You are on page 1of 12

Nondomestic, Exotic, Wild and Zoo Animals–Original Article

Veterinary Pathology
2019, Vol. 56(1) 106-117
Distribution of Viral Antigen and ª The Author(s) 2018
Article reuse guidelines:
sagepub.com/journals-permissions
Inflammatory Lesions in the Central DOI: 10.1177/0300985818798112
journals.sagepub.com/home/vet
Nervous System of Cockatiels (Nymphicus
hollandicus) Experimentally Infected with
Parrot Bornavirus 2

Jeann Leal de Araujo1 , Aline Rodrigues-Hoffmann1 , Paula R. Giaretta1,


Jianhua Guo1, Jill Heatley2, Ian Tizard1, and Raquel R. Rech1

Abstract
Neurotropism is a striking characteristic of bornaviruses, including parrot bornavirus 2 (PaBV-2). Our study evaluated the dis-
tribution of inflammatory foci and viral nucleoprotein (N) antigen in the brain and spinal cord of 27 cockatiels (Nymphicus hol-
landicus) following experimental infection with PaBV-2 by injection into the pectoral muscle. Tissue samples were taken at 12
timepoints between 5 and 114 days post-inoculation (dpi). Each experimental group had approximately 3 cockatiels per group and
usually 1 negative control. Immunolabeling was first observed within the ventral horns of the thoracic spinal cord at 20 dpi and in
the brain (thalamic nuclei and hindbrain) at 25 dpi. Both inflammation and viral antigen were restricted to the central core of the
brain until 40 dpi. The virus then spread quickly at 60 dpi to both gray and white matter of all analyzed sections of the central
nervous system (CNS). Encephalitis was most severe in the thalamus and hindbrain, while myelitis was most prominent in the gray
matter and equally distributed in the cervical, thoracic, and lumbosacral spinal cord. Our results demonstrate a caudal to rostral
spread of virus in the CNS following experimental inoculation of PABV-2 into the pectoral muscle, with the presence of viral
antigen and inflammatory lesions first in the spinal cord and progressing to the brain.

Keywords
Psittaciform 1 orthobornavirus (PaBV), Avian bornavirus, cockatiel, Nymphicus hollandicus, central nervous system, viral distribu-
tion, encephalomyelitis

The family Bornaviridae of the order Mononegavirales animals presented clinical signs or at the end of the experiments.
includes mammalian viruses including Borna disease viruses Therefore, crucial information about lesion distribution and viral
(BoDV-1 and 2) and a novel zoonotic species transmitted progression in the early timepoints was consequently missed.
by variegated squirrels (variegated squirrel bornavirus 1; Nevertheless, recent studies using only oculonasal or oral routes
VSBV-1)47 as well as viruses of reptiles and aquatic, psit- of infection were not able to establish persistent infection of
tacine, and passerine birds.1 Parrot bornaviruses (PaBVs), PaBV-2 in experimentally inoculated cockatiels.21
including Parrot bornavirus-2 (PaBV-2), are avian borna- Neurotropism is an important feature of mammalian borna-
viruses that belong to the species Psittaciform 1 or 2 orthor- viruses and has been extensively studied in natural and
bornavirus and are the causative agents of proventricular
dilatation disease (PDD), a lethal disease of captive psitta-
cine birds.2,25,31 1
Department of Veterinary Pathobiology, Texas A&M University, College
Several aspects of the pathogenesis of PDD are still not well Station, TX, USA
2
understood, including its natural route of infection.26,33,43,53 Department of Small Animal Clinical Sciences, Texas A&M University, College
Station, TX, USA
Experimental inoculations using mixed routes of infection (oral,
oculonasal, subcutaneous, and intramuscular)15,37,41,46 or single Supplemental material for this article is available online.
routes (intramuscular, intravenous, or intracerebral)18,42,43 were
Corresponding Author:
able to induce clinical disease and characteristic lesions. How- Jeann Leal de Araujo, Department of Veterinary Pathobiology, Texas A&M
ever, histopathology and immunohistochemistry were imple- University, 307C Manuel Drive, College Station, TX 77840, USA.
mented to analyze late timepoints of infection when the Email: lealjeann@gmail.com
de Araujo et al 107

