Sei sulla pagina 1di 9

Review TRENDS in Neurosciences Vol.29 No.

7 July 2006

INMED/TINS special issue

Viral infections in the developing


and mature brain
Anthony N. van den Pol
Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA

A number of different RNA and DNA viruses can invade infect the brains of humans or animals and cause
the brain and cause neurological dysfunction. These neurological dysfunction.
range from the tiny polio picornavirus, which has only With increasingly open trade in food, animal products
7 kb of RNA genetic code that preferentially infects and pets, and faster human transportation in the
motor neurons, to the relatively large cytomegalovirus, shrinking world, exotic viruses can potentially move into
which has O100 genes in its 235 kb DNA genome and new geographical locations and expand both their
causes various neurological problems in the developing geographical range and host species, as has happened
brain but is comparatively harmless to adults. This brief with human immunodeficiency virus (HIV) in the past
overview of some aspects of neurovirology addresses three decades from Africa, with West Nile virus (WNV) in
the complex problems that underlie an appreciation of the past decade from the middle east, and with H5N1
the contribution of viral infections to brain disease. [This avian influenza from east Asia currently. HIV and WNV
review is part of the INMED/TINS special issue Nature can cause neurological problems in humans, and H5N1
and nurture in brain development and neurological can do so in animals [3]. These three viruses have each
disorders, based on presentations at the annual passed to humans from non-human species: HIV from
INMED/TINS symposium (http://inmednet.com/).] chimpanzees or other non-human primates, WNV from
birds via mosquitoes, and H5N1 from birds directly. If we
Introduction look at the demographics for HIV, the virus was unknown
Despite years of study, and a clear view of which regions in 30 years ago, and now the world count of infected humans
the brain are involved, we still do not know what causes is approaching 40 million (http://www.cdc.gov/hiv/
most neurological and psychiatric diseases. The specific resources/factsheets/index.htm); HIV infections that pro-
characteristics and symptoms for different CNS diseases gress to AIDS have a 50% probability of inducing
are for the most part determined by the identity of the neurological dysfunction, ranging from neuropathy to
brain cells or pathways that are damaged or lost. Several full dementia.
potential factors have been identified that contribute to Viruses can cause neurological problems not only by
CNS disease, including environmental neurotoxins, cytolytic actions on neurons or glia, but also by inducing
genetic substrates, developmental trauma and nutrition. apoptosis, damaging the blood–brain barrier, initiating
Viruses that invade the brain have been postulated to autoimmune attack on specific cells, expressing viral
cause some CNS diseases, but the evidence to support this genes and repressing cellular genes, causing fusion of
view is complicated, and the data are sometimes based cells, altering neuronal migration, attenuating neural
more on correlation than on causation. One problem that progenitor replication, and blocking cerebrospinal fluid
underlies any hypothesis about viral mechanisms of brain generation and flow. The multiple mechanisms of viral
dysfunction relates to the large number of different induction of CNS dysfunction further complicate an
viruses that can infect the brain, each with a unique understanding of viral agents in brain disease.
mechanism of entry, replication, defense against the
immune system, and dissemination, and each with Genetic predisposition to viral infection
preferred target cells and developmental stages of It is difficult to divorce viral infections of the brain from
infection [1,2] (Table 1). Some viruses are common (e.g. potential genetic sensitivity to such infections. A crucial
herpes simplex or influenza), and although the majority of factor in whether a virus gets into the brain relates to
humans have been infected by them only a small how efficient the immune system is in eliminating the
percentage show serious neurological symptoms due to virus from the whole body. A virus that is rapidly
these infections. Other viruses are rare, and might be controlled by the immune system is less likely to gain
encountered only in discrete or isolated regions of the access into the CNS. Examples in studies with rodents
world (e.g. viruses with exotic names such as Kemerova, underline the crucial importance and interaction of the
Lebombo, Bluetongue, Nyabira, Pootvoot, Kenai, Nipah genetic background with specific viruses. Cytomegalo-
and Changuinola), but on occasion have been found to virus (CMV) is an enveloped (i.e. external lipid
Corresponding author: van den Pol, A.N. (anthony.vandenpol@yale.edu). membrane) double-stranded DNA virus in the herpes
family that can cause substantial damage to the
www.sciencedirect.com 0166-2236/$ - see front matter Q 2006 Elsevier Ltd. All rights reserved. doi:10.1016/j.tins.2006.06.002
Review TRENDS in Neurosciences Vol.29 No.7 July 2006 399

Table 1. Properties of seven viruses that infect the nervous system and cause neurological dysfunctiona,b

Name CMV HSV Rabies HIV Polio WNV LCMV


Virus family Beta herpesvirus Alpha herpesvirus Rhabdovirus Retrovirus Picorna virus Flavivirus Arenavirus
Genome (C) DNA (C) DNA (–) RNA (C) RNA (C) RNA (C) RNA (C/–) RNA
Double- Double- Single- Single- Single- Single- Ambisense
stranded stranded stranded stranded stranded stranded single-stranded
linear linear linear linear (2x) linear linear circular

