Sunteți pe pagina 1din 11

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/26244663

Human Hendra virus infection causes acute and


relapsing encephalitis

Article in Neuropathology and Applied Neurobiology · July 2009


DOI: 10.1111/j.1365-2990.2008.00991.x · Source: PubMed

CITATIONS READS

47 41

8 authors, including:

Kum Thong Wong Beng Beng Ong


University of Malaya Queensland Government
164 PUBLICATIONS 3,470 CITATIONS 22 PUBLICATIONS 393 CITATIONS

SEE PROFILE SEE PROFILE

Koon-Chu Yaiw Lin-Fa Wang


Apodemus AB The Commonwealth Scientific and Industrial …
29 PUBLICATIONS 318 CITATIONS 388 PUBLICATIONS 9,152 CITATIONS

SEE PROFILE SEE PROFILE

All content following this page was uploaded by Koon-Chu Yaiw on 06 July 2016.

The user has requested enhancement of the downloaded file. All in-text references underlined in blue are added to the original document
and are linked to publications on ResearchGate, letting you access and read them immediately.
Neuropathology and Applied Neurobiology (2009), 35, 296–305 doi: 10.1111/j.1365-2990.2008.00991.x

Human Hendra virus infection causes acute and


relapsing encephalitis
K. T. Wong*, T. Robertson†, B. B. Ong‡, J. W. Chong*, K. C. Yaiw*, L. F. Wang§, A. J. Ansford‡ and
A. Tannenberg‡
*Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia, †Royal Brisbane &
Women’s Hospital, ‡Queensland Health Pathology & Scientific Services, Brisbane, Queensland, and §CSIRO Livestock
Industries, Australian Animal Health Laboratory and Australian Biosecurity Cooperative Research Center, Geelong,
Victoria, Australia

K. T. Wong, T. Robertson, B. B. Ong, J. W. Chong, K. C. Yaiw, L. F. Wang, A. J. Ansford and A. Tannenberg (2009)
Neuropathology and Applied Neurobiology 35, 296–305
Human Hendra virus infection causes acute and relapsing encephalitis

Aim: To study the pathology of two cases of human Cytoplasmic viral inclusions and antigens and viral RNA
Hendra virus infection, one with no clinical encephalitis in neurones and ependyma suggested viral replication. In
and one with relapsing encephalitis. Methods: Autopsy the case of relapsing encephalitis, there was severe wide-
tissues were investigated by light microscopy, immunohis- spread meningoencephalitis characterized by neuronal
tochemistry and in situ hybridization. Results: In the loss, macrophages and other inflammatory cells, reactive
patient with acute pulmonary syndrome but not clinical blood vessels and perivascular cuffing. Antigens and
acute encephalitis, vasculitis was found in the brain, lung, viral RNA were mainly found in neurones. Vasculitis was
heart and kidney. Occasionally, viral antigens were dem- absent in all the tissues examined. Conclusions: The case
onstrated in vascular walls but multinucleated endothelial of acute Hendra virus infection demonstrated evidence
syncytia were absent. In the lung, there was severe inflam- of systemic infection and acute encephalitis. The case of
mation, necrosis and viral antigens in type II pneumocytes relapsing Hendra virus encephalitis showed no signs of
and macrophages. The rare kidney glomerulus showed extraneural infection but in the brain, extensive inflam-
inflammation and viral antigens in capillary walls and mation and infected neurones were observed. Hendra
podocytes. Discrete necrotic/vacuolar plaques in the brain virus can cause acute and relapsing encephalitis and the
parenchyma were associated with antigens and viral findings suggest that the pathology and pathogenesis are
RNA. Brain inflammation was mild although CD68+ similar to Nipah virus infection.
microglia/macrophages were significantly increased.