experimental cases of Borna disease.20,34,48,55 BoDV-1 infec- laboratory cell collection) cultured for 7 passages in minimum
tion occurs in horses and sheep in central Europe49 and causes a essential medium (MEM) supplemented with 10% fetal bovine
lymphoplasmacytic meningoencephalomyelitis with predilec- serum (FBS, Gibco, ThermoFisher Scientific, Walthan, MA),
tion for the gray matter. It affects primarily the hippocampus, and subsequently maintained in MEM supplemented with 2%
caudate nucleus, substantia nigra, midbrain, and hypothala- FBS, as previously described.19 The cell debris were removed
mus.52,54 Eosinophilic intranuclear inclusion bodies, known by centrifugation at 3000  g for 10 minutes. Serial 10-fold
as Joest-Degen bodies, are a sporadic finding in the neurons dilutions of 3 stocks of virus aliquots were analyzed by focus-
of animals affected by Borna disease and particularly promi- forming assays to determine viral titration. Titers above 8 
nent in the hippocampus.4 Additionally, retinal degeneration 104 focus forming units per milliliter (FFU/ml) were consid-
and lymphoplasmacytic optic neuritis have been reported as a ered acceptable for the experimental inoculation as previously
cause of blindness in horses infected with BoDV-1.5,14 BoDV- described.18,19
2 was isolated from a horse with severe neurological disease in
Austria, and although it differs from BoDV-1 by more than Experimental Animals, Virus Inoculation,
15% of nucleotide identity, the clinical and pathological pre-
and Infection Timeline
sentation were identical to BoDV-1 infection.38
The distribution of inflammatory lesions of natural and Thirty-four cockatiels (Nymphicus hollandicus) originated
experimental bornaviral diseases in other mammals such as from 2 PaBV-negative aviaries were used for experimental
variegated squirrels, humans, and immunocompetent rodents inoculations of PaBV-2 (CK1-34), as previously described.33
are similar to those observed in horses and sheep.16,52 In con- Briefly, cockatiels were divided into 12 groups that corre-
trast, neonatal rodents experimentally inoculated with BoDV-1 sponded to different euthanasia timepoints (5, 10, 20, 25, 30,
lack evidence of encephalitis but suffer significant develop- 35, 40, 60, 80, 95, 100, and 114 days post-inoculation; dpi).
mental injury in the central nervous system (CNS), particularly Each group had 3 experimental cockatiels, except for time-
in the hippocampus and cerebellum, including dentate gyrus points 5 dpi (2 cockatiels), 10 dpi (2 cockatiels), 25 dpi
involution and the loss of cerebellar granular cells, (4 cockatiels), 35 dpi (5 cockatiels), 95 dpi (1 cockatiel), and
respectively.28,54,55 100 dpi (2 cockatiels), as described elsewhere.33 Twenty-seven
Neurons, glial, and ependymal cells are the main target cells cockatiels were inoculated with PaBV-2 while 7 cockatiels
of the brains infected by both mammalian and avian borna- were inoculated only with PaBV-free DEF and served as neg-
viruses.8–11,58 Immunolabeling is present in the nuclei alone ative controls. Negative controls were distributed in groups
or nuclei and cytoplasm of these cells, a reflection of the intra- correspondent to timepoints for 20 dpi, 25 dpi, 30 dpi, 35 dpi,
nuclear replication of bornaviruses,27 a characteristic shared 40 dpi, 60 dpi, and 114 dpi (1 negative control for each of these
among only 3 other families of animal RNA viruses: the Ortho- timepoints). Cockatiels were inoculated using intramuscular
myxoviridae, Retroviridae, and Nyamiviridae.12,23,54 injection in the right pectoral muscle with 4  104 FFU of
Although neurotropism is a prominent characteristic of virus. Any clinical signs observed during the experimental
PaBVs, studies on the precise localization of inflammatory period were documented. At the corresponding timepoints,
lesions and viral distribution are limited and lack a systematic cockatiels were anesthetized with isoflurane (IsoThesia, Henry
approach.40,44 The aim of this study was to analyze the distri- Schein, Melville, NY) and then euthanized with carbon dioxide
bution of inflammatory lesions and PaBV nucleoprotein anti- (CO2). The results of the histological and immunohistochem-
gen localization in a chronologic manner to map the CNS areas ical analyses of other organs from the same experimentally
affected by this virus. infected animals are reported elsewhere.33

Histology
Material and Methods
Brain and spinal cord of each bird were placed in 4% neutral
Ethics Statement formaldehyde for 48 hours. Coronal sections of the brain were
All procedures in this study were conducted using protocols obtained from the cerebrum at the level of the hyperpalium and
approved by the Texas A&M Biosafety and Animal Use Com- nucleus basorostralis (Level I), cerebrum at the level of the
mittees (IACUC 20150-0045) and Institutional Biosafety Com- striatum (Level II), and cerebrum at the level of the arcopalium,
mittee (IBC2015-021 and IBC2015-142) and meet all federal thalamus, and midbrain (Level III). Additionally, a sagittal
requirements as defined in the Animal Welfare act (AWA), section was taken encompassing the hindbrain and cerebellum
Public Health Service Policy (PHS), and Humane Care and Use (Level IV) (Supplemental Figure S1). Three transversal serial
of Laboratory Animals. samples from cervical spinal cord between C1 and C13, thor-
acic spinal cord between T1 and T7, lumbosacral spinal cord at
the level of the synsacrum, and the 5 caudal vertebrae (Supple-
Viral Culture and Inoculum Preparation mental Figure S2) were collected from all birds. Samples of
Parrot bornavirus 2 (PaBV-2 CK1-34) was inoculated into duck spinal cord were collected with intact vertebrae and decalci-
embryo fibroblasts (DEF, Schubot Exotic Birds health center fied, which prevented the destruction of the adjacent spinal
108 Veterinary Pathology 56(1)

Figure 1. Brain, cockatiel. Schematic map of histological landmarks in the 4 selected brain levels (I, II, III, IV). Luxol-fast blue/periodic acid-Schiff
(PAS) stain. Abbreviations: AL, ansa lnticularis; AP, arcopallium; Bas, nucleus basorostralis; Cbl, nucleus cerebellaris intermedius; CbM, nucleus
cerebellaris intermedius; CP, comissura posterior; DBC, nucleus decussationis brachiorum conjunctivorum; DIVA, dorsalis intermedius pars
ventralis anterior; DLP, nucleus dorsolateralis posterior thalami; DMP, nucleus dorsomedialis posterior thalami; GCt, substantia grisea centralis;
HP, hyperpallium; La, nucleus laminaris; Mc, nucleus magnocellularis; NP, nidopallium; OS, nucleus olivaris superior; Ote, optic tectum; Ov,
nucleus ovoidalis; OVH, organum vasculosum of the hypothalamus; P, pallidum; PvT, paraventricular nuclei of the thalamus; Rt, nucleus rotundus;
S, nucleus solitarius; SL, nucleus septalis lateralis; SM, nucleus septalis medialis; SRt, nucleus subrotundus; St, striatum; VeD, nucleus vestibularis
descendens; VeL, nucleus vestibularis lateralis; VMH, ventromedial nucleus of hypothalamus.