Genome size 235 kb 152 kb 11 kb 9.2 kb 7.5 kb 11 kb 3.5C7 kbc


Number of genes O100 w100 5 9 1d 1d 2C2c
Virus size w200 nm w200 nm 75!180 nm 100 nm 28 nm 45 nm 50–200 nm
Envelope Yes Yes Yes Yes No Yes Yes
Route of Cell-mediated Trigeminal nerve Retrograde Cell-mediated Retrograde Direct BBB BBB crossing
brain entry BBB crossing Olfactory nerve axonal BBB crossing axonal crossing
BBB crossing
Neurological Deafness Encephalitis Guillain–Barre- Dementia Paralysis Encephalitis Developmental
Developmental Herpetic neuralgia like syndrome Encephalitis Respiratory Meningitis brain defects
brain defects Meningitis Progressive Myelitis arrest Myopathy Encephalitis
Encephalitis Myelitis encephalitis Neuropathy Hydrocephalus
Epilepsy Paralysis Meningitis
Guillain–Barre
syndrome
Retinitis
a
Major routes of entry are listed, but as for most viruses, other routes are not excluded. Similarly, the list of symptoms is not exhaustive, and many other symptoms can be
produced by infection of the CNS by each virus [1,2,23,31,62].
b
Abbreviations: BBB, blood–brain barrier; CMV, cytomegalovirus; HIV, human immunodeficiency virus; HSV, herpes simplex virus; LCMV, lymphocytic choriomeningitis
virus; WNV, West Nile Virus. DNA genomes are blue, negative-stand RNA is pink, and positive-strand RNA is green.
c
Short and long segments.
d
Encoding a single large polyprotein. Protease cleavage yields as many as 11 (polio virus) or 10 (WNV) virally encoded proteins

developing brain. Different strains of mice show strik- but the virus remained highly infective for most human
ingly different responses to viral infections. For instance, tumor cells [9].
the C57Bl/6 strain is resistant to CMV infections, Many neurological or psychiatric diseases, including
whereas Balb/c mice are susceptible; a difference in a Parkinson’s disease, schizophrenia, autism, obsessive
single mouse gene that regulates natural killer cells compulsive disorder, Tourette’s syndrome and narcolepsy,
seems to be responsible [4,5]. The difference in genetic show a genetic contribution [10–12] (Persico and Bour-
sensitivity to a virus is not simply due to generally weak geron, in this issue). However, the genetic substrate
immune surveillance by Balb/c mice, because when appears to have only a partial, sometimes minor, role in
infected by a different virus the tables are reversed. the manifestation of many of these diseases, indicating
For instance, when infected by intranasal application of other factors, probably environmental and sometimes
a neuro-adapted Sindbis virus, a positive-strand RNA viral, have crucial roles in concert with an underlying
virus, 100% of the C57Bl/6 mice died from fatal genetic substrate. A genetic contribution to neurological
encephalitis; by contrast, all of the Balb/cBy mice dysfunction might generate mistakes in brain develop-
infected by the virus in this way survived [6]. Similar ment or wiring for instance, but could also alter sensitivity
sensitivities are found in human viral infections. to viral infections within the brain that might lead to
destruction of select cell types.
Humans with a mutated chemokine receptor CCR5, a
co-receptor for HIV, show a substantial reduction in the
probability of developing AIDS; by contrast, there is a Many infected, few show symptoms
higher incidence of West Nile encephalitis associated One of the biggest complications in trying to understand
with the mutated receptor [7]. Although outside the the role of viruses in human neurological dysfunction is
scope of the present review, minor shifts in viral genomes the striking difference in symptoms of those infected.
can also have an important role in neurovirulence: for This is perhaps best exemplified by polio virus, a small,
example, a single amino acid shift from cysteine to serine non-enveloped positive-strand RNA virus. Although
at position 102 of WNV substantially attenuated eliminated in many parts of the world today by
neurovirulence [8]. A single nucleotide mutation at immunization, there are geographical pockets where
position M51 in vesicular stomatitis virus (VSV, polio still exists. Unfortunately, polio seems to be
Figure 1), a bullet-shaped RNA virus, reduced the ability returning into regions in which it had been eradicated,
of the virus to infect normal interferon-responsive cells, in part because of regional political instability leading to
www.sciencedirect.com
400 Review TRENDS in Neurosciences Vol.29 No.7 July 2006

Figure 1. Electron micrograph of neurons infected by vesicular stomatitis virus (VSV). Bullet-shaped VSV is shown infecting hippocampal neurons in vitro; rabies in the same
family of rhabdoviruses has a similar ultrastructural morphology. The long arrow points to VSV being taken up in clathrin coated pits in a dendrite. Short arrows point to
viruses that are outside the cell, with one arrow showing the shape of the virus in a longitudinal dimension and the other showing the virus cut in cross-section. Scale bar,
200 nm.