Keywords: encephalitis, Hendra virus, Henipavirus, human, infection, pathogenesis, pathology

Because it shares a high degree of similarity in genome


Introduction
organization and sequence and other biological character-
Hendra virus (HeV), an emerging paramyxovirus from the istics to another recently discovered virus, the Nipah virus
family Paramyxoviridae was first discovered in Australia in (NiV), both viruses have been placed in the newly created
1994 following an outbreak in horses and humans [1]. genus Henipavirus [2,3]. There is good evidence that the
natural hosts of HeV and NiV are pteropid fruit bats [4–6],
Correspondence: Kum Thong Wong, Department of Pathology, whose range includes northern Australia, southeast
Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur,
Malaysia. Tel: +603 7967 5762; Fax: +603 7955 6845; E-mail: Asia, the Indian subcontinent and eastern Africa. Hence,
wongkt@um.edu.my it was not surprising that after the first NiV outbreaks in
296 © 2009 Blackwell Publishing Ltd
Table 1. Clinicopathological findings in four cases of human Hendra virus infection

Positive IHC† and/or ISH staining in Final


Probable autopsy tissues pathological‡
Case incubation or clinical
# Clinical presentation* period Light microscopy of autopsy tissues CNS tissues Non-CNS tissues diagnosis

1 49-year-old man, presented with influenza-like 6 days Blood vessels: Brain Lung Acute Hendra
illness, followed by respiratory and renal failure Vasculitis/endotheliitis in brain, lung, (neurones, ependyma, (type II pneumocytes, infection with
and cardiac irritability. Died after about 12 days heart and kidney blood vessels) blood vessels) encephalitis§
of admission Brain: Kidney
Small necrotic/vacuolar plaques in the (glomeruli, capillaries,
cerebrum and cerebellum, in addition to tubules)
mild meningitis and mild parenchymal
inflammation
Lung:
Severe parenchymal inflammation and
necrosis. Incidental fungal infection in the
lungs was also noted
Other organs:
Generally mild or focal inflammation in
the parenchyma of other organs

2 35-year-old man who first presented with 12 days Brain: Brain All tissues negative Relapsing
headache, drowsiness and meningitis but after 13 (13 months) Mainly confluent geographic areas of (neurones mainly; Hendra
months was readmitted with fever, seizures, neuronal loss, gliosis and parenchymal possibly also glial and encephalitis
neurological deficits and coma. Died after 25 days and perivascular inflammation were inflammatory cells)
of admission observed mainly in the cerebral cortex

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
Other non-CNS organs:
No vasculitis, necrosis or significant
inflammation in the lung, heart, kidney,
liver and spleen

3 40-year-old man presented with influenza-like 5 days NA¶ NA NA Acute Hendra


illness, myalgia, headache and vertigo of 5-day infection
duration. Recovered apparently well after 6 weeks
4 Adult female (age unknown) presented with dry 7 days NA NA NA Acute Hendra
cough and sore throat, cervical infection
lymphadenopathy, fever, body aches and
tiredness. Recovered and apparently well

*Information (age, sex, clinical presentation, incubation period) compiled from references [12–14].
†IHC, immunohistochemistry; ISH, in situ hybridization; CNS, central nervous system. ISH was not performed on non-CNS tissues of Case #1 due to insufficient tissues.
Human Hendra virus infection

‡Pathological diagnosis available for Cases # 1 and 2 only.


§Acute encephalitis based on our pathological evidence only; clinical encephalitis was not reported.
¶NA = tissues not available for study.
297
298 K. T. Wong et al.

