting that viral reactivation resulted in either
been used to define effective coping strategies in                     asymptomatic shedding or an abortive reactivation.
psychotherapy patients (Esterling et al, 1994).                        It is unlikely that these increases occur in indivi-
These types of studies suggest that the observed                       duals who regularly reactivate HSV, since their
increases in viral antibody titers is due to reactiva-                 antibody levels would be extremely high at base-
tion of latent virus which acts to trigger the immune                  line. The notion of an abortive reactivation that
response. A similar type of study has also demon-                      results in increases in HSV antibody titers without
HSV in neuroscience
                                            SL Turner and FJ Jenkins
120
      viral shedding or recurrent disease is particularly                 The most common endpoint for detecting viral
      intriguing. In these instances, antibody levels could            reactivation has been the isolation of infectious
      be used as a marker for stress-induced reactivation              virus from either ganglia or tissues. Although this is
      in individuals who do not routinely present with                 a fairly sensitive method, it fails to detect abortive
      recurrent HSV infections. Since a majority of                    replication and therefore may not detect a large
      individuals in the general population are felt to                percentage of reactivation events. In order to further
      have latent HSV in their sensory ganglia while only              define distinct reactivation triggers, analyses utiliz-
      a small percentage actually demonstrate recurrent                ing methods such as immunohistochemistry, in situ
      disease (as reviewed in Whitley and Gnann, 1993),                hybridization and RNA PCR may be necessary.
      titers to HSV antibodies may serve as a useful                      Another reactivation approach involves modula-
      marker for stress-induced abortive reactivations in              tion of the host immune response. When mice are
      most people.                                                     preimmunized with HSV specific antibodies latency
         The investigation of the role(s) of psychological             is established in the absence of observable pathology
      or physical stress in reactivation of latent herpes-             and the hosts immune system remains naive to viral
      viruses needs an appropriate animal model. While                 antigen (Birmanns et al, 1993). Subsequent exposure
      animal models for the establishment of herpes                    to reactivation stimuli leads to reactivation as
      latency exist, useful models for reactivation are                identified by virus recovery and increased antibody
      lacking. Existing in vivo animal models usually                  titers. This preimmunization protocol results in
      reactivate at low frequencies and require stimuli                increased reactivation percentages as compared to
      that are often difficult to associate with human                 non-immunized mice (Birmanns et al, 1993).
      phenomena. Animal models for HSV latency                         Although direct mechanisms for this phenomena
      include both mouse and rabbit models in which                    have yet to be elucidated, increased levels of viral
      peripheral inoculation with wild-type HSV (eg. on                DNA were observed in ganglia of preimmunized
      scarified corneas or in footpads) results in the                 mice. It should be considered that this facilitation of
      establishment of latency in neurons of the sensory               reactivation may be a function of increased detect-
      ganglia inervating the site of inoculation (such as              ability due to an unprimed immune response, and
      the trigeminal or dorsal root ganglia). The most                 not altered triggering stimuli or a prevention of
      common method for reactivation of latent HSV                     reactivation by an active component of the immune
      (from sensory ganglia) is the explanation of                     response. With this in mind, the importance of
      latently-infected tissue into cell culture medium                effective and sensitive measures of reactivation
      (Devi-Rao et al, 1994; Tanaka et al, 1994; Cai et al,            becomes apparent.
      1993b). Following explant, infectious virus can be                  Stress and its effects on the body and on different
      detected in these tissues 1 ± 5 days later. Some                 systems in the body have recently become more
      studies have utilized this method to evaluate viral              thoroughly defined in biological respects. Key
      mutants and different strains for reactivation                   factors, including the hypothalamic-pituitary-adre-
      competency in efforts designed to identify viral                 nal axis and glucocorticoid and catecholamine
      genes that are involved in reactivation (for exam-               regulation, have been shown to affect the nervous,
      ple, see Cai et al, 1993b). This procedure, although             endocrine and immune systems (for reviews, see
      highly efficient at reactivation (generally 80 ± 95%             Besedovsky and Del Rey, 1991; Dunn, 1989; Black,
      effective), represents an in vitro method which                  1994; Blalock, 1989). A combined goal of behavioral
      should not be interpreted as an in vivo reactivation             scientists, neuroscientists and herpes virologists
      protocol, even though it results in the reactivation             should be to define these responses with respect to
      of virus from latently-infected host tissue.                     viral reactivation. In addition, systemic responses
         Reactivation procedures representing in vivo                  to psychological stressors need to be defined locally
      protocols include epinephrine-iontophoresis in rab-              at the site of viral latency and reactivation. These
      bits, intravenous administration of cyclophospha-                goals will only be met using multi-disciplinary
      mide and dexamethasone, corneal scarification,                   approaches and collaborations.
      ultraviolet light exposure, surgical trauma, adminis-
      tration of cadmium sulfate, and hyperthermia                     Conclusions
      (Sawtell and Thompson, 1992; Fawl and Roizman,                   Herpes simplex virus, as a result of its rather unique
      1993; Harbour et al, 1983; Blyth et al, 1976; Bloom et           life cycle in humans, is a useful tool for many areas
      al, 1994). Most of these suffer from the criticism that          of research. Its use in the field of neuroscience
      they do not have an analogous use in humans.                     represents the newest and certainly one of the most
      Ideally, what is needed is a reactivation protocol               interesting and complex directions of research.
      that is based on a psychological stressor and results            Studies utilizing HSV in gene therapy, neuronal
      in significant levels of viral reactivation. However, at         tracings, demyelinating diseases, neuro-immune
      the present time, such a protocol does not exist. The            interactions and health related effects of psycholo-
      major stumbling block may be the simple fact that                gical stress will continue for many years and should
      HSV is a human pathogen which doesn't behave                     provide important insights in areas that up to now
      identically in non-human animal models.                          have been most difficult to study.
You can also read