impaired immunization programs. Similar to smallpox, cutting the axons that innervate rabies-infected muscles
the possibility of eradicating polio virus completely is or using colchicine to stop axonal transport blocks
greatly increased by the absence of an animal reservoir movement of the virus into the brain, and neurological
for the virus. An important take-home message from illness is prevented [21,22] (Figure 2). Once within the
polio is that the typical neurological symptoms of full or CNS, rabies spreads rapidly (Figure 3). Unlike many
partial motor paralysis are generally found in !1% of other viruses, rabies might not cause cytolytic death of
non-immunized humans infected with the virus. In other the infected neurons; rather, it takes command of the
words, the great majority of those infected seldom even translational pathways in neurons, blocks the synthesis
know that they are, or have been, infected with this of proteins that neurons normally make, and conscripts
potentially paralyzing virus [13]. Similar to most other these pathways into synthesizing proteins encoded by the
viruses that can infect the brain, polio virus is not viral RNA genome. This renders the neuron unable to
restricted to brain cells and can infect other cell types carry out its normal signaling function, because it cannot
outside the CNS. Polio is primarily a virus of the generate neuronal molecules [23]. Remarkably, neuro-
gastrointestinal tract. Although the receptor for polio pathological examination of a rabies-infected brain might
virus has been identified as a member of the immu- show little tissue necrosis or cellular loss, whereas
noglobulin superfamily [14,15], the crucial mechanism immunocytochemical detection of viral antigens will
that enables polio to spread into and replicate within the reveal widespread viral infection of neurons [18].
motor neuron system in the spinal cord has not yet been Similarly, lymphocytic choriomeningitis virus (LCMV)
determined. An emerging virus that can cause neuro- and Borna disease virus (BDV) can infect glia and
logical symptoms similar to polio is enterovirus 71 neurons without killing them, and can lead to long-term
(which causes hand, foot and mouth disease) [16,17], infections; BDV causes severe behavioral disturbances in
another small RNA virus from the same family. Similar many animal species, and has been associated with
to polio virus, only a small percentage of humans increased incidence of psychiatric disease, including
infected with WNV show neurological symptoms (http:// schizophrenia and bipolar disorder, in humans [24,25].
www.cdc.gov/ncidod/dvbid/westnile/). In humans, LCMV fetal infection can lead to serious
In contrast to polio and WNV, infections by rabies neurological complications; infections in adults generally
virus, a negative-strand RNA virus, are much more likely do not. In a rodent brain exposed to LCMV, the virus
to be dangerous. Although the probability of infection infects cerebellar neurons and periventricular neurons,
with rabies virus is only 15% after a bite from a rabid dog but only the cerebellar neurons die [26]. Interestingly,
[2], humans with symptomatic rabies rarely survive. The one mechanism for cell death is a direct attack on
crucial event here is whether the rabies virus is taken up infected cerebellar cells by CD8CT cells; in rodents
by motor axons and retrogradely transported back into with a weakened T-cell immune response, cerebellar
the spinal cord. Three possible neuronal receptors for cells are spared [27]. This is another example of the
rabies have been identified: the ACh receptor [18], neural genetic substrate for immune surveillance having a
cell adhesion molecule (NCAM) [19] and the low-affinity crucial role in neurological damage resulting from viral
nerve growth factor (NGF) receptor P75 [20]. Either infections; in this case, a mouse with a vigorous T-cell
www.sciencedirect.com
Review TRENDS in Neurosciences Vol.29 No.7 July 2006 401

(a) Anterograde transport into brain from (c) Direct BBB crossing
peripheral cell body via olfactory nerve e.g. WNV, EEE
e.g. VSV, HSV, influenza

(d)
‘Trojan Horse’ crossing
e.g. HIV, CMV
(b) Axon terminal uptake and transport
back to cell body in CNS or PNS

DRG neuron Motorneuron


e.g. VZV, HSV e.g. Rabies

Figure 2. Virus entry into nervous system. (a) Several viruses (VSV, herpes, influenza, pseudorabies virus) can enter the brain by traveling in an anterograde direction along
the olfactory nerve into the olfactory bulb [47,53,63]. (b) Herpes viruses such as varicella zoster virus (VZV) and herpes simplex virus (HSV) commonly enter the trigeminal
nerve or dorsal root ganglion sensory nerves, traveling toward the cell body during primary infection and, if awakened from latency in the cell body, back to the periphery
[62]. Some viruses such as rabies, and in some cases polio, enter the brain after retrograde axonal transport from muscles or other tissue. (c) West Nile virus (WNV) and
eastern equine encephalitis (EEE) can cross the blood–brain barrier (BBB) directly, or can infect endothelial cells and subsequently cross the BBB. (d) Human
immunodeficiency virus (HIV) and cytomegalovirus (CMV) cross the BBB hidden in ‘Trojan Horse’ macrophages or monocytes, and subsequently are released to infect other
cells within the brain. All viruses in this diagram are illustrated as small green hexagons.

response to LCMV might lose more CNS neurons than not clearly understood, although it might relate in part to
one with a weak immune response. the developmental period of infection, with infections in
the first trimester being the most problematic. In contrast
One virus, multiple neurological syndromes: to its effects on the developing brain, although CMV
developmental perspective infection is fairly common in adults it causes relatively
Several viruses that can cause neurological dysfunction little neurological problem, except in immunocompro-
show development-dependent preferences. CMV is a mised individuals such as AIDS patients. It is one of the
member of the herpes family, with a genome of w235 kb opportunistic viruses that invade the brain when the
that encodes O100 genes. Similar to the other seven immune system is weakened [31,35].
members of the herpes family that infect humans, This raises the question of why CMV is a problem in
infection can lead to viral latency, both within [28] and the developing brain but not in the mature brain. One
outside the brain. In latency, the viral genome remains reason is that in early development, the systemic immune
within the nucleus of infected cells, and can be reactivated system is not fully functional, and the T and B
to produce replication-competent virus (Figure 4). As with lymphocytes, natural killer cells and macrophages/mono-
other herpes viruses, CMV can remain latent for a human cytes are not mature enough to eliminate a viral infection
lifetime. CMV is particularly dangerous to the developing effectively and rapidly [29,36–38]. The developing brain
human or mouse fetus, and has been suggested as the also has a poor blood–brain barrier, so if a virus enters
number one infectious agent causing birth defects and the developing fetus its passage into the brain is not
neurological dysfunction in the developing human brain impeded as it would be in the adult brain. CMV appears
[29–31]. CMV can cause various neurological problems to have an intrinsic preference for developing brain cells
including epilepsy, blindness, deafness, polymicrogyria, [39,40], perhaps related to the inability of developing
microcephaly, hydrocephalus and mental retardation [32– neurons or glia to effect an efficient cellular immune
34]. The probability of CMV-mediated neurological pro- response. When a CMV promoter such as CMV ie1 is used
blems has been estimated at 0.1% of births, with a to drive reporter genes, transgenic mice show greater
possibility of additional subtle problems such as learning expression during early brain development, suggesting
deficits in infected children [2,33]. As with some of the that transcriptional activation of some of the viral genes
other examples described here, only a fraction (!10%) of is selectively enhanced in the developing brain [41,42].
intrauterine CMV infections result in neurological dys- Depending on which part of the brain is infected, CMV
function. Why some fetuses show no CNS debilitation is causes different neurological syndromes [39,43].
www.sciencedirect.com
402 Review TRENDS in Neurosciences Vol.29 No.7 July 2006