Malaysia and Singapore [7–9], sporadic NiV outbreaks characteristics that are distinguishable from acute NiV
continued to be seen in these regions, notably in Bang- encephalitis [16,18]. Pathological findings suggested a
ladesh and India [10,11]. Although so far HeV infection true recurrence of infection rather than postinfectious
has only been reported from Australia, like NiV, there is encephalomyelitis as viral antigens/RNA in neurones
every possibility that future outbreaks may occur in other could be demonstrated. However, vasculitis and multi-
countries as well, given the wide range of fruit bats. nucleated endothelial cells or syncytia were not demon-
HeV-infected horses acted as intermediate hosts for strated [15,16].
human infections [1,6]. Of the four human cases that Of the two HeV human fatalities included in this study
have been reported to date (Table 1), two were fatal and all (Table 1), Case # 1 presented clinically as an acute HeV
had close contact with infected horses before developing infection characterized by a predominant pulmonary syn-
the disease [12–14]. Previous publications on human drome with no apparent clinical encephalitis [12,14].
HeV infection have concentrated mainly on epidemiologi- However, studies of acute HeV infection in horses, guinea
cal, virological and clinical aspects with no detailed data pigs and other animals [1,19,20] suggest that there is some
on pathological features [1,3,12,14]. Moreover, the target similarity to acute NiV infection in that vasculitis and
cells of HeV in natural human infections have not been encephalitis can occur [15]. Hence, we hypothesize that
fully investigated. there may be histological evidence of CNS involvement in
Studies of acute human NiV infection have demonst- Case # 1. In the case of relapsing HeV encephalitis (Table 1,
rated that blood vessels and a broad range of parenchymal Case # 2), it was previously reported that in addition to
cells were susceptible to infection [15]. The most severely parenchymal inflammation and viral antigens, occasional
involved tissues were in the central nervous system (CNS), multinucleated endothelial cells were observed in blood
but the lung, kidney, lymphoid and other organs were also vessels in the brain and other organs [14]. Our objective is
susceptible. Two distinct clinicopathological forms of to undertake a more detailed study of the neuropathology
encephalitis were recognized, viz. acute and relapsed/late- of human HeV infection by studying the histopathological
onset NiV encephalitis [15,16]. Acute NiV encephalitis features, in particular CNS pathology and by investigating
appears as part of the acute infection that occurs 1–2 viral localization with immunohistochemistry (IHC) and
weeks after viral exposure [17]. Pathologically, it is charac- in situ hybridization (ISH) in infected tissues.
terized by necrotic plaques believed to be associated with
vasculitis-induced thrombosis and neuronal infection.
Materials and methods
This dual pathogenetic mechanism of tissue damage con-
sisting of microinfarction and direct neuronal infection Formalin-fixed, paraffin-embedded autopsy tissues of
appears to be unique among viral encephalitides [15]. various major organs were sectioned at 5–7 microns and
On the other hand, relapsed/late-onset NiV encephalitis routinely stained with haematoxylin and eosin. These
usually presents several weeks after the acute infection organs/tissues included brain, lung, heart, kidney, liver
has subsided, with new episodes of neurological manifes- and spleen (Table 1: Cases # 1 and 2); pancreas, thyroid,
tations. Relapsed encephalitis occurs in survivors who adrenal, lymph node and intestine (Case # 1 only) and
initially present with acute encephalitis and late- spinal cord (Case # 2 only). IHC to detect HeV antigens was
onset encephalitis is found in those with acute, mild, performed on further sections. A standard immunoperoxi-
nonencephalitic infection earlier. Relapsed/late-onset dase protocol was followed [21], using a primary mouse
encephalitis generally have similar clinicopathological anti-Hendra polyclonal antibody (1:1000 dilution; gift

Figure 1. Acute Hendra virus infection: vasculitis and endotheliitis involving blood vessels in the meninges (A, arrows), brain parenchyma
(D, arrows; E), lung (B, arrow) and heart (C, arrow), with intramural or subendothelial inflammatory cells. Viral antigens demonstrated in
endothelial cells lining a meningeal capillary (F, arrow; inset). Inflammation in the lung (G) characterized by intra-alveolar inflammation,
necrosis, macrophages and the rare multinucleated giant cell (inset, arrow). Proliferating type II pneumocytes and alveolar macrophages
were positive for viral antigens (H). Kidney glomerulus with surrounding inflammation and thrombotic plug (I, arrow) and viral antigens
in capillary walls, podocytes and other cells (J). A–E, G, I: haematoxylin & eosin stain. F, H, J: immunohistochemistry/DAB/Mayer’s
haematoxylin. Magnification: A–F, H–J, G (inset): ¥40 obj; G: ¥10 obj. Bar: A–F, I, J = 50 microns; G = 100 microns; G (inset), H = 25
microns.

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
Human Hendra virus infection 299

A B C

D F G

H I J

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
300 K. T. Wong et al.

A B

C D

E F G H

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
Human Hendra virus infection 301

Figure 2. Acute Hendra virus infection in the central nervous system. (A) Subtle vacuolar plaque (thin arrows) in the cerebellar molecular
layer consisting of fine neuropil vacuolation with adjacent eosinophilic Purkinje cells (thick arrow). (B) Necrotic/vacuolar plaque in the
dentate nucleus, showing neuronal loss, coarse neuropil vacuolation and mild inflammation. Some surviving/degenerate neurones were still
present (arrows). Fine granular viral antigens in the neuropil (C) within a vacuolar plaque in the cerebellar molecular layer. (D) Viral
antigens in the cytoplasm (thin arrows) and processes (thick arrow) of neurones found within or adjacent to a necrotic/vacuolar plaque in
the dentate nucleus. Eosinophilic viral inclusions (E) and viral antigens (F, thick arrow) and viral RNA (G) in the cytoplasm of neurones in
the hippocampus pyramidal layer. Viral protein was also detected in the nucleus (F, thin arrow). (H) Plaque-like cluster of neurones and
adjacent ependymal cells (arrow) in the periaqueductal grey matter of the midbrain staining positive for viral antigens. A, B, E:
haematoxylin & eosin stain; C, D, F, H: immunohistochemistry/DAB/Mayer’s haematoxylin; G: in situ hybridization/NBT/BCIP/Mayer’s
haematoxylin. Magnification: A, B, H: ¥10 obj; C–G: ¥40 obj. Bar: A–D = 50 microns; E–G = 25 microns, H = 100 microns.

