2005 Volume 46 2 171 177

background image

Introduction

Bovine mastitis is the most important infectious
disease of dairy cattle, affecting both the quality
and quantity of milk produced in the world. Masti-
tis occurs with the highest frequency around partu-
rition, when protective inflammatory responses at
the blood-mammary barrier are delayed or hin-
dered (Burton and Erskine 2003; Diez-Fraile et al.
2003; Paape et al. 2002). Mastitis can cause de-
struction of milk-synthesizing tissues, resulting in
decreased milk production and altered milk com-

position. Depending on the duration and severity
of disease, the productive performance of infected
dairy cattle may be diminished for the remainder
of the lactation cycle (Wilson et al. 2004). On av-
erage, losses associated with mastitis cost Ameri-
can dairy producers about 2 billion dollars
annually (Sordillo and Streicher 2002), and world-
wide losses are estimated at 25 billion dollars an-
nually.

The innate immune system represents the first

line of defense in the host response to infection
and is poised to recognize and respond immedi-

J Appl Genet 46(2), 2005, pp. 171-177

Immunorelevant gene expression in LPS-challenged bovine
mammary epithelial cells

Ravi Pareek

1

, Olga Wellnitz

2

, Renate Van Dorp

3

, Jeanne Burton

3

, David Kerr

1

1

Department of Animal Science, University of Vermont, Burlington, USA

2

Department of Physiology, Technical University of Munich, Germany

3

Department of Animal Science, Immunogenetics Laboratory and Center for Animal Functional Genomics, Michigan State

University, East Lansing, Michigan, USA

Abstract. Infection of the bovine mammary gland, in addition to causing animal distress, is a major economic
burden of the dairy industry. Greater understanding of the initial host response to infection may lead to more accu-
rate selection of resistant animals or to novel prophylactic or therapeutic intervention strategies. The epithelial
cell plays a role in the host response by alerting the immune system to the infection and providing a signal as to
where the infection is located. To understand this process better, a cDNA microarray approach was used to search
for potential signals produced by mammary epithelial cells in response to exposure to

Escherichia coli

lipopolysaccharide (LPS). Total RNA from separate cultures of epithelial cells from 4 Holstein cows was har-
vested 6 h after LPS challenge or control conditions. For each cow, RNA from control or LPS-exposed cells was
transcribed to cDNA and labeled with Cy3 or Cy5, then pooled and applied to a bovine total leukocyte (BOTL)
microarray slide containing 1278 unique transcripts. Dye reversal was used so that RNA from two of the control
cultures was labeled with Cy3 while RNA from the other two control cultures was labeled with Cy5. From the re-
sulting microarray data we selected 4 of the 9 genes significantly (P < 0.02) induced (>1.25-fold) in response to
LPS exposure for more detailed analysis. The array signal intensity for 3 of these genes,

RANTES/CCL5, IL-6 and

T-PA, was relatively low, but quantitative real-time RT-PCR (Q-RT-PCR) analysis revealed that they were in-
duced 208-fold, 10-fold and 3-fold, respectively. The gene that showed the greatest fold induction by microarray
analysis (2.5-fold) was

CXCL5. This gene had a relatively strong signal intensity on the array and was easily de-

tected by northern blot analysis, which indicated a 10-fold induction. This cell culture model system provides evi-
dence for an important role of the mammary epithelial cell in initiating the innate response to infection.

Key words: bovine total leukocyte microarray, BOTL, lipopolysaccharide, mammary gland.

Received: September 12, 2004. Revised: February 26, 2005. Accepted: March 03, 2005.
Correspondence: D.E. Kerr, Department of Animal Science, Terrill Hall, University of Vermont, 570 Main Street, Burlington,
VT 05405, USA; e-mail: david.kerr@uvm.edu

background image

ately to the earliest stages of infection (Hoffmann
et al. 1999). The inherent capability of the innate
immune system to respond to a vast number of
pathogens is mediated by its ability to recognize
highly conserved motifs shared by diverse patho-
gens. These motifs, commonly referred to as
pathogen-associated molecular patterns (PAMPs),
include some bacterial cell wall components, like
lipopolysaccharide (LPS), peptidoglycan and
lipoteichoic acid (Aderem and Ulevitch 2000).