(a)
Rabies, PRV (Bartha strain):
Retrograde trans-synaptic transmission

2 3
1

(b)
PRV:
Anterograde trans-synaptic transmission

2
1

(c)
CMV, VSV:
Cell-to-cell diffusion
2

2
2

Figure 3. Virus spread within the nervous system. Viruses (red shading) are disseminated by various mechanisms. (a) Some viruses such as pseudorabies virus (PRV Bartha
strain; a pig herpes virus that can infect several mammalian species) and rabies (a rhabdovirus) can be transported retrogradely by the axon and released by the cell body and
dendrites to be subsequently taken up and transported by synaptically coupled neurons. (b) Some viruses (e.g. PRV) are transported anterogradely from the cell body to the
axon terminal where the virus is released (i.e. wild-type PRV is transported along the axon in both retrograde and anterograde directions). (c) Other viruses such as CMV and
VSV are spread to a large degree by local diffusion from one cell to another, or by long distance by diffusion through the cerebrospinal fluid [1,2,25,31,39,62,64].

Although at the cellular level the mechanisms of cytolytic to HIV and to secondary opportunistic infections. The
infection might be the same in different regions of the destructive actions of HIV on the brain are complicated.
brain, completely different symptoms will be generated The current view is that neurological dysfunction is
by infections of different brain regions, with motor largely due to invasion of the brain by HIV-infected
deficits from infections of motor regions of the brain monocytes; this leads to infection of microglia or
and sensory loss from infection of either the sensory pericytes, and with the monocytes these cause secondary
receptors or central sensory processing regions of the damage to the blood–brain barrier or release cytotoxic
brain. Some viruses such as polio show a high degree of molecules that evoke secondary damage in neurons and
selectivity in which regions of the brain are infected, glia [44,45].
whereas others such as CMV can and do infect many Several other viruses, including positive-strand RNA
regions of the brain. Which region of the developing brain alpha viruses such as Sindbis [46] and the negative-
is damaged by CMV may depend on the stage of strand RNA VSV [40,47], show a preference for infecting
development during which CMV first enters the brain. and replicating in developing rodent brain cells. Some
In addition to CMV, three other viruses that are viruses selectively infect dividing neuronal precursors.
transmitted from mother to fetus are rubella (German For instance, feline panleucopenia virus (FPV) is an
measles), HIV and LCMV. Although most people are now autonomous parvovirus, related to B19 that infects
vaccinated against rubella, in the non-vaccinated, fetal humans. FPV and related rodent parvoviruses infect
infection with rubella or LCMV, particularly in the first replicating granule cells in the developing cerebellum,
trimester (a window of increased vulnerability to viral leading to cerebellar ataxia [48]. Other viruses such as
damage) can lead to microcephaly, reduced mitosis of LCMV also preferentially infect replicating cells within
developing brain cells, deafness and motor or mental the brain [26].
retardation [2]. Maternal transmission of HIV is not
uncommon in infected mothers; estimates of the rates of Childhood infection, adult manifestations
transmission vary, but 25% transmission is probable in Some viruses can infect us at a young age and cause
mothers not receiving antiviral therapy. Children born minor discomfort, and then reappear from a latent
with HIV show various neurological symptoms, due both genome that has been quiet for years or even decades.
www.sciencedirect.com
Review TRENDS in Neurosciences Vol.29 No.7 July 2006 403

(a) (b)
CMV VSV
DNA genome RNA genome
replication in nucleus replication in cytoplasm

Transcription Transcription Replication

mRNA

Translation
mRNA
Replication

Figure 4. Replication of CMV and VSV. This diagram shows the replication pathways of two unrelated viruses: a DNA virus in the herpes family (CMV) and a negative-strand
RNA virus in the rhabdovirus family (VSV). The temporal sequence in both panels is top to bottom, with the free virus attaching to the cell at the top, and the progeny virus
exiting the cell at the bottom of the figure. (a) CMV attaches to a cell, and its lipid envelope fuses with the plasma membrane, enabling the virus nucleocapsid to enter the
cytoplasm. At the nuclear envelope, the linear double-strand DNA enters the nucleus, and circularizes, and the genome replicates. The origin of the CMV envelope and how
the nucleocapsid moves from the nucleus is still being studied, but one current view is shown here. The final virus is contained in an endosome-like organelle that releases
the progeny virus after fusion of the organelle to the cell plasma membrane. CMV expresses many genes that counteract attempts by the cell to block viral replication. (b) VSV
binds to its cellular receptor, and then is brought into the cytoplasm by endocytosis. The single negative-strand RNA escapes the vacuole after acidification, and is copied into
a positive-strand RNA. This RNA serves as a template for additional negative-strand viral genomes, and initiates protein synthesis. The virus captures its envelope from the
cell plasma membrane, in which viral proteins such as the VSV G protein are already positioned. Viruses such as VSV that have RNA genomes show high mutation rates; that
for VSV is 1 in 10 000 nucleotides, or w1 mutation per viral genome [23,65]. Mutation rates of DNA viruses are lower.

