• Keine Ergebnisse gefunden

Human-Restricted Bacterial Pathogens Block Shedding of Epithelial Cells by Stimulating Integrin Activation

N/A
N/A
Protected

Academic year: 2022

Aktie "Human-Restricted Bacterial Pathogens Block Shedding of Epithelial Cells by Stimulating Integrin Activation"

Copied!
5
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Human-Restricted Bacterial Pathogens Block Shedding of Epithelial Cells by Stimulating Integrin Activation

Petra Muenzner/ Verena Bachmann/ Wolfgang Zimmermann,2 Jochen HentscheL,1,3 Christof R. Hauck1,4*

CoLonization of mucosaL surfaces is the key initiaL step in most bacteriaL infections. One meehanism proteeting the mueosa is the rapid shedding of epitheLiaL eeILs, aLso termed exfoLiation, but it is uncLear how pathogens eounteraet this proeess. We found that eareinoembryonie antigen (CEA)-binding baeteria eoLonized the urogenitaL traet of CEA transgenic mice, but not of wild-type mice, by suppressing exfoLiation of mucosaL eeILs. CEA binding triggered de novo expression of the transforming growth faetor reeeptor CDIOS, ehanging foeaL adhesion eomposition and aetivating ß1 integrins. This manipuLation of integrin inside-out signaLing promotes effieient mueosaL coLonization and represents a potentiaL target to prevent or eure baeteriaL infeetions.

D

wing colonization of mucosal surfaces, incoming microbes must cope with mul- tiple host defenses (I). One protectlve mechanism ofthe mucosa in stratified and squa- mous tissues is the accelerated tumover anel shedding of sllperficial epithelial cells, also re- felTed to as exfoliation (2-4). Althollgh in vitro

studies have suggested that microbes modulate cell detachment (5, 6), it is cUITently unknown how s'uccessful mucosal pathogens deal with the ex- foliation response in vivo. Neisseria gOllorrllOeae.

a Gram-negative microorganism, causes one of the most common sexllally transmitted diseases worldwide (7). Even though these bacteria can

induce the exfoliation of host cells upon contact (8-11), they are able to establish themselves on virtually every mllcosal surface of the human body.

To investigate how gonococci manage to col- onize the urogenital mucosa efficiently, we per- fonned vaginal infection of fernale mice (12). In line with the innate capacity of epithelial cells to re- spond to this bacterial challenge, N. gonorrhoeae tIiggered detachment of superticial epithelial cells within 20 hours (Fig. JA) and only small numbers of gonococci could be re-isolated from wild-type mice (Fig. I B). Gonococci are aelapted to humans as therr sole natural host. One of the host-specific vimlence traits that gonococci share with olher specialized mucosal colonizers, including Hae- moplzilus injluenzae. Moraxella calarrhalis, and N meningitidis, is the ability to recognize hu-

lLehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, 784S7 Konstanz, Germany. 'Tumor Immunology Laboratory, LlFE Center, Ludwig-Maximilians-Universität Mün- chen, 81377 München, Germany. 3EM Service, Fachbereich Biologie, Universität Konstanz, 78457 Konstanz, Germany.

'Konstanz Research School Chemical Biology, Universität Konstanz, 78457 Konstanz, Germany.

*To whom correspondence should be addressed. E-mai!:

eh ristol. ha uck@uni-konstanz.de

1197 First publ. in: Science 329 (2010), 5996, pp. 1197-1201

(2)

man carcinoembryonic antigen-related cell adhe- sion molecules (CEACAMs) (/3,14). To engage CEACAMs, gonococci use outer membrane ad- hesins of the colony opacity-associated (Opa) protein family [OpaCEA; for a review see (15)].

Bacterial engagement ofhuman CEACAMs can block detachment of infected epithelial cells in vitro, which suggests that gonococci and other CEACAM-binding bacteria might use CEACAMs to modulate exfoliation (5).

To test the hypothesis that CEACAM en- gagement by bacteria attenuates epithelial exfo- liation and promotes mucosal colonization, we used a humanized mouse model of gonococcal urogenital tract infection: CEA transgenic (CEAtg) mice harboring the human CEACAM5 gene (/6).

In CEAtg mice, CEA can be detected on mucosal surfaces including the female urogenital tract (fig.