ganglia during the trimming process. All samples were pro- inflammatory lesions was represented by a different color.
cessed routinely for histologic examination, and when neces- Lesions were considered minimal when 1 single layer of peri-
sary, decalcification was performed using Formical-4 vascular cuffing and no more than 1 glial nodule was in the
(American master tech, Lodi, CA) as recommended by the analyzed section, mild when vessels had 2 layers of perivascu-
manufacturer. The histological evaluation was done blindly lar cuffing and/or 2 to 3 glial nodules, moderate when vessels
by a board-certified pathologist (R. R. Rech). had 3 layers of perivascular cuffing and/or 4 to 7 glial nodules,
The neuroanatomic location of each inflammatory focus and severe when vessels had 4 or more layers of perivascular
was mapped in a diagram, and the description of the regions cuffing and/or more than 7 glial nodules. All sections were
of the brain was adapted for this purpose and followed what analyzed separately for each bird and then overlapped to gen-
has been suggested by the avian brain nomenclature consor- erate the final diagram using a graphic software (Photoshop
tium,45 avian brain atlases,7,36 and avian medicine researchers elements, Adobe systems, San Jose, CA). Lymphoplasmacytic
(Fig. 1).13,39,51 A single blood vessel surrounded by layers of inflammation was scored as follows: – (absent), þ (minimal),
lymphocytes and plasma cells (lymphoplasmacytic perivascu- þþ (mild), þþþ (moderate), þþþþ (severe).
lar cuffing) or a focus of inflammation in the neuroparenchyma
(glial nodule) was interpreted as a single inflammatory focus
(Figs. 2, 3) and indicated by a dot in the diagram. The severity Immunohistochemistry
of inflammation was represented by 4 different dot sizes (min- Immunohistochemistry was performed using polyclonal anti-
imal, mild, moderate, or severe), and each bird with bodies against a specific region of the PaBV N-protein, as
de Araujo et al 109

cervical segment, 95 and 100 dpi in the thoracic, and 40 dpi


in the lumbosacral spinal cord. Ganglioneuritis was more
severe and frequent in the lumbosacral spinal segment. Menin-
gitis in the spinal cord was only observed at 80, 95, 100, and
114 dpi, and it was also more frequent and severe in the lum-
bosacral segment. No inflammation was observed in the glyco-
gen body.
At 25 dpi, the first areas of the brain to present inflammation
were the hindbrain and the thalamus. More specifically, ence-
Figures 2–3. Parrot bornavirus 2 infection, brain, cockatiel. Figure 2. phalitis was more prominent in the gray matter and first spread-
Lymphoplasmacytic perivascular cuffing. HE. Figure 3. Glial focus in the ing through the nucleus magnocellularis, nucleus olivaris
gray matter. HE. superior, and nucleus laminaris in the hindbrain and through
the paraventricular nucleus of the thalamus. By 30 dpi, inflam-
previously described,33 on serial tissue sections, mounted on mation had spread laterally through thalamic nuclei, including
charged slides, and examined by light microscopy. Due to the the nucleus ovoidalis and nucleus rotundus, and dorsally to the
intranuclear replication of PaBVs, only cells with intranuclear commisura posterior, which was the main region of the white
or intranuclear and intracytoplasmic immunolabeling were matter affected by inflammation. The nuclei septalis and scat-
considered positive. Cells with only intracytoplasmic immuno- tered areas in the cerebrum at the level of the striatum and
labeling were considered negative. Immunolabeling was scored nidopallium were also affected at 30 dpi. At 35 dpi, inflamma-
as previously described,33 and the progression for each CNS tory foci reached the optic tectum of the midbrain. Inflamma-
section was recorded in schematic plates. Briefly, slides were tion in the hindbrain was consistently present in the areas
scored based on the number of N-protein positive cells in the corresponding to the nucleus olivaris superior, nucleus solitar-
whole section of brain or spinal cord as follows: – (absent), þ ius, nucleus laminaris, and nucleus decussationis. Only 5 had
(1%–3% affected cells), þþ (4%–10% affected cells), þþþ inflammatory foci in the cerebellum, which only started at 40
(11%–25% affected cells), and þþþþ (more than 25% dpi and were concentrated at the nucleus cerebellaris interme-
affected cells). The immunohistochemical evaluation was done dius and nucleus cerebellaris internus. Inflammation reached
blindly by a board-certified pathologist (A. Rodrigues- the most dorsal areas of the caudal cerebrum comprising the
Hoffmann). mesopallium and hyperpallium at 40 dpi and cranial cerebrum
at 60 dpi affecting both gray and white matter. Hindbrain and
midbrain continuously presented inflammatory foci throughout
Results 60, 80, 95, and 100, until 114 dpi. The cerebrum did not show
inflammation at 95 dpi. At 60, 80, 100, and 114 dpi, inflam-
Distribution of the Inflammatory Infiltrate matory foci were mainly concentrated in the mesopallium and
In the spinal cord, lesions were equally distributed among the 3 hyperpallium. Meningitis in the brain was only observed at 60,
segments with wide distribution within ventral and dorsal horns 100, and 114 dpi, and its severity ranged from minimal to mild.
(Fig. 4). For infected birds, inflammation was more prominent
in the central core of the brain, mainly in the thalamus and
hindbrain (Fig. 5).
Distribution of Viral Antigen
The progression and severity of the inflammation across Immunolabeling was observed in the nuclei and/or nuclei and
different timepoints in the examined CNS sections are summar- cytoplasm of predominantly neurons, but glial and ependymal
ized in Table 1. The first spinal cord segment to develop cells were also intermittently positive (Figs. 6–8). Immunola-
inflammatory foci was the thoracic spinal cord at 25 dpi. beling was observed as early as 20 dpi unilaterally in the neu-
Inflammation was predominant in the ventral horns accompa- rons and glial cells of the ventral horn and adjacesnt ganglia of
nied by ganglioneuritis until 35 dpi, when it started to spread to the thoracic spinal cord and at 25 dpi in the cervical and lum-
the dorsal horns. Both cervical and lumbosacral segments of bosacral segments. At 40 dpi, the immunolabeling spread to the
the spinal cord first developed inflammation at 30 dpi, affecting ependymal cells, adjacent neurons, and glial cells, including
both dorsal and ventral horns as well as adjacent spinal ganglia. cells in the dorsal horns in all 3 segments of the spinal cord.
All sections presented inflammation, primarily in the gray mat- Immunolabeling in the glial cells of the white matter was first
ter, throughout all timepoints until 114 dpi. Although less fre- observed at 80 dpi. In all following timepoints (80, 95, 100, and
quent and severe than inflammation in the gray matter, 114 dpi), a diffuse labeling involving neurons, glial, and epen-
inflammatory foci in the white matter were first observed close dymal cells in both gray and white matter as well as neurons of
to nerve insertions of the ventral funiculus at 30 dpi, followed the adjacent ganglia was observed (Fig. 9).
by the dorsal funiculus at 80 dpi. Then white matter inflamma- In the brain, the thalamic nuclei and the hindbrain were
tion spread to ventral, dorsal, and lateral funiculi until 114 dpi. simultaneously the first areas of the brain with PaBV immuno-
Ganglioneuritis was observed at all timepoints from 25 to 114 labeling at 25 dpi. From 25 to 30 dpi, scattered neurons of the
dpi, with the following exceptions: 25 and 114 dpi in the paraventricular nuclei of the thalamus and glial cells scattered
110 Veterinary Pathology 56(1)

