10
Mesangial cell complement receptor 1-related protein y limits complement-dependent neutrophil accumulation in immune complex glomerulonephritis Introduction The complement system is tightly regulated by more than 30 plasma and cell membrane proteins to protect self tissue from injury. 1 In humans, decay-accelerating factor (DAF; CD55) and membrane cofactor protein (MCP; CD46) are the main membrane-bound intrinsic regulators of C3 activation. In rodents, complement receptor 1 (CR1) related gene/protein y (Crry) has combined decay- accelerating and factor I cofactor activity for C3b and C4b; therefore, it combines functionalities of both DAF and MCP. 2 It has been shown using transgenic and recombinant protein techniques that Crry is protective in different immune complex-mediated kidney diseases. 3–6 Deficiency of Crry led to early embryonic lethality as a result of unrestricted complement activation and concomitant pla- cental inflammation, which could be averted by coexisting C3 deficiency in Crry )/) C3 )/) mice. 7 Because of the resultant C3 deficiency, the role of Crry in protecting Lihua Bao, 1 Ying Wang, 1 Peili Chen, 1 Menaka Sarav, 1 Mark Haas, 2 Andrew W. Minto, 1 Miglena Petkova 1 and Richard J. Quigg 1 1 Section of Nephrology, The University of Chicago, Chicago, IL, and 2 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA doi:10.1111/j.1365-2567.2009.03102.x Received 16 January 2009; revised 6 March 2009; accepted 11 March 2009. Correspondence: L. Bao, MD, Section of Nephrology, The University of Chicago, 5841 S. Maryland Avenue, MC5100 Chicago, IL 60637, USA. Email: [email protected] Senior author: Richard J. Quigg, email: [email protected] Summary The absence of complement receptor 1 (CR1) related gene/protein y (Crry) leads to embryonic lethality as a result of unrestricted complement activation and concomitant neutrophil infiltration. Here we used Crry )/) C3 +/) mice to investigate the role of Crry in the pathogenesis of immune complex glomerulonephritis (GN). After 3 weeks of immunization with horse spleen apoferritin, six of nine Crry )/) C3 +/) mice and none of the six control C3 +/) mice developed proliferative GN (P =0010). After 5 weeks of immunization, GN scores in Crry )/) C3 +/) mice were 067 ± 022 mean ± standard error of the mean (SEM), compared with 032 ± 016 in C3 +/) mice. Glomerular hypercellularity was attributable to neutrophil infiltration in mice with GN (17±03/glomerulus) compared with those without GN (04±01/glomerulus) (P =0001). Absent stain- ing for a-smooth muscle actin and proliferating cell nuclear antigen sug- gested that mesangial cell proliferation did not play a significant role in this model. Serum C3 levels in Crry )/) C3 +/) mice were approximately 20% and 30% those of wild-type mice and C3 +/) mice, respectively. To determine whether this acquired hypocomplementaemia was relevant to this GN model system, Crry )/) C3 +/) mouse kidneys were transplanted into wild-type mice followed by immunization with apoferritin for 1 or 2 weeks. Surprisingly, none of the Crry )/) C3 +/) mouse kidneys developed GN at these early time-points, indicating that increasing circulating C3 levels several-fold did not increase susceptibility to GN. Renal expression of decay-accelerating factor was not different among any of the groups studied. Thus, our data indicate that mesangial cell Crry limits comple- ment activation and subsequent neutrophil recruitment in the setting of local immune complex deposition. Keywords: complement; inflammation; knockout mice Please cite this article in press as: Bao L. et al. Mesangial cell complement receptor 1-related protein y limits complement-dependent neutrophil accumulation in immune complex glomerulonephritis, Immunology (2009) doi: 10.1111/j.1365-2567.2009.03102.x Abbreviations: CR, complement receptor; Crry, CR1-related gene/protein y; GN, glomerulonephritis; hpf, high-power field; IF, immunofluorescence. Ó 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e895 IMMUNOLOGY ORIGINAL ARTICLE

