ArticlePDF Available

Influenza A Viruses Grow in Human Pancreatic Cells and Cause Pancreatitis and Diabetes in an Animal Model

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

Influenza A viruses commonly cause pancreatitis in naturally and experimentally infected animals. In this study we report the results of in vivo investigations carried out to establish whether influenza infection could cause metabolic disorders linked to pancreatic infection. In addition, in vitro tests in human pancreatic islets and in human pancreatic cell lines were performed to evaluate viral growth and cell damage.Infection of an avian model with two low pathogenicity avian influenza isolates caused pancreatic damage resulting in hyperlipasemia in over 50% of subjects, which evolved into hyperglycemia and subsequently diabetes. Histopathology of the pancreas showed signs of an acute infection resulting in severe fibrosis and disruption of the structure of the organ. Influenza nucleoprotein was detected by IHC in the acinar tissue.Human seasonal H1N1 and H3N2 viruses and avian H7N1 and H7N3 influenza isolates were able to infect a selection of human pancreatic cell lines. Human viruses were also shown to be able to infect human pancreatic islets. In situ hybridization assays indicated that viral nucleoprotein could be detected in beta cells. The cytokine activation profile indicated a significant increase of MIG/CXCL9, IP-10/CXCL10, RANTES/CCL5, MIP1b/CCL4, Groa/CXCL1, IL8/CXCL8, TNFa and IL-6.Our findings indicate that influenza infection may play a role as causative agent of pancreatitis and diabetes in humans and other mammals.
Content may be subject to copyright.
Influenza A Viruses Grow in Human Pancreatic Cells and Cause
Pancreatitis and Diabetes in an Animal Model
Ilaria Capua,
a
Alessia Mercalli,
b
Matteo S. Pizzuto,
a
Aurora Romero-Tejeda,
a
Samantha Kasloff,
a
Cristian De Battisti,
a
Francesco Bonfante,
a
Livia V. Patrono,
a
Elisa Vicenzi,
c
Valentina Zappulli,
d
Vito Lampasona,
b
Annalisa Stefani,
e
Claudio Doglioni,
f,g
Calogero Terregino,
a
Giovanni Cattoli,
a
Lorenzo Piemonti
b
Department of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Padova, Italy
a
; Diabetes Research Institute-DRI, San Raffaele
Scientific Institute, Milan, Italy
b
; Viral Pathogens and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute,
Milan, Italy
c
; Department of Public Health, Comparative Pathology and Veterinary Hygiene, Faculty of Veterinary Medicine, Agripolis, Legnaro, Padova, Italy
d
; Animal
Health and Welfare Department, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Padova, Italy
e
; Unit of Pathology, San Raffaele Scientific Institute, Milan, Italy
f
;
Università Vita-Salute, Milan, Italy
g
Influenza A viruses commonly cause pancreatitis in naturally and experimentally infected animals. In this study, we report the
results of in vivo investigations carried out to establish whether influenza virus infection could cause metabolic disorders linked
to pancreatic infection. In addition, in vitro tests in human pancreatic islets and in human pancreatic cell lines were performed
to evaluate viral growth and cell damage. Infection of an avian model with two low-pathogenicity avian influenza isolates caused
pancreatic damage resulting in hyperlipasemia in over 50% of subjects, which evolved into hyperglycemia and subsequently dia-
betes. Histopathology of the pancreas showed signs of an acute infection resulting in severe fibrosis and disruption of the struc-
ture of the organ. Influenza virus nucleoprotein was detected by immunohistochemistry (IHC) in the acinar tissue. Human sea-
sonal H1N1 and H3N2 viruses and avian H7N1 and H7N3 influenza virus isolates were able to infect a selection of human
pancreatic cell lines. Human viruses were also shown to be able to infect human pancreatic islets. In situ hybridization assays
indicated that viral nucleoprotein could be detected in beta cells. The cytokine activation profile indicated a significant increase
of MIG/CXCL9, IP-10/CXCL10, RANTES/CCL5, MIP1b/CCL4, Groa/CXCL1, interleukin 8 (IL-8)/CXCL8, tumor necrosis factor
alpha (TNF-), and IL-6. Our findings indicate that influenza virus infection may play a role as a causative agent of pancreatitis
and diabetes in humans and other mammals.
I
nfluenza A viruses (IAVs) originate from the wild-bird reservoir
and infect a variety of hosts, including domestic birds. These
viruses are also able to infect a significant number of mammals, in
which they may become established. Among these are pigs,
equids, mustelids, sea mammals, canids, felids, and humans. IAVs
cause systemic or nonsystemic infection, depending on the strain
involved. The systemic disease occurs mostly in avian species and
is known as highly pathogenic avian influenza (HPAI). It is char-
acterized by extensive viral replication in vital organs and death
within a few days after the onset of clinical signs in the majority of
infected animals. The nonsystemic form, which is by far the most
common, occurs in birds and mammals and is characterized by
mild respiratory and enteric signs. To differentiate it from HPAI,
in birds it is known as low-pathogenicity avian influenza (LPAI).
The different clinical presentation results from the fact that non-
systemic influenza A viruses are able to replicate only in the pres-
ence of trypsin or trypsin-like enzymes, and thus, their replication
is believed to be restricted to the respiratory and enteric tracts.
Avian IAVs have a tropism for the pancreas (14). Necrotizing
pancreatitis is a common finding in wild and domestic birds in-
fected with HPAI virus (58), and the systemic nature of HPAI is
in line with these findings. In contrast, it is difficult to explain
pancreatic colonization by LPAI viruses, which is a common find-
ing in infected chickens and turkeys (914). Previous studies have
reported that certain IAVs can also cause pancreatitis in mammals
following natural or experimental infection (1518). Recently,
there have been reports of pancreatic damage in human cases
associated with H1N1pdm influenza A virus infection, including
both acute pancreatitis and the onset of type 1 diabetes (T1D)
(1923).
To date, there has been no attempt to establish whether influ-
enza viruses are able to grow in pancreatic cells in vitro, and no
data are available on the consequences of influenza virus replica-
tion in the pancreas in vivo. In this study, we explored the impli-
cations of influenza virus infection for pancreatic endocrine func-
tion in an animal model, and we performed in vitro experiments
aiming to establish the occurrence, extent, and implications of
influenza A virus infection in human cells of pancreatic origin. For
the in vivo studies, we selected the turkey as a model, due to the fact
that turkeys are highly susceptible to influenza virus infection and
pancreatic damage is often observed as a postmortem lesion. Ex-
perimental infections were performed with two LPAI viruses,
A/turkey/Italy/3675/1999 (H7N1) and A/turkey/Italy/2962/2003
(H7N3), as both viruses had been associated with pancreatic le-
sions in naturally infected birds. For the in vitro studies, in addi-
tion to the previously mentioned avian strains, we selected A/New
Caledonia/20/99 (H1N1) and A/Wisconsin/67/05 (H3N2), as
Received 27 March 2012 Accepted 17 October 2012
Published ahead of print 24 October 2012
Address correspondence to Ilaria Capua, icapua@izsvenezie.it.
Supplemental material for this article may be found at http://dx.doi.org
/10.1128/JVI.0714-12.
Copyright © 2013, American Society for Microbiology. All Rights Reserved.
doi:10.1128/JVI.00714-12
January 2013 Volume 87 Number 1 Journal of Virology p. 597– 610 jvi.asm.org 597
these viruses have circulated for extensive periods in humans, and
existing epidemiological data would be suitable for a retrospective
study. The strains were used to infect two established human pan-
creatic cell lines (including human insulinoma and pancreatic
duct cell lines) and primary cultures of human pancreatic islets.
MATERIALS AND METHODS
In vivo experiment. The aim of this study was to establish whether two
natural nonsystemic avian influenza viruses obtained from field out-
breaks, without prior adaptation, could cause endocrine or exocrine pan-
creatic damage following experimental infection of young turkeys. The
study was performed in strict accordance with the relevant national and
local animal welfare bodies [Convention of the European Council no. 123
and National Guidelines (Legislative Decree 116/92)]. The protocol was
approved by the Committee on the Ethics of Animal Experiments of the
Istituto Zooprofilattico Sperimentale delle Venezie (IZSVe) (permit
number CE.IZSVE.022012).
Animals. Sixty-eight female meat turkeys obtained at 1 day of age
from a commercial farm were used in this study. The birds were housed in
negative-pressure, HEPA-filtered isolation cabinets for the duration of
the experimental trial. Before carrying out the infection, the animals were
housed for 3 weeks to allow adaptation and growth, received feed and
water ad libitum, and were identified by means of wing tags.
Viruses. Two LPAI viruses isolated during epidemics in Italy were
used for the experimental infection: A/turkey/Italy/3675/1999 (H7N1)
and A/turkey/Italy/2962/2003 (H7N3). Stocks of AI viruses were pro-
duced by a single passage in 9-day-old embryonated specific-pathogen-
free (SPF) chicken eggs via the allantoic cavity, according to EU Council
Directive 2005/94/EC (24). The allantoic fluid was harvested 48 h postin-
oculation, aliquoted, and stored at 80°C until use. For viral titration, 100
l of 10-fold-diluted viral suspension was inoculated in SPF embryonated
chicken eggs, and the median embryo infectious dose (EID
50
) was calcu
-
lated according to the Reed and Muench formula (54).
Experimental design. Birds were divided into three experimental
groups (A [H7N1], B [H7N3], and K [control]). Groups A and B, each
consisting of 24 animals, were infected via the oral-nasal route with 0.1 ml
of allantoic fluid containing 10
6.83
EID
50
of the A/turkey/Italy/3675/1999
(H7N1) virus and 10
6.48
EID
50
of the A/turkey/Italy/2962/2003 (H7N3)
virus. Group K, consisting of 20 animals, received 0.1 ml of negative al-
lantoic fluid via the oral-nasal route and served as a negative control. All
birds were observed twice daily for clinical signs. On days 0, 3, 6, 9, 13, 15,
20, 23, 27, 31, 34, 41, and 45 postinfection (p.i.), blood was collected from
the brachial veins of all animals using heparinized syringes in order to
determine glucose and lipase levels in plasma. On days 2 and 3 p.i., tra-
cheal swabs were collected to evaluate viral replication. On day 3 p.i.,
blood was also collected to determine the presence of viral RNA in the
blood. On days 4 and 7 p.i., two birds from each infected group were
humanely sacrificed, and the pancreas and lungs were processed for the
detection of viral RNA and for histopathology and immunohistochemis-
try (IHC). Similarly, on days 8 and 17 p.i., one subject from each experi-
mental group was euthanized, and the pancreas was collected for histo-
logical and immunohistochemical studies. For this purpose, we selected
hyperglycemic subjects that had also shown an increase in lipase levels.
Biochemical analyses. Blood samples were collected in Gas Lyte 23 G
pediatric syringes containing lyophilized lithium heparin as an anticoag-
ulant. At each sampling, 0.3 ml of blood was collected and refrigerated at
4°C until it was processed. To obtain plasma, samples were immediately
centrifuged at 1,500 g for 15 min at 4°C. To determine the levels of
glucose and lipase in plasma, commercially available kits (Glucose HK
and LIPC; Roche Diagnostics GmbH, Mannheim, Germany) were applied
to the computerized system Cobas c501 (F. Hoffmann-La Roche Std.,
Basel, Switzerland). The Glucose HK test is based on a hexokinase enzy-
matic reaction. The linearity of the reaction is 0.11 to 41.6 mmol/liter (2 to
750 mg/dl), and its analytic sensitivity is 0.11 mmol/liter (2 mg/dl). The
LIPC test is based on a colorimetric enzymatic reaction with a linearity of
3 to 300 U/liter and an analytic sensitivity of 3 U/liter.
Molecular tests. Tracheal swabs, blood samples, and organs (pancreas
and lungs) were tested for viral RNA by means of real-time reverse trans-
criptase PCR (RRT-PCR) for the identification of the influenza virus ma-
trix (M) gene.
RNA extraction. Viral RNA was extracted from 100 l of blood using
the commercial NucleoSpin RNA II kit (Macherey-Nagel) and from 50 l
of phosphate-buffered saline (PBS) containing a tracheal-swab suspen-
sion using the Ambion MagMax-96 AI-ND Viral RNA Isolation Kit for
the automatic extractor. One hundred fifty milligrams of homogenized
lung and pancreas tissues was centrifuged, and viral RNA was extracted
from 100 l of clarified suspension using the NucleoSpin RNA II kit
(Macherey-Nagel).