from Dr SR Zaki, CDC, Atlanta, USA), with slight modifica- blood vessels in the brain, lung, kidney and heart
tions in that our assay utilized the ENVISION detection (Figure 1A–E). In contrast to perivascular cuffing, vascu-
method (DakoCytomation, Copenhagen, Denmark). The litis was characterized by karyorrhexis and intramural or
slides were counter-stained with haematoxylin, dehy- subendothelial inflammatory cells. However, no convinc-
drated through serial ethanols and xylene and cover- ing endothelial syncytia or multinucleated giant cells were
slipped before microscopic examination. IHC to CD68 to found. Occasional thrombotic plugs were detected in the
detect microglia/macrophages were performed as previ- pulmonary blood vessel and rarely in glomerular capillar-
ously described [22]. ies (Figure 1I). Viral antigens were observed in some
The DNA probes were prepared from a plasmid contain- vascular endothelium and possibly smooth muscle cells,
ing the whole N gene of HeV, using a standard polymerase in the brain (Figure 1F), lung and kidney (Figure 1J).
chain reaction (PCR) that incorporated digoxigenin-11- Choroid plexus showed no evidence of vasculitis or virus.
dUTP to produce a digoxigenin-labelled DNA probe of In the lung, there was severe parenchymal inflamma-
271 bp, as previously described [22]. The forward and tion, necrosis, intra-alveolar macrophages/inflammatory
reverse primers for the PCR were, respectively, 5′-gca-atg- cells, prominent type II pneumocyte proliferation and
gct-gac-aga-gat-ga-3′ (N gene nucleotide position: occasional alveolar membranes (Figure 1G). The kidney
745–764, Genbank accession number: AF017149) and showed the rare focal glomerulitis (Figure 1I) and focal
5′-gct-cga-ggc-cct-att-tct-ct-3′ (nucleotide position: inflammation around necrotic tubules. There was evi-
1035–1016). The PCR conditions were 95°C for 3 min; dence of emperipolesis and the occasional bizarre multi-
followed by 35 cycles of 94°C for 30 s, 56°C for 1 min, nucleated giant cell in the lymph nodes. Viral antigens
72°C for 30 s; and 72°C for 10 min, final extension. The were demonstrated in alveolar type II pneumocytes, intra-
ISH protocol used was as described previously [23] and alveolar macrophages and the occasional glomeruli and
performed on all tissues except for the non-CNS tissues of tubules (Figure 1H,J).
Case # 1 (Table 1), as these tissues were insufficient for In the brain parenchyma, there were both subtle and
this purpose. more obvious discrete necrotic or vacuolar plaques, in the
For IHC and ISH assays, HeV-infected hamster tissues cerebellum (Figure 2A,B), the gray and white matter of
(courtesy of Dr Fabian Wild, France) served as positive the cerebral cortex (Figure 3A), corpus callosum, hippoc-
controls. Negative control tissues included normal CNS ampus, thalamus, external capsule and globus pallidus.
tissues and CNS tissues known to be infected by Enterovi- The most obvious necrotic plaques were found in the
rus 71 [22] and measles virus [24]. For both assays, dupli- white matter consisting of eosinophilic axonal spheroid-
cate procedures that omitted the primary antibody and like material (Figure 3A). The vacuolar plaques comprised
probe, respectively, were performed as additional negative neuropil vacuolation, mild parenchymal infiltration by
controls. mononuclear inflammatory cells and mild perivascular
cuffing. In addition, neuronal loss may be evident in
neuronal areas (Figure 2B). In the cerebellar molecular
Results
layer, vacuolar plaques consisted of small fine vacuoles
(Figure 2A). Occasional neuronal bodies at the peri-
Acute HeV infection (Table 1, Case # 1)
phery of a plaque may show cytoplasmic eosinophilia
The light microscopic features are summarized in the (Figure 2A). Eosinophilic cytoplasmic viral inclusions
Table 1. Vasculitis and endotheliitis were observed in were detected in neurones in the hippocampus (pyramidal
© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
302 K. T. Wong et al.