throughout the thalamus were positive. At 30 dpi, few neurons


of the nucleus basorostralis and striatum in the cerebrum were
positive. At 35 dpi, the labeling rapidly spread throughout the
central core reaching the nuclei septalis in the subpallium and
neurons of the nuclei cereberallis and also in the granule cell
layer of the cerebellum. Ependymal cells were also positive at
35 dpi, particularly in the organum vasculosum of the hypotha-
lamus, surrounding the third ventricle. At 40 dpi, immunola-
beling was strongly positive in the pallial areas, particularly
prominent in the arcopallium and nidopallium. Also at 40 dpi,
PaBV-positive neurons and glial cells were identified in the
optic tectum and hyperpallium. The most widespread immuno-
labeling for hindbrain was first observed at 60 dpi. A diffuse
pattern affecting virtually all regions of the cerebrum was
observed from 60 to 114 dpi. A widespread pattern affecting
not only the cerebellar nuclei but also the white matter and the
Purkinje cells was first observed at 80 dpi and remained pos-
itive until 114 dpi (Fig. 10).
No inflammatory foci or antigen immunolabeling were
observed in uninfected control birds. Individual data of the
inflammatory infiltrates and viral antigen distribution for the
CNS of each one of the experimental cockatiels are available in
Supplemental Table S1.

Discussion
After 20 dpi, all infected birds experimentally inoculated with
PaBV-2 in the pectoral muscle developed a lymphoplasmacytic
myelitis or lymphoplasmacytic encephalitis or a combination
of both. This was associated with consistent intranuclear immu-
nolabeling of PaBV N protein antigen mainly in neurons, com-
patible with what has been described in natural cases of PDD.40
Although previous studies have been able to identify PaBV
N-protein antigen and inflammatory lesions in the CNS of
natural and experimental cases of PDD,15,18,41,42,43,46 the dis-
tribution of the inflammation and/or immunolabeling were
broadly determined with minimal neuroanatomic mapping.40,44
In this study, immunolabeling for PaBV N-protein preceded the
development of inflammation throughout all timepoints. The
ventral horns of the thoracic spinal cord were the first pos-
itive CNS areas at 20 dpi. The hindbrain and the thalamus
were the first areas of the brain to simultaneously present
viral antigen and inflammation at 25 dpi. The interval
between 25 and 60 dpi was crucial for the viral spread;
during this period, several areas in the gray and white mat-
ter were progressively and consistently affected, and both
inflammation and viral distribution were more concentrated
to the ventral areas of the brain.
Figure 4. Distribution and severity of inflammation in the cervical
(top), thoracic (middle), and lumbosacral (bottom) spinal cord of
PDD has been often described as a gastrointestinal disease,
PABV-2-inoculated cockatiels. Animals with inflammatory lesions and signs may include proventricular dilation, regurgitation,
are represented by different colored dots and numbers. Each dot crop impaction, anorexia, weight loss, and undigested seeds
represents an inflammatory focus, and the 4 different dot sizes in the feces.53 Experimental studies in cockatiels have shown
represent different degrees of severity (minimal, mild, moderate, gastrointestinal signs prior to the development of neurological
and severe). signs,42,43 and ganglioneuritis has been reported as a more
de Araujo et al 111

Figure 5. Distribution and severity of inflammation in 4 selected levels (I, II, III, and IV) of the brain of PABV-2–inoculated cockatiels. Animals
with inflammatory lesions are represented by different colors. Each dot represents an inflammatory focus, and the 4 different dot sizes represent
different degrees of severity (minimal, mild, moderate, and severe).

consistent finding than meningoencephalomyelitis.15,42,43,46 The anatomical localization of lesions in the CNS is a
However, it is important to note that PDD is primarily a neu- crucial step for developing differential diagnoses of neuro-
rological disease, and gastrointestinal signs are in fact caused logical diseases13 and consequently, the understanding of
by ganglia and nerve dysfunction.53 As previously demon- the pathogenesis of neurotropic viruses. Lesions in different
strated,33 lesions in the CNS may develop prior to peripheral anatomical regions of the CNS may lead to distinct neuro-
lesions, although this does not necessarily mean that the logical conditions,34 hence the importance of mapping the
affected bird will have clinically evident neurological disease, main areas affected in neurological diseases such as PDD.
especially in the early timepoints of infection. In experimental Although the correlation between affected areas and clinical
studies, the route of inoculation, tissues analyzed, and metho- presentation of PaBV-infected birds has not been thoroughly
dology implemented must be considered to explain the discre- explored, the neuroanatomical localization of lesions in
pancy between the development of neurological and rodents experimentally infected with BoDV has provided a
gastrointestinal signs. good understanding of the clinical presentation. In Lewis
112
Table 1. Progression of the inflammatory lesions and immunolabeling for parrot bornavirus-2 in the central nervous system of infected cockatiels across 12 different timepoints.*