The most well-known example is varicella zoster virus projecting axons are lost. Because the virus emerges
(VZV or chicken pox). Prior to immunization, 95% of the from select ganglia, the appearance of the VZV-induced
US population was infected in early childhood. VZV is in skin lesions are restricted to select dermatomes inner-
the herpes family, and during the initial 1–2-week long vated by the infected ganglia, and often occur on only
illness, often occurring in young children, a fever and the left or right half of the body.
itchy dermal lesions might be the primary symptoms. Polio is another virus that can cause secondary
Transport of herpes from the skin back to the dorsal root problems decades after the primary infection, a condition
ganglia (DRG) might occur at the time of primary know as post-polio syndrome. In this syndrome, those
infection. But 30 or 40 years later, a replication- regions of the body that showed weakness or paralysis
competent virus re-emerges from the viral genome that during the primary infection show a further decline of
has waited dormant in the nucleus of neurons in the function decades later. Unlike VZV, most reports have
DRG. The virus then travels back out the axon to the failed to localize infectious virus during the post-polio
axon terminal, where it is released to again infect other syndrome [13]. The mechanism for the late decline in
cells in the skin, leading to dermal discomfort or pain function is not clear, but it might relate to the plasticity of
(post-herpetic neuralgia) that can last for long periods surviving motor neurons that reinnervated the virally
(herpes zoster or shingles) [49]. The post-herpetic pain denervated muscles, or to the fact that normal neuron loss
might be due directly to skin infection, or in some cases in old age might take a larger toll when many motor
might relate to central reorganization of pain pathways neurons have already been killed by the polio virus.
in the spinal cord as DRG cells and their centrally- Alternately, an autoimmune cause has been suggested.
www.sciencedirect.com
404 Review TRENDS in Neurosciences Vol.29 No.7 July 2006

Not all viruses cause problems primarily during early that, contrary to Koch’s postulate that an infectious agent
development. WNV, for instance, is more likely to cause should cause the disease in all those infected, not everyone
neurological problems in the elderly [3]. The mechanism infected with a particular virus (e.g. polio) shows
for this is not clear, but it might be based on a reduced symptoms. This is where animal models are particularly
immune response to WNV. valuable, because the genetic background, developmental
stage of exposure and type of virus can be controlled.
Altered viral targets dependent on brain development In experimental animal models, viruses can initiate
Some viruses have specific targets within the CNS. For MS-like symptoms. But which virus might cause MS?
instance, polio shows a cellular preference for the motor Interestingly, several viruses can induce MS-like symp-
system. Some viruses show a developmental shift in which toms in animals, including VSV, measles, vaccinia, mouse
cells are infected. Intranasal inoculation of VSV causes hepatitis, Theiler’s, Chandipura, Semliki Forest (SFV)
widespread lethal encephalitis in neonatal mice and rats. and Venezuelan equine encephalitis (VEE) [2]. When mice
In rodents 2–3 weeks old, it can selectively infect the lacked CD8CT cells, SFV-induced demyelination was
noradrenergic neurons of the locus coeruleus and the prevented [58]; similarly, the use of immunocompromised
serotonergic neurons of the dorsal raphe, after which the mice prevented VEE-induced demyelination [59], indicat-
virus is eliminated from the CNS by the immune system; ing that the demyelination was due to the host auto-
this leads to a permanent reduction in brain serotonin and immune response to viral infection and not to viral
noradrenaline levels, and long-term changes in behavior. cytolysis. That many different viruses can precipitate
In adult rodents, VSV inoculation of the olfactory nerves MS-like symptoms in rodents suggests that it might be
can lead to infection of the GABAergic neurons of the difficult to identify which virus (or viruses) have a similar
olfactory bulb, particularly the periglomerular and gran- role in the human disease. But if MS can be caused by an
ule cells. But the excitatory projection neurons, the mitral infection, it raises the question of whether MS in humans
cells, appear resistant to infection, and in most cases the must be restricted to a single virus. If an autoimmune
virus does not proceed caudally from the olfactory bulb to response can be initiated by infection of oligodendrocytes
the rest of the brain [47,50–54]. in rodents, multiple viruses might similarly precipitate
It is not clear what crucial differences in developing and such an immune response in humans. As already noted
mature brains cause a shift in the cell preference of VSV here for other viral syndromes, a genetic predisposition is
infection. It is probably not a change in the viral receptor, important in models of virally induced MS-like symptoms.
possibly phosphatidyl serine, a molecule found on the For instance, measles virus caused Lewis rats to show
surface of most developing and mature cells [23]. More substantial lesions in the brain similar to those induced by
likely, the difference relates to the ability of cells within injection of myelin basic protein to generate experimental
the brain to resist or block VSV replication through a host allergic encephalitis, an animal model of MS; lesions
of mechanisms including activation of interferon continued even in the absence of the replicating virus,
pathways, Toll-like receptors, interleukin-12, or nitric suggesting an autoimmune response had been initiated.
oxide [47,55,56]. Although VSV poses little threat to By contrast, brown Norway rats treated similarly showed
humans [57], VSV studies are potentially important little evidence of brain lesions [60,61]. MS is more
because they demonstrate that a virus can invade the prevalent in certain geographic regions of the world, for
brain, selectively destroy specific groups of cells depending instance in higher latitudes. That this prevalence is not
on the stage of brain development, and then disappear solely based on genetic predisposition is suggested by
from the brain after only a few days, leaving no genomic indications that immigrants take on the MS risk of the
trace but causing a permanent attenuation of transmitter area into which they move (http://www.nationalmssociety.
levels and alteration in behavior. These VSV studies might org/Sourcebook-Epidemiology.asp). It is important to note
reflect parallel events in the human brain, in which a that, although viruses might cause some cases of MS,
virus can infect a subset of neurons or glia, and then be other cases of MS might be entirely independent of viral
eliminated, leaving a neurological or psychiatric disease induction. As with other neurological disease, there is no
with no detectable cause. reason that every case must have the same exact cause,
although some general similarity in induction might
One disease, many viruses be expected.
Multiple sclerosis (MS) is characterized by the loss of
axonal myelination over time, leading to impairments in Concluding remarks
neuronal signaling and function. It is generally associated Our view of viral causes of CNS dysfunction has evolved
with an autoimmune imbalance involving an immune from a simple cause-and-effect paradigm into one that
attack on the myelin-forming oligodendrocytes. Could the recognizes multiple factors that influence the final
autoimmune targeting be initiated by a virus? This is a neurological outcome. The presence of a neurotropic
complicated question, and numerous viruses have been virus in the brain might be brief, but can result in long-
isolated from MS patients, including herpes simplex, lasting effects. Depending on which region of the brain is
measles, CMV, Epstein–Barr, rhabdovirus and human infected, a single virus can generate different neurological
herpes 6 [2]. But a substantial problem is that correlation or psychiatric symptoms; or a single set of symptoms
does not mean causation. Particularly when many of these might be generated by multiple viruses that share a
viruses are commonly found in humans who show no sign common target. Together, these factors underline the
of neurological problem. A further complicating factor is complexity of determining a clear viral responsibility for a
www.sciencedirect.com
Review TRENDS in Neurosciences Vol.29 No.7 July 2006 405