Although milk macrophages may play a role in

the triggering of the inflammatory response, stud-
ies at various epithelial sites strongly suggest that
epithelial cells are capable of responding to bacte-
rial intrusion and play a major part in the initiation
of inflammation. A better knowledge of the medi-
ators involved in the initial recognition and signal-
ing of bacterial invasion could help in devising
strategies to modulate the defense of the udder
(Rainard and Riollet 2003).

We, and others, have used a bovine mammary

epithelial cell culture system to investigate factors
produced by these cells in response to infection by
Staphylococcus aureus or exposure to Escherichia

coli LPS (Wellnitz and Kerr 2004; Boudjellab et
al. 1998; Talhouk et al. 1990; Okada et al. 1997).
These studies demonstrated that epithelial cells re-
spond with enhanced production of IL-8, TNFá,
and lactoferrin. The role of IL-8 in attracting neu-
trophils to the site of infection is well known, and
may be a key feature of the epithelial cell re-
sponse. In an effort to uncover additional epithe-
lial cell signals of infection, we employed a
microarray-based screening approach. We were
particularly interested in the early responses and
thus examined the RNA from our bovine epithelial
cell model following a 6-hour exposure to

E. coli

LPS.

Material and methods

LPS treatment of mammary epithelial cells

The procedures for cell culture and cryopreserva-
tion have been previously described (Wellnitz and
Kerr 2004). In brief, mammary tissue was col-
lected at slaughter from 4 lactating Holstein cows.
Epithelial cells were obtained after collagen-
ase-based digestion of the tissue. Cell numbers
were multiplied by culture on a plastic substratum,
passaged several times, and then cryopreserved.
Aliquots of cells (passage 3) from all 4 cows were
thawed for culture, and grown separately to con-
fluence in a growth medium consisting of

DMEM/F12,

10%

FBS,

ITS

supplement

(5 µg mL

–1

insulin, 5 µg mL

–1

transferrin and

0.005 µg mL

–1

sodium selenite; Invitrogen,

Carlsbed, CA), penicillin G (100 u mL

–1

) and

streptomycin (100 µg mL

–1

). They were then split

into two 75-mL flasks/cow (15 × 10

6

cells/flask)

in challenge media (3% FBS medium). When
~80% confluence was obtained, the medium was
replaced with fresh challenge media, with or with-
out 1 µg mL

–1

LPS (0111:B4; Sigma). Total RNA

was harvested after 6 hrs by TRIzol (Invitrogen,
Carlsbed, CA) and mRNA was isolated by
oligo(dT)-latex beads with the NucleoTrap Kit
(BD Biosciences, San Jose, CA).

Bovine total leukocyte (BOTL) microarray

In the present study we used 4 BOTL microarray
slides (one for each cow), in which samples de-
rived from control and LPS-treated cultures com-
peted for hybridization to the arrayed cDNA spots.
The

spots

on

the

BOTL

slides

represent

1278 genes, spotted in triplicate, drawn from a bo-
vine leukocyte library and additionally supple-
mented

with

candidate

genes

involved

in

apoptosis and inflammation. A listing of the genes
is available on the Center for Animal Functional
Genomics (CAFG) website (www.nbfgc.msu.edu).
The general cDNA spotting design used on
the BOTL microarray is described in detail on
the website of the Center for Biotechnology Infor-
mation Gene Expression Omnibus (NCBI GEO;
http://www.ncbi.nlm.nih.gov/geo/, platform ac-
cession number GPL 363). Control genes spotted
within and across the 48-patch microarrays (9 × 9
series of spots per patch) included 96 spots of syn-
thetic Q cDNA (external controls), 144 spots of
GAPDH, 75 spots of ß-actin and 75 spots of

RPL-19 (internal controls).

For probe synthesis, 1 µg of polyA

+

-enriched

RNA was converted to cDNA (42°C for 65 min,
then 70° for 5 min) using the Atlas PowerScript
fluorescent labeling kit (BD Biosciences, San
Jose, CA). Dye couplings and labeled cDNA
purifications

were

performed

according

to

the manufacturer’s instructions (Amersham Bio-
sciences,

Piscataway,

NJ).

For

two

cows

the LPS-challenged cultures were labeled with
CY5 and control cultures with CY3. Dye labeling
was reversed for the other two cows.

