2014 Russell & Tregoning RSV dSH

1
CONCLUSION RSV infection leads to an IL- 1β response, from a range of cells. The SH protein of RSV inhibits inflammasome function. Deletion of the SH gene modulates the outcome of secondary RSV infection. RSV ΔSH is a vaccine candidate strain which is protective to RSV infection We propose that this is via its pore structure altering the K + flux. INHIBITION OF THE INFLAMMASOME BY THE SMALL HYDROPHOBIC PROTEIN OF RESPIRATORY SYNCYTIAL VIRUS Ryan F. Russell 1 , Jacqueline U. McDonald 1 , Ziyun Zhong 1 , Alex Bukreyev 2 , Peter L. Collins 2 , John S. Tregoning 1 1 Mucosal Infection and Immunity group, St Mary’s Campus, Imperial College London, UK. 2 Laboratory of Infectious Diseases, NIAID/NIH, MD, USA. INTRODUCTION Respiratory syncytial virus (RSV) infects most children in the first year of life and is a major single cause of hospitalization in infants and young children. The small hydrophobic (SH) protein of RSV has recently been described as a viral pore protein (viroporin) which forms a proton-selective channel (figure 1). The function of SH is unknown, but SH knockout viruses are partially attenuated in vivo and are potential vaccine candidates. Here, we present evidence that SH is involved in evasion of the host immune response, inhibiting the inflammasome. Figure 1 . Computational analysis by Gan et al (2013) suggests that the SH protein forms a pentamer containing a pore forming transmembrane region, whose diameter allows only for the passage of small molecules and ions. Further studies reveal potential ion channel activity. The SH proteins of other related viruses show inflammasome modulating activity. FIGURE 1. The SH protein of RSV encodes a viral pore protein (viroporin) that can modulate the inflammasome. GURE 2. RSV ΔSH is protective against RSV infection. FIGURE 3. RSV ∆SH induces a greater IL-1B response than wild type in vitro. FIGURE 4. RSV ∆SH is attenuated in vivo but induces a greater IL-1B response than wild type. FIGURE 5. IL-1B is produced by neutrophils and macrophages in vivo. Figure 2 . Mice were infected with either RSV A2, ΔSH or control treated, 4 weeks later all mice were challenged with RSV A2. Anti-RSV IgG before RSV challenge (A). Weight change (B), lung viral load (C), lung IL-1B (D) and lung cell number (E) after infection. Lung CD4+ (F) and CD8+ (G) T cells on day 4 and day 7. Lung RSV specific CD8 T cells on day 7 (H). Anti-RSV IgG day 7 (I). Figure 4 . Mice were infected intranasally with RSV A2 (wild type) or RSV lacking the SH gene (RSV ∆SH). Weight loss (A), lung viral load (B) and lung cell number (C) were measured after infection. Lung CD4 T (D), CD8 T (E) and DX5+ NK (G) cells were measured by flow cytometry. IL-6 (G), CXCL1/KC (H) and IL-1B (I) were measured in lung supernatants. Figure 3 . HEp-2 cells were infected with *** MOI RSV A2 or RSV ∆SH, viral load was assessed by plaque assay (A). Supernatants were collected and analysed for IL-1B level by ELISA following infection of HEp-2 cells (B), PHBE cells (C), THP-1 cells (D), and neutrophils (F). Points represent mean +/- SEM of n=3 repeats of HEp2, PHBE and THP1 cells and 3 individual PBMC and neutrophil donors. Figure 5 . Mice were infected