One-step RRT-PCR. The isolated RNA was amplified using the pub-
lished primers and probes from Spackman et al. (25), targeting the con-
served matrix (M) gene of type A influenza virus. Five microliters of RNA
was added to the reaction mixture, composed of 300 nM forward and
reverse primers (M25F and M124-R, respectively) and 100 nM fluores-
cent-label probe (M64). The amplification reaction was performed in a
final volume of 25 l using the commercial QuantiTect Multiplex RT-
PCR kit (Qiagen, Hilden, Germany). The PCR was performed using the
following protocol: 20 min at 50°C and 15 min at 95°C, followed by 40
cycles at 94°C for 45 s and 60°C for 45 s. In vitro-transcribed target RNA
was obtained using Mega Short Script 7 (high-yield transcription kit; Am-
bion) according to the manufacturer’s instructions, quantified by Nano-
Drop 2000 (Thermo Scientific), and used to create a standard calibration
curve for viral-RNA quantification. To check the integrity of the isolated
RNA, the -actin gene was also amplified using a set of primers designed
in house (primer sequences are available upon request). The reaction
mixture was composed of 300 nM forward and reverse primers and 1
EvaGreen (Explera, Jesi, Italy). The amplification reaction was performed
in a final volume of 25 l using the commercial Superscript III kit (Invit-
rogen, Carlsbad, CA). The PCR was performed using the following pro-
tocol: 30 min at 55°C and 2 min at 94°C, followed by 45 cycles at 94°C for
30 s and 60°C for 1 min.
Histology and immunohistochemistry. Formalin-fixed, paraffin-
embedded pancreas sections were cut (3 m thick). Slides were stained
with hematoxylin and eosin (H&E) (Histoserv, Inc., Germantown, MD).
Representative photographs were taken with SPOT Advanced software
(version 4.0.8; Diagnostic Instruments, Inc., Sterling Heights, MI). The
reagents and methodology for influenza virus IHC were as follows: poly-
clonal antibody anti-type A influenza virus nucleoprotein (NP) and
mouse anti-influenza virus A (NP subtype A, clone EVS 238; European
Veterinary Laboratory), 1:100 in PBS-2.5% bovine serum albumin (BSA)
for1hatroom temperature; secondary antibody, goat anti-mouse IgG2a
horseradish peroxidase (HRP) (Southern Biotech), 1/200 in PBS-2.5%
BSAfor1hatroom temperature. Antigen retrieval was performed by
incubating the slides for 10 min at 37°C in trypsin (Digest-all Kit; Invit-
rogen). Endogenous peroxidase was blocked with 3% H
2
O
2
for 10 min at
room temperature. Before incubation with primary antibody, a blocking
step was performed with PBS-5% BSA for 20 min at room temperature.
Diaminobenzidine (DAB) was applied as a chromogen (DakoCytoma-
tion; code K3468). IHC for insulin and glucagon was performed with
polyclonal guinea pig anti-swine insulin, 1:50 (A0564; Dako, Carpinteria,
CA), and polyclonal rabbit anti-glucagon, 1:200 (NCL-GLUC; Novocas-
tra, Newcastle, United Kingdom), using as a detection system the En Vi-
sion Ap (K1396; Dako, Carpinteria, CA) and nuclear fast red (K139;6
Dako) for influenza virus A staining and En VisionSystem HRP-labeled
polymer anti-rabbit (K4002; Dako, Carpinteria, CA) and DAB (K3468;
Dako, Carpinteria, CA) for insulin and glucagon staining.
In vitro experiment. The aim of these experiments was to establish
whether human and avian influenza viruses could grow on cell lines de-
rived from the human pancreas and to investigate the effect of human
influenza virus replication in human pancreatic islets.
Capua et al.
598 jvi.asm.org Journal of Virology
Cell lines. Madin-Darby canine kidney (MDCK) cells were main-
tained in Alpha’s modified Eagle medium (AMEM) (Sigma) supple-
mented with 10% fetal bovine serum (FBS), 1% 200 mM
L-glutamine, and
a 1% penicillin-streptomycin-nystatin (pen-strep-nys) solution. The hu-
man insulinoma cell line hCM (26) and immortalized human ductal ep-
ithelial cell line HPDE6 (27) were maintained in RPMI (Gibco) supple-
mented with 1%
L-glutamine, 1% antibiotics, and FBS (5% and 10%,
respectively). MDCK and HPDE6 cells were passaged twice weekly at a
subcultivation ratio of 1:10 and 1:4, while hCM cells were split three times
per week at a ratio of 1:4. All cells were maintained in a humidified incu-
bator at 37°C with 5% CO
2
.
Primary cells. Pancreatic islets were isolated and purified at San Raf-
faele Scientific Institute from pancreases of multiorgan donors according
to Ricordi’s method. Briefly, after cannulation of the pancreatic duct,
collagenase solution (2,000 U; Serva, Germany) at 4°C was injected
through the duct (perfusion). Subsequently, the pancreas was cut into
small pieces and loaded into a digestion chamber, named Ricordi’s cham-
ber, for an enzymatic and mechanical digestion at 37°C. Final purification
of digested pancreas was performed using a continuous gradient (Ficoll;
Biochrom, Berlin) in a computerized centrifuge system (COBE 2991 cell
processor). Islet preparations with purities of 80% 8% (mean
standard deviation [SD]; n 6) not suitable for transplantation were used
after approval by the local ethical committee. Cells were seeded in 24-well
plates and 25-cm
2
flasks at 150 islets/ml and maintained in final wash
culture medium (Mediatech, Inc., Manassas, VA) at 37°C with 5% CO
2
.
Sialic acid receptor characterization on hCM and HPDE6 cells. The
presence of alpha-2,3- and alpha-2,6-linked sialic acid residues was deter-
mined by flow cytometry. Following trypsinization, 1 10
6
cells were
washed twice with PBS-10 mM HEPES (PBS-HEPES) for 5 min at 1,200
rpm and then treated with an avidin/biotin-blocking kit (Vector Labora-
tories) according to the manufacturer’s instructions, with cells incubated
for 15 min with 100 l of each solution. Alpha-2,3 and alpha-2,6 sialic acid
linkages were detected by incubating cells for 30 min with 100 l of bio-
tinylated Maackia amurensis (MAA) lectin II (Vector Laboratories; 5 g/
ml), followed by 100 l of phycoerythrin (PE)-streptavidin (BD Biosci-
ences; 10 g/ml) for 30 min at 4°C in the dark or with 100 lof
fluorescein-conjugated Sambucus nigra (SNA) lectin (Vector Laborato-
ries; 5 g/ml). Cells were washed twice with PBS-HEPES between all
blocking and staining steps and resuspended in PBS with 1% formalin
prior to analysis. To confirm the specificity of lectins, cells were pretreated
with 1 U per ml of neuraminidase (NA) from Clostridium perfringens
(Sigma) for 1 h prior to the avidin/biotin block. Samples were analyzed on
a BD Facscalibur or BD LSR II (BD Biosciences), and a minimum of 5,000
events were recorded.
Viruses and viral titration. Stocks of A/New Caledonia/20/99 (H1N1)
and A/Wisconsin/67/05 (H3N2) viruses were kindly provided to San Raf-
faele Hospital (HSR) by Nadia Naffakh, Pasteur Institute, CNR Virus
Influenza (Paris, France), and were serially expanded 2 times in MDCK
cells prior to use.
To propagate IAV, monolayer-cultured MDCK cells were washed
twice with PBS and infected with A/NewCaledonia/20/99 (H1N1) or
A/Wisconsin/67/05 (H3N2) at a multiplicity of infection (MOI) of 0.001.
After virus adsorption for1hat35°C, the cells were washed twice and
incubated at 35°C with Dulbecco’s modified Eagle’s medium (DMEM)
without serum supplemented with tosylsulfonyl phenylalanyl chlorom-
ethyl ketone (TPCK)-treated trypsin (1 g/ml; Worthington Biomedical
Corporation, Lakewood, NJ). Supernatants were recovered 48 h postin-
fection.
LPAI H7N1 A/turkey/Italy/3675/1999 and H7N3 A/turkey/Italy/
2962/2003 viruses isolated during epidemics in Italy were grown in 9-day-
old embryonated SPF chicken eggs as described above. The sequences for
A/New Caledonia/20/99 (H1N1), A/Wisconsin/67/05 (H3N2), and
A/turkey/Italy/3675/1999 (H7N1) isolates were previously published.
For viral titration, plaque assays were performed as previously de-
scribed (28). Briefly, MDCK monolayer cells, plated in 24-well plates at
2.5 10
5
cells/well, were washed twice with DMEM without serum, and
serial dilutions of virus were adsorbed onto cells for 1 h. The cells were
covered with 2 minimal essential medium (MEM)-Avicel (FMC Biopo-
lymer, Philadelphia, PA) mixture supplemented with TPCK-treated tryp-
sin (1 g/ml). Crystal violet staining was performed 48 h postinfection,
and visible plaques were counted.
Virus replication kinetics in pancreatic-cell lines. Semiconfluent
monolayers of HPDE6 and hCM cells seeded on 24-well plates were
washed twice with PBS and then infected with human viruses (H1N1 and
H3N2) at an MOI of 0.001 and with avian viruses (H7N1 and H7N3) at an
M.O.I of 0.01 using 200 l of inoculum per well. The inoculum was
removed after1hofabsorption and replaced with 1 ml of serum-free
medium containing 0.05 g/ml TPCK-trypsin (Sigma). At 1, 24, 48, and
72 h postinfection, supernatants from three infected wells and one control
well were harvested, and viral titers were determined by virus isolation
using the 50% tissue culture infectious dose (TCID
50
) assay, as well as by
real-time RT-PCR detection of the matrix gene. All replication kinetics
experiments were repeated three times.
TCID
50
. Confluent monolayers of MDCK cells seeded onto 96-well
plates were washed twice in serum-free medium and inoculated with 50 l
of 10-fold serially diluted samples in serum-free MEM. After1hofad-
sorption, an additional 50 l of serum-free medium containing 2 g/ml
TPCK-trypsin was added to each well. Cytopathic-effect (CPE) scores
were determined after 3 days of incubation at 37°C by visual examination
of infected wells using a light microscope. The TCID
50
value was deter
-
mined using the method of Reed and Muench.
Growth assay in pancreatic islets. Islets were infected with H1N1 and
H3N2 influenza viruses by adding 4.8 10
2
or 4.8 10
3
PFU/well, re
-
spectively. Viral growth was performed with and without the addition of
TPCK-trypsin (Sigma) (1 g/ml). Uninfected islets were left as a negative
control. Samples were collected every 48 h from the day of infection (time
zero [t
0
]) until day 10 (t
5
, the fifth time point that occurred at 10 days
postinfection). Each sample was centrifuged at 150 g for 5 min. The
supernatant was collected and stored at 80°C for quantitative real-time
PCR, virus titration, and cytokine expression profiling. The pellet was
washed twice with PBS, stored at 80°C, and subsequently processed for
real-time PCR. All pellets and supernatants were tested by real-time PCR
in triplicate.
Detection of viral RNA from pancreatic tissue. The total RNAs from
pancreatic islet pellets and supernatants were isolated using the commer-
cial NucleoSpin RNA II kit (Macherey-Nagel) according to the manufac-
turer’s instructions. The RNAs were eluted in 60 l of elution buffer and
tested using one-step RRT-PCR for the influenza virus matrix gene to
evaluate viral growth.
A quadratic regression model (C
T
[threshold cycle] ⫽␤
0
⫹␤
1
TPCK-
trypsin ⫹␤
2
time ⫹␤
3
time
2
⫹␤
4
time TPCK-trypsin ⫹␤
5
time
2
TPCK-trypsin) for each virus and specimen was used to analyze the trend
of C
T
values over time. The influence of TPCK-trypsin presence and the
interaction between its presence and the time point were evaluated. The
regression model took into account the influence of the intragroup cor-
relation among repeated measurements for each observed time in the
confidence interval (CI) calculation. A residual postestimation analysis
was performed to verify the validity of the model.
One-step RRT-PCR. Quantitative real-time PCR targeting the con-
served M gene of type A influenza virus was applied according to the
protocol described above. To check the integrity of the isolated RNA, the
-actin gene was also amplified using the primers and probe previously
described (29). The reaction mixture consisted of 400 nM forward and
reverse primers (Primer-beta act intronic and Primer-beta act reverse,
respectively) and 200 nM fluorescent-label probe (5= Cy5 or 3= black hole
quencher 1 [BHQ1]). The amplification reaction was performed in a final
volume of 25 l using the commercial QuantiTect Multiplex RT-PCR kit
(Qiagen, Hilden, Germany). The PCR protocol was 20 min at 50°C and 15
min at 95°C, followed by 45 cycles at 94°C for 45 s and 55°C for 45 s.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 599
Western blot analysis. Cellular pellets were resuspended in lysis buf-
fer (50 mM Tris-HCl, pH 8, 1.0% SDS, 350 mM NaCl, 0.25% Triton-X,
proteases inhibitor cocktail) and then mixed and incubated on ice for 30
min. The suspension was sonicated three times for 5 min each time and
then centrifuged at maximum speed for 10 min. A Bradford test was
performed in order to calculate the total protein concentration for each
sample. Based on this calculation, the same amount of protein per sample
was loaded and electrophoresed in 12% polyacrylamide gels. Following
SDS-PAGE, the proteins were transferred from the gel onto immunoblot
polyvinylidene difluoride (PVD) membranes (Bio-Rad) by electro-
blotting. The membranes were saturated overnight at 4°C in 5% grade
blocker nonfat dried milk (Bio-Rad) in PBS and then incubated for1hat
room temperature under constant shaking in PBS containing 0.05%
Tween 20 (Sigma), 5% dried milk, and mouse monoclonal influenza A
virus nucleoprotein antibody (Abcam). -Actin antibody (Abcam) was
used as a loading control. After incubation with the primary antibody, the
membranes were exposed for1htoHRP-rabbit polyclonal secondary
antibody to mouse IgG (Abcam), followed by visualization of positive
bands by enhanced chemiluminescence (ECL) using Hyperfilm ECL
(Amersham Biosciences).