5 dpi 10 dpi 15 dpi 20 dpi 25 dpi 30 dpi 35 dpi 40 dpi 60 dpi 80 dpi 95 dpi 100 dpi 114 dpi

Level IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF IHC/INF

Brain - Level I -/- -/- -/- -/- -/- þ/- þ/- þ/- þþþ/þþþ þþþ/þþ þþ/þ þþþ/þþþþ þþþ/þþþþ
Brain - Level II -/- -/- -/- -/- -/- þ/þþþ þ/þþ þþ/þþþ þþþ/þþþ þþþ/þþþ þ/- þþþ/þþþþ þþþ/þþþþ
Brain - Level III -/- -/- -/- -/- þ/þ þþ/þþþ þþ/þþ þþ/þþþ þþþ/þþþ þþþ/þþþ þþþ/þþþ þþþþ/þþþþ þþþþ/þþþþ
Brain - Level IV -/- -/- -/- -/- þþ/þ þþ/þþþ þþþ/þþ þþ/þþþ þþþ/þþþ þþþ/þþþ þþþ/þþþ þþþþ/þþþþ þþþþ/þþþþ
Cervical spinal cord -/- -/- -/- -/- þþ/- þ/þþ þ/þþþ þþ/þþþ þþþ/þþ þþþ/þþþ þþþ/þþ þþþþ/þþþ þþþþ/þþþþ
Thoracic spinal cord -/- -/- -/- þ/- þþ/þ þ/þþþ þ/þþþ þþþ/þþþ þþþ/þþþ þþþ/þþþþ þþþ/þþþþ þþþþ/þþþþ þþþþ/þþþþ
Lumbosacral spinal -/- -/- -/- -/- þ/- þ/þþ þ/þþ þþ/þ þþ/þ þþþ/þþþ þþþ/þþ þþþ/þþþ þþþþ/þþþþ
cord
Abbreviations: -, absent; þ, minimal; þþ, mild; þþþ, moderate; þþþþ, severe. INF, inflammation score; IHC, immunolabeling score.
*The score of histopathology and immunohistochemistry for each timepoint was based on the average of the results of all birds in the respective timepoint. However, when only one bird presented inflammation or was
positive by IHC, its score was considered representative for the timepoint
de Araujo et al 113

Figures 6–8. Parrot bornavirus 2 infection, cerebrum, cockatiel, immunohistochemistry (IHC) for PaBV-2. Figure 6. Intranuclear and intra-
cytoplasmic immunolabeling for PaBV-2 nucleoprotein in neurons. Figure 7. Intranuclear immunolabeling for PaBV-2 nucleoprotein in glial cells.
Figure 8. Intranuclear immunolabeling for PaBV-2 nucleoprotein in ependymal cells.

Figure 9. Spinal cord, cockatiel. Progression of immunolabeling for PaBV-2 nucleoprotein in the thoracic spinal cord. Immunolabeling was
observed as early as 20 dpi unilaterally in few neurons and glial cells of the ventral horn and adjacent ganglia of the thoracic spinal cord. At 30 dpi,
immunolabeling was more intense and affecting more neurons and glial cells but still focally and unilaterally concentrated in the ventral horns.
Immunolabeling progressively involved all areas of white and grey matter by 80 dpi. IHC for PaBV-2.

rats, experimental infection of BoDV usually causes a wide- which results in a neuroendocrine disorder and consequent
spread lymphoplasmacytic meningoencephalitis with viral obesity.22
antigen dissemination in all brain regions, which results in The most common neurological signs seen in birds affected
a biphasic neurobehavioral disorder. However, when the rats by PDD correspond to both behavioral and motor disorders,
are infected with the BoDV-obese variant, inflammation and including lethargy, depression, ataxia, seizures, paresis, and
viral antigen localization is restricted to the septum, hippo- paralysis.26,53 In our study, the concentration of inflammatory
campus, amygdala, and ventromedial tuberal hypothalamus, lesions in viscero-limbic components (eg, ventral striatum and
114 Veterinary Pathology 56(1)

Figure 10. Brain, cockatiel. Progression of immunolabeling for PaBV-2 nucleoprotein in 4 brain levels at 4 timepoints. Immunolabeling was
readily visible at 35 dpi in nuclei of the central core, which can be observed at levels III and IV of the brain. The immunolabeling in these areas
progressively spread throughout these sections until 114 dpi. Immunolabeling in sections I and II were more prominent only starting at 80 dpi
and remained widespread until 114 dpi. IHC for PaBV-2.

pallidum), associated nuclei (eg, nucleus dorsolateralis poster- degeneration and necrosis have been reported in some PDD
ior thalami, nucleus dorsomedialis posterior thalami), and cases3,40 but were not observed in our study. Glial foci were
nuclei involved in reward and motivation (eg, paraventricular sporadically seen in the CNS of our experimental cockatiels,
nuclei of thalamus)32 are in agreement with the regions that are which might have been associated with neuronal death at some
usually affected by behavioral disorders that involve the limbic point of the course of the disease and therefore might play a
system in mammals. Lesions in these areas are usually associ- role in the pathogenesis of neurological manifestations com-
ated with lethargy, depression, obtundation, and stupor.13 monly seen in PDD cases. The wide distribution of lesions in
In mammals, the widespread inflammatory lesions in the the gray matter of the spinal cord seen in our study explain
cerebrum may explain signs such as seizures, while lesions in clinical signs such as paresis and paralysis, sporadically seen in
the cerebellum might be associated with ataxia. Although not PDD cases. Additionally, the high diversity of parrot borna-
well documented in birds, similar circumstances might explain viruses can potentially be associated with variable target cells
these clinical manifestations in PDD cases. Purkinje cell and lesions and consequently, distinct clinical presentations.42
de Araujo et al 115