Mesangial cell complement receptor 1-related protein y limits complement-dependent neutrophil accumulation in immune complex glomerulonephritis

Embed Size (px)

Citation preview

Mesangial cell complement receptor 1-related protein y limitscomplement-dependent neutrophil accumulation in immune complex

glomerulonephritis

Introduction

The complement system is tightly regulated by more than

30 plasma and cell membrane proteins to protect self

tissue from injury.1 In humans, decay-accelerating factor

(DAF; CD55) and membrane cofactor protein (MCP;

CD46) are the main membrane-bound intrinsic regulators

of C3 activation. In rodents, complement receptor 1

(CR1) related gene/protein y (Crry) has combined decay-

accelerating and factor I cofactor activity for C3b and

C4b; therefore, it combines functionalities of both DAF

and MCP.2

It has been shown using transgenic and recombinant

protein techniques that Crry is protective in different

immune complex-mediated kidney diseases.3–6 Deficiency

of Crry led to early embryonic lethality as a result of

unrestricted complement activation and concomitant pla-

cental inflammation, which could be averted by coexisting

C3 deficiency in Crry)/) C3)/) mice.7 Because of the

resultant C3 deficiency, the role of Crry in protecting

Lihua Bao,1 Ying Wang,1 Peili

Chen,1 Menaka Sarav,1 Mark Haas,2

Andrew W. Minto,1 Miglena

Petkova1 and Richard J. Quigg1

1Section of Nephrology, The University of

Chicago, Chicago, IL, and 2Department of

Pathology, Johns Hopkins University School

of Medicine, Baltimore, MD, USA

doi:10.1111/j.1365-2567.2009.03102.x

Received 16 January 2009; revised 6 March

2009; accepted 11 March 2009.

Correspondence: L. Bao, MD, Section of

Nephrology, The University of Chicago, 5841

S. Maryland Avenue, MC5100 Chicago, IL

60637, USA. Email:

[email protected]

Senior author: Richard J. Quigg,

email: [email protected]

Summary

The absence of complement receptor 1 (CR1) related gene/protein y

(Crry) leads to embryonic lethality as a result of unrestricted complement

activation and concomitant neutrophil infiltration. Here we used Crry)/)

C3+/) mice to investigate the role of Crry in the pathogenesis of immune

complex glomerulonephritis (GN). After 3 weeks of immunization with

horse spleen apoferritin, six of nine Crry)/) C3+/) mice and none of the

six control C3+/) mice developed proliferative GN (P = 0�010). After

5 weeks of immunization, GN scores in Crry)/) C3+/) mice were

0�67 ± 0�22 mean ± standard error of the mean (SEM), compared with

0�32 ± 0�16 in C3+/) mice. Glomerular hypercellularity was attributable to

neutrophil infiltration in mice with GN (1�7 ± 0�3/glomerulus) compared

with those without GN (0�4 ± 0�1/glomerulus) (P = 0�001). Absent stain-

ing for a-smooth muscle actin and proliferating cell nuclear antigen sug-

gested that mesangial cell proliferation did not play a significant role in

this model. Serum C3 levels in Crry)/) C3+/) mice were approximately

20% and 30% those of wild-type mice and C3+/) mice, respectively. To

determine whether this acquired hypocomplementaemia was relevant to

this GN model system, Crry)/) C3+/) mouse kidneys were transplanted

into wild-type mice followed by immunization with apoferritin for 1 or

2 weeks. Surprisingly, none of the Crry)/) C3+/) mouse kidneys developed

GN at these early time-points, indicating that increasing circulating C3

levels several-fold did not increase susceptibility to GN. Renal expression

of decay-accelerating factor was not different among any of the groups

studied. Thus, our data indicate that mesangial cell Crry limits comple-

ment activation and subsequent neutrophil recruitment in the setting of

local immune complex deposition.