with RSV A2 intranasally. Expression of IL-1B by Ly6G+ (neutrophils: A) and CD11c/MHCII lo/F480+ (alveolar macrophages: B) was measured by flow cytometry at various time points after infection. Mice were treated with anti-Ly6G depleting antibody (1A8) or control antibody (Con) intraperitoneally, and clodronate liposomes (CL) or empty liposomes (PL) intranasally prior to RSV A2 infection. Neutrophil (C) and alveolar macrophage (D) numbers were analysed by flow cytometry, RSV L gene (E) by RT-PCR and lung IL-1B (F), KC (G) and IL-6 (H) were measured on day 1 after infection. SH K + Casp1 NLRP3 Pro IL-1β Meet the authors! 4 7 10 0 10 1 10 2 10 3 10 4 10 5 10 6 10 7 10 8 10 9 D ays afterC hallenge R SV L gene copies per g Lung R N A Control RSV SH RSV A2 0 5 10 15 20 Anti-R SV IgG g/ml *** *** *** Control R SV SH RSV A2 0 25 50 75 100 125 Anti-R SV IgG g/ml *** *** *** 0 1 2 3 4 5 6 7 85 90 95 100 105 C ontrol RSV SH RSV A2 D ays afterC hallenge % OriginalWeight *** *** ### ### * ### * 4 7 0 5 10 6 1 10 7 2 10 7 D ays afterC hallenge Lung cell num ber * ** 4 7 0 2 4 6 8 D ays afterC hallenge C D 3+C D 8% ** ** 4 7 0 50 100 150 200 250 D ays afterC hallenge Lung IL-1 pg/ml *** *** Control R SV SH RSV A2 0 10 20 30 40 50 R SV (M 2)Specific CD 8 (% total CD8) *** * 4 7 0 2 4 6 8 D ays afterC hallenge CD3 + CD4 + (%) * ** A B C D E F G H I 0 1 2 3 4 5 6 7 90 95 100 105 110 D ays afterinfection % O riginal W eight RSV SH RSV A2 * * * 0 1 2 3 4 5 6 7 10 4 10 5 10 6 10 7 10 8 10 9 D ays afterinfection R SV L gene copies per g Lung R N A 0 1 2 3 4 5 6 7 0 100 200 300 400 500 D ays afterinfection C XC L1 pg/m l ** 0 1 2 3 4 5 6 7 0 100 200 300 400 500 D ays afterinfection IL-6 pg/m l ** 0 1 2 3 4 5 6 7 0 200 400 600 800 1000 D ays afterinfection IL-1 pg/ml 0 1 2 3 4 5 6 7 10 6 10 7 D ays afterinfection Lung cell num ber 0 1 2 3 4 5 6 7 10 4 10 5 10 6 D ays afterinfection Lung C D 4 T cells * 0 1 2 3 4 5 6 7 10 4 10 5 10 6 D ays afterinfection Lung C D 8 T cells * 0 1 2 3 4 5 6 7 10 4 10 5 10 6 D ays afterinfection Lung N K cells * A B D E F C G H I 0 20 40 60 80 0 200 400 600 800 1000 ** * * * H ours afterinfection IL-1 pg/ml 0 20 40 60 80 0 500 1000 1500 ** * * H ours afterinfection IL-1 pg/ml 0 20 40 60 80 0 5 10 15 20 H ours afterinfection IL-1 pg/ml RSV A2 RSV SH C ontrol 0 50 100 150 200 IL-1 pg/ml * 0 20 40 60 80 0 5.0 10 5 1.0 10 6 1.5 10 6 2.0 10 6 H ours afterinfection R SV pfu/m l RSV A2 RSV SH A B C D E PL + C on P L + 1A 8 C L + C on C L + 1A8 0 100 200 300 400 500 Lung IL-1 pg/ml ** *** *** PL + C on P L + 1A 8 C L + C on C L + 1A8 0 100 200 300 400 Lung IL-6 pg/m l ** ** PL + C on PL + 1A8 C L + C on C L + 1A8 0 200 400 600 800 1000 Lung KC pg/m l ** ** PL + C on PL + 1A8 C L + C on C L + 1A8 10 4 10 5 10 6 10 7 10 8 R SV L gene copies per g Lung R NA PL + C on P L + 1A 8 C L + C on C L + 1A8 0 50000 100000 150000 Ly6G + cell count *** *** 0 1 2 3 4 5 6 7 0 10000 20000 30000 40000 50000 Days afterinfection IL-1 Positive Ly6G+ *** ** *** RSV A2 Control 0 1 2 3 4 5 6 7 0 500 1000 1500 Days afterinfection IL-1 + C D 11c + M HCIloF4/80 + Cell count *** *** P L + C on PL + 1A8 C L + C on C L + 1A8 0 2000 4000 6000 C D 11c + M H C IIlo F480 + cell count ** ** * A B C D E F G H