SIA). Wild-type and CEAtg female mice were

Fig. 1. CEA binding facilitates mucosal coloniza- tion and blocks exfoliation of superficial epithelial cells. (A) Female wild-type mice were infected for 24 hours with piliated, non-opaque gonococci, or remained uninfected. Genital tracts were excised, fixed, and pracessed for SEM. Micrographs show the luminal surface of the upper vaginal and cervical region. Exfoliating cells are highlighted by arrows. (8) Schematic representation of the in- fection pratocol used with respect to pretreatment of the mice, vaginal infection, and re-isolation of bacteria (i. indicates the time point of lentiviral transduction). The graph shows colonization of wild-type (0) or CEAtg (. ) female mice with the indicated bacterial strains. Each circle reflects the number of bacteria re-isolated fram an individual animal (n = 8 unless indicated otherwise); da ta were compiled fram six experiments. The dotted line indicates the lower detection limit (40 bacteria per anima\). The median for each experimental group of animals is noted; groups were compared against numbers isolated from Ngo OpaCEA-infected CEAtg mice by two-tailed Mann-Whitney U-test (*P <

0.001). (C) Genital tracts infected as in (8) were excised and processed for SEM. Pictures (at two different magnifications, as indicated by the scale bars) show the luminal surface of the upper vaginal and cervical regions. Inset shows adherent bacteria (black arrowhead). (0) Quantification of exfoliating epithelial cells. Bars represent mean ± SD of exfoliating cells in an area of about 0.075 mm2 (n = 24); groups were compared against uninfected mice by one-tailed Mann-Whitney U-test (*P <

0.01).

vaginally infected with I O~ bacteria, and coloniz- ing bacteria were recovered by urogenital swabs 24 hours later. Consistent with the initial observa- tions, only a few gonococci could be re-isolated from wild-type mice irrespective of the bacterial phenotype, whereas the numbers of gonococci ex- pressing OpacEA proteins recovered from CEAtg mice were higher by a factor of about 50 to 100 (Fig. I B). The OpacEA protein expression by re- isolated bacteria was unaltered (fig. SI B). Some re-isolates expressed additional Opa proteins, further pointing to an in vive advantage of opaque phenotypes. Isogenic gonococcal strains lacking Opa protein expression (Opa-), strains expressing type IV pili (Opa-jp"'), or strains expressing a heparan sulfate proteoglycan-binding Opa pro- tein (Opar-rspc;) (/7, 18) could not be isolated in increased numbers from CEAtg mice (Fig. I B).

Scanning electron microscopy (SEM) ofthe upper

A

'tI GI

-

(,)

.!

c:

'c

::l

o Cl Z

B

; *

vaginal tract revealed massive exfoliation of the superficial epithelial layer in wild-type animals infected with OpacEA-expressing N. gonorrhoeae (Ngo OpaCEA) as weil as in CEAtg animals in- fected with non-CEACAM-binding gonococci (Fig. I C). Vast exfoliation was already evident at low magnification; at higher magnification, the detachment of multiple superficial epithelial cells was apparent (Fig. I C). In contrast, exfoliation was not elevated in CEAtg mice infected with Ngo OpaCEA (Fig. I, C and 0). Thus, the OpaCEA- CEA interaction suppresses exfoliation and fa- cilitates mucosal colonization.

In the urogenital tract of infected mice, gonococ- ci colocalized with CEA-positive cells on the vagi- nal surface of CEAtg mice (Fig. 2, A and B).

Gonococci lacking Opa protein expression (Ngo Opa-) were rarely detected on the mucosal sur- face ofCEAtg mice, and gonococci were absent in

Pretreatment wlth 17ß.estradlol and Irlmethoprlm

o 2

wildtype

3!

* * *

88

~ 0 ~o

*

-

8 00

0 0 ~

1

* •

••

4 days

CEAtll

g :J:

I

... I ...

.L - ... *

...

I

- .. *

...

,.~

.. ..

~~

... , ....

~.~.~

.... .

~.?

...

! ... ~ ... .

c

"

Ol-~

Oll!

:(0 Wo 00>

Z

+ Ol-e:

0>"

. . 0.

~o Wo 0",

z

n=17 n=11

o

80

*

70 *

CQ

~ 60

CQ

.!!l 60 4i ~ 40

~ c:: 30

~

20

CI>

10

Wildtype 'i' CEAtg 'i'

(3)

Fig. 2. CEA engagement by bacteria triggers CD10S expression in vivo. (A) Genital tracts from wild-type or CEAtg mice infected for 24 hours with OpaCEA- expressing (Ngo OpaCEA) or non-opaque (Ngo Opa-) gonococci were excised, and cryosections were co- stained with antibodies against N. gonorrhoeoe (green) and CEA (red). Cell nuclei were visualized by Hoechst (blue). Ngo OpaCEA bound to the CEA-positive mucosal surface of CEAtg mice are indicated by arrowheads. (8) Quantification of cell-associated bacteria in CEAtg mice.