Keywords: complement; inflammation; knockout mice

Please cite this article in press as: Bao L. et al. Mesangial cell complement receptor 1-related protein y limits complement-dependent

neutrophil accumulation in immune complex glomerulonephritis, Immunology (2009) doi: 10.1111/j.1365-2567.2009.03102.x

Abbreviations: CR, complement receptor; Crry, CR1-related gene/protein y; GN, glomerulonephritis; hpf, high-power field; IF,immunofluorescence.

� 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e895

I M M U N O L O G Y O R I G I N A L A R T I C L E

kidneys from complement activation cannot be readily

determined in unmanipulated DKO (Crry)/)C3)/)) mice.

We previously generated a Crry-deficient mouse kidney

model by transplanting Crry)/) C3)/) kidneys into Crry-

and C3-sufficient wild-type mice. Without the potent

complement-regulating functions of Crry, spontaneous

unrestricted complement activation occurred in the

tubulointerstitium of Crry-deficient kidneys, which

quickly led to fibrosis and renal failure.8

Other groups have utilized creative approaches to gen-

erate Crry-deficient mice, exploiting the fact that fetal

lethality is attributable to maternal alternative pathway

activation and formation of proinflammatory C5 prod-

ucts.9–11 In one such instance, it was interesting that

Crry)/) C3+/) mice showed substantial alternative path-

way turnover which led to reduced C3 concentration sys-

temically, suggesting that Crry has a critical role in

maintaining homeostasis of the complement system.10

While both Crry- and factor H-deficient mice have con-

siderable systemic complement consumption, the former

do not develop de novo renal pathology as do factor

H-deficient mice, suggesting either functional differences

between these two important complement regulators in

restricting complement activation within the kidney, and/

or fundamental differences between the two strains.11,12

The current study focuses on the role of Crry deficiency

in the pathogenesis of glomerulonephritis (GN).

In addition to the tubulointerstitium of the kidneys,

Crry is also present in the mouse glomerular mesan-

gium.13,14 In distinct contrast to the tubulointerstitium

Crry, the mesangial cell Crry appears to be of little con-

sequence in normal mice.8 To examine its relevance when

there are immune complex deposits, we utilized a chronic

serum sickness model induced with horse spleen apoferr-

itin in native Crry)/) C3+/) mice,15,16 as well as Crry)/)

C3+/) kidneys transplanted into wild-type mice.

Materials and methods

Animal work protocols

Crry+/) and C3+/) mice were kindly provided by Dr Hec-

tor Molina (Washington University School of Medicine,

St Louis, MO). The C57BL/6 mice used in the backcross-

ing procedure were purchased from Harlan Sprague Daw-

ley (Indianapolis, IN). Crry+/) and C3+/) mice had been

backcrossed onto the C57BL/6 background for at least

eight generations. Crry+/) and C3+/) mice were inter-

crossed to generate Crry)/) C3)/) mice, followed by cross-

ing with C3+/) mice to generate the Crry)/) C3+/)

animals employed in these studies. As controls, C3+/)

mice on a C57BL/6 background were used.

Experimental serum sickness was induced by daily

intraperitoneal injections of 4 mg of horse spleen apoferr-

itin (Calzyme Laboratories, Inc., San Luis Obispo, CA).

Crry)/) C3+/) mice were studied after 3 weeks (n = 9)

and 5 weeks (n = 9), and control C3+/) mice received the

same injection schedule for 3 weeks (n = 6) and 5 weeks

(n = 8). At the termination of the study, all mice were

killed and the kidneys were harvested for further studies.

These animal experiments were approved by the Univer-

sity of Chicago Animal Care and Use Committee.

Kidneys from a separate group of Crry)/) C3+/) mice

were transplanted into normal C57BL/6 mice (n = 9)

according to a protocol described previously.17 In brief,

donor mice were anaesthetized and the donor left kidney

was removed with artery, vein and ureter attached, and

preserved in cold saline on ice. The recipient was then

anaesthetized and the left kidney was excised. End-to-side

anastomoses of the donor renal vein, artery and ureter to

the recipient inferior vena cava, aorta and bladder, respec-

tively, were performed. Mice were allowed to recover

from the surgery for about 1 week. Serum sickness was

then induced for 1 week (n = 3) and 2 weeks (n = 3), as

described above. Controls were similarly transplanted

mice receiving saline instead of apoferritin (n = 3).