Cow-specific pairs of labeled cDNA were in-

cubated at 70°C for 5 min just prior to application
to the array under a cover slip. Hybridization was
allowed to proceed at 50°C overnight in a humidi-
fied chamber (HYBAID; Labnet, Woodbridge,

172

R. Pareek et al.

background image

NJ). The cover slips were then removed and the ar-
rays

washed

twice

for

15

min

at

me-

dium-stringency (45°C, 1 × SSC, 0.05% SDS) on
an orbital shaker. An additional wash (0.1 × SSC)
was applied for 5 min. Finally, arrays were re-
moved, rinsed with H

2

O, and centrifuged in open

50-mL conical tubes (500 g for 3 min at room tem-
perature) to dry. Array scanning was done using
a GeneTAC LS IV scanner (Genomic Solutions,
Ann Arbor, MI) and accompanying software (ver-
sion 3.01). Spot analyses were performed using
GeneTAC Integrator 3.3.0 microarray analysis
software (Genomic Solutions, Ann Arbor, MI).

Northern blot and Q-RT-PCR analysis

Verification of altered expression of

CXCL5 was

pursued through the use of northern blot. Total
RNA (8

mg/lane) was separated on agarose gel

containing 1.1% formaldehyde and blotted to
Gene

Screen

hybridization

membranes

(NEN/Perkin Elmer) in 1 M ammonium acetate.
Membranes

were

sequentially

probed

with

32

P-labeled

CXCL5, and bovine ß-actin PCR prod-

ucts. Differences in gene expression were ana-
lyzed with a phosphoimager (STORM, Molecular
Dynamics). Expression of

CXCL5 was normalized

to ß-actin expression levels.

Microarray-indicated changes in

RANTES,

IL-6, and T-PA expression were verified with
quantitative real-time RT-PCR (Q-RT-PCR). To-
tal RNA (1.5 g) was treated with DNase
(DNAfree; Ambion) according to the manufac-
turer’s protocol, then reverse-transcribed to cDNA
with random hexamer primers by using the Super-
script First-Strand Synthesis System (Invitrogen,
Carlsbed, CA) according to the manufacturer’s
protocols. Real-time PCR was performed with
the SYBR Green JumpStart Taq Ready Mix
(Sigma) on an ABI Prism 7700 Sequence Detector
at the Vermont Cancer Center core facility. Bo-
vine specific primers for

T-PA were 5’-CAA TGA

CAT CGC GCT GCT-3’ for forward; and 5’-TGG
GTA CAA CCT GAC GTG AGC-3’ for reverse;
primers for

RANTES were 5’-GCC AAC CCA

GAG AAG AAGTG-3’ for forward; and 5’-CTG
CTT AGG ACA AGA GCG AGA-3’ for reverse;
primers

for

IL-6 were 5’-TGAGGGAAA

TCAGGAAAATGT-3’

for

forward;

and

5’-CAGTGTTTGTGGCTGGAGTG-3’ for re-
verse; primers for ß-actin were 5’-GCA AAT
GCT TCT AGG CGG ACT-3’ for forward; and
5’-CAA TCT CAT CTC GTT TTC TGC G-3’ for
reverse. The above primers flanked 205, 119, 110

and 85-bp regions, respectively. For each PCR
product the melting curve was determined.
The comparative threshold cycle number (2

DDCt

)

method was used after a validation experiment
demonstrated that efficiencies of target and refer-
ence (ß-actin) were approximately equal. Ct val-
ues define the threshold cycle of PCR, at which
amplified products were detected. Our results are
represented as

DCt values, where DCt is the differ-

ence in threshold cycles for target and ß-actin as an
internal control. Fold changes in expression for
LPS relative to control (

DDCt) were calculated

from the arithmetic formula 2

DDCt

as described in

detail by Livak and Schmittgen (2001).

Statistical analysis

After the BOTL microarray experiment, total in-
tensity values for each dye channel were loaded
into SAS for data normalization and analysis. Data
normalization was performed considering a robust
local regression technique (Yang et al. 2002) using
the LOWESS (Locally-weighted Regression) pro-
cedure of SAS 8 (SAS/STAT Software, Cary,
NC). The normalized data were analyzed with
a 2-step mixed model advocated by Wolfinger et
al. (2001). Significance of values of expressed
genes was measured by Student’s

t-test in LPS

versus control spots.

Differences between control and LPS chal-

lenge in the 4 genes selected for further validation
by Q-RT-PCR or northern blot analysis were
tested for significance by paired

t-test. A P value

of <0.01 was considered significant. One sample
for the

T-PA gene expression was lost due to

a technical error.