Bars represent the mean number ± SO of bacteria associated with 100 (ells (n = 3). (C) Cryosections as in (A) were costained with antibodies against N. gonor- rhoeae (green), a rat monoclonal antibody against mu- rine CD10S (red), and nuclei (blue). CD10S expression (arrowheads) on the mucosal surface of CEAtg mice is seen in the vicinity of cell-associated Ngo OpaCEA.

A

E

C

.~C\O m. 1 .c .. GI

"

.,c

..

(JO

~~

c

'"

.a c(

'"

'>

2 a.. u..

CI

~ '"

°2 ....

0>

. -

()

B

2.6

... E'

o:>.C 2

'.::~ cO

1';'1.6 .- g

..

"

.~-e 1

'1)0

<11

c( 0.6 0

A wildtype ~

B 80

o

~<f ",*.,.~~o ~

o<:l~ o<:l CEAtg

2.6 ... c E

0:>.0 2

.=:11

li

1.6

=;

~H 1

1-11

c( 0.6 0

"'GFP

virus

. w/oMn2+

D+ Mn2+

C0105wt Cö'1ö6635 virus virus

D 3

E 2.6

.~~ c 2

~t!!.

=6 ~ 1.6

=~ ....

()O

~

0.6

C0105

o - + - +

7 T

(j ~

"'~

- +

Fig. 3. C010S enhances in- tegrin activity by delocalization of zyxin. (Al Human vaginal epithe- lial cells (hVECs) were transduced with lentivirus encoding GFP, wild-type C010S (CD10Swt), or

CD10S~3S. Cells were used in adhesion assays, and adherent cells were quantified after crystal violet staining. Bars represent mean ± SO of five wells. (8) Cells transduced as in W were replated onto collagen and stimulated with Mn2+ (a global activator of integrin activity), or rernained unstimulated, before analysis with an activation- specifi( integrin ßl antibody (clone 9EG7; active integrin ßl) or an activation-independent integrin ßl antibody (clone AIIB2; total integrin ßl)' Bars represent the mean ± SO of five wells of a rep- resentative experiment repeated twice with similar results. (C) hVECs were transduced as in (A), and parallel sam pIes were stained for zyxin or vinculin. Small arrowheads highlight zyxin at focal adhesions, which is missing in CD10Swt- expressing cells. Vinculin-positive focal adhesions (arrows) are found in all cells. (D) HeLa cells were transfected with vectors encoding GFP, GFP-zyxin, or GFP-zyxin ~2/3L1M,

in the presence or absence of CDlOS. Cells were used in adhesion assays as in (A). Bars represent mean ± SO of five wells.

(4)

wild-type mice, even ifinfected with Ngo Op!lc:Ei\

(Fig. 2, A and B). How can the engagement of api- cally expressed CEA lead to reduced detachment of infected cells? CEACAM-binding by bacteria triggers de novo expression ofCD I 05, a member of the transforming growth factor-ß I receptor (TGFß I R) family, which prornotes cell-matrix ad- hesion and blocks cell migration (5, 19). lndeed, CEA engagement by Ngo OpaCEi\ in vitro stim- ulated the expression ofCDIOS and increased the adhesiveness of infected cells to several extracellu- lar matrix proteins (fig. S2). Moreover, CEACAM- binding bacteria, but not Opa-bacteria, triggered de novo expression ofCDIOS by vaginal epithe- lial cells ofCEAtg mice in vivo (Fig. 2C and fig. S3).

CD I 05, a well-characterized endothelial marker, was present on small blood vessels in the sub- mucosa of wild-type (fig. S4) and CEAtg mice, but was absent !fom uninfected vaginal epithelial cells (Fig. 2C and fig. S3). Thus, CD I 05 expres- sion in response to CEA stimulation by gonococci could be responsible for enhanced matrix adhesion of infected epithelial cells and the suppression of exfoliation.

Cell adhesion to the extracellular matrix is mediated by integrins, and TGFß I Rs influence integrin expression (20). However, CD I 05, a type III nonsignaling member of the TGFßI R family, does not increase the amount of integrin ß I or integrin-associated cytoplasmic proteins (/9, 21). In HeLa cells and 293 cells, CDIOS promoted cell adhesion by stimulating integrin inside-out signaling, leading to enhanced integrin activity (fig. SS). The adhesion-promoting activ-

A

.,

~ 26

0

....