Visualization of viral growth in pancreatic cell lines. HPDE6 and
hCM cells were grown on slides to 80% confluence and infected with
either H1N1or H3N2 virus at an MOI of 0.1 with 0.05 mg/ml of TPCK-
trypsin. The cells were fixed and permeabilized at 0, 24, 48, and 72 h p.i.
with chilled acetone (80%). After blocking with PBS containing 1% BSA,
the cells were incubated for1hat37°C in a humidified chamber with
mouse monoclonal antibody to fluorescein isothiocyanate (FITC)-conju-
gated influenza A virus nucleoprotein (Abcam) in PBS containing 1%
BSA and 0.2% Evan’s Blue. The staining solution was decanted, and the
cells were washed three times. Nuclei of negative-control cells were
stained with DAPI (4=,6-diamidino-2-phenylindole) (Sigma) and then
washed with PBS and observed under UV light.
In situ visualization of viral RNA in pancreatic islets. To visualize
viral RNA localized within cells, purified human pancreatic islets were
harvested at 2, 5, and 7 days postinfection with human influenza viruses.
The islets were then incubated for 24 h in methanol-free 10% formalin,
deposited at the bottom of flat-bottom tubes, embedded in agar to immo-
bilize them, dehydrated, and finally embedded in paraffin. Islet samples
were sectioned at 4 m. For colocalization experiments, islets were enzy-
matically digested into single cells with a trypsin-like enzyme (12605-01;
TrypLE Express; Invitrogen, Carlsbad, CA) and cytocentrifuged onto
glass slides. In situ hybridization was performed using the Quantigene
ViewRNA technique, based on multiple oligonucleotide probes and
branched-DNA signal amplification technology, according to the manu-
facturer’s instructions (Affymetrix, Santa Clara, CA). The probe set used
was designed to hybridize the H1N1/A/New Caledonia/20/99 virus (Gen-
Bank sequence DQ508858.1). Due to sequence homology in the region
covered by the probes, the same set also recognized the H3N2 virus RNA,
as confirmed in pilot experiments. To identify cell types within islets, the
following Quantigene probes were used: insulin for beta cells (INS gene;
NCBI reference sequence NM_000207), alpha-amylase 1 for exocrine
cells (AMY1A gene; NCBI reference sequence NM_004038), and cytoker-
atin 19 for duct cells (KRT19 gene; NCBI reference sequence NM_002276).
Quantification of cells positive for each probe was performed within 8 ran-
domly chosen fields using the IN Cell Investigator software (GE Healthcare
United Kingdom Ltd.).
Determination of insulin secretion in infected islets. Aliquots of 100
islet equivalents (uninfected or infected with H1N1/A/New Caledonia/
20/99 and H3N2/A/Wisconsin/67/05) per column were loaded onto Sep-
hadex G-10 columns with medium at low glucose concentration (2 mM)
and preincubated at 37°C for 1 h. After preincubation, the islets were
exposed to sequential 1-h incubations at low (2 mM) and high (20 mM)
glucose concentrations. Supernatants were collected with protease inhib-
itor cocktail (Roche Biochemicals, Indianapolis, IN) and stored at 80°C
at the end of each incubation. The insulin content was determined with an
insulin enzyme-linked immunoassay kit (Mercodia AB, Uppsala, Swe-
den) following manufacturer’s instructions. Insulin secretion indices
were calculated as the ratio between the insulin concentration at the end of
high-glucose incubation and the insulin concentration at the end of low-
glucose incubation.
Cytokine expression profile. The capability of H1N1 and H3N2 vi-
ruses to induce cytokine expression in human pancreatic islets was mea-
sured using multiplex bead-based assays based on xMAP technology (Bio-
Plex; Bio-Rad Laboratories, Hercules, CA). The parallel wells of
pancreatic islets were infected with viruses or were mock infected. The
culture medium supernatant was collected before infection and 2, 4, 6, 8,
and 10 days postinfection and assayed for 48 cytokines. Selected cytokines,
the limits of detection, and the coefficients of variability (intra-assay per-
cent coefficient of variation [%CV] and interassay %CV) of the cytokine/
chemokine are reported in Table S5 in the supplemental material.
Evaluation of cell death following infection (LIVE/DEAD assay).
The viability of islet cells after infection with human viruses was measured
using the LIVE/DEAD cell assay kit (L-3224; Molecular Probes, Inc., Le-
iden, The Netherlands). The assay is based on the simultaneous determi-
nation of live and dead cells with two fluorescent probes. Live cells are
stained green by calcein due to their esterase activity, and nuclei of dead
cells are stained red by ethidium homodimer 1. Islets harvested after 5
days of culture were further enzymatically digested into single cells with
trypsin-like enzyme (12605-01; TrypLE Express; Invitrogen, Carlsbad,
CA). According to the manufacturer’s instructions, single cells were incu-
bated with the labeling solution for 30 min at room temperature in the
dark, cytocentrifuged onto glass slides, and assessed with a Carl Zeiss
Axiovert 135TV fluorescence microscope. Analysis of dead cells was per-
formed on cytospin preparations using IN Cell Investigator software (GE
Healthcare United Kingdom Ltd.). Positive cells in each category were
quantified with 10 systematically random fields.
Statistical analysis. Data were generally expressed as mean stan-
dard deviation or median (minimum-maximum). Differences between
parameters were evaluated using Student’s t test when parameters were
normally distributed and a Mann-Whitney U test when parameters were
not normally distributed. Kaplan-Meier and/or Cox regression analysis
was used to analyze the incidence of events during the time. A P value of
less than 0.05 was considered an indicator of statistical significance. Anal-
ysis of data was done using the SPSS statistical package for Windows (SPSS
Inc., Chicago, IL).
Nucleotide sequence accession numbers. The sequences of all eight
influenza virus genome segments belonging to H7N3 A/turkey/Italy/
2962/2003 were submitted to GenBank and assigned accession numbers
JX515660, JX515661, JX515662 , JX515663, JX515664, JX515665,
JX515666, and DQ090062.
RESULTS
In vivo experiment. (i) Clinical disease. Turkeys from both
H7N1-challenged (A) and H7N3-challenged (B) groups showed
clinical signs typical of LPAI infection, such as conjunctivitis, si-
nusitis, diarrhea, ruffled feathers, and depression on day 2 p.i.
Mild symptoms regressed by day 20 p.i. Only two subjects from
group A showed sinusitis until day 30 p.i. Mortality rates were low
in both groups: one subject in group A died on day 8 p.i., and one
subject in group B died on day 19 p.i.
(ii) Detection of viral RNA. Viral RNA was detected from the
tracheal swabs collected from 17/20 birds infected with H7N1 vi-
rus and 19/20 birds infected with H7N3 virus on day 2 p.i. and in
all animals on day 3 p.i. Viral RNA was also detected from the
blood of two birds of group A H7N1 and four birds of group B
H7N3 on day 3 p.i. and from the pancreases and lungs collected on
days 4 and 7 p.i. (see Table S4 in the supplemental material). No
viral RNA was detected from the uninfected controls.
(iii) Biochemical analyses. In blood samples collected intra
Capua et al.
600 jvi.asm.org Journal of Virology
vitam to reveal metabolic alterations, a significant increase in
plasma lipase levels (10 to 100 times the values of the control
birds) was evident in H7N1-challenged (12/20) and H7N3-chal-
lenged (10/20) turkeys between days 3 and 9 p.i. (Fig. 1A), while
no uninfected controls showed modification of lipase levels (20/
20; P 0.001; Pearson chi-squared test). A clear trend between the
presence of viral RNA in blood at day 3 and the increase in lipase
was evident in infected animals (hazard ratio, 2.51; 95% confi-
dence interval, 0.92 to 6.81; P 0.07). Lipase levels within the
normal range were rapidly reestablished in all cases, and it was
decided to no longer evaluate this parameter on day 23 (see Tables
S1 and S2 in the supplemental material). After day 9 p.i., 5 birds in
group A and 5 birds in group B developed hyperglycemia
(Fig. 1B). Of these, two birds maintained the hyperglycemic status
throughout the experiment, while in all the other birds, the levels
of blood glucose returned to levels similar to those of controls (see
Table S3 in the supplemental material). A clear association be-
tween the increase in lipase between days 3 and 9 p.i. and the
development of hyperglycemia after day 9 p.i. was evident. In fact,
hyperglycemia was present only in the birds that developed high
lipase values postinfection and never appeared in birds with nor-
mal lipase levels (10/22 and 0/18, respectively; P 0.001), with a
median time between development of hyperlipasemia and hyper-
glycemia of 4.5 days (minimum-maximum, 3 and 7).
(iv) Histopathology and immunohistochemistry. None of
the control turkeys showed significant histological changes or pos-
itive immunohistological reactions to avian influenza virus (AIV)
(Fig. 2A). In all infected birds, histopathologic lesions were evi-
dent, although markedly different in samples collected at different
times postinfection. At early stages (days 4 to 8 p.i.), acute pancre-
atitis with necrotic acinar cells and massive inflammatory infiltra-
tion composed of macrophages, heterophils, lymphocytes, and
plasma cells dominated over areas of healthy/uninvolved/spared
tissue (Fig. 2B). From day 8 p.i., these necrotic inflammatory le-
sions/necrotic inflammatory areas were gradually replaced by
ductule hyperplasia and lymphocytic infiltration with a mild de-
gree of fibroplasia. At later stages (day 17 p.i), extensive fibrosis
with lymphoid nodules replaced pancreatic parenchyma, and dis-
ruption of the normal architecture of the organ was evident (Fig.
2C). Variable numbers of necrotic acinar cells were observed dur-
ing the entire experimental period. Obstructive ductal lesions
were not seen in any case or stage.
By immunohistochemistry, degenerating and necrotic acinar
cells showed specific reaction to virus nucleoprotein antigen anti-
body during the experimental period (Fig. 2D). Some of the vas-
cular endothelial cells also showed a positive reaction, as well as
occasional ductal epithelial cells. In uninfected controls, the insu-
lin-positive tissues of the pancreas were scattered singly or in small
groups of islets of various shapes and sizes in the interstitium of
the exocrine part (Fig. 3A). At day 8 p.i., the normal structure of
FIG 1 Biochemical analysis. Kaplan-Meier analysis for the appearance of hyperlipasemia (A) and hyperglycemia (B) (plasma glucose, 27.78 mmol/liter)
among mock-, H7N1-, and H7N3-infected turkeys. Differences were tested using the log rank statistic. The bar graphs show the frequency of events in relation
to hyperlipasemia, hyperglycemia, and presence of viremia.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 601
islets was partially destroyed and the number of islet cells was
reduced. The remaining islets were smaller and distorted, with
irregular outlines or dilated intraislet capillaries; the number of
cells staining for insulin was also reduced, and these cells pre-
sented enlarged cytoplasm and sometimes appeared to have gran-
ular degeneration and even necrosis. Fibrous bands appeared in-
side the islet, with islet fragmentation and dislocation of small and
large clusters of endocrine cells (Fig. 3B). At day 17 p.i., separate
large clusters of endocrine insulin-positive cells were evident em-
bedded in or close to the extensive fibrosis that replaced the acinar
component (Fig. 3C). Beyond day 17 p.i., groups of very large
(200 m in diameter), usually irregular islet-like areas of mainly
insulin immunoreactivity were clearly present scattered in exten-
sive acinar fibrosis (Fig. 3D and E).
In vitro experiment. (i) Susceptibility of human pancreatic
cell lines to human influenza A viruses. The susceptibility of en-
docrine (hCM insulinoma) and ductal (HPDE6) cell lines to
H1N1/A/New Caledonia/20/99 and H3N2/A/Wisconsin/67/05
infections was investigated.
(ii) Receptor distribution. Our investigation using lectin
staining for receptor detection on both the hCM and HPDE6 cell
lines revealed high levels of alpha-2,6 sialic acid-linked sialic acid
molecules (required by human-tropic viruses), as well as alpha-
2,3-linked residues (used by avian-tropic viruses). The mean peak
intensities of hCM incubated with MAA lectin II (alpha-2,3 spe-
cific) and SNA lectin (alpha-2,6 specific) were nearly identical at
approximately 2.6 10
4
for both receptors. HPDE6 also had
high-level expression of both receptor types, with 3.7 10
4
for
SNA and 1.6 10
4
for MAA. MDCK cells were also included as a
positive-control line for both receptor types, as these cells are
widely used for the isolation of viruses of human and avian origin.