A E

C D F

G H I J

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
Human Hendra virus infection 303

Figure 3. Acute and relapsing Hendra encephalitis. Acute Hendra encephalitis (A–D): necrotic/vacuolar plaque in cerebral white matter
consisting of eosinophilic, axonal spheroid-like material (A, inset). Increased number of CD68+ microglia/macrophages in necrotic/vacuolar
plaques in the cerebellar dentate nucleus (B) and cerebellar molecular layer (C) and around neurones, neuronophagia (D). Severe
meningoencephalitis in relapsing Hendra encephalitis (E–J): large confluent areas of inflammation in the cerebral cortex (E, arrows)
consisting of perivascular cuffing, reactive blood vessels (F), parenchymal inflammation, necrosis and neuronal loss. Inflammatory cells
comprised mainly of macrophages, lymphocytes and some plasma cells (G). Focal areas of viral antigens were found either within or at the
edge of inflammatory lesions (H, arrows), mainly in neurones and possibly glial and inflammatory cells (I). Viral RNA was demonstrated
mainly in neurone-like cells (J, arrows). A, E–G: haematoxylin & eosin; B–D, H, I: immunohistochemistry/DAB/Mayer’s haematoxylin; J: in
situ hybridization/NBT/BCIP/Mayer’s haematoxylin. Magnification: A, D, G, I, J: ¥40 obj; B, C, F: ¥10 obj; E, H: ¥4 obj. Bar: A, D = 50
microns; A (inset), B, C, E, F, H = 100 microns, G, I, J = 25 microns.

layer) (Figure 2E) and thalamus and ependymal cells. Within inflamed areas, focal viral antigens/RNA were
There were no nuclear inclusions detected. Overall, by demonstrated mainly in surviving neurones (Figure 3H–J)
light microscopy, parenchymal and meningeal inflamma- and possibly in some glial/inflammatory cells as well.
tion and perivascular cuffing were mild and prominent Overall, inflammation was much more extensive than
microglial nodules were not observed. However, CD68 viral antigens/RNA. Severe meningitis was found over
IHC showed an increased number of microglial cells/ many areas of the cerebral cortex (Figure 3E). All the
macrophages in and around necrotic/vacuolar plaques non-CNS organs were uninflamed and in particular, no
(Figure 3B,C) and surrounding some neurones, neu- vasculitis, multinucleated endothelial syncytia or viral
ronophagia (Figure 3D). antigens/RNA were detected.
Viral antigens and/or RNA were detected in the neu-
ropil and in single or plaque-like groups of neurones
Discussion
(both soma and processes) in the cerebellum (dentate
nucleus, molecular and granular layers), cerebral cortex, Pathological findings in the two cases confirmed that HeV
midbrain (periaqueductal gray area, substantia nigra), was neuronotropic and could cause CNS infection giving
hippocampus (pyramidal layer, dentate gyrus, entorhinal rise to acute and relapsing encephalitis, respectively. In the
cortex), thalamus and ependymal and subependymal patient with acute HeV infection without apparent clinical
cells (Figure 2C,D,F–H). Positive neurones were often, encephalitis (Case # 1), mild vasculitis, meningitis, paren-
but not always, found in or near necrotic/vacuolar chymal inflammation, necrotic/vacuolar plaques and
plaques (Figure 2C,D). Conversely, not all plaque-like neuronal infection were found in the CNS, features that
neuronal virus antigen positivity was associated with have not been previously reported in acute infection.
necrosis or prominent vacuolation (Figure 2H). Necrotic It was postulated that the characteristic and well-defined
plaques in the white matter were not associated with necrotic plaque described in acute NiV encephalitis is the
viral antigens/RNA. result of a combination of neuronal infection and micro-
infarction following vasculitis-induced thrombosis [15].
This was because in the vicinity of necrotic plaques, vas-
Relapsing HeV infection (Table 1, Case # 2)
culitis, thrombus-occluded vessels and/or infected neu-
Only the CNS tissues and mainly the cerebral cortex rones were often observed together [15]. In our case
showed evidence of inflammation although very focal of acute HeV encephalitis, the necrotic/vacuolar plaques
areas in the pons, cerebellum and spinal cord were also may be equivalent to the necrotic plaque in acute NiV
inflamed. Inflammatory lesions were usually extensive encephalitis. There was evidence that the necrotic/
and consisted of intense infiltration of macrophages, lym- vacuolar plaques were associated with neuronal infection
phocytes and some plasma cells, with prominent perivas- by HeV. Vasculitis and viral antigens could be demonstrated
cular cuffing (Figure 3E–G). There was severe neuronal in cerebral vessels, although admittedly, these abnormal
loss, glial proliferation and an increased number of vessels are not necessarily found near the plaques.
reactive blood vessels (Figure 3F). More discrete, plaque- Thrombus-occluded blood vessels were also absent.
like, but otherwise similar inflammatory lesions, were Vasculitis was also noted in the lung, heart and kidney
occasionally observed. There was no evidence of vasculi- and vascular thrombi were identified in the lung and glom-
tis, endothelial syncytia or thrombosis and viral inclusions eruli. Hence, we think it is possible that vasculitis-induced
were not prominent. thrombosis and microinfarction could have occurred in the
© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
304 K. T. Wong et al.