In this study, the inflammatory lesions bear some similari- cells that is reported in the hippocampus of BoDV-infected rats
ties to what has been described for mammalian bornaviruses. difficult to assess in birds.30,35
Lymphoplasmacytic meningoencephalomyelitis with predilec- The pathognomonic feature of mammalian BoDV infection,
tion for gray matter, including areas such as the thalamus and the eosinophilic intranuclear inclusion bodies known as Joest-
hypothalamus, and relative sparing of the cerebellum and dor- Degen bodies,4 were not observed in this study. Although these
sal cerebrum was our main histological finding as reported for inclusion bodies have been reported in few PDD cases,6,18,57
horses, sheep, variegated squirrels, humans, and immunocom- demonstration of the inclusion bodies in hematoxylin and
petent rodents.16,20,21,38,44,55 In contrast, the predilection for eosin–stained sections as observed for mammals56 has not been
olfactory bulb and hippocampus in mammals, a feature that consistent, and a careful analysis of the morphological and
fits the olfactory route of infection proposed for BoDV patho- staining characteristics of the inclusion bodies must be taken
genesis8 and follows a rostral-caudal progression of BoDV in consideration to differentiate them from fragmented nucleoli
infection, was not observed in our study. or artifacts. The immunohistochemical pattern observed in
The natural route of infection for PaBVs remains to be samples from humans, squirrels, shrews, horses, sheep, and
elucidated. Experimental studies have been able to reproduce rodents infected with mammalian bornaviruses17,24,56 has a
clinical disease using different routes of inoculation, including similar presentation to the immunolabeling in the nuclei, cyto-
single intramuscular, intracerebral, or intravenous plasm, and neuronal and glial processes noted in the CNS of
routes,18,42,43 and also by combined intramuscular, oral, ocu- birds infected with PaBVs.40,44,58
lonasal, and subcutaneous routes.15,37,41 Nonetheless, experi- Our results suggest that the common practice of collecting
ments using combined routes have nebulous results in regards and assessing only a single section of the brain could lead to a
to which one of the routes was responsible for causing clinical false-negative diagnosis in PaBV-infected birds, especially in
disease. Recently, experimental studies using oral and oculo- the early timepoints of infection. We observed inflammation
nasal routes of infection have failed to induce disease and/or in the hindbrain and midbrain from early to late timepoints
PDD lesions in cockatiels after 6 months or experiment,21 (25–114 dpi). On the other hand, the cerebrum at the level of
which may indicate that these routes are unlikely to be associ- the hyperpalium remained as the least affected area until 100
ated with PaBVs’ natural route of transmission. Additionally, dpi. Therefore, sampling limited to rostral areas of cerebrum
skin injuries due to squirrel bites and scratches have been would be prone to miss the vast majority of inflammatory
reported by family members of the 3 patients who died with lesions, which might result in the misleading assumption that
variegated squirrel bornavirus-1 (VSBV-1)–induced encepha- gastrointestinal ganglioneuritis was present prior to
litis in Germany.24 The successful experiments using intramus- meningoencephalomyelitis.
cular routes of inoculation in psittacine birds and the recent In conclusion, our results suggest that thalamus and hind-
data about skin injuries in VSBV-1 cases reiterate the impor- brain were considered the most appropriate sections of the
tance of investigating the association of tegument injuries and brain for sampling to determine PaBV-2 infection spread and
intramuscular lesions as a port of entry for PaBVs. However, it inflammation. Little or no difference was observed between the
is important to note that although intramuscular inoculation of 3 segments of the spinal cord in our study of experimental
PaBV-2 successfully caused PDD in cockatiels of our experi- PaBV-2 infection, based on distribution of viral antigen and
ment, there is still not enough data to support that this is the inflammatory lesions. Further research is needed to understand
route of infection in natural PDD cases. whether a similar process happens in naturally infected birds
A potential intramuscular route of infection (as was done in and elucidate the mode of viral spread from animal to animal as
our experiment33) creates the possibility of PaBVs being spread well as more fully understand the physiologic and behavioral
first to the spinal cord and only reaching the rostral areas of the effects of this temporal progression in the distribution of viral
late in the course of infection, resulting in a caudal-rostral antigen and inflammation. We advocate sampling the spinal
progression of infection and explaining the reduced inflamma- cord with the vertebral bodies in birds because this results in
tion and presence of viral N-protein antigen seen in the more outstanding visualization of the spinal ganglia and the decalci-
rostral areas during the late timepoints. fication process does not interfere with viral antigen detection.
Although the hippocampus is associated with spatial mem-
ory in both birds and mammals, its localization and structure
Acknowledgements
differ enormously between these classes of animals.50 The
We acknowledge the excellent technical assistance provided by Debra
mammalian hippocampus is located rostral to the midbrain and
Turner, Luan Henker, Cintia Queiroz, Caitlin Older, Courtney Smith,
ventral to the thalamus, while the avian hippocampus has a Anna Blick, Mackenzie Branco, Kelly Mallet, Brittany Kerr, Robin
dorsal location in the pallial region of the cerebrum.29 This Katchko, Randi Gold, Sierra Guidry, and Kristen Streeter.
area was not affected in the cockatiels of this study, probably
due the predilection of the virus for the central core of the brain
and consequent inflammation concentrated in the ventral areas Declaration of Conflicting Interests
of the brain. Additionally, birds lack a distinguishable dentate The author(s) declared no potential conflicts of interest with respect to
gyrus, which makes the progressive cell death of the granule the research, authorship, and/or publication of this article.
116 Veterinary Pathology 56(1)