brain disease that might manifest at substantial time 23 Rose, J.K. and Whitt, M.A. (2001) Rhabdoviridae: the viruses and
after the initial infection is resolved. Given the many their replication. In Field’s Virology (4th edn) (Knipe, D.M. and
Howley, P.M., eds), pp. 1221–1244, Lippincott Williams & Wilkins
variables that can have a role in whether a viral infection
24 Bode, L. and Ludwig, H. (2003) Borna disease virus infection, a human
leads to CNS involvement and dysfunction in humans, the mental-health risk. Clin. Microbiol. Rev. 16, 534–545
use of carefully controlled animal models is crucial for 25 Schneider, U. et al. (2005) Genome trimming: a unique strategy for
understanding and combating viral infections that replication control employed by Borna disease virus. Proc. Natl. Acad.
underlie CNS dysfunction. Sci. U. S. A. 102, 3441–3446
26 Bonthius, D.J. et al. (2002) Critical role for glial cells in the
propagation and spread of lymphocytic choriomeningitis virus in the
developing rat brain. J. Virol. 76, 6618–6635
Acknowledgements 27 Pearce, B.D. et al. (1999) Lymphocytic responses and the gradual
I thank Dr G. Wollmann for suggestions and artistic contribution to the hippocampal neuron loss following infection with lymphocytic
schematic figures. choriomeningitis virus (LCMV). J. Neuroimmunol. 101, 137–147
28 Tsutsui, Y. et al. (1995) Prolonged infection of mouse brain neurons
with murine cytomegalovirus after pre- and perinatal infection. Arch.
References Virol. 140, 1725–1736
1 Flint, S.J. et al. (2004) Principles of Virology, ASM Press 29 Alford, C.A. and Britt, W.J. (1996) Cytomegalovirus. In Field’s
2 Johnson, R.T. (1998) Viral Infections of the Nervous System, Virology (3rd edn) (Fields, B.N. et al., eds), pp. 2493–2534,
Lippincott-Raven Lippincott-Raven
3 Govorkova, E.A. et al. (2005) Lethality to ferrets of H5N1 influenza 30 White, D.O. and Fenner, F.J. (1994) Medical Virology, Academic Press
viruses isolated from humans and poultry in 2004. J. Virol. 79, 31 Mocarski, E. (1996) Cytomegaloviruses and their replication. In
2191–2198 Field’s Virology (3rd edn) (Fields, B.N. et al., eds), pp. 2447–2492,
4 Lee, S.H. et al. (2001) Susceptibility to mouse cytomegalovirus is Lippincott-Raven
associated with deletion of an activating natural killer cell receptor of 32 Bale, J.F. et al. (1985) Neuroradiographic abnormalities in congenital
the C-type lectin superfamily. Nat. Genet. 28, 42–45 cytomegalovirus infection. Pediatr. Neurol. 1, 42–47
5 Krmpoti1, A. et al. (2002) MCMV glycoprotein gp40 confers virus 33 Hicks, T. et al. (1993) Congenital cytomegalovirus infection and
resistance to CD8C T cells and NK cells in vivo. Nat. Immunol. 3, neonatal auditory screening. J. Pediatr. 123, 779–782
529–535 34 Perez-Jimenez, A. et al. (1998) Epilepsy and disorders of cortical
6 Thach, D.C. et al. (2000) Differences between C57BL/6 and BALB/cBy development in children with congenital cytomegalovirus infection.
mice in mortality and virus replication after intranasal infection with Rev. Neurol. 26, 42–49
neuroadapted Sindbis virus. J. Virol. 74, 6156–6161 35 Ho, M. (1982) Human cytomegalovirus infections in immunosup-
7 Glass, W.G. et al. (2006) CCR5 deficiency increases risk of sympto- pressed patients In Cytomegalovirus, Biology and Infection. Current
matic West Nile virus infection. J. Exp. Med. doi: 10.1084/jem. Topics in Infectious Disease, pp. 171–204, Plenum Medical Books
20051970 36 Britt, W.J. and Vugler, L.G. (1990) Antiviral antibody responses in
8 Wicker, J.A. et al. A single amino acid substitution in the central mothers and their newborn infants with clinical and subclinical
portion of the West Nile virus NS4B protein confers a highly congenital cytomegalovirus infections. J. Infect. Dis. 161, 214–219
attenuated phenotype in mice. Virology (in press) 37 Booss, J. et al. (1989) Host defense response to cytomegalovirus in the
9 Stojdl, D.F. et al. (2003) VSV strains with defects in their ability to central nervous system. Am. J. Pathol. 134, 71–78
shutdown innate immunity are potent systemic anti-cancer agents. 38 Reuter, J.D. et al. (2004) Systemic immune deficiency necessary for
Cancer Cell 4, 263–275 cytomegalovirus invasion of the mature brain. J. Virol. 78, 1473–1487
10 Persico, A.M. and Bourgeron, T. (2006) Searching for ways out of the 39 van den Pol, A.N. et al. (1999) Cytomegalovirus cell tropism,
autism maze: genetic, epigenetic and environmental clues. Trends replication, and gene transfer in brain. J. Neurosci. 19, 10948–10965
Neurosci. (in this issue) 40 van den Pol, A.N. et al. (2002) Relative neurotropism of a recombinant
11 Abelson, J.F. et al. (2005) Sequence variants in SLITRK1 are rhabdovirus expressing a green fluorescent envelope glycoprotein.
associated with Tourette’s syndrome. Science 310, 317–320 J. Virol. 76, 1309–1327
12 Selkoe, D.J. and Podlisny, M.B. (2002) Deciphering the genetic basis of 41 Koedood, M. et al. (1995) Human cytomegalovirus (CMV) immediate-
Alzheimer’s disease. Annu. Rev. Genomics Hum. Genet. 3, 67–99
early enhancer/promoter specificity during embryogenesis defines
13 Pallansch, M.A. and Roos, R.P. (2001) Eteroviruses: polioviruses,
target tissue of congenital HCMV infection. J. Virol. 69, 2194–2207
coxsackieviruses, echoviruses, and newer enteroviruses. In Field’s
42 van den Pol, A.N. and Ghosh, P. (1998) Selective neuronal expression
Virology (4th edn) (Knipe, D.M. and Howley, P.M., eds), pp. 723–775,
of green fluorescent protein with cytomegalovirus promoter reveals
Lippincott Williams & Wilkins
entire neuronal arbor in transgenic mice. J. Neurosci. 18,
14 Mendelsohn, C. et al. (1989) Cellular receptor for poliovirus: molecular
10640–10651
cloning, nucleotide sequence, and expression of a new member of the
43 Shinmura, Y. et al. (1999) Migration of virus infected neuronal cells in
immunoglobulin superfamily. Cell 56, 855–865
cerebral slice cultures of developing mouse brains after in vitro
15 Ohka, S. and Nomoto, A. (2001) Recent insights into poliovirus
infection with murine cytomegalovirus. Acta Neuropathol. (Berl.) 98,
pathogenesis. Trends Microbiol. 9, 501–505
16 Palacios, G. and Oberste, M.S. (2005) Enteroviruses as agents of 590–596
emerging infectious diseases. J. Neurovirol. 11, 424–433 44 Johnson, R.T. (1999) Nervous system viruses. In Encyclopedia of
17 Shekhar, K. et al. (2005) Deaths in children during an outbreak of Virology (Granoff, A. and Webster, R., eds), pp. 1013–1020, Elsevier
hand, foot and mouth disease in Peninsular Malaysia – clinical and 45 Glass, J.D. and Johnson, R.T. (1996) Human immunodeficiency virus
pathological characteristics. Med. J. Malaysia 60, 297–304 and the brain. Annu. Rev. Neurosci. 19, 1–26
18 Lentz, T.L. et al. (1982) Is the acetylcholine receptor a rabies virus 46 Vernon, P.S. and Griffin, D.E. (2005) Characterization of an in vitro
receptor? Science 215, 182–184 model of alphavirus infection of immature and mature neurons.
19 Thoulouze, M.I. et al. (1998) The neural cell adhesion molecular is a J. Virol. 79, 3438–3447
receptor for rabies. J. Virol. 72, 7181–7190 47 Reiss, C.S. et al. (1998) Viral replication in olfactory receptor neurons
20 Tuffereau, C. et al. (1998) Low-affinity nerve-growth factor receptor and entry into the olfactory bulb and brain. Ann. N. Y. Acad. Sci. 855,
(P75NTR) can serve as a receptor for rabies virus. EMBO J. 17, 751–761
7250–7259 48 Bloom, M.E. and Young, N.S. (2001) Parvoviruses. In Field’s Virology
21 Tsiang, H. (1979) Evidence for an intraaxonal transport of fixed and (4th edn) (Knipe, D.M. and Howley, P.M., eds), pp. 2361–2379,
street rabies virus. J. Neuropathol. Exp. Neurol. 38, 286–299 Lippincott Williams & Wilkins
22 Jackson, A.C. (2003) Rabies virus infection: an update. J. Neurovirol. 49 Gilden, D. (2004) Varicella zoster virus and central nervous system
9, 253–258 syndromes. Herpes 11, 89A–94A
www.sciencedirect.com
406 Review TRENDS in Neurosciences Vol.29 No.7 July 2006