description

INHIBITION OF THE INFLAMMASOME BY THE SMALL HYDROPHOBIC PROTEIN OF RESPIRATORY SYNCYTIAL VIRUS

Transcript of 2014 Russell & Tregoning RSV dSH

Page 1: 2014 Russell & Tregoning RSV dSH

CONCLUSION• RSV infection leads to an IL-1β

response, from a range of cells.

• The SH protein of RSV inhibits inflammasome function.

• Deletion of the SH gene modulates the outcome of secondary RSV infection.

• RSV ΔSH is a vaccine candidate strain which is protective to RSV infection

• We propose that this is via its pore structure altering the K+ flux.

INHIBITION OF THE INFLAMMASOME BY THE SMALL HYDROPHOBIC PROTEIN OF RESPIRATORY SYNCYTIAL VIRUS

Ryan F. Russell1, Jacqueline U. McDonald1, Ziyun Zhong1, Alex Bukreyev2, Peter L. Collins2, John S. Tregoning1

1Mucosal Infection and Immunity group, St Mary’s Campus, Imperial College London, UK.2 Laboratory of Infectious Diseases, NIAID/NIH, MD, USA.

INTRODUCTION• Respiratory syncytial virus (RSV) infects most children in the first year of life and is a major single

cause of hospitalization in infants and young children.

• The small hydrophobic (SH) protein of RSV has recently been described as a viral pore protein (viroporin) which forms a proton-selective channel (figure 1).

• The function of SH is unknown, but SH knockout viruses are partially attenuated in vivo and are potential vaccine candidates.

• Here, we present evidence that SH is involved in evasion of the host immune response, inhibiting the inflammasome.

Figure 1. Computational analysis by Gan et al (2013) suggests that the SH protein forms a pentamer containing a pore forming transmembrane region, whose diameter allows only for the passage of small molecules and ions. Further studies reveal potential ion channel activity. The SH proteins of other related viruses show inflammasome modulating activity.

FIGURE 1. The SH protein of RSV encodes a viral pore protein (viroporin) that can modulate the inflammasome.

4 7100

101

102

103

104

105

106

107

108

109

Days after Challenge

RSV

L ge

ne co

pies

per

g Lu

ng R

NA

Control RSV SH RSV A20

5

10

15

20

Anti-

RSV

IgG

g/m

l

*********

Control RSV SH RSV A20

25

50

75

100

125

Anti-

RSV

IgG

g/m

l

*********

0 1 2 3 4 5 6 785

90

95

100

105

ControlRSV SHRSV A2

Days after Challenge

% O

rigin

al W

eigh

t

*** ***### ###

*###

*

4 70

5106

1107

2107

Days after Challenge

Lung

cell n

umbe

r

***

4 70

2

4

6

8

Days after Challenge

CD3+

CD8%

****

4 70

50

100

150

200

250

Days after Challenge

Lung

IL-1

pg

/ml

******

Control RSV SH RSV A20

10

20

30

40

50

RSV

(M2)

Spe

cific

CD8

(% to

tal C

D8)

****

4 70

2

4

6

8

Days after Challenge

CD3+ CD

4+ (%)