Results

In general, the results from the microarray analysis
lacked sensitivity. However, cells from the indi-
vidual cows appeared to yield consistent results,
which can be seen by a representative northern
blot analysis of

CXCL5 (Figure 1). In total, the sta-

tistical analysis revealed that 54 genes were signif-
icantly induced by LPS, but the overall fold
changes were quite low. Considering the robust-
ness of the system, we concentrated our efforts on
the 9 genes that had the greatest fold changes
(Table 1). Of these genes, BLASTn analysis and
gene ontology information as well as Genecards™
studies for functional genomics in humans indi-

Gene expression in LPS-challenged cells

173

background image

cated that 5 genes were likely components of
the innate immune system, and 4 genes were re-
lated to apoptosis regulation.

In accordance with our goal of screening for

signals produced by epithelial cells to indicate
LPS exposure, we selected 4 genes from
the microarray data for further analysis. Based on
the

relatively

strong

signal

intensity

from

the microarray chips, the expression of

CXCL5

was examined by traditional northern blot analy-
sis. This technique revealed that

CXCL5 was con-

sistently induced in LPS-treated cells of all 4 cows
(Figure 1). Subsequent quantification of band in-
tensity indicated that LPS challenge caused
a 10.1-fold increase (P < 0.01) in the expression of

CXCL5, which encodes an epithelial-derived
chemokine (Figure 2).

Three other genes, which had a relatively low

signal intensity on the microarray slides, were se-
lected for further examination by the very sensi-
tive Q-RT-PCR technique. The expression of
RANTES, which encodes a CC cytokine, was dra-
matically increased (P < 0.01) by LPS exposure.
The mean

DCt for RANTES declined from

11.1 ± 1.0 to 3.4 ± 0.3 in response to LPS.
This corresponds to a 208-fold, LPS-mediated, in-
duction in RANTES gene expression (Figure 3a).
With a similar analysis, the

DCt of IL-6, whose

product is known for its cytokine activity in
the acute-phase response, declined (P < 0.01) from
10.0 ± 0.3 to 6.7 ± 0.3, which corresponds to
a 10.5-fold induction (Figure 3b). The expression
of

T-PA, whose product is involved in

174

R. Pareek et al.

Table 1. List of top ten up-regulated genes in mammary epithelial cells in response to LPS treatment for 6 hours, as
detected by microarray analysis (P

< 0.02)

Gene name

Gene product

Ontology and/or molecular function of gene product

Fold

change

CXCL5

epithelial-derived neutrophil-activating
protein ENA-78

neutrophil activator

2.46

IL-1ß converting en-
zyme/Caspase-13B

apoptosis-related cysteine protease

caspase activator

1.44

IL-6

B-cell stimulatory factor 2

cytokine activity for acute-phase response; final dif-
ferentiation of B-cells into Ig-secreting cells

1.39

T-PA

plasminogen activator (tissue type)

binds to fibrin with high affinity, activates plasmin

1.36

IAP-1

NSD inhibitor of apoptosis protein 1

apoptotic suppressor

1.36

Caspase-13A

apoptosis-related cysteine protease

caspase activator

1.33

KIAA1554

double cortin domain-containing pro-
tein 2 (RU2S protein).

intracellular signaling cascade

1.27

RANTES/CCL5

member 5 of small inducible cytokine
subfamily A (Cys-Cys)

chemoattractant for blood monocytes

1.26

Beta 2-microglobulin

beta 2-microglobulin (beta2M)

beta-chain of major histocompatibility complex
class I molecules, acting as MHC class I receptor

1.25

PIAP

putative inhibitor of apoptosis

unknown

1.25

Figure 1. Northern blot analysis of CXCL5 gene
expression in RNA (8µg/lane) from control (CTL) and
LPS-treated mammary epithelial cells, derived from
4 cows, and treated for 6 hours with LPS (1µg mL

–1

).

ß-actin expression was used as a lane loading control.

Bands were visualized with a phosphoimager.

Figure 2. Quantification of northern blot analysis of
CXCL5 gene expression in control and LPS-treated
mammary epithelial cells derived from 4 cows and treated
for 6 hours with LPS (1µg mL

–1

). *Treatment means are

significantly different (P<0.01).

background image

plasminogen activity, was also induced by LPS
treatment, although to a much lesser extent. Nor-
malized cycle threshold values of

T-PA declined

(P < 0.01) from 9.65 ± 0.7 to 8.2 ± 0.8 due to LPS
exposure, corresponding to a 3-fold induction of
T-PA gene expression (Figure 3c).