50 Mohammed, A.K.H. et al. (1990) Behavioural deficits and serotonin 58 Subak-Sharpe, I. et al. (1993) In vivo depletion of CD8C T cells
depletion in adult rats after transient infant nasal viral infection. prevents lesions of demyelination in Semliki Forest virus infection.
Neuroscience 35, 355–363 J. Virol. 67, 7629–7733
51 Mohammed, A.K. et al. (1992) Persistent changes in behaviour and 59 Dal Canto, M.C. and Rabinowitz, S.G. (1981) Murine central nervous
brain serotonin during aging in rats subjected to infant nasal virus infection by a viral temperature sensitive mutant. Am. J. Pathol. 102,
infection. Neurobiol. Aging 13, 83–87 412–426
52 Lundh, B. et al. (1988) Non-lethal infection of aminergic reticular core 60 Liebert, U.G. (2001) Slow and persistent virus infections of neurons –
neurons: age-dependent spread of ts mutant vesicular stomatitis virus a compromise for neuronal survival. In The Mechanism of Neuronal
from the nose. J. Neuropathol. Exp. Neurol. 47, 497–506 Damage in Virus Infections of the Nervous System (Gosztonyi, G., ed.),
53 van den Pol, A.N. et al. (2002) Enhanced cytomegalovirus infection of pp. 35–59, Springer
the developing brain independent of the adaptive immune system. 61 Liebert, U.G. and ter Meulen, V. (1987) Virological aspects of measles
J. Virol. 76, 8842–8854 virus-induced encephalomyelitis in Lewis and BN rats. J. Gen. Virol.
54 Sabin, A.B. and Olitsky, P.K. (1937) Influence of host factors on 68, 1715–1722
neuroinvasiveness of vesicular stomatitis virus. J. Exp. Med. 66, 62 Whitley, R.J. (2001) Herpes simplex viruses. In Field’s Virology (4th
15–34 edn) (Knipe, D.M. and Howley, P.M., eds), pp. 2461–2509, Lippincott
55 Trottier, M.D. et al. (2005) VSV replication in neurons is Williams & Wilkins
inhibited by type I IFN at multiple stages of infection. Virology 63 Mori, I. et al. (2005) Olfactory transmission of neurotropic viruses.
333, 215–225 J. Neurovirol. 11, 129–137
56 Griffin, D.E. and Hardwick, J.M. (1999) Perspective: virus infections 64 Enquist, L.W. and Card, J.P. (2003) Recent advances in the use of
and the death of neurons. Trends Microbiol. 7, 155–160 neurotropic viruses for circuit analysis. Curr. Opin. Neurobiol. 13,
57 Tesh, R.B. et al. (1969) Ecologic studies of vesicular stomatitis virus. I. 603–606
Prevalence of infection among animals and humans living in an area 65 Drake, J.W. and Holland, J.J. (1999) Mutation rates among RNA
of endemic VSV activity. Am. J. Epidemiol. 90, 255–261 viruses. Proc. Natl. Acad. Sci. U. S. A. 96, 13910–13913