Fluorescence-activated cell sorter (FACS) analysis showed that
MDCK cells expressed levels of alpha-2,3 receptors similar to
those of HPDE6 cells, with the mean peak intensity near 1.8 10
4
,
while alpha-2,6 expression was equal to that of hCM cells, with
mean fluorescence at 2.5 10
4
. Therefore, both pancreatic cell
lines can be said to express sialic acid receptors at levels compara-
ble to those of MDCK cells, and in the case of hCM cells, expres-
sion of the human-virus receptors was even higher (see Fig. S1 in
the supplemental material). Pretreatment of all cells with 1 U/ml
of NA from Clostridium perfringens resulted in decreased fluores-
cence for both lectin types, confirming specificity (data not
shown).
(iii) Virus replication kinetics in pancreatic cell lines. hCM
and HPDE6 cells were infected with human influenza H1N1 and
H3N2 viruses at an MOI of 0.001. Visual examination of the in-
FIG 2 Histopathology and immunohistochemistry. (A) Turkey pancreas. Normal tissue is shown. Acinar cells containing zymogen granules in their cytoplasm
are evident, associated with two nests of normal islet cells and a ductal structure (H&E). (B) Turkey pancreas. Shown is diffuse and severe necrosis of acinar cells
(arrows) with severe inflammatory infiltrate (asterisks) (H&E). (C) Turkey pancreas. Most of the pancreas has been replaced by foci of fibrous connective tissue
and lymphoid nodules, with some ductular proliferation. (D) Turkey pancreas. Shown is immunohistochemistry for avian influenza virus NP, with an areaof
necrosis, and positive nuclei and cytoplasm in both acinar and ductal cells.
Capua et al.
602 jvi.asm.org Journal of Virology
fected cells by light microscopy revealed no cytopathic effect at
any time point postinfection on hCM or HPDE6 cells. TCID
50
results revealed a continued increase in viral titers in HPDE6 cells
over the 72-h course, though the H1N1 viral titers were only
slightly higher at 72 h than at 48 h postinfection. In contrast, viral
titers reached in hCM cells remained quite similar from 48 to 72 h
postinfection for both H1N1 and H3N2 isolates (Fig. 4A and B).
An examination of viral-RNA replication by qRRT-PCR showed a
continued increase in viral replication up to 72 h postinfection in
both cell lines and for both human viruses tested (Fig. 4C and D).
Despite the higher MOI used to perform the infections (MOI
0.01), both H7N1 and H7N3 avian influenza viruses showed lower
levels of replication in both pancreatic cell lines than the human
viruses (see Fig. S2 in the supplemental material), with a trend
characterized by steady levels of virus RNA up to 48 h p.i. and a
decrease for both cell lines at 72 h p.i. Based on the RRT-PCR
results, hCM cells appeared to be more sensitive to avian viruses,
since the total amount of “M gene” RNA on average was 2 log units
higher than in HPDE6 cells (see Fig. S2A and B in the supplemen-
tal material). This was also confirmed by TCID
50
results (see Fig.
S2C and D in the supplemental material), which for both viruses
reached higher titers in hCM. In the latter, however, the H7N1
strain exhibited a higher replication efficacy in hCM than in
H7N3. This result is not reflected in the RRT-PCR results, in
which comparable amounts of viral RNA were detected for both
viruses.
No significant differences in viral replication between the two
avian viruses were observed in HPDE6 cells.
(iv) Western blot analysis for detection of virus nucleopro-
tein. Results of H1N1 and H3N2 influenza virus nucleoprotein
expression in the hCM and HPDE6 cell lines are reported in Fig.
5a, b, e, and f. No differences relating to the viral strain or the cell
type were shown in the trend of NP expression. As expected, in-
fluenza virus nucleoprotein was not observed at t
0
(before infec
-
tion), while it was detected at 24 (t
24
), 48 (t
48
), and 72 (t
72
) hours
p.i. for both viruses in hCM and HPDE6 cells. Comparing the
bands obtained from samples at t
24
to those obtained at t
48
and t
72
,
an increase in the NP expression was observable. On the other
hand, the amount of -actin, used as loading control, was at the
same level in all the samples tested (Fig. 5c, d, g, and h).
(v) Immunofluorescence targeting the NP protein. Human
influenza virus replication was also detected by a fluorescent sig-
nal derived from FITC conjugate in hCM cells at 24 h p.i. (see Fig.
S3A and B in the supplemental material) for both viruses tested,
which increased at 48 and 72 h p.i. No differences were observed
between the viral strains tested. The fluorescent signal for both
viruses was also observed at 24 h postinfection in HPDE6 cells (see
Fig. S3C and D in the supplemental material). In this case also, the
number of cells marked continued to increase at 48 and 72 h p.i.,
demonstrating the enhancement of the nucleoprotein expression
over time (data not shown).
(vi) Susceptibility of human pancreatic islets to human influ-
enza A viruses. The regression model indicated that the C
T
values
for both viruses, in the presence or absence of TPCK-trypsin,
tested both in pellets and in supernatant specimens, decreased
significantly over time (P 0.05) (Fig. 6). The statistical analysis
showed that the virus titer increased over time independently of
the virus subtype and type of sample (pellet or supernatant). In-
terestingly, C
T
values for the viruses grown with TPCK-trypsin
decreased significantly more than those obtained without the ex-
ogenous proteases (P 0.01) only for H1N1 pellets and superna-
tant samples (Fig. 6A and C). TPCK-trypsin seemed to enhance
H3N2 virus growth, but the difference did not reach statistical
significance (P 0.10) (Fig. 6B and D). The residual postestima-
tion analysis indicated that the model used was appropriate (data
not shown). Analysis of data was done using STATA 12.0 statisti-
cal software.
In situ hybridization was performed to visualize viral RNA lo-
FIG 3 Immunohistochemistry for insulin. Shown is turkey pancreas with representative islet structures before and after H7N3 infection at different time points.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 603
calized within islet cells. The results clearly demonstrate the pres-
ence of viral RNA after both H1N1 and H3N2 virus infection (Fig.
7A). Since human islet primary cultures contain both endocrine
and exocrine cells, a fluorescence-based multiplex in situ hybrid-
ization strategy was applied to determine which and how many
cells were infected in the islets. For this purpose, distinctly labeled
probes were combined to analyze viral RNA and insulin, amylase,
or cytokeratin in 19 transcripts simultaneously, and after hybrid-
ization, human islets were disaggregated and cell positivity was
quantified. Five days after infection, 0%, 10.8%, and 4.3% of total
cells were positive for viral RNA in mock-, H1N1-, and H3N2-
infected islets, respectively (P 0.001) (Fig. 7B). Of the H1N1-
positive cells, 49% 27% stained positive for insulin, 26% 16%
for amylase, and 1.6 2.4% for CK19, and 25% 21% were
negative for the tested transcripts. Of the H3N2-positive cells,
40% 23% stained positive for insulin, 20% 20% for amylase,
and 2.3 5% for CK19, and 41% 45% were negative for the
tested transcripts (Fig. 7C). On the other hand, of the insulin-
positive cells, 14% 10% and 8% 8% were positive for viral
RNA 5 days after H1N1 and H3N2 infection, respectively (P
0.023). Of the amylase-positive cells, 27% 9% and 9% 6%
were positive for viral RNA after H1N1 and H3N2 infection, re-
FIG 5 Western blot analysis for the detection of viral nucleoprotein in pan-
creatic cell lines. Western blot analysis of H1N1 (a and b) and H3N2 (e and f)
influenza virus NP (56 kDa) in hCM and HPDE6 cells. (c, d, g, and h) Samples
were collected before infection (t
0
) and 24 (t
24
), 48 (t
48
), and 72 (t
72
) hours
postinfection. -Actin (42 kDa) was used as a loading control in order to
ensure that the same amount of protein was tested for each sample.
FIG 4 Human influenza virus replication kinetics in pancreatic cell lines. Shown is the replication kinetics of A/New Caledonia/20/99 (H1N1) and A/Wisconsin/
67/2005 (H3N2) in hCM and HPDE6 cells. hCM and HPDE6 cells were infected with each virus at an MOI of 0.001, and at 24, 48, and 72 h postinfection,
supernatants from three infected and one mock-infected control well were harvested for virus isolation and qRRT-PCR. (A) Virus isolation results of H1N1 in
hCM and HPDE6 cells. (B) Virus isolation results for H3N2 in hCM and HPDE6 cells. (C) qRRT-PCR results for H1N1 in hCM and HPDE6 cells. (D) qRRT-PCR
results for H3N2 in hCM and HPDE6 cells. All results represent means plus standard deviations for three independent experiments.
Capua et al.
604 jvi.asm.org Journal of Virology
spectively (P 0.001). Of the CK19-positive cells, 3% 4% and
1.3% 3% were positive for viral RNA after H1N1 and H3N2
infection, respectively (P 0.36) (Fig. 8).
(vii) Modulation of survival, insulin secretion, and innate
immunity in human pancreatic islets infected with human in-
fluenza A viruses in vitro. Visual examination of the infected
islets by light microscopy and LIVE/DEAD assay revealed no sig-
nificant cytopathic effect at any time point postinfection (days 0 to
7). Five days after infection, uninfected cells showed an overall
mortality of 3.26%, H3N2-infected cells 5.21%, and H1N1-in-
fected cells 7.38% (P, nonsignificant versus mock-infected cells)
(see Fig. S4A in the supplemental material). Moreover, exposure
of islets to either H1N1 or H3N2 virus did not affect their ability to
respond to high glucose, as tested in a static perfusion system (see
Fig. S4B in the supplemental material).
The capability of H1N1 and H3N2 viruses to induce cytokine/
chemokine expression in human pancreatic islets was shown by
detectable expression of all but three (interleukin 1b [IL-1b], IL-5,
and IL-7) of the cytokines tested. In mock-infected cells, the high-
est concentrations were detected for CCL2/MCP1 (maximum,
25,558 pg/ml, day 4), ICAM-1 (maximum, 14,063, day 10),
CXCL8/IL-8 (maximum, 11,635 pg/ml, day 10); IL-6 (8,452 pg/
ml, day 4), CXCL1/GRO- (maximum, 8,581 pg/ml, day 4),
VCAM-1 (maximum, 5,566 pg/ml, day 6), VEGF (maximum,
3,225 pg/ml, day 10), SCGF-b (maximum, 1,427 pg/ml, day 6),
hepatocyte growth factor (HGF) (maximum, 1,195 pg/ml, day 6).
MIF (maximum, 806 pg/ml, day 6), granulocyte colony-stimulat-
ing factor (G-CSF) (maximum, 794 pg/ml day 6), CXCL9/MIG
(maximum, 448 pg/ml, day 6), granulocyte-macrophage colony-
stimulating factor (GM-CSF) (maximum, 280 pg/ml, day 4), IL-
2Ra (maximum, 230 pg/ml, day 6), IL-12p40 (maximum, 215
pg/ml, day 6), macrophage colony-stimulating factor (M-CSF)
(maximum, 212 pg/ml, day 10), LIF (maximum, 185 pg/ml, day
6), and CXCL4/SDF1 (maximum, 121 pg/ml, day 6) showed lower
but consistent expression. CXCL10/IP-10, PDGF-BB, IL-1Ra, IL-
12p70, CCL11/eotaxin, FGFb, CCL5/RANTES, CCL4/MIP-1,
CCL7/MCP-3, IL-3, IL-16, SCF, TRAIL, alpha 2 interferon (IFN-
2), IFN-, and CCL27/CTAK showed low but consistent expres-
sion (maximum, between 10 and 100 pg/ml). Very low (maxi-
mum, 10 pg/ml) but detectable expression was present for IL-2,
IL-4, IL-9, IL-10, IL-13, IL-15, CCL3/MIP-1, tumor necrosis fac-
tor alpha (TNF-), IL-17, IL-18, IL-1a, -NGF, and TNF-. Two
inflammatory cytokines (IL-6 and TNF-) and six inflammatory
chemokines (CXCL8/IL-8, CXCL1/GRO-, CXCL9/MIG,
CXCL10/IP-10, CCL5/RANTES, and CCL4/MIP-1) showed
over 5-fold increase in influenza virus-infected cell supernatants
compared to mock-infected controls (Fig. 9A). Among these, the
IFN--inducible chemokines CXCL9/MIG and CXCL10/IP-10
FIG 6 Viral-RNA detection by RRT-PCR of the M gene in human pancreatic islets. Shown are two-way quadratic prediction plots with CIs for RRT-real-time
C
T
values (ct) obtained from H1N1 (A and C) and H3N2 (B and D) (4.8 10
3
PFU/well) pancreatic islet infection. For each virus, the C
T
trends in pancreatic
islet pellets and supernatants from the day of infection (t
0
) until day 10 (t
5
) in the presence (left) or absence (middle) of TPCK-trypsin and as an average of the
previous two conditions (right) are represented.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 605
showed the strongest response to H1N1 or H3N2 infection (over
100-fold increase). Both peaked 6 to 8 days p.i. and showed a
stronger response to higher doses of the viruses (Fig. 9B).