~ 20 ii E

'c ~

16

~ ..

10

~

"

! 6

CI >

0

"

CI

It: 0

ity ofCDIOS was located in the cytoplasmic do- main of CD I 05, wh ich was necessary (fig. S6) and sufficient (fig. S7) for this process. In line with these results, transduction with wild-type CD 105 (CD I OSwt), but not with CD I 05 carrying a 3S-amino acid truncation of the cytoplasmic domain (CD I OSL'13S), enhanced ce li-matrix ad- hesion and increased integrin activity of human vaginal epithelial cells (hVECs) (Fig. 3, A and B). One ofthe cellular factors that associate with the cytoplasmic part of CD 105 is zyxin, a focal adhesion protein that binds CD I 05 via three UM (Lin-Il Isl-1 Mec-3) domains (/9). Zyxin-deficient fibroblasts show enhanced integrin activity and matrix adhesion (22), wh ich was not further stim- ulated by CD I 05 expression (fig. S8). The de- pendence ofCDIOS-stimulated integrin activity on zyxin, which by itself acts as a negative regulator of cell adhesion, could be explained by a scenario where de novo expression of CD 105 interferes with zyxin's inhibitory function at focal adhe- sion sites. In h VECs, wh ich express zyxin but not CD I 05 (fig. S9), zyxin was located at peripheral contact sites (Fig. 3C). However, upon expres- sion of CD 10Swt, but not CD I OSL'13S, zyxin was delocalized (Fig. 3C and fig. S9). In contrast, the localization ofthe focal adhesion protein vinculin was unaltered (F ig. 3C). lncreasing the cellular pool of zyxin by overexpression, but not by ex- pression of a zyxin mutant lacking the second and third UM domains (zyxin L'12/3LIM), abrogated the increased adhesion of CD I OS-transfected cells (Fig. 3D). Thus, CDIOS-mediated delocalization of endogenous zyxin positively influences integrin

wildtype<j> CEAtg<j>

*

*

t>"

~~ !<<l.

»"

~~ !<<l.

Ci Fig. 4. Bacterial colonization is blocked

by inhibition of C010S expression in vaginal epithelial cells or by mucosal delivery of zyxin. (A) Female mice were transduced with GFP, shCOlOS-encoding virus, or left without virus (w/o virus).

Twenty-four hours later, animals were infected with Ngo OpaCEA' and coloniz- ing gonococci were enumerated the next day. Bars represent the mean ± 5D of bacteria re-isolated from six individual animals. Groups were compared against numbers isolated from untransduced CEAtg mice by one-tailed Mann-Whitney U-test (*P< 0.01). (8) 5EM of the mucosal epithelium of mice transduced and in- fected as indicated. Exfoliating epithelial cells are marked by arrows; cell-associated bacteria are indicated by arrowheads.

(C) Quantification of exfoliating epithe- lial cells as in Fig. 10; groups were com- pared against GFP-transduced wild-type mice by one-tailed Mann-Whitney U-test (*P< 0.01). (D and E) Wild-type or CEAtg mice were transduced with lentiviral particles encoding GFP, GFP-zyxin, or GFP-zyxinL'1213l1M, or left without virus (w/o virus) before infection with Ngo OpaCEA' Colonizing gonococci in (D) and exfoliating cells in (E) were enumerated as in (A) and (C), respectively.

.i~ 9"

~o ~C;

.:i' "

Ci .i~ 9"

1\0 ~C;

.:i' "

C wildtype<j> CEAtg<j>

80

~

!!

60 Gi

"

g'40

~

~

20

w

o

wildtype<j>

activity and could be responsible for the enhanced cell adhesion and the suppression of epithelial ex- foliation triggered by CEACAM-binding bacteria.