Discussion

The use of microarrays in conjunction with other
genomic and proteomic methods should provide
complementary tools for selection of livestock
with superior health and performance attributes.
In the present study, we further exploit our model
system (Wellnitz and Kerr 2004) to examine
the direct effect of LPS on bovine mammary epi-
thelial cells by gene expression profiling as well as
independent confirmation of expression changes
by quantitative techniques. A similar approach
was used to profile parturition-induced changes in
expression of genes of bovine blood neutrophils
(Madsen et al. 2004).

A key component of the host innate immune re-

sponse to mastitis is a rapid increase in concentra-
tions of cytokines in milk (Bannerman et al.
2004a; 2004b). Bovine lymph also contains in-
creased levels of cytokines, such as IL-8 and
TNF-á (Persson Waller et al. 2003) in response to
LPS exposure. However, the contribution of
the mammary epithelial cell to this response is un-
known. Our microarray and Q-RT-PCR results in-
dicated that there was a marked induction of IL-6
production by bovine epithelial cells after stimula-
tion with LPS. This cytokine, along with IL-1ß
and TNFá, are known to play major roles in the in-
flammatory and febrile responses to infection
(Virta et al. 2002; Dinarello 1996). Furthermore,
we have previously demonstrated a rapid induc-
tion of TNFá in response to LPS and

S. aureus in-

fection in this epithelial cell culture system
(Wellnitz and Kerr 2004).

Our microarray gene list indicated that the pro-

duction of two novel chemokines, RANTES and
CXCL5,

was

also

induced

by

LPS.

The chemokines are grouped into 4 families ac-
cording to the number and arrangement of con-
served N-terminal cysteine motifs: C, CC, CXC,
and CX

3

C. The CXC chemokines are subdivided

into two classes based on the presence or absence
of a tripeptide motif, Glu-Leu-Arg (ELR), which
is N-terminal to the conserved CXC region. Mem-
bers that contain the motif (ELR

+

) are potent

chemoattractants for neutrophils and promoters of
angiogenesis, whereas those that do not contain
the motif (ELR

) are potent chemoattractants for

mononuclear cells. Representatives of the ELR

+

CXC chemokines are structurally similar, includ-
ing IL-8/CXCL8, and ENA-78/CXCL5 (Cole et
al. 2001). RANTES is a member of the CC family
of chemokines.

The remarkable induction of chemokine gene

expression by the epithelial cell lends strong sup-
port to its role in stimulating migration of
polymorphonuclear

neutrophil

leukocytes

(PMNs) into the mammary gland to provide de-
fense against invading mastitis pathogens (Paape
et al. 2003). These neutrophil-attracting che-
mokines, known to be produced by epithelial cells,
could lead to cell-specific markers for immune re-
sponse in future

in vitro studies. A recent study

(Ruddy et al. 2004) has demonstrated that IL-17
and TNFá cooperatively induce the production of
the

LPS-inducible

chemokine

CXCL5

in

the preosteoblast murine cell line, MC3T3. Sig-
naling deficiency of IL-17 in mice causes dramatic
reductions in CXCL5 and subsequent neutrophil
chemotaxis, resulting in an increased susceptibil-
ity to bacterial infection. The structurally similar
IL-8 is also known to be produced by mammary

Gene expression in LPS-challenged cells

175

Figure 3.

RANTES (a), IL-6 (b), and T-PA (c) gene expression in control (CTL) and LPS-treated mammary epithelial

cells, as measured by quantitative RT-PCR. ÄCt values represent Ct values of

RANTES, IL-6 and T-PA after subtraction

of Ct values for ß-actin (internal control). *P < 0.01 between control and treated cells.

background image

epithelial cells in response to LPS (Wellnitz and
Kerr 2004, Boudjellab et al. 1998).

Further evidence of epithelial signaling of in-

fection is indicated by the >200-fold induction of
RANTES gene expression. RANTES/CCL5 is one
of several CC cytokine genes involved in
immunoregulatory and inflammatory processes.
The CC cytokines are proteins characterized by
two adjacent cysteines. The cytokine encoded by
this gene functions as a chemoattractant for blood
monocytes,

memory

T

helper

cells

and

eosinophils.