DISCUSSION
The objective of our work was to assess IAV replication in pancre-
atic cells and to evaluate its consequences both at the cellular level
in vitro and at the tissue level in vivo. In fact, despite previous
reports on a potential role of IAV in pancreatic damage in both
animals and humans (24, 1923), to date, there has been no
attempt to establish whether IAVs are able to grow in pancreatic
cells, and no data are available on the consequences of IAV repli-
cation in the pancreas. Our studies have generated novel in vitro
data indicating that human influenza A viruses are able to grow in
human pancreatic primary cells and cell lines. The addition of
exogenous trypsin appears to enhance but not to be essential for
viral replication in human pancreatic islets. In vitro studies are
corroborated and become of greater significance if combined with
animal studies, in which two nonsystemic strains of IAVs were
able to colonize the pancreases of experimentally infected young
turkeys and to cause metabolic consequences reflecting endocrine
and exocrine damage.
Colonization of the pancreas by IAV has been reported follow-
ing a number of natural and experimental infections of animals,
primarily in birds undergoing both systemic and nonsystemic in-
fection. To date, there is no direct evidence of influenza virus
infection of the pancreas in humans; however, several reports
yield indirect evidence of its occurrence. Here, we have for the first
time demonstrated that two nonsystemic avian influenza viruses
cause severe pancreatitis resulting in a dysmetabolic condition
comparable with diabetes as it occurs in birds. Literature is avail-
able on the clinical implications of endocrine and exocrine dys-
functions of the pancreas in birds, including poultry. With refer-
ence to endocrine function, several studies indicate that with a
total pancreatectomy, birds suffer severe hypoglycemic crisis lead-
ing to death (30). If a residual portion of the pancreas as small as
1% of the pancreatic mass is left in situ, a transient (or reversible)
hyperglycemic condition is observed in granivorous birds, in
which normal glycemia is reestablished within a couple of weeks
(31, 32). This indicates that the pancreatic tissue of birds has sig-
nificant compensatory potential and is also influenced by the fact
that there is evidence of the presence of some endocrine tissue able
to secrete insulin outside the pancreas (33). Insulin is the domi-
nant hormone in well-fed birds, while glucagon is the dominant
hormone in fasting birds. In our experiment, which was carried
FIG 7 Viral-RNA detection by in situ hybridization in human pancreatic islets. Islets were infected with H1N1 and H3N2 by adding 100 l of viral suspension
containing a viral dilution of 4.8 10
3
PFU/well. Mock-infected islets were left as a negative control. (A) Two days after infection, the presence of the viral-RNA
molecules was detected on cytoembedded pancreatic islets upon addition of the Fast Red alkaline phosphatase substrate due to the formation of a colored
precipitate. Bound viral mRNA was then visualized using either a standard bright-field or a fluorescence microscope (magnification, 40). Arrows, viral-mRNA-
positive cells. (B and C) Five days after infection, multiplex fluorescence-based in situ hybridization was performed, and after disaggregation, islet cells were
cytocentrifuged onto glass slides. Viral-RNA-, insulin-, amylase-, and CK19-positive cells were assessed with a Carl Zeiss Axiovert 135TV fluorescence micro-
scope. Quantification was performed using IN Cell Investigator software. Each dot represents the percentage of positive cells quantified on one systematically
random field. Results from two experiments are shown. A Mann-Whitney U test was used for statistical analysis.
Capua et al.
606 jvi.asm.org Journal of Virology
out with food ad libitum, damage to the endocrine component of
the pancreas would most likely manifest itself as hyperglycemia.
With reference to the exocrine function, pancreatitis in birds is
characterized by malaise, reluctance to feed, enteritis, and depres-
sion. Intra vitam investigations have been based on increased he-
matic lipase concentration (32). In our study, pancreatitis was
evaluated both in vivo, by measuring the lipase concentration in
the blood, and postmortem, by histopathologic examination of
pancreases collected at different time points. As it occurs in mam-
mals, pancreatic damage results in a rapid increase of the hematic
lipase levels that is transient, and values returned to normal by day
15 p.i. Interestingly, the birds that had shown increased lipase
levels in the blood and thus supposedly the most severe pancreatic
damage exhibited high blood glucose levels in subsequent days,
which in only a few cases persisted until the termination of the
experiment. This is in keeping with the clinical and metabolic
presentation of diabetes in birds. The histological investigations
clearly demonstrated viral replication in the exocrine portion of
the pancreas, resulting in fibrosis and disruption of the architec-
ture of the organ. While it is clear that both isolates under study
replicated extensively in the acinar component of the pancreas, we
were unable to determine whether viral replication also occurred
in the islets. In any case, it is reasonable to believe that viral infec-
tion resulted in severe acute pancreatitis, which impaired both the
endocrine and exocrine functions.
Current knowledge of influenza virus replication indicates that
influenza viruses that do not exhibit a multibasic cleavage site on
the hemagglutinin (HA) protein do not become systemic. How-
ever, in our in vivo experiments, the virus reached the pancreas,
and we detected viral RNA on day 3 p.i. in the blood of 2/20 (group
A, H7N1) and 4/20 (group B, H7N3) infected turkeys. It is there-
fore possible that following replication in target organs, such as
the lung and the gut, in some individuals, a small amount of virus
reaches the bloodstream and thus the pancreas. Although the de-
tected C
T
values indicate low levels of viral RNA, this often re
-
sulted in the development of pancreatitis (detected in vivo by hy-
perlipasemia). This, in turn, in our experimental model, has
resulted in a hyperglycemic condition, in keeping with the presen-
tation of diabetes in granivorous birds.
The results of our in vitro experiments show that all IAVs
tested, both of avian (H7N1A/turkey/Italy/3675/1999 and H7N3
A/turkey/Italy/2962/2003) and of human (H1N1 Caledonia/
20/99 and H3N2 A/Wisconsin/67/2005) origin, are able to grow in
established pancreatic cell lines and that human viruses also grow
in pancreatic islets. Viral replication occurs in cells of both endo-
crine and exocrine origin. Our investigations also show that both
alpha-2,3 and alpha-2,6 receptors are present in pancreatic cells,
indicating that both human and avian influenza viruses could find
suitable receptors in the organ. The human viruses used in this
study did not induce significant mortality of islet cells, and insulin
secretion did not appear to be affected by infection in this system.
Whether other strains, such as H1N1pdm or viruses of high
FIG 8 Virus RNA and insulin/amylase/CK19 localization. Islets were infected with H1N1 and H3N2 by adding 100 l of viral suspension containing a viral
dilution of 4.8 10
3
PFU/well. Five days after infection, multiplex fluorescence-based in situ hybridization was performed using the Quantigene ViewRNA
technique, based on multiple oligonucleotide probes and branched-DNA signal amplification technology, according to the manufacturer’s instructions. (Left)
The red signal corresponds to the presence of influenza virus RNA, and the green signal corresponds to the presence of insulin, amylase, or CK18 transcripts
(magnification, 63). White arrows, double-positive cells. (Right) Viral-RNA-, insulin-, amylase-, and CK19-positive cells were assessed with a Carl Zeiss
Axiovert 135TV fluorescence microscope. Quantification was performed using IN Cell Investigator software. Each dot represents the percentage of positive cells
quantified on one systematically random field. Results from two experiments are shown.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 607
pathogenicity, may cause more significant damage remains to be
established. On the other hand, it was clear from the cytokine
expression profile that IAV infection is able to induce a strong
proinflammatory response in human pancreatic islets. The IFN-
-inducible chemokines MIG/CXCL9 and IP-10/CXCL10
showed the highest increase after infection. Large amounts of
RANTES/CCL5, MIP1b/CCL4, Gro/CXCL1, IL-8/CXCL8,
TNF-, and IL-6 were also released.
Of interest, many of these factors are described as key media-
tors in the pathogenesis of type 1 diabetes (34). Recently IP10/
CXCL10 was identified as the dominant chemokine expressed in
vivo in the islet environment of prediabetic animals and type 1
diabetic patients, whereas RANTES/CCL5 and MIG/CXCL9 pro-
teins were present at lower levels in the islets of both species (35).
The chemokine IP-10/CXCL10 attracts monocytes, T lympho-
cytes, and natural killer (NK) cells, and islet-specific expression of
CXCL10 in a mouse model of autoimmune diabetes caused by
viruses (rat insulin promoter [RIP]-lymphocytic choriomeningi-
tis virus [LCMV]) accelerates autoimmunity by enhancing the
migration of antigen-specific lymphocytes (36). This is in line
with other findings, in which neutralization of IP-10/CXCL10
(37) or its receptor (CXCR3) (38) prevents autoimmune disease
in the same mouse model (RIP-LCMV). Studies in nonobese di-
abetic (NOD) mice have demonstrated elevated expression of IP-
10/CXCL10 mRNA and/or protein in pancreatic islets during the
prediabetic stage (39). Increased levels of MIP1b/CCL4 and IP-10/
CXCL10 are present in the sera of patients who have recently been
diagnosed as having type 1 diabetes mellitus (T1D) (4042).
We speculate that if influenza virus finds its way to the pan-
creas, either through viremia, as detected in human patients (43
45), or through reflux from the gut through the pancreatic duct,
the virus would find a permissive environment. Here, the virus
would find appropriate cell receptors and susceptible cells belong-
ing to both the endocrine and exocrine components of the organ.
Viral replication would result in cell damage due to the activation
of a cytokine storm similar to the one associated with various
conditions linked to diabetes. Thus, we believe that influenza virus
infections should be investigated as potential agents of pancreatic
damage as reported previously (19, 20), resulting in acute pancre-
atitis and the onset of type 1 diabetes (2123). The rapid world-
wide increase in the incidence of T1D suggests a major role for
environmental factors in the etiology. According to cross-sec-
tional and prospective studies on T1D patients and/or prediabetic
individuals, virus infections may be one of these. Possible viral
involvement in the etiology of T1D has been suggested by several
authors. Viruses associated with human T1D include enterovi-
ruses, such as coxsackievirus B (CVB) (4648), but also measles
virus, congenital rubella virus, mumps virus, cytomegalovirus
(4951), and influenza B virus (52). Rotavirus and reovirus have
additionally been shown to be diabetogenic in mice (49, 53).
In conclusion, our findings suggest that the pancreas could be
a potential target of IAV infection, and thus, we propose that IAV
could play a role in the etiopathogenesis of pancreatitis and dia-
betes in humans and perhaps in other mammals. Clearly, a thor-
ough analysis of existing clinical data and prospective collection of
human samples will be needed to fully address this issue. In the
FIG 9 Cytokine/chemokine expression profile modification induced by human influenza A virus infection. Islets were infected with H1N1 and H3N2 by adding
100 l of viral suspension containing two viral dilutions of 4.8 10
3
or 4.8 10
2
PFU/well. Mock-infected islets were left as a negative control. Samples were
collected every 48 h from the day of infection (t
0
) until day 10 (t
10
). The supernatant was collected and assayed for 50 cytokines. (A) Virus-induced modification
in the islet cytokine/chemokine profile. The data are expressed as the maximum fold increase for each factor detected during culture in relation to mock-infected
islets (n 2). Dashed line, 5-fold-increase threshold. The error bars indicate standard deviations. Red items are factors with a 5-fold increased threshold. (B)
IFN--inducible chemokine CXCL9/MIG and CXCL10/IP-10 concentrations during 10 days of culture in the presence () or absence () of H1N1 and H3N2.
Capua et al.
608 jvi.asm.org Journal of Virology
meantime, a mammalian preclinical model should be tested to
evaluate the role of IAV infection directly or in the context of other
immune-mediated conditions in etiopathogenesis of islet damage
and beta cell autoimmunity leading to T1D. Similarly, IAV infec-
tion should be ruled out when clinical cases with symptoms of
endocrine or exocrine pancreatic damage of unknown etiology
occur.
ACKNOWLEDGMENTS
We acknowledge H. L. Shivaprasad (University of California, Davis) for
his support with histopathology and immunohistochemistry and Marzia
Mancin (IZSVe, Legnaro, Padova, Italy) for her support with statistical
analysis of data.
Preliminary studies were funded through the EU FP6 project “Train-
ing and Technology Transfer of Avian Influenza Diagnostics and Disease
Management Skills” (FLUTRAIN) (project no. 044212).
REFERENCES
1. Capua I, Alexander DJ. 2009. Avian influenza and Newcastle Disease, a
field and laboratory manual. Springer, Berlin, Germany.
2. Harder TC, Vahlenkamp TW. 2010. Influenza virus infections in dogs
and cats. Vet. Immunol. Immunopathol. 134:54 60.
3. Kuiken T, Rimmelzwaan G, van Riel D, van Amerongen G, Baars M,
Fouchier R, Osterhaus A. 2004. Avian H5N1 influenza in cats. Science
306:241.
4. Rimmelzwaan GF, van Riel D, Baars M, Bestebroer TM, van Ameron-
gen G, Fouchier RA, Osterhaus AD, Kuiken T. 2006. Influenza A virus
(H5N1) infection in cats causes systemic disease with potential novel
routes of virus spread within and between hosts. Am. J. Pathol. 168:176
183.
5. Abolnik C, Londt BZ, Manvell RJ, Shell W, Banks J, Gerdes GH, Akol
G, Brown IH. 2009. Characterisation of a highly pathogenic influenza A
virus of subtype H5N2 isolated from ostriches in South Africa in 2004.