CNS at some stage of the acute infection. Multinucleated tion. After 13 months, vasculitis present initially, subsided
endothelial syncytia, a unique feature of endothelial infec- and healed, but the virus infection recurred to spread
tion, is observed in acute NiV infection but this was not throughout the CNS. This phenomenon has also been
observed in this HeV case. The reasons for this disparity suggested for relapsed/late-onset NiV encephalitis [16].
could include the rarity of multinucleated endothelial syn- Further investigations are needed to determine if the
cytia (only found in 27% of acute NiV encephalitis) [15], a immune response and other host factors, or viral muta-
relatively mild CNS involvement, disintegration of throm- tions were responsible for this interesting phenomenon.
botic plugs over time or inadequate sampling. Within or In the case of measles, another important human
near the discrete white matter necrotic plaques, there was paramyxovirus, relapsing encephalitis has not been
no evidence of infection of glial or other cells whatsoever, reported. However, late-onset NiV encephalitis might
suggesting that microinfarction may be solely involved in arguably be represented by subacute sclerosing panen-
their pathogenesis. If this is correct, focal infarction and cephalitis and measles inclusion body encephalitis as both
death of oligodendroglial cells surrounding axons may be present months to years after the acute infection. As
responsible for this phenomenon. relapsed NiV encephalitis could occur even after 4 years or
As vasculitis was so widespread, we postulate that more [25], it is theoretically possible that survivors of
viraemia must have occurred, spreading the infection to acute HeV infection could still develop encephalitis long
multiple organs; hence, acute HeV infection should be after 13 months. Fortunately, relapsed/late-onset NiV
regarded as a systemic infection. Neuronal and other encephalitis is not uniformly fatal, suggesting that HeV
parenchymal cell infection was probably facilitated by vas- encephalitis may not be invariably fatal as well [16].
culitic damage to the blood brain barrier and other vessels. Nonetheless, long-term follow-up of Cases # 3 and 4
Like meningeal vessel vasculitis, involvement of the (Table 1) is essential and could be invaluable to determine
ependyma could enable virus to enter cerebrospinal fluid future disease course. Remarkably, in asymptomatic or
and contribute to further viral spread in the CNS via this mildly symptomatic NiV-infected patients, despite absence
route. It is difficult to determine if clinical encephalitis was of neurological manifestations, brain scans may show a
missed as a result of the patient’s severe clinical condition few discrete lesions similar to acute NiV encephalitis [26].
at admission, or whether clinical encephalitis was not
severe enough to be detected.
Acknowledgement
Apart from blood vessels, the target cells/tissues in
acute HeV infection seemed to be similar to NiV infection This work was partly supported by Malaysian Government
[15]. Like NiV, HeV was neuronotropic and to a lesser IRPA Grant (06-02-03-0199-PR0060-/04). We are
extent, ependyma were susceptible to infection. Viral grateful to Dr Sherif R Zaki, Centers for Disease Control
inclusions, and antigens/RNA in neurones and ependyma and Prevention, Atlanta, USA for the generous gift of
in particular, suggest that active viral replication occurs in anti-Hendra antibodies and to Dr F. Wild, INSERM, Lyon,
these cells. Likewise, type II pneumocytes and alveolar France for the infected hamster tissues.
macrophages may be able to support viral replication.
Overall, like NiV, involvement of glial cells appears to be
References
rare in HeV infection.
The second fatal case of HeV infection with a relapse of 1 Murray K, Selleck P, Hooper P, Hyatt A, Gould A, Gleeson
neurological manifestations 13 months after acute disease L, Westbury H, Hiley L, Selvey L, Rodwell B, Ketterer P. A
morbillivirus that caused fatal disease in horses and
seems to be similar to relapsed NiV encephalitis [16]. The
humans. Science 1995; 268: 94–7
pathological findings confirmed that the patient had 2 Wang LF, Harcourt BH, Yu M, Tamin A, Rota PA, Bellini
recurrent infection rather than postinfectious encephalo- WJ, Eaton BT. Molecular biology of Hendra and Nipah
myelitis. At variance to a previous report [14], we found viruses. Microbes Infect 2001; 3: 279–87
no evidence of multinucleated endothelial syncytia or vas- 3 Wang LF, Yu M, Hansson E, Pritchard LI, Shiell B, Michal-
ski W, Eaton BT. The exceptionally large genome of
culitis in any of the organs examined, nor any evidence of
Hendra virus: support for creation of a new genus within
viral antigens/RNA in the non-CNS organs. We postulate the family Paramyxoviridae. J Virol 2000; 74: 9972–9
that recurrence originated from viral foci that were dis- 4 Chua KB, Koh CL, Hooi PS, Wee KF, Khong JH, Chua BH,
seminated to the CNS by viraemia during the acute infec- Chan YP, Lim ME, Lam SK. Isolation of Nipah virus from