Several

molecules

along

with

RANTES, which mediate leukocyte trafficking in
the immune system, are expressed in the mam-
mary gland (Nishimura 2003) and found in milk
(Bannerman et al. 2004a; 2004b). The present
study implicates the epithelial source of this
chemokine.

Our results indicate that LPS-challenged mam-

mary cells may contribute indirectly to reduced
milk

quality

and

tissue

damage

through

up-regulation of

T-PA gene expression. Conver-

sion of plasminogen to plasmin generates a source
of proteolytic activity in the bovine mammary
gland (Zavizion et al.1996). Confirmation of

T-PA

gene expression by Q-RT-PCR supports previous
studies (Zachos et al. 1992) on the effects of masti-
tis and stage of lactation on plasminogen activator
(PA) activity in milk, where PA activity is in-
creased during severe mastitis.

In conclusion, the present study has used

microarray technology to uncover a remarkable
induction of

CXCL5 and RANTES gene expression

in bovine mammary epithelial cells in response to
LPS. Without the array data, it is unlikely that we
would have chosen these two candidate genes for
further investigations. Marked induction of

IL-6

and

T-PA expression by bovine epithelial cells af-

ter stimulation with LPS confirms that the prod-
ucts of those genes act as a pro-inflammatory
cytokine and as an agent stimulating tis-
sue-degradation, respectively.

Acknowledgements. This study was supported by
the grant VT-AS-034CG (D.E.K.) from the Vermont
Agricultural Experiment Station, USDA.

REFERENCES

Aderem A, Ulevitch RJ, 2000. Toll-like receptors in

the induction of the innate immune response. Na-
ture 406:782–787.

Bannerman DD, Paape MJ, Lee JW, Zhao X, Hope JC,

Rainard P, 2004a.

Escherichia coli and Staphylo-

coccus aureus elicit differential innate immune re-

sponses following intramammary infection. Clin
Diagn Lab Immunol 11:463–472.

Bannerman DD, Paape MJ, Hare WR, Hope JC, 2004b.

Characterization of the bovine innate immune re-
sponse to intramammary infection with

Klebsiella

pneumoniae. J Dairy Sci 87: 2420–2432.

Boudjellab N, Chan-Tang HS, Li X, Zhao X, 1998.

Interleukin 8 response by bovine mammary epithe-
lial cells to lipopolysaccharide stimulation. Am J
Vet Res 59: 1563–1567.

Burton JL, Erskine RJ, 2003. Mastitis and immunity:

some new ideas for an old disease.

Veterinary

Clinics of North America: Food Animal Practice
19: 1–45.

Cole AM, Ganz T, Liese AM, Burdick MD, Liu L,

Strieter RM, 2001. Cutting edge: IFN-inducible
ELR

-

CXC chemokines display defensin-like

antimicrobial activity. J Immuno 167: 623–627.

Diez-Fraile

A,

Meyer

E,

Burvenich

C,

2003.

Sympathoadrenal and immune system activation
during the periparturient period and their associa-
tion with bovine coliform mastitis. A review. Vet Q
25: 31–44.

Dinarello CA, 1996. Cytokines as mediators in

the pathogenesis of septic shock. Curr Top
Microbiol Immunol 216: 133–165.

Hoffmann JA, Kafatos FC, Janeway CA, Ezeko-

witz RA, 1999. Phylogenetic perspectives in innate
immunity. Science 284: 1313–1318.

Livak KJ, Schmittgen TD, 2001. Analysis of relative

gene expression data using real-time quantitative
PCR and the 2(-Delta Delta C(T)) method. Methods
25: 402–408.

Madsen SA, Chang LC, Hicky MC, Rosa GJ,

Coussens PM, Burton JL, 2004. Microarray analy-
sis of gene expression in blood neutrophils of
parturient cows. Physiol Genomics 16: 212–221.

Nishimura T, 2003. Expression of potential lympho-

cyte trafficking mediator molecules in the mam-
mary gland. Vet Res 34: 3–10.

Okada H, Ito T, Ohtsuka H, Kirisawa R, Iwai H,

Yamashita K, Yoshino T, Rosol TJ, 1997. Detection
of interleukin-1 and interleukin-6 on cryopreserved
bovine mammary epithelial cells in vitro. J Vet Med
Sci 59: 503–507.