Funding 20. Hallensleben W, Schwemmle M, Hausmann J, et al. Borna disease virus-


induced neurological disorder in mice: infection of neonates results in immu-
The author(s) disclosed receipt of the following financial support for
nopathology. J Virol. 1998;72:4379–4386.
the research, authorship, and/or publication of this article: We thank
21. Heckmann J, Enderlein D, Piepenbring AK, et al. Investigation of different
the Avian Health Complex, the Schubot Center, Texas A&M Univer-
infection routes of parrot bornavirus in cockatiels. Avian Dis. 2017;61:90–95.
sity, and the CAPES-Science without borders program for supporting
22. Herden C, Herzog S, Richt JA, et al. Distribution of Borna disease virus in the
this study. brain of rats infected with an obesity-inducing virus strain. Brain Pathol. 2000;
10:39–48.
23. Herrel M, Hoefs N, Staeheli P, et al. Tick-borne Nyamanini virus replicates in
ORCID iD
the nucleus and exhibits unusual genome and matrix protein properties. J Virol.
Jeann Leal de Araujo http://orcid.org/0000-0001-8688-9864 2012;86:10739–10747.
Aline Rodrigues-Hoffmann http://orcid.org/0000-0001-6148-2531 24. Hoffmann B, Tappe D, Hoper D, et al. A variegated squirrel bornavirus asso-
ciated with fatal human encephalitis. N Engl J Med. 2015;373:154–162.
25. Honkavuori KS, Shivaprasad HL, Williams BL, et al. Novel borna virus in
References
psittacine birds with proventricular dilatation disease. Emerg Infect Dis.
1. Afonso CL, Amarasinghe GK, Banyai K, et al. Taxonomy of the order Mono- 2008;14:1883–1886.
negavirales: update 2016. Arch Virol. 2016;161:2351–2360. 26. Hoppes S, Gray PL, Payne S, et al. The isolation, pathogenesis, diagnosis,
2. Amarasinghe GK, Arechiga Ceballos NG, Banyard AC, et al. Taxonomy of the transmission, and control of avian bornavirus and proventricular dilatation dis-
order Mononegavirales: update 2018. Arch Virol. 2018;163:2283–2294. ease. Vet Clin North Am Exot Anim Pract. 2010;13:495–508.
3. Berhane Y, Smith DA, Newman S, et al. Peripheral neuritis in psittacine birds 27. Horie M, Kobayashi Y, Suzuki Y, et al. Comprehensive analysis of endogenous
with proventricular dilatation disease. Avian Pathol. 2001;30:563–570. bornavirus-like elements in eukaryote genomes. Philos Trans R Soc Lond B Biol
4. Bestetti G. [Microscopic and ultrastructural studies on Joest-Degen inclusion Sci. 2013;368:20120499.
bodies in spontaneous Borna disease of the horse]. Schweiz Arch Tierheilkd. 28. Hornig M, Solbrig M, Horscroft N, et al. Borna disease virus infection of adult
1976;118:493–498. and neonatal rats: Models for neuropsychiatric disease. Curr Top Microbiol
5. Bilzer T, Grabner A, Stitz L. [Immunopathology of Borna disease in the horse:
Immunol. 2001;253:157–177.
clinical, virological and neuropathologic findings]. Tierarztl Prax. 1996;24:
29. Jarvis ED, Gunturkun O, Bruce L, et al. Avian brains and a new understanding
567–576.
of vertebrate brain evolution. Nat Rev Neurosci. 2005;6:151–159.
6. Bourque L, Laniesse D, Beaufrere H, et al. Identification of avian bornavirus in
30. Kempermann G. New neurons for “survival of the fittest.” Nat Rev Neurosci.
a Himalayan monal (Lophophorus impejanus) with neurological disease. Avian
2012;13:727–736.
Pathol. 2015;44:323–327.
31. Kistler AL, Gancz A, Clubb S, et al. Recovery of divergent avian bornaviruses
7. Brauth S, Heaton J, Roberts T, et al. Budgegerigar brain atlas. http://www.
from cases of proventricular dilatation disease: identification of a candidate
brauthlab.umd.edu/atlas.htm. Accessed November 2017.
etiologic agent. Virol J. 2008;5:88.
8. Carbone KM. Borna disease virus and human disease. Clin Microbiol Rev.
32. Kuenzel WJ. The avian subpallium and autonomic nervous system A2. In:
2001;14:513–527.
Scanes CG, ed. Sturkie’s Avian Physiology. 6th ed. San Diego: Academic Press;
9. Carbone KM, Duchala CS, Narayan O. Borna disease virus and antigens in glial
2015:135–163.
and neuronal cell-populations of the nervous-system. Annals of Neurology.
33. Leal de Araujo J, Rech RR, Heatley JJ, et al. From nerves to brain to
1988;24:476–482.
gastrointestinal tract: A time-based study of parrot bornavirus 2 (PaBV-2)
10. Carbone KM, Moench TR, Lipkin WI. Borna disease virus replicates in astro-
pathogenesis in cockatiels (Nymphicus hollandicus). PLoS One. 2017;12:
cytes, schwann-cells and ependymal cells in persistently infected-rats—location
e0187797.
of viral genomic and messenger-rnas by insitu hybridization. J Neuropathol Exp
34. Ludlow M, Kortekaas J, Herden C, et al. Neurotropic virus infections as the
Neurol. 1991;50:205–214.
cause of immediate and delayed neuropathology. Acta Neuropathol. 2016;131:
11. Carbone KM, Trapp BD, Griffin JW, et al. Astrocytes and schwann-cells are
virus-host cells in the nervous-system of rats with Borna disease. J Neuropathol 159–184.
Exp Neurol. 1989;48:631–644. 35. Mayer D, Fischer H, Schneider U, et al. Borna disease virus replication in
12. Cros JF, Palese P. Trafficking of viral genomic RNA into and out of the nucleus: organotypic hippocampal slice cultures from rats results in selective damage
influenza, Thogoto and Borna disease viruses. Virus Res. 2003;95:3–12. of dentate granule cells. J Virol. 2005;79:11716–11723.
13. de Lahunta A, Glass EG, Kent M. The neurologic examination. In: de Lahunta 36. Mikula S, Stone JM, Jones EG. BrainMaps.org—interactive high-resolution
A, Glass EG, Kent M, eds. Veterinary Neuroanatomy and Clinical Neurology. digital brain atlases and virtual microscopy. Brains Minds Media. 2008;3:
4th ed. St. Louis, MO: Elsevier Saunders; 2015:525–539. bmm1426.
14. Dietzel J, Kuhrt H, Stahl T, et al. Morphometric analysis of the retina from 37. Mirhosseini N, Gray PL, Hoppes S, et al. Proventricular dilatation disease in
horses infected with the Borna disease virus. Vet Pathol. 2007;44:57–63. cockatiels (Nymphicus hollandicus) after infection with a genotype 2 avian
15. Gancz AY, Kistler AL, Greninger AL, et al. Experimental induction of proven- bornavirus. J Avian Med Surg. 2011;25:199–204.
tricular dilatation disease in cockatiels (Nymphicus hollandicus) inoculated 38. Nowotny N, Kolodziejek J, Jehle CO, et al. Isolation and characterization of a
with brain homogenates containing avian bornavirus 4. Virol J. 2009;6:100. new subtype of Borna disease virus. J Virol. 2000;74:5655–658.
16. Gosztonyi G. Natural and experimental Borna disease virus infections—neuro- 39. Orosz S, Antinoff N. Clinical avian neurology and neuroanatomy. In: Speer B,
pathology and pathogenetic considerations. APMIS Suppl. 2008:53–57. ed. Current Therapy in Avian Medicine and Surgery. St. Louis, MO: Elsevier;
17. Gosztonyi G, Ludwig H. Borna disease—neuropathology and pathogenesis. In: 2015:363–377.
Koprowski H, Lipkin WI, eds. Borna Disease. Berlin, Springer Verlag KG; 40. Ouyang N, Storts R, Tian Y, et al. Histopathology and the detection of avian
1995:39–73. bornavirus in the nervous system of birds diagnosed with proventricular dilata-
18. Gray P, Hoppes S, Suchodolski P, et al. Use of avian bornavirus isolates to tion disease. Avian Pathol. 2009;38:393–401.
induce proventricular dilatation disease in conures. Emerg Infect Dis. 2010;16: 41. Payne S, Shivaprasad HL, Mirhosseini N, et al. Unusual and severe lesions of
473–479. proventricular dilatation disease in cockatiels (Nymphicus hollandicus) acting
19. Guo J, Payne S, Zhang S, et al. Avian bornaviruses: diagnosis, isolation, and as healthy carriers of avian bornavirus (ABV) and subsequently infected with a
genotyping. Curr Protoc Microbiol. 2014;34:15I1.1–15I1.33. virulent strain of ABV. Avian Pathol. 2011;40:15–22.
de Araujo et al 117