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305
Human Hendra virus infection 305

Malaysian island flying-foxes. Microbes Infect 2002; 4: 17 Goh KJ, Tan CT, Chew NK, Tan PSK, Kamarulzaman A,
145–51 Sarji SA, Wong KT, Abdullah BJ, Chua KB, Lam SK
5 Reynes JM, Counor D, Ong S, Faure C, Seng V, Molia S, . Clinical features of Nipah virus encephalitis among pig
Walston J, Georges-Courbot MC, Deubel V, Sarthou JL. farmers in Malaysia. N Engl J Med 2000; 342: 1229–35
Nipah virus in Lyle’s flying foxes, Cambodia. Emerg Infect 18 Sarji SA, Abdullah BJJ, Goh KJ, Tan CT, Wong KT. Mag-
Dis 2005; 11: 1042–6 netic resonance imaging features of Nipah encephalitis.
6 Halpin K, Young PL, Field HE, Mackenzie JS. Isolation of AJR Am J Roentgenol 2000; 175: 437–42
Hendra virus from pteropid bats: a natural reservoir of 19 Williamson MM, Hooper PT, Selleck PW, Westbury HA,
Hendra virus. J Gen Virol 2000; 81: 1927–32 Slocombe RFS. A guinea-pig model of Hendra virus
7 Chua KB, Bellini WJ, Rota PA, Harcourt BH, Tamin A, encephalitis. J Comp Path 2001; 124: 273–9
Lam SK, Ksiazek TG, Rollin PE, Zaki SR, Shieh WJ, 20 Hooper P, Zaki S, Daniels P, Middleton D. Comparative
Goldsmith CS,Gubler DJ, Roehrig JT, Eaton B, Gould AR, pathology of the diseases caused by Hendra and Nipah
Olson J, Field H, Daniels P, Ling AE, Peters CJ, Anderson viruses. Microbes Infect 2001; 3: 315–22
LJ, Mahy BWJ. Nipah virus: a recently emergent deadly 21 Wong KT, Grosjean I, Brisson C, Blanquier B, Fevre-
paramyxovirus. Science 2000; 288: 1432–5 Montange M, Bernard A, Loth P, Georges-Courbot M,
8 Paton NI, Leo YS, Zaki SR, Auchus AP, Lee KE, Ling AE, Chevallier M, Marianneau P, Akaoka H, Lam SK, Wild F,
Chew SK, Ang B, Rollin PE, Umapathi T, Sng I, Lee CC, Deubel V. A golden hamster model for human acute
Lim E, Ksiazek TG. Outbreak of Nipah-virus infection Nipah virus infection. Am J Pathol 2003; 163: 2127–37
among abattoir workers in Singapore. Lancet 1999; 354: 22 Wong KT, Badmanthan M, Ong KC, Kojima H, Nagata
1253–6 N, Chua KB, Ong BB, Nagashima K. The distribution
9 Chua KB, Goh KJ, Wong KT, Kamarulzaman A, Tan PSK, of inflammation and virus in human Enterovirus 71
Ksiazek TG, Zaki SR, Paul G, Lam SK, Tan CT. Fatal encephalomyelitis suggest possible viral spread by neural
encephalitis due to Nipah virus among pig-farmers in pathways. J Neuropathol Exp Neurol 2008; 67: 162–9
Malaysia. Lancet 1999; 354: 1257–9 23 Jessie K, Fong MY, Devi S, Lam SK, Wong KT. Localization
10 Hsu VP, Hossain MJ, Parashar UD, Ali MM, Ksiazek TG, of Dengue virus in naturally infected human tissues by