Influenza Other Respi. Viruses 3:63– 68.
6. Bertran K, Perez-Ramirez E, Busquets N, Dolz R, Ramis A, Darji A,
Abad FX, Valle R, Chaves A, Vergara-Alert J, Barral M, Hofle U, Majo
N. 2011. Pathogenesis and transmissibility of highly (H7N1) and low
(H7N9) pathogenic avian influenza virus infection in red-legged partridge
(Alectoris rufa). Vet. Res. 42:24.
7. Tanimura N, Tsukamoto K, Okamatsu M, Mase M, Imada T, Naka-
mura K, Kubo M, Yamaguchi S, Irishio W, Hayashi M, Nakai T,
Yamauchi A, Nishimura M, Imai K. 2006. Pathology of fatal highly
pathogenic H5N1 avian influenza virus infection in large-billed crows
(Corvus macrorhynchos) during the 2004 outbreak in Japan. Vet. Pathol.
43:500–509.
8. Teifke JP, Klopfleisch R, Globig A, Starick E, Hoffmann B, Wolf PU,
Beer M, Mettenleiter TC, Harder TC. 2007. Pathology of natural infec-
tions by H5N1 highly pathogenic avian influenza virus in mute (Cygnus
olor) and whooper (Cygnus cygnus) swans. Vet. Pathol. 44:137–143.
9. Condobery PK, Slemons RD. 1992. Biological properties of waterfowl-
origin type A influenza viruses in chickens. Avian Dis. 36:17–23.
10. Morales AC, Jr, Hilt DA, Williams SM, Pantin-Jackwood MJ, Suarez
DL, Spackman E, Stallknecht DE, Jackwood MW. 2009. Biologic char-
acterization of H4, H6, and H9 type low pathogenicity avian influenza
viruses from wild birds in chickens and turkeys. Avian Dis. 53:552–562.
11. Mutinelli F, Capua I, Terregino C, Cattoli G. 2003. Clinical, gross, and
microscopic findings in different avian species naturally infected during
the H7N1 low- and high-pathogenicity avian influenza epidemics in Italy
during 1999 and 2000. Avian Dis. 47:844 848.
12. Okamatsu M, Saito T, Yamamoto Y, Mase M, Tsuduku S, Nakamura K,
Tsukamoto K, Yamaguchi S. 2007. Low pathogenicity H5N2 avian influ-
enza outbreak in Japan during the 2005–2006. Vet. Microbiol. 124:35– 46.
13. Shinya K, Awakura T, Shimada A, Silvano FD, Umemura T, Otsuki K.
1995. Pathogenesis of pancreatic atrophy by avian influenza a virus infec-
tion. Avian Pathol. 24:623– 632.
14. Zanella A. 2003. Avian influenza attributable to serovar H7N1 in light
layers in Italy. Avian Dis. 47:1177–1180.
15. Klopfleisch R, Wolf PU, Wolf C, Harder T, Starick E, Niebuhr M,
Mettenleiter TC, Teifke JP. 2007. Encephalitis in a stone marten (Martes
foina) after natural infection with highly pathogenic avian influenza virus
subtype H5N1. J. Comp. Pathol. 137:155–159.
16. Lipatov AS, Kwon YK, Pantin-Jackwood MJ, Swayne DE. 2009. Patho-
genesis of H5N1 influenza virus infections in mice and ferret models dif-
fers according to respiratory tract or digestive system exposure. J. Infect.
Dis. 199:717–725.
17. Thiry E, Zicola A, Addie D, Egberink H, Hartmann K, Lutz H, Poulet
H, Horzinek MC. 2007. Highly pathogenic avian influenza H5N1 virus in
cats and other carnivores. Vet. Microbiol. 122:25–31.
18. Yingst SL, Saad MD, Felt SA. 2006. Qinghai-like H5N1 from domestic
cats, northern Iraq. Emerg. Infect. Dis. 12:1295–1297.
19. Blum A, Podvitzky O, Shalabi R, Simsolo C. 2010. Acute pancreatitis
may be caused by H1N1 influenza A virus infection. Isr. Med. Assoc. J.
12:640641.
20. Calore EE, Uip DE, Perez NM. 2011. Pathology of the swine-origin
influenza A (H1N1) flu. Pathol. Res. Pract. 207:86 –90.
21. Cano M, Iglesias P, Perez G, Diez JJ. 2010. Influenza A virus (H1N1)
infection as a cause of severe diabetic ketoacidosis in type 1 diabetes. En-
docrinol. Nutr. 57:37–38.
22. Nenna R, Papoff P, Moretti C, Pierangeli A, Sabatino G, Costantino F,
Soscia F, Cangiano G, Ferro V, Mennini M, Salvadei S, Scagnolari C,
Antonelli G, Midulla F. 2011. Detection of respiratory viruses in the 2009
winter season in Rome: 2009 influenza A (H1N1) complications in chil-
dren and concomitant type 1 diabetes onset. Int. J. Immunopathol. Phar-
macol. 24:651– 659.
23. Watanabe N. 2011. Conversion to type 1 diabetes after H1N1 influenza
infection: a case report. J. Diabetes 3:103.
24. European Union. 2006. Community measures for the control of avian
influenza and repealing directive 92/40/EEC. Council directive 2005/
94/EC of 20 December 2005. Official J. Eur. Union Legis. L 49:16 65.
25. Spackman E, Senne DA, Myers TJ, Bulaga LL, Garber LP, Perdue ML,
Lohman K, Daum LT, Suarez DL. 2002. Development of a real-time
reverse transcriptase PCR assay for type A influenza virus and the avian H5
and H7 hemagglutinin subtypes. J. Clin. Microbiol. 40:3256 –3260.
26. Baroni MG, Cavallo MG, Mark M, Monetini L, Stoehrer B, Pozzilli P.
1999. Beta-cell gene expression and functional characterisation of the hu-
man insulinoma cell line CM. J. Endocrinol. 161:59 68.
27. Ouyang H, Mou L, Luk C, Liu N, Karaskova J, Squire J, Tsao MS. 2000.
Immortal human pancreatic duct epithelial cell lines with near normal
genotype and phenotype. Am. J. Pathol. 157:1623–1631.
28. Matrosovich M, Matrosovich T, Garten W, Klenk HD. 2006. New
low-viscosity overlay medium for viral plaque assays. Virol. J. 3:63.
29. Wakeley PR, Johnson N, McElhinney LM, Marston D, Sawyer J, Fooks
AR. 2006. Development of a real-time, differential RT-PCR TaqMan assay
for lyssavirus genotypes 1, 5 and 6. Dev. Biol. (Basel) 126:227–236, 326
327.
30. Hazelwood RL. 2000. Pancreas, p 539 –555. In Whittow GC (ed), Sturkie’s
avian physiology, 5th ed. Academic Press, San Diego, CA.
31. Laurent F, Mialhe P. 1978. Effect of free fatty acids and amino acids on
glucagon and insulin secretions in normal and diabetic ducks. Diabetolo-
gia 15:313–321.
32. Hoffmann WE, Solter PF. 2008. Diagnostic enzymology of domestic
animals, p 365–366. In Kaneko J, Harvey JW, Bruss ML (ed), Clinical
biochemistry of domestic animals, 6th ed. Elsevier Inc., New York, NY.
33. Colca JR, Hazelwood RL. 1982. Persistence of immunoreactive insulin,
glucagon and pancreatic polypeptide in the plasma of depancreatized
chickens. J. Endocrinol. 92:317–326.
34. Eizirik DL, Colli ML, Ortis F. 2009. The role of inflammation in insulitis
and beta-cell loss in type 1 diabetes. Nat. Rev. Endocrinol. 5:219 –226.
35. Sarkar SA, Lee CE, Victorino F, Nguyen TT, Walters JA, Burrack A,
Eberlein J, Hildemann SK, Homann D. 2012. Expression and regulation
of chemokines in murine and human type 1 diabetes. Diabetes 61:436
446.
36. Rhode A, Pauza ME, Barral AM, Rodrigo E, Oldstone MB, von Herrath
MG, Christen U. 2005. Islet-specific expression of CXCL10 causes spon-
taneous islet infiltration and accelerates diabetes development. J. Immu-
nol. 175:3516 –3524.
37. Christen U, McGavern DB, Luster AD, von Herrath MG, Oldstone MB.
2003. Among CXCR3 chemokines, IFN-gamma-inducible protein of 10
kDa (CXC chemokine ligand (CXCL) 10) but not monokine induced by
IFN-gamma (CXCL9) imprints a pattern for the subsequent development
of autoimmune disease. J. Immunol. 171:6838 6845.
38. Frigerio S, Junt T, Lu B, Gerard C, Zumsteg U, Hollander GA, Piali L.
Pancreatic Damage by Influenza A Virus
January 2013 Volume 87 Number 1 jvi.asm.org 609
2002. Beta cells are responsible for CXCR3-mediated T-cell infiltration in
insulitis. Nat. Med. 8:1414 –1420.
39. Cardozo AK, Proost P, Gysemans C, Chen MC, Mathieu C, Eizirik DL.
2003. IL-1beta and IFN-gamma induce the expression of diverse chemo-
kines and IL-15 in human and rat pancreatic islet cells, and in islets from
pre-diabetic NOD mice. Diabetologia 46:255–266.
40. Hanifi-Moghaddam P, Kappler S, Seissler J, Muller-Scholze S, Martin S,
Roep BO, Strassburger K, Kolb H, Schloot NC. 2006. Altered chemokine
levels in individuals at risk of type 1 diabetes mellitus. Diabet. Med. 23:
156–163.
41. Nicoletti F, Conget I, Di Mauro M, Di Marco R, Mazzarino MC,
Bendtzen K, Messina A, Gomis R. 2002. Serum concentrations of the
interferon-gamma-inducible chemokine IP-10/CXCL10 are augmented
in both newly diagnosed type I diabetes mellitus patients and subjects at
risk of developing the disease. Diabetologia 45:1107–1110.
42. Shimada A, Morimoto J, Kodama K, Suzuki R, Oikawa Y, Funae O,
Kasuga A, Saruta T, Narumi S. 2001. Elevated serum IP-10 levels ob-
served in type 1 diabetes. Diabetes Care 24:510 –515.
43. Likos AM, Kelvin DJ, Cameron CM, Rowe T, Kuehnert MJ, Norris PJ,
National Heart Lung Blood Institute Retrovirus Epidemiology Donor
Study-II (REDS-II). 2007. Influenza viremia and the potential for blood-
borne transmission. Transfusion 47:1080 –1088.
44. Oughton M, Dascal A, Laporta D, Charest H, Afilalo M, Miller M. 2011.
Evidence of viremia in 2 cases of severe pandemic influenza A H1N1/09.
Diagn. Microbiol. Infect. Dis. 70:213–217.
45. Tse H, To KK, Wen X, Chen H, Chan KH, Tsoi HW, Li IW, Yuen KY.
2011. Clinical and virological factors associated with viremia in pandemic
influenza A/H1N1/2009 virus infection. PLoS One 6:e22534. doi:10.1371/
journal.pone.0022534.
46. Goldberg E, Krause I. 2009. Infection and type 1 diabetes mellitus—a two
edged sword? Autoimmun. Rev. 8:682– 686.
47. Pino SC, Kruger AJ, Bortell R. 2010. The role of innate immune pathways
in type 1 diabetes pathogenesis. Curr. Opin. Endocrinol. Diabetes Obes.
17:126–130.
48. Richer MJ, Horwitz MS. 2009. Coxsackievirus infection as an environ-
mental factor in the etiology of type 1 diabetes. Autoimmun. Rev. 8:611–
615.
49. Jun HS, Yoon JW. 2003. A new look at viruses in type 1 diabetes. Diabetes
Metab. Res. Rev. 19:8 –31.
50. Richer MJ, Horwitz MS. 2008. Viral infections in the pathogenesis of
autoimmune diseases: focus on type 1 diabetes. Front. Biosci. 13:4241–
4257.
51. von Herrath MG, Holz A, Homann D, Oldstone MB. 1998. Role of
viruses in type I diabetes. Semin. Immunol. 10:87–100.
52. Sano H, Terasaki J, Tsutsumi C, Imagawa A, Hanafusa T. 2008. A case
of fulminant type 1 diabetes mellitus after influenza B infection. Diabetes
Res. Clin. Pract. 79:e8 e9. doi:10.1016/j.diabres.2007.10.030.
53. Graham KL, Sanders N, Tan Y, Allison J, Kay TW, Coulson BS. 2008.
Rotavirus infection accelerates type 1 diabetes in mice with established
insulitis. J. Virol. 82:6139 6149.
54. Reed LJ, Muench H. 1938. A simple method of estimating fifty percent
endpoints. Am. J. Hyg. 27:493– 497.
Capua et al.
610 jvi.asm.org Journal of Virology

Supplementary resources (7)

... Several in vivo and in vitro studies have been conducted to gain a better insight into the disease pathophysiology [10,11]. In the mechanism of influenza A virus infection, sialic acid (SA) receptors, located on the surface of cells, act as an integral part by facilitating attachment and entry of viral particles into the cell. ...