42. Piepenbring AK, Enderlein D, Herzog S, et al. Parrot Bornavirus (PaBV)-2 50. Striedter GF. Evolution of the hippocampus in reptiles and birds. J Comp
isolate causes different disease patterns in cockatiels than PaBV-4. Avian Neurol. 2016;524:496–517.
Pathol. 2016;45:156–168. 51. Swayne DE, Barnes HJ, Abdul-Aziz T, et al. Nervous system. In: Abdul-Aziz T,
43. Piepenbring AK, Enderlein D, Herzog S, et al. Pathogenesis of avian bornavirus Fletcher OJ, Barnes HJ, eds. Avian Histopathology 2016. 4th ed. Jacksonville,
in experimentally infected cockatiels. Emerg Infect Dis. 2012;18:234–241. Fl: Amer Asoc Avian Pathology; 2016:469–519.
44. Raghav R, Taylor M, Delay J, et al. Avian bornavirus is present in many tissues 52. Tizard I, Ball J, Stoica G, Payne S. The pathogenesis of bornaviral diseases in
of psittacine birds with histopathologic evidence of proventricular dilatation mammals. Anim Health Res Rev. 2016;17:92–109.
disease. J Vet Diagn Invest. 2010;22:495–508. 53. Tizard I, Shivaprasad HL, Guo J, et al. The pathogenesis of proventricular
45. Reiner A, Perkel DJ, Bruce LL, et al. The avian brain nomenclature forum: dilatation disease. Anim Health Res Rev. 2016;17:110–126.
terminology for a new century in comparative neuroanatomy. J Comp Neurol. 54. Tomonaga K, Kobayashi T, Ikuta K. Molecular and cellular biology of Borna
2004;473:E1–E6. disease virus infection. Microbes Infect. 2002;4:491–500.
46. Rubbenstroth D, Brosinski K, Rinder M, et al. No contact transmission of avian 55. Watanabe M, Lee BJ, Kamitani W, et al. Neurological diseases and viral
bornavirus in experimentally infected cockatiels (Nymphicus hollandicus) and dynamics in the brains of neonatally borna disease virus-infected gerbils. Vir-
domestic canaries (Serinus canaria forma domestica). Vet Microbiol. 2014;172: ology. 2001;282:65–76.
146–156. 56. Weissenbock H, Bago Z, Kolodziejek J, et al. Infections of horses and shrews
47. Schlottau K, Jenckel M, van den Brand J, et al. Variegated squirrel bornavirus with Bornaviruses in Upper Austria: a novel endemic area of Borna disease.
1 in squirrels, germany and the Netherlands. Emerg Infect Dis. 2017;23: Emerg Microbes Infect. 2017;6:e52.
477–481. 57. Weissenbock H, Bakonyi T, Sekulin K, et al. Avian bornaviruses in psittacine
48. Sobbe M, Bilzer T, Gommel S, et al. Induction of degenerative brain lesions birds from Europe and Australia with proventricular dilatation disease. Emerg
after adoptive transfer of brain lymphocytes from Borna disease virus-infected Infect Dis. 2009;15:1453–1459.
rats: presence of CD8þ T cells and perforin mRNA. J Virol. 1997;71: 58. Wunschmann A, Honkavuori K, Briese T, et al. Antigen tissue distribution of
2400–2407. Avian bornavirus (ABV) in psittacine birds with natural spontaneous proven-
49. Staeheli P, Sauder C, Hausmann J, et al. Epidemiology of Borna disease virus. tricular dilatation disease and ABV genotype 1 infection. J Vet Diagn Invest.
J Gen Virol. 2000;81:2123–2135. 2011;23:716–726.

You might also like