Kuzmin I, Niezgoda M, Rupprecht C, Bresee J, Breiman RF. immunohistochemistry and in situ hybridization. J Infect
Nipah virus encephalitis emergence, Bangladesh. Emerg Dis 2004; 189: 1411–18
Infect Dis 2004; 10: 2082–7 24 Chong HT, Ramli N, Wong KT, Looi LM, Iwasaki T,
11 Harit AK, Ichhpujani RL, Gupta S, Gill KS, Lal S, Ganguly Nagata N, Ramasundram V, Tan CT. Subacute measles
NK, Agarwal SP. Nipah/Hendra virus outbreak in encephalitis: a case of long term survival with follow-up
Siliguri, West Bengal, India in 2001. Indian J Med Res MR brain scans. Neurology Asia 2007; 12: 121–5
2006; 123: 553–60 25 Chong HT, Tan CT. Relapsed and late-onset Nipah
12 Selvey LA, Wells RM, McCormack JG, Ansford AJ, Murray encephalitis, a report of three cases. Neurol J Southeast
K, Rogers RJ, Lavercombe PS, Selleck P, Sheridan JW. Asia 2003;8:109–12
Infection of humans and horses by a newly described 26 Tan KS, Sarji SA, Tan CT, Abdullah BJ, Chong HT, Thaya-
morbillivirus. Med J Aust 1995; 162: 642–5 paran T, Koh CN. Patients with asymptomatic Nipah
13 Hanna JN, McBride WJ, Brookes DL, Shield J, Taylor CT, virus infection may have abnormal cerebral MR imaging.
Smith IL, Scott BC, Smith GA. Hendra virus infection in a Neurol J Southeast Asia 2000; 5: 69–73
veterinarian. Med J Aust 2006; 185: 562–4
14 O’Sullivan JD, Allworth AM, Paterson DL, Snow TM, Received 29 January 2008
Boots R, Gleeson LJ, Gould AR, Hyatt AD, Bradfield J. Fatal Accepted after revision 30 September 2008
encephalitis due to novel paramyxovirus transmitted Published online Article Accepted on 2 October 2008
from horses. Lancet 1997; 349: 93–5
15 Wong KT, Shieh WJ, Kumar S, Norain K, Abdullah W, Addendum
Guarner J, Goldsmith CS, Chua KB, Lam SK, Tan CT, Goh
KJ, Chong HT, Jusoh R, Rollin PE, Ksiazek TG, Zaki SR. In July 2008, a further two human cases were reported;
Nipah virus infection: pathology and pathogenesis of an one subsequently succumbed to the infection [ProMED-
emerging paramyxoviral zoonosis. Am J Pathol 2002; mail. Hendra virus, human, equine, Australia (07):
161: 2153–67
(Queensland). ProMED-mail 2008; 21 Aug: 20080821.
16 Tan CT, Goh KJ, Wong KT, Sarji SA, Chua KB, Chew NK,
Murugasu P, Loh YL, Chong HT, Tan KS, Thayaparan T, 2606. http://www.promedmail.org. Accessed 21 August
Kumar S, Jusoh MR. Relapsed and late-onset Nipah 2008]. Both cases had contact with infected horses treated
encephalitis. Ann Neurol 2002; 51: 703–8 in a veterinary facility in Brisbane, Australia.

© 2009 Blackwell Publishing Ltd, Neuropathology and Applied Neurobiology, 35, 296–305

S-ar putea să vă placă și