Paape M, Bannerman D, Zhao X, Lee J, 2003. The bo-

vine neutrophil: Structure and function in blood and
milk. Vet Res 34: 597–627.

Paape M, Mehrzad J, Zhaom X, Detilleuxm J,

Burvenich C, 2002. Defense of the bovine mam-
mary gland by polymorphonuclear neutrophil leu-
kocytes.

J Mammary Gland Biol Neoplasia 7:

109–121.

Persson Waller K, Colditz IG, Lun S, Ostensson K,

2003. Cytokines in mammary lymph and milk dur-
ing endotoxin-induced bovine mastitis. Res Vet Sci
74: 31–36.

Rainard P, Riollet C, 2003. Mobilization of neutrophils

and defense of the bovine mammary gland. Reprod
Nutr Dev 43: 439–457.

176

R. Pareek et al.

background image

Ruddy MJ, Shen F, Smith JB, Sharma A, Gaffen SL,

2004.

Interleukin-17

regulates

expression

of

the CXC chemokine LIX/CXCL5 in osteoblasts:
implications for inflammation and neutrophil re-
cruitment. J Leukoc Biol 76: 135–144.

SAS Institute. (2000) SAS/STAT Software (ver. 8).

Cary, NC: SAS Institute.

Sordillo LM, Streicher KL, 2002. Mammary gland im-

munity and mastitis susceptibility. Mammary
Gland Biol Neoplasia 7: 135–146.

Talhouk RS, Neiswander RL, Schanbacher FL, 1990.

In vitro culture of cryopreserved bovine mammary
cells on collagen gels: synthesis and secretion of ca-
sein and lactoferrin. Tissue Cell 22: 583–599.

Virta M, Murme M, Helminen M, 2002. Increased

plasma levels of pro- and anti-inflammatory
cytokines in patients with febrile seizures. Epilepsia
43: 920–923.

Wellnitz O, Kerr DE, 2004. Cryopreserved bovine

mammary cells to model epithelial response to in-
fection. Vet Immunol Immunopathol 101: 191–202.

Wilson DJ, Gozalezr N, Hertl J,

Schulte HF,

Bennett GJ, Schukken YH, Grohn YT, 2004. Effect
of clinical mastitis on the lactation curve: A mixed
model estimation using daily milk weights. J Dairy
Sci 87: 2073–2084.

Wolfinger RD, Gibson G, Wolfinger ED, Bennett L,

Hamadeh H, Bushel P, Afshari C, Paules RS. 2001.
Assessing

gene

significance

from

cDNA

microarray expression data via mixed models.
J Comput Biol 8: 625–637.

Yang YH, Dudoit S, Luu P, Lin DM, Peng V, Ngai J,

Speed

TP,

2002.

Normalization

for

cDNA

microarray data: a robust composite method ad-
dressing single and multiple slide systematic varia-
tion. Nucleic Acids Res. 30:e15.

Zachos T, Politis I, Gorewit RC, Barbano DM, 1992.

Effect of mastitis on plasminogen activator activity
of milk somatic cells. J Dairy Res 59: 461–467.

Zavizion B, Heegard CW, White J, Cheli F, Politis I,

1996. Synthesis of plasminogen activator inhibitor
1 by bovine mammary epithelial and myoepithelial
cell lines. J Dairy Res 63: 451–458.

Gene expression in LPS-challenged cells

177


Wyszukiwarka

Podobne podstrony:
2005 01 46
kk, ART 177 KK, III KK 270/04 - wyrok z dnia 10 maja 2005 r
Axiom Volume 1Tutorial (2005)
kpk, ART 553 KPK, III KK 177/05 - wyrok z dnia 7 listopada 2005 r
dr dobbs journal volume 30 issue 2 number 369 february 2005 een www!osiolek!com LJHWNJQZ7RJVY7VF3ROE
auto swiat 2005 46 dodatek polish magazine ebook unk RUOYRYATV3ZURY2S3DE6IKSV2FVUAG4EJG7QLYY
OP 2005 3 177
46 Phys Rev B 72 Rapid Commun 201401 2005
BS EN ISO 1133 2005 Plastics Determination of the melt mass flow rate (MFR) and the melt volume flow
Axiom Volume 1Tutorial (2005)
AM1 2005 W1upg
Wytyczne ERC 2 2005
BYT 2005 Pomiar funkcjonalnosci oprogramowania
Wyklad3 2005

więcej podobnych podstron