***

A B C D

E F G H I

0 1 2 3 4 5 6 790

95

100

105

110

Days after infection

% O

rigin

al W

eigh

t

RSV SHRSV A2

* **

0 1 2 3 4 5 6 7104

105

106

107

108

109

Days after infection

RSV

L g

ene

copi

espe

r g

Lung

RN

A

0 1 2 3 4 5 6 70

100

200

300

400

500

Days after infection

CX

CL1

pg/

ml **

0 1 2 3 4 5 6 70

100

200

300

400

500

Days after infection

IL-6

pg/

ml

**

0 1 2 3 4 5 6 70

200

400

600

800

1000

Days after infection

IL-1

pg

/ml

0 1 2 3 4 5 6 7106

107

Days after infection

Lung

cel

l num

ber

0 1 2 3 4 5 6 7104

105

106

Days after infection

Lung

CD

4 T

cells

*

0 1 2 3 4 5 6 7104

105

106

Days after infection

Lung

CD

8 T

cells

*

0 1 2 3 4 5 6 7104

105

106

Days after infection

Lung

NK

cel

ls *

A B

D E F

C

G H I

0 20 40 60 800

200

400

600

800

1000

**

*

*

*

Hours after infection

IL-1

pg

/ml

0 20 40 60 800

500

1000

1500

**

*

*

Hours after infection

IL-1

pg

/ml

0 20 40 60 800

5

10

15

20

Hours after infection

IL-1

pg

/ml

RSV A2 RSV SH Control0

50

100

150

200

IL-1

pg

/ml

*

0 20 40 60 800

5.0105

1.0106

1.5106

2.0106

Hours after infection

RSV

pfu

/ml

RSV A2RSV SH

A B C D E

PL + Con PL + 1A8 CL + Con CL + 1A80

100

200

300

400

500

Lung

IL-1

pg

/ml **

******

PL + Con PL + 1A8 CL + Con CL + 1A80

100

200

300

400

Lung

IL-6

pg/

ml

****

PL + Con PL + 1A8 CL + Con CL + 1A80

200

400

600

800

1000

Lung

KC

pg/

ml

****

PL + Con PL + 1A8 CL + Con CL + 1A8104

105

106

107

108

RS

V L

gen

e co

pies

per

g Lu

ng R

NA

PL + Con PL + 1A8 CL + Con CL + 1A80

50000

100000

150000Ly

6G+

cell

coun

t

******

0 1 2 3 4 5 6 70

10000

20000

30000

40000

50000

Days after infection

IL-1

P

ositi

ve L

y6G

+

*** **

***RSV A2Control

0 1 2 3 4 5 6 70

500

1000

1500

Days after infection

IL-1

+C

D11

c+ MH

CI l

oF4/

80+

Cel

l cou

nt

******

PL + Con PL + 1A8 CL + Con CL + 1A80

2000

4000

6000

CD

11c+

MH

CII

lo F

480+

cell

coun

t

****

*

A B

C D

E F

G H

FIGURE 2. RSV ΔSH is protective against RSV infection.

FIGURE 3. RSV ∆SH induces a greater IL-1B response than wild type in vitro.

FIGURE 4. RSV ∆SH is attenuated in vivo but induces a greater IL-1B response than wild type.

FIGURE 5. IL-1B is produced by neutrophils and macrophages in vivo.

Figure 2. Mice were infected with either RSV A2, ΔSH or control treated, 4 weeks later all mice were challenged with RSV A2. Anti-RSV IgG before RSV challenge (A). Weight change (B), lung viral load (C), lung IL-1B (D) and lung cell number (E) after infection. Lung CD4+ (F) and CD8+ (G) T cells on day 4 and day 7. Lung RSV specific CD8 T cells on day 7 (H). Anti-RSV IgG day 7 (I).

Figure 4. Mice were infected intranasally with RSV A2 (wild type) or RSV lacking the SH gene (RSV ∆SH). Weight loss (A), lung viral load (B) and lung cell number (C) were measured after infection. Lung CD4 T (D), CD8 T (E) and DX5+ NK (G) cells were measured by flow cytometry. IL-6 (G), CXCL1/KC (H) and IL-1B (I) were measured in lung supernatants.

Figure 3. HEp-2 cells were infected with *** MOI RSV A2 or RSV ∆SH, viral load was assessed by plaque assay (A). Supernatants were collected and analysed for IL-1B level by ELISA following infection of HEp-2 cells (B), PHBE cells (C), THP-1 cells (D), and neutrophils (F). Points represent mean +/- SEM of n=3 repeats of HEp2, PHBE and THP1 cells and 3 individual PBMC and neutrophil donors.

Figure 5. Mice were infected with RSV A2 intranasally. Expression of IL-1B by Ly6G+ (neutrophils: A) and CD11c/MHCII lo/F480+ (alveolar macrophages: B) was measured by flow cytometry at various time points after infection. Mice were treated with anti-Ly6G depleting antibody (1A8) or control antibody (Con) intraperitoneally, and clodronate liposomes (CL) or empty liposomes (PL) intranasally prior to RSV A2 infection. Neutrophil (C) and alveolar macrophage (D) numbers were analysed by flow cytometry, RSV L gene (E) by RT-PCR and lung IL-1B (F), KC (G) and IL-6 (H) were measured on day 1 after infection.

SHK+

Casp1

NLRP3

Pro IL-1β

Meet the authors!