... These findings explain the mechanism of pancreatic cell injury consequent to influenza A virus infection. Further studies have been able to establish this finding by demonstrating that certain influenza viruses have a tropism for pancreatic acinar cells [11]. These observations can be used to explain the manifestation of acute pancreatitis in influenza infection. ...
Article
Full-text available
Background Acute hemorrhagic pancreatitis is a life-threatening condition leading to shock and multiorgan failure. Although prevalent in the general population, the incidence during pregnancy is low, with a high maternal and fetal mortality rate. The highest incidence is in the third trimester/early postpartum period. Infectious etiology for acute hemorrhagic pancreatitis is rare with only a handful of cases following influenza infection being documented in the literature. Case presentation A 29-year-old Sinhalese pregnant lady in the third trimester presented with an upper respiratory tract infection and abdominal pain, for which she was managed with oral antibiotics. An elective caesarean section was done at 37 weeks gestation due to a past section. On postoperative day 3 she developed a fever with difficulty in breathing. Despite treatment, she succumbed to death on the sixth postoperative day. The autopsy revealed extensive fat necrosis with saponification. The pancreas was necrosed and hemorrhagic. The lungs showed features of adult respiratory distress syndrome and necrosis was observed in the liver and kidneys. Polymerase chain reaction of lungs detected influenza A virus (subtype H3). Conclusion Although rare, acute hemorrhagic pancreatitis from an infectious etiology carries risk of morbidity and mortality. Therefore, a high level of clinical suspicion must be upheld among clinicians to minimize adverse outcomes.
... The relation between AP and some viruses such as CMV, HIV, HSV, EBV, VZV, mumps virus, coxsackievirus, and some others has been proven. 9 Based on the Revised Atlanta Classification System for AP Diagnosis, at least two of the three following criteria should be found-abdominal pain (defined as acute onset, persistent, severe epigastric pain often radiating to the patients' back), increased serum lipase or amylase levels to greater than 3 times the upper limit of normal value, and characteristic findings of AP on contrast-enhanced CT. AP was diagnosed in our patient based on the Atlanta criteria. ...
Article
The present study comprised of 48 patients with COVID-19 infection of both genders. All patients were subjected to blood cytology, biochemistry, and inflammatory indicators. Pancreatic injury was defined as any abnormality in amylase and lipase level. Results: Common symptoms were fever in 90%, breathlessness in 75%, cough in 68%, fatigue in 76%, headache in 42% and diarrhoea in 38%. The difference was significant (P< 0.05). Leukocytes (X109/L) count was 5.26, neutrophils (X109/L) were 3.78, platelets (X109/L) were 195.2, monocytes (X109/L) were 0.45 and lymphocytes (X109/L) were 0.92.
... On the other hand, protective effects can be achieved by suppressing autoimmune phenomena through regulatory immune responses [27]. Influenza viruses and EBV infection were considered as central roles in the pathogenesis of pSS through the autoimmunity induced by different mechanisms in previous literature [28][29][30][31][32][33]. ...
Article
Full-text available
Introduction Primary Sjögren’s syndrome (pSS) is an autoimmune disease characterized by inflammatory infiltration, and dysfunction of the salivary and lacrimal glands. This research aimed to explore the disease pathogenesis and improve the diagnosis and treatment of pSS by mining inflammation-associated biomarkers. Methods Five pSS-related datasets were retrieved from the Gene Expression Omnibus (GEO) database. Inflammation-associated biomarkers were determined by the least absolute shrinkage and selection operator (LASSO) and support vector machines recursive feature elimination (SVM-RFE). Single sample gene set enrichment analysis (ssGSEA) was implemented to profile the infiltration levels of immune cells. Real-time quantitative PCR (RT-qPCR) verified the expression of biomarkers in clinical samples. Results Four genes (LY6E, EIF2AK2, IL15, and CXCL10) were screened as inflammation-associated biomarkers in pSS, the predictive performance of which were determined among three pSS-related datasets (AUC > 0.7). Functional enrichment results suggested that the biomarkers were involved in immune and inflammation-related pathways. Immune infiltration analysis revealed that biomarkers were notably connected with type 2 T helper cells, regulatory T cells which were significantly expressed between pSS and control. TESTOSTERONE and CYCLOSPORINE were predicted to take effect by targeting CXCL10 and IL15 in pSS, respectively. Conclusion Four inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were explored, and the underlying regulatory mechanisms and targeted drugs associated with these biomarkers were preliminarily investigated according to a series of bioinformatics methods based on the online datasets of pSS, which provided a reference for understanding the pathogenesis of pSS. Key Points• Inflammation-associated biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were firstly identified in Sjögren’s syndrome based on LASSO and SVM-RFE analyses. • CXCL10, EIF2AK2 and LY6E were prominently positively correlated with immature B cells, while IL15 were significantly negatively correlated with memory B cells in Sjögren’s syndrome. • LY6E, EIF2AK2, IL15, and CXCL10 were significantly more highly expressed in clinical Sjögren’s syndrome samples compared to healthy control samples, which was consistent with the analysis results of the GEO database. • LY6E, EIF2AK2, IL15, and CXCL10 might be used as the biomarkers for the treatment and diagnosis of Sjögren’s syndrome.
... The distribution of heterophilic antigens is not only individually variable, but also species-specific. One study found that infection of turkeys with influenza A virus induced pancreatitis and diabetes in the animals (23). Infection with lymphocytic choroid plexus meningitis virus or polyomavirus stimulates the development of multiple sclerosis disease, with NZB mice being the preferred choice (24). ...
Article
Full-text available
Introduction Microbial infections are associated with the occurrence of autoimmune diseases, but the mechanisms of microbial infection inducing autoimmune diseases are not fully understood. The existence of heterophilic antigens between microorganisms and human tissues may explain part of the pathogenesis of autoimmune diseases. Here, we investigate the distribution of heterophilic antigens and its relationship with autoimmune diseases. Methods Monoclonal antibodies against a variety of microorganisms were prepared. The titer, subclass and reactivity of antibodies with microorganisms were identified, and heterophilic antibodies that cross-reacted with human tissues were screened by human tissue microarray. The reactivity of these heterophilic antibodies with different individuals and different species was further examined by immunohistochemistry. Results In this study, 21 strains of heterophilic antibodies were screened. The results showed that these heterophilic antibodies were produced due to the existence of heterophilic antigens between microorganism and human body and the distribution of heterophilic antigens had individual, tissue and species differences. Conclusion Our study showed that heterophilic antigens exist widely between microorganisms and human body, and the heterophilic antigens carried by microorganisms may break the immune tolerance of the body through carrier effect and initiate immune response, which may be one of the important mechanisms of infection inducing autoimmune diseases.
... Our functional enrichment analysis revealed the nine pSS-related and in ammationassociated DEGs involved in multiple virus infections, which were reported in previous research. In uenza viruses can induce autoimmunity by several mechanisms, such as causing T cell bystander activation and direct beta cell damage in diabetes [24,25]. Research about pSS patients immunized with an H1N1 in uenza vaccine showed pSS patients developed higher H1N1 IgG titers than healthy controls after inoculation, and off-target B cells also produced increased antibody levels against Epstein-Barr virus (EBV) and autoantibodies [26]. ...
Preprint
Full-text available
Background: Primary Sjögren’s syndrome (pSS) is an autoimmune disease characterized by inflammatory infiltration and dysfunction of the salivary and lacrimal glands. This research aimed to explore the disease pathogenesis and improve the diagnosis and treatment of pSS by mining inflammatory biomarkers. Methods: Five pSS-related datasets were retrieved from the Gene Expression Omnibus (GEO) database. Inflammatory biomarkers were determined by Least absolute shrinkage and selection operator (LASSO) and support vector machines recursive feature elimination (SVM-RFE). Single sample gene set enrichment analysis (ssGSEA) was implemented to profile the infiltration levels of immune cells. The expression of biomarkers in clinical samples was verified by Real-Time Quantitative PCR. Results: Four genes (LY6E, EIF2AK2, IL15, and CXCL10) were confirmed as inflammatory biomarkers in pSS. Functional enrichment suggested that the biomarkers were involved inimmune and inflammation-related pathways. Immune infiltration analysis revealed that biomarkers were notably connected with some differential immune cells between pSS and control. Also, the RT-qPCR results of clinical samples further affirmed the results of the public database. Conclusion: Four inflammatory biomarkers (LY6E, EIF2AK2, IL15, and CXCL10) were defined and regulatory mechanisms and targeted drugs were investigated in pSS, which provided a basis for understanding the pathogenesis and improving clinical diagnosis and treatment for the disease.
... The incidence of DKA in hospitalized COVID-19 patients without previous insulin dependence (1.4%) was signi cantly higher than those in hospitalized in uenza (0.8%) whereas the demographics and comorbidities were similar between groups, suggesting virus speci c activity or disease severity likely contribute to glucose dysregulation and insulin dependency. SARS-CoV-2 has proposed tropism for ACE2 receptors of pancreatic beta cells [32], and there is some evidence of tropism of the in uenza virus for pancreatic beta cells, with an ability to replicate in human pancreatic cells, most often resulting in pancreatitis [33,34]. Although in uenza is also a known contributor to DKA, its relationship to DKA is likely nonspeci c as an infectious process, and more likely to affect ketosis-prone diabetic patients [35,36]. ...
Preprint
Full-text available
Whether SARS-CoV-2 infection triggers diabetic ketoacidosis (DKA) is unclear. This study characterized incidence, patient profiles, risk factors, and outcomes of in-hospital DKA in COVID-19 patients without prior insulin dependence and compared with influenza. This cohort consisted of 13,383 hospitalized COVID-19 patients (March 2020 to July 2022) and 19,165 hospitalized influenza patients (January 2018 to July 2022) in Bronx, NY. Patients with prior DKA and prior insulin use were excluded. Primary outcomes were in-hospital mortality and new-insulin use 3-month post-infection. The incidence of DKA in hospitalized COVID-19 patients was significantly higher than hospitalized influenza patients (1.4% vs. 0.8%, p < 0.05). COVID-19 patients with DKA were more likely to be intubated, receive steroid treatment, and die (mortality OR = 6.178, p < 0.05) than those without DKA. DKA patients without pre-existing diabetes were more likely to die than DKA patients with pre-existing diabetes (OR = 7.56, p < 0.05). Steroid use, pre-existing type-2 diabetes, and male sex were risk factors for DKA. Patients with DKA had a higher rate of insulin use 3 months post SARS-CoV-2 infection compared to those without DKA (8.2% vs. 1.6%, p < 0.05), suggesting SARS-CoV-2 infection could trigger new insulin dependence. Identification of risk factors for DKA and new insulin-dependency could enable careful monitoring and timely intervention.
... In addition, viral infection associated pancreatitis have been reported to other virus infections [245][246][247], yet, the mechanism of its development depends on the type of the viral infection [248]. Others have considered that SARS-COV2 gastrointestinal infection contributes to the elevated blood levels of pancreatic lipase as well as amylase enzymes during the onset of the infection [249] due to the increased GIT permeability [250,251], hence, they have hypothesized that over 20% of SARS-COV2 hospitalized patients with abdominal pain are misdiagnosed as developing acute pancreatitis or even developing multiple organs failure [242,252]. Some have attributed such cases of acute pancreatitis to the SARS-COV2 experienced cytokines storm, microvascular thrombosis or acute respiratory distress, thus for such reasons 20% of SARS-COV2 infected individuals with GIT symptoms are reported to have acute pancreatitis [242]. ...
Article
Full-text available
Since the emergence of SARS-COV2 infection, accumulating reports as well as evidences have been reported to the complex disease-disease interactions between this viral infection and the pre-existing diabetes co-morbidity. Hyperglycemia accompanied the onset of infection is repeatedly reported to be associated with the disease severity, prognosis and mortality rate elevation. Various mechanism has been speculated to lay behind the reported hyperglycemia including direct pancreatic tissues tropism besides the indirect cytokine storm overwhelming inflammatory immune response. In another perspective, other collection of studies has reported and emphasized the involvement of SARS-COV2 infection in the development of acute pancreatitis as one of this infection complications during the onset of infection, hence increasing the morbidity of such infection including the admission of the patients to the intensive care unit. Some have related the diagnosis of acute pancreatitis to the serum amylase and lipase levels manifested with the experienced GI symptoms, while, others consider CT scan images to confirm diagnosis. Finally, some reporters has hypothesized that SARS-COV2 infection may develop autoantibodies that probably precipitate type I diabetes mellitus long after infection. Therefore, due to significance of hyperglycemia/ diabetes to fate / severity of the infection as well as long term complications of SARS-COV2 this survey have covered aspects related to these issues.