5th INMED/TINS Conference


September 9–12 2006, La Ciotat, France

Physiogenic and pathogenic oscillations: beauty and the beast

Neuronal networks generate a variety of oscillations and patterns that are involved in sensory integrative processes as well as
memory processes. Thus, sensory binding is thought to involve the generation of oscillations in different parts of the brain that are
related to various sensory modalities and somehow help in reconstructing the image. Oscillations are also involved in a wide range
of pathogenic patterns and provide a signature of the neurological disorder. For example, in Parkinson’s disease, abnormal
patterns are likely to be involved in akinetic disorders, and epilepsies are associated with high-frequency oscillations that also
transform a naive structure to one that generate seizures, on the principle that ’seizures beget seizure’.

Using a wide range of recordings and preparations, imaging techniques and mathematical models, we are beginning to
understand how the brain generates these patterns and which neuronal population is dominant in the generation of a given
pattern. What is now needed is to confront the various results and concepts and to generate an organised model of the physiogenic
and pathogenic patterns: what are the similarities and differences in terms of mechanisms, of generators, etc? We shall invite to the
meeting experts in the clinical, physiological, imaging and theoretical aspects of this question. We expect attendees to include
students and senior researchers in neurology and neurobiology, as well as in mathematics and modelling.

Past INMED/TINS conferences


4th INMED/TINS Conference: Nature and nurture in brain development and neurological disorders
3rd INMED/TINS Conference: Multiple facets of GABAergic synapses
2nd INMED/TINS Conference: Nature and nurture in brain development
1st INMED/TINS Conference: Nature and nurture in neuroscience

More information: http://inmednet.com/2006-conference.html

www.sciencedirect.com

Potrebbero piacerti anche