To test a causal relationship arnong CEACAM- dependent up-regulation of CD 105, suppression of epithelial exfoliation, and irnproved colonization ofthe urogenital mucosa by pathogenic gonococci, we generated a GFP (green fluorescent protein)- and short hairpin RNA-encoding lentivirus tar- geting murine CDIOS (shCDIOS) (fig. SIO). ln vivo, vaginal application of shCD I 05 or a GFP control virus resulted in strong expression ofGFP in the superficial epitheliallayer (fig. SI 0). To in- vestigate the role of CD 1 05 expression for mu- cosal colonization in vivo, we applied shCDIOS or GFP control virus 24 hours before vaginal in- fection of mice. Application of shCD I 05 virus suppressed the ability ofNgo OP3cEi\ to colonize the urogenital tract of CEAtg mice, whereas the GFP control virus had no effect (Fig. 4A). More- over, SEM ofthe mucosa revealed that transduc- tion with shCD 105 virus reversed the phenotype of the infected epithelial cells and allowed pro- nounced exfoliation in response to infection with CEACAM-binding bacteria (Fig. 4, Band C). In CEAtg mice treated with the GFP control virus, Ngo OpaCEi\ were still able to block exfoliation (Fig. 4, Band C). Thus, up-regulation ofCDIOS on mucosal epithelial cells interferes with exfo- Iiation and prornotes gonococcal colonization of the urogenital tract. To further confirm the causal relationship between CD 10S-initiated molecular processes and the suppression of epithelial exfo- liation, we transduced CEAtg mice with lentiviral

CEAtg<j>

E

..

80

~ ! 60

Gi

"

11140

~

c 20

~

w

wildtype<j> CEAtg<j>

* *

(5)

particles driving the expression of zyxin-GFP, zyxin-ö2/3LIM-GFP, or GFP, respective\y (fig.

SI I). Oelivery ofzyxin to the vaginal epithelium did not result in increased exfoliation of the un- infected mucosa (fig. S 11). However, zyxin, but not zyxin-ö2/3LIM, overexpression blocked the colonization ofCEAtg mice by OpaCEA -expressing gonococci (Fig. 40). The overexpression ofzyxin, but not ofzyxin-ö2/3LIM, allowed exfoliation of CEA-positive cells infected with Ngo 0p!lc;EA and reestablished the responsiveness of the vagi- nal mueosa (Fig. 4E and fig. SI I). Thus, CEA- initiated up-regulation of COI05 on superfieial epithelial cells and the resulting delocalization of zyxin fTom integrin-rieh foeal adhesion sites are the eritical moleeular events that allow CEACAM- binding mieroorganisms to eounteraet the exfo- Iiation response.

Although symptomatie gonoeoceal infeetion in humans might be a multistep proeess orches- trated by additional virulence faetors (J 3,23), our results establish a speeifie role of OpaCI'A pro- teins in promoting mueosal eolonization. In vive challenge experiments with male volunteers have revealed that after infeetion of the urethra with non-opaque gonoeoeei, baeteria re-isolated fTom these volunteers almost invariably eonverted to an Opa protein-expressing phenotype (24, 25). In addition to the urogenital traet, members of the CEACAM family are present on all mueosal surfaces including the nasopharynx (26). These mueosal habitats are eolonized by several Gram- .

negative baeterial speeies, whieh make use of unrelated protein adhesins to engage human CEACAMs (27-31). The bloekage of epithelial exfoliation afforded by CEACAM binding might have driven this eonvergent evolution that allows speeialized baeteria to transfonn the mueosa into a dependable platfonn for eolonization.

References and Notes

1. ]. P. Pearson, I. A. Brownlee, in Colonizalion af Mumml Surfaees, ]. P. Nataro, P. S. Cohen, H. l. T. Mobley, ]. N. Weiser, Eds. (ASM Press, Washington, De. 2005), pp. 3-16.

2. M. A. Mulvey el 01., Scienee 282, 1494 (1998).

3. M. A. Mulvey, ]. D. Schilling, ]. ]. Martinez, S. ). Hultgren, Proc. Natl. Acad. Sei. U.S.A. 97, 8829 (2000).

4. I. U. Mysorekar, M. A. Mulvey, S. ]. Hultgren, ]. I. Gordon, ]. Biol. Chem. 277, 7412 (2002).

5. P. Muenzner, M. Rohde, S. Kneitz, C. R. Hauck,]. Cell Bial. 170, 825 (2005).

6. M. Kim el 01., Nalure 459, 578 (2009).

7. WHO, Global Prevalenee and Incidenee 0/ Seleeted Curable Sexually Transmitted Infeetions-Overview and Estimales (World Health Drganization, Geneva, 2001).

8. M. A. Melly, C. R. Gregg, Z. A. McGee, ]. In/eet. Dis. 143, 423 (1981).

9. Z. A. McGee, A. P. ]ohnson, D. Taylor-Robinson,]. Infeel.

Dis. 143, 413 (1981).