Article
Aims: This study characterized incidence, patient profiles, risk factors and outcomes of in-hospital diabetic ketoacidosis (DKA) in patients with COVID-19 compared with influenza and pre-pandemic data. Methods: This study consisted of 13 383 hospitalized patients with COVID-19 (March 2020-July 2022), 19 165 hospitalized patients with influenza (January 2018-July 2022) and 35 000 randomly sampled hospitalized pre-pandemic patients (January 2017-December 2019) in Montefiore Health System, Bronx, NY, USA. Primary outcomes were incidence of in-hospital DKA, in-hospital mortality, and insulin use at 3 and 6 months post-infection. Risk factors for developing DKA were identified. Results: The overall incidence of DKA in patients with COVID-19 and influenza, and pre-pandemic were 2.1%, 1.4% and 0.5%, respectively (p < .05 pairwise). Patients with COVID-19 with DKA had worse acute outcomes (p < .05) and higher incidence of new insulin treatment 3 and 6 months post-infection compared with patients with influenza with DKA (p < .05). The incidence of DKA in patients with COVID-19 was highest among patients with type 1 diabetes (12.8%), followed by patients with insulin-dependent type 2 diabetes (T2D; 5.2%), non-insulin dependent T2D (2.3%) and, lastly, patients without T2D (1.3%). Patients with COVID-19 with DKA had worse disease severity and higher mortality [odds ratio = 6.178 (4.428-8.590), p < .0001] compared with those without DKA. Type 1 diabetes, steroid therapy for COVID-19, COVID-19 status, black race and male gender were associated with increased risk of DKA. Conclusions: The incidence of DKA was higher in COVID-19 cohort compared to the influenza and pre-pandemic cohort. Patients with COVID-19 with DKA had worse outcomes compared with those without. Many COVID-19 survivors who developed DKA during hospitalization became insulin dependent. Identification of risk factors for DKA and new insulin-dependency could enable careful monitoring and timely intervention.
Article
Our previous studies found that the H1-50 monoclonal antibody (mAb) of influenza A virus hemagglutinin (HA) cross-reacted with pancreatic tissue and islet β-cells, and further studies showed that H1-50 mAb binds to prohibitin (PHB) protein of islet β-cells. These suggest that there are heterophilic epitopes between influenza virus HA and pancreatic tissue, which may be involved in the pathogenesis of type 1 diabetes. To further investigate these heterophilic epitopes, we screened binding epitopes of H1-50 mAb using a phage 12-peptide library. DNA sequencing and comparative analysis were performed on specific positive phage clones, and the sequence of 12-peptide binding to H1-50 mAb was obtained. The binding epitopes of H1-50 mAb in influenza virus HA were determined by sequence analysis and experimental verification, and their distribution within the three-dimensional structure was assessed by PyMOL. The results showed that H1-50 mAb specifically binds to polypeptides (306-SLPFQNIHPITIGK-319) of influenza A virus HA, located in the stem of the HA protein. However, there is no specific binding sequence between H1-50 mAb and the PHB protein of islet β-cells in the primary structure, and we speculate that the binding of H1-50 mAb to islet β-cells may depend on the spatial conformation. The identification of the heterophilic epitopes of H1N1 influenza virus hemagglutinin provides a new perspective on type 1 diabetes that may be caused by influenza virus infection, which may contribute to the prevention and control of influenza.
Article
Full-text available
Clinical epidemiological studies have reported that viral infections cause autoimmune pathology in humans. Host-pathogen protein sequences and structure-based molecular mimicry cause autoreactive T cells to cross-activate. The aim of the current study was to implement im- munoinformatics approaches to infer sequence- and structure-based molecular mimicry between viral and human proteomic datasets. The protein sequences of all the so far known human-infecting viruses were obtained from the VIPR database, and complete human proteome data were retrieved from the NCBI repository. Based on a predefined, stringent threshold of comparative sequence analyses, 24 viral proteins were identified with significant sequence similarity to human proteins. PathDIP identified the enrichment of these homologous proteins in nine metabolic pathways with a p-value < 0.0001. Several viral and human mimic epitopes from these homologous proteins were predicted as strong binders of human HLA alleles, with IC50 < 50 nM. Downstream molecular docking analyses identified that lead virus-human homologous epitopes feasibly interact with HLA and TLR4 types of immune receptors. The vast majority of these top-hit homolog epitopic peptides belong to the herpes simplex and poxvirus families. These lead epitope biological sequences and 3D structural-based molecular mimicry may be promising for interpreting herpes simplex virus and poxvirus infection-mediated autoimmune disorders in humans.
Book
This book contains comprehensive and updated information on how to perform Avian Influenza and Newcastle Disease diagnosis from the suspicion in the field to the characterisation of isolates. It provides guidelines to outbreak management, field investigation, necropsy techniques, sampling methods and complete laboratory diagnosis, including molecular methods. The outstanding images collected from field outbreaks, including clinical and pathological findings, and the selection of laboratory protocols make this publication unique. It will therefore be an invaluable tool for all veterinarians, scientists, animal health professionals, and public health officials involved in the management of these infections.
Chapter
Similar to the compact pancreas of the dog, cat, man, and other primates, and dissimilar to the diffuse tissue in rats and rabbits, the avian pancreas is a discrete lobular structure that usually has well-defined clefts or morphologic folds (Figure 21–1). This organ, weighing between 2.5 and 4.0 g in adult chickens, is suspended within the U-shaped duodenal loop of the small intestine and is responsible for two families of secretions, the digestive enzymes (exocrine) and the protein-aceous hormones (endocrine). The exocrine (acinar) tissue synthesizes and releases enzymes into at least three discrete pancreatic ducts, conveying them to the duodenal loop where all semiliquid foodstuffs are subjected to the powerful lipolytic, proteolytic, and amylolytic enzymes (Chapter 10). The avian pancreas also synthesizes and releases directly into the bloodstream peptide hormones which emanate from scattered islet (endocrine) tissue embedded within the sea of acinar tissue.
Chapter
Since the appearance of the previous (third) edition of this text, several excellent monographs have appeared that impact and extend our knowledge of avian carbohydrate metabolism. While not restricted to avian forms, “The Evolution of Pancreatic Islets” (Grillo et al., 1976) contains sufficient chapter material devoted to birds to place these organisms properly in their phylogenetic strata with regard to embryogenesis, secretory regulation, and functional significance of the islet products in embryos as well as adult life. “Avian Endocrinology” (Epple and Stetson, 1980) is an overview of those endocrine mechanisms (many of which are unique) important in avian metabolism. “Recent Advances in Avian Endocrinology” (Pethes et al., 1981) extends and broadens the aforementioned sources and, in addition, adds a utilitarian flavor to the wealth of information contained therein as far as the relationship of didactic material to the physiology of production is concerned.
Chapter
This chapter presents an overview of the diagnostic enzymology of domestic animals. Clinical enzymology is the discipline that studies and tests enzyme activity in serum, plasma, urine, or other body fluids to help establish the diagnosis and prognosis of disease, and to screen for abnormal organ function. The automation of enzyme assays, and the popularity of the serum chemistry profile in veterinary medicine, has allowed retrospective studies to be conducted and has given veterinarians an opportunity to critically evaluate the diagnostic function of the common assays in a large number of animals on a regular basis, as well as gain a "feel" for the results, thereby allowing for more subtle clinical interpretations. This chapter explores the universal factors affecting changes in enzyme content of bodily fluids. The chapter then delves into specific details relevant to particular enzymes. Discussion of basic concepts in enzymology, such as enzyme structure, kinetics, or analysis, is limited to those that are of clinical relevance or that add insight into interpreting changes in body fluid enzyme activity.
Article
Animal insulinoma cell lines are widely used to study physiological and pathophysiological mechanisms involved in glucose metabolism and to establish in vitro models for studies on ‚-cells. In contrast, human insulinoma cell lines are rarely used because of diYculties in obtaining and culturing them for long periods. The aim of our study was to investigate, under diVerent experimental conditions, the capacity of the human insulinoma cell line CM to retain ‚-cell function, particularly the expression of constitutive ‚-cell genes (insulin, the glucose transporters GLUT1 and GLUT2, glucokinase), intracellular and secreted insulin, ‚-cell granules, and cAMP content. Results showed that CM cells from an early-passage express specific ‚-cell genes in response to glucose stimulation, in particular the insulin and GLUT genes. Such capacity is lost at later passages when cells are cultured at standard glucose concentrations. However, if cultured at lower glucose concentration (0·8 mM) for a longer time, CM cells re-acquire the capacity to respond to glucose stimulation, as shown by the increased expression of ‚-cell genes (insulin, GLUT2, glucokinase). Nonetheless, insulin secretion could not be restored under such experimental conditions despite the presence of intracellular insulin, although cAMP response to a potent activator of adenylate cyclase, forskolin, was present indicating a viable system. In conclusion, these data show that the human insulinoma cell line CM, at both early-passage and late-passage, posseses a functional glucose-signalling pathway and insulin mRNA expression similar to normal ‚-cells, representing, therefore, a good model for studies concerning the signalling and expression of ‚-cells. Furthermore, we have previously shown that it is also a good model for immunological studies. In this respect it is important to note that the CM cell line is one of the very few existing human ‚-cell lines in long-term culture.
Article
T cell–mediated loss of insulin-secreting β cells in the islets of Langerhans is the hallmark of type 1 diabetes. The molecular basis for the directed migration of autoreactive T cells leading to insulitis is presently unknown. Here we demonstrate that in response to inflammation, β cells secrete the chemokines CXC ligand 10 and CXC ligand 9, which specifically attract T-effector cells via the CXC chemokine receptor 3. In mice deficient for this receptor, the onset of type 1 diabetes is substantially delayed. Thus, in the absence of known etiological agents, CXC receptor 3 represents a novel target for therapeutic interference early in type 1 diabetes.
Article
Caracterización biológica en pollos y pavos de los virus de la influenza aviar de baja patogenicidad subtipos H4, H6 y H9 detectados en aves silvestres. Se evaluó en pollos libres de patógenos específicos y en pavos, la patogénesis, la eliminación viral y la respuesta serológica contra virus de la influenza aviar de baja patogenicidad subtipos H4, H6, y H9 que fueron aislados de aves silvestres. Pollos de cuatro semanas y pavos de tres semanas de edad fueron inoculados directamente en las coanas con virus de la influenza aviar de baja patogenicidad con una dosis por ave de 1 × 106 dosis infectantes para embrión de pollo50% (EID50) y se examinaron en sus signos clínicos por tres semanas. Se recolectaron muestras de hisopos orofaríngeos y cloacales a los dos, cuatro, y siete días postinoculación para la detección por RT-PCR en tiempo real. Se recolectaron muestras para suero a los siete, catorce y 21 días después de la inoculación y se analizaron para detectar anticuerpos contra el virus de la influenza aviar mediante un ensayo de inmunoabsorción con enzimas ligadas (ELISA) y por la prueba de inhibición de la hemoaglutinación. Se recolectaron muestras de tejidos para histopatología de tres aves por grupo a los tres días después de la inoculación. Se secuenciaron los genes de la hemaglutinina de los virus estudiados y se llevó a cabo el análisis filogenético. La signología clínica incluyó desde la ausencia de signos clínicos a depresión moderada, disminución de la actividad y disminución en el consumo de alimento y agua. Con base en los resultados de detección, los pollos libres de patógenos específicos mostraron mayor eliminación viral en la orofarínge y en cloaca en comparación con los pavos comerciales. Los resultados del estudio de las lesiones microscópicas en ambas especies mostró la predominancia de lesiones en los tractos respiratorio y gastrointestinal, que fueron consistentes con los virus de baja patogenicidad. En pollos y pavos, la eliminación orofaríngea estuvo fuertemente correlacionada con las lesiones encontradas en el tracto respiratorio superior. Los pavos presentaron menos lesiones en el tracto respiratorio y mayores lesiones en el tracto gastrointestinal en comparación con los pollos. Trece de los virus de la influenza de baja patogenicidad causaron seroconversión en pavos comerciales, mientras que solo seis virus de baja patogenicidad causaron seroconversión en pollos libres de patógenos específicos. El análisis filogenético de los genes HA mostró que los virus H4, H6 y H9 evaluados en este estudio representaron completamente a la diversidad genética observada con los virus de la influenza en Norteamérica y sus subtipos respectivos. Estos datos son importantes para los sistemas de vigilancia y el control porque algunos de los virus de la influenza aviar de baja patogenicidad (con origen en aves silvestres examinados en este estudio) que pueden infectar y pueden ser eliminados por pollos y pavos pudieran ser difíciles de detectar en la avicultura comercial. Específicamente, la detección es difícil porque estos virus no causan una enfermedad clínica evidente o mortalidad, sino solo inducen lesiones microscópicas leves y mostraron una seroconversión baja. Abbreviations: AI = avian influenza; AIV = avian influenza virus; BALT = bronchial associated lymphoid tissue; BSL = biosafety level; C = cloacal; CT = cycle threshold; EID50 = 50% embryo infectious dose titer; ELISA = enzyme-linked immunosorbant assay; F = fecal; GALT = gut associated lymphoid tissue; HA = hemagglutination; HI = hemagglutination inhibition; HPAI = highly pathogenic avian influenza; LPAI = low pathogenicity avian influenza; LRE = linear regression equation; NVSL = National Veterinary Services Laboratory; OP = oropharyngeal; PBS = phosphate-buffered saline; PI = postinoculation; RT-PCR = reverse transcriptase-polymerase chain reaction; SPF = specific-pathogen-free