10. K. F. Tjia, ]. P. van Putten, E. Pels, H. C. Zanen, Grae/es Areh. Clin. Exp. Ophlhalmol. 226, 341 (1988).

11. I. M. Mosleh, H. ]. Boxberger, M. ). Sessler, T. F. Meyer, Infeet. Immun. 65, 3391 (1997).

12. See supporting material on Scienee Online.

13. M. Virji, Nal. Rev. Micrabiol. 7, 274 (2009).

14. M. Voges, V. Bachmann, R. Kammerer, U. Gophna, C. R. Hauck, BMC Micrabiol. 10, 117 (2010).

15. C. R. Hauck, T. F. Meyer, Curr. Opin. Microbial. 6, 43 (2003).

16. A. M. Eades-Perner el 01., Cancer Res. 54,4169 (1994).

17. ]. P. van Putten, S. M. Paul, EMBO]. 14, 2144 (1995).

18. T. Chen, R. ). Belland, ]. Wilson, ). Swanson,]. Exp. Med.

182, 511 (1995).

19. B. A. Conley el 01., ]. Biol. Chem. 279, 27440 (2004).

20. G. Zambruno el 01., ]. Cell Bial. 129, 853 (1995).

21. M. Guerrero-Esteo el 01., Eur. ]. Cell Biol. 78, 614 (1999).

22. L. M. Hoffman el 01., ]. Cell Bial. 172, 771 (2006).

23. A.]. Merz, M. So, Annu. Rev. Cell Dev. Biol. 16, 423 (2000). 24. ]. Swanson, O. Barrera, ]. Sola, ]. Boslego,]. Exp. Med.

168, 2121 (1988).

25. A. E. ]erse el 01.,]. Exp. Med. 179, 911 (1994).

26. S. Hammarström, Sem in. Cancer Bial. 9, 67 (1999).

27. M. Virji, K. Makepeace, D. ). P. Ferguson:S. M. Watt, Mol. Microbiol. 22, 941 (1996).

28. M. Virji el 01., Mol. Micrabiol. 36, 784 (2000).

29. D.]. Hili, M. Virji, Mol. Microbiol. 48, 117 (2003).

30. c. N. Berger, O. Billker, T. F. Meyer, A. L. Servin, I. Kansau, Mal. Micrabiol. 52, 963 (2004).

31. M. Toleman, E. Aho, M. Virji, Cell. Microbiol. 3, 33 (2001).

32. We thank M. C. Beckerle for zyxin-deficient fibroblasts and antibodies; M. Chudakov for the mKate cDNA;

]. W. Greiner for sending the CEAtg mice; T. F. Meyer for bacterial strains; D. W. Piston for eDNA of mCerulean;

A. ]. Sehaeffer for hVEC cells; D. Vestweber for providing antibody; C. Hentschel, ]. Scharrer, and R. Mak'anyengo for assistance with SEM; and B. Planitz for expert animal care. Supported by Deutsche Forsehungsgemeinsehaft grant Ha2B56/6-1 (C.R.H.).

Supporting Online Material

www.sciencemag .0rg/cgi/contenUful V3 2 9/5 9 9 61119 7IDC 1 Materials and Methods

Figs. SI to S10 References

14 April 2010; accepted 30 ]une 2010 1O.1126/scienee.1190892

Referenzen

ÄHNLICHE DOKUMENTE

Accumulating evidence suggests that intracellular macromolecules such as proteins and nucleic acids are released into the extracellular milieu where they may serve as alarmins and

Our data show that the virus can break through the polarized airway epithelial cells and initiate BVDV infection via both the apical and basolateral domain without any

Here, two well-differentiated respiratory epithelial cell culture systems, porcine precision-cut lung slices (PCLS) and porcine airway air-liquid interface (ALI)

The sialic acids can act as receptor determinants for the HA protein of influenza viruses and mediate the binding of bacteria to viral glycoproteins exposed on the surface

To determine the virulence of A(H1N1)pdm09 influenza viruses, strains isolated in the years following the 2009 influenza pandemic were selected to infect ALI cultures

In the second part of my thesis, I established two primary well-differentiated ALI cultures, porcine tracheal epithelial cells (PTEC) and porcine bronchial epithelial

The effect of Streptococcus suis co-infection on the infection of well- differentiated porcine respiratory epithelial cells by swine influenza

We investigated the susceptibility of bovine airway epithelial cells (BAEC) to infection by the three major viruses associated with the BRDC: bovine respiratory