Measurements of blood urea nitrogen (BUN) andurinary albumin

BUN concentrations were detected with a Beckman

Autoanalyzer (Beckman Coulter, Fullerton, CA). Urinary

albumin concentrations were measured with a mouse

albumin enzyme-linked immunosorbent assay (ELISA) kit

(Bethyl Laboratories, Montgomery, TX) and normalized

to urinary creatinine as described previously.18

Measurements from tissue

To evaluate renal pathological changes, kidney tissues

were fixed in 4% paraformaldehyde and embedded in

paraffin. Four-lm sections were stained with periodic

acid-Schiff and examined by a renal pathologist (MH) in

a blinded manner. For each slide, the severity of GN,

glomerulosclerosis, interstitial nephritis, and arteritis was

graded in a semiquantitative (0–4) manner as previously

described.4

For immunofluorescence (IF) microscopy, 4-lm cryo-

stat sections were fixed in ether-ethanol and stained with

fluorescein isothiocyanate (FITC)-conjugated antibodies

to mouse C3 and immunoglobulin G (IgG) (Cappel Phar-

maceuticals, Inc., Aurora, OH). A semiquantitative score

of glomerulus staining intensity and distribution from 0

to 4 was given in a blinded manner as described previ-

ously.18 To visualize the co-localization of these different

targets, FITC-conjugated anti-mouse C3 (shown in green)

and tetramethyl rhodamine isothiocyanate (TRITC)-con-

jugated anti-mouse IgG (shown in red) (Cappel Pharma-

ceuticals) were used on the same renal sections from

different groups of mice.

e896 � 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904

L. Bao et al.

To assess different renal-infiltrating cells, immunohisto-

chemistry was performed on 4% paraformaldehyde-fixed

and paraffin-embedded kidney sections. Antigen was

retrieved by heating slides in a citrate buffer (pH 6�0) in

a microwave oven for 10 min. Endogenous peroxidases

and biotin were blocked using 0�3% hydrogen peroxide

and the Avidin/Biotin Blocking Kit (Vector Laboratories,

Burlingame, CA). Normal goat serum (10%, volume/vol-

ume) was also used as a separate blocking step. The slides

were then incubated with rat anti-mouse neutrophil

(AbD Serotec, Oxford, UK), rat anti-mouse B220 (BD

Pharmingen, San Jose, CA) for B cells, rat anti-mouse

Thy-1�2 (AbD Serotec) for T cells, or rat anti-mouse F4/

80 (AbD Serotec) for macrophages/monocytes, followed

by mouse anti-rat IgG (BD Pharmingen) and streptavi-

din-peroxidase (Sigma, St Louis, MO). Specifically bound

antibodies (Abs) were detected using the 3, 30-diamino-

benzidine (DAB) Substrate Kit (Vector Laboratories). To

quantify each type of leucocyte, at least 20 glomeruli or

20 high-power fields (hpfs) (·400) of the renal cortex in

each mouse kidney section were examined in a blinded

manner.

Proliferating cell nuclear antigen (PCNA) and

a-smooth muscle actin (a-SMA) were used as markers of

mesangial cell proliferation and activation.19,20 These were

identified in tissues stained for neutrophils as follows.

Paraffin-embedded slides were first stained with anti-

mouse neutrophil Abs, as described above. Following

DAB development (generating a brown colour), the per-

oxidases in the first staining were blocked with 0�3%

hydrogen peroxide. Slides were subsequently incubated

with rabbit anti-PCNA (Santa Cruz Biotechnology, Santa

Cruz, CA), or anti-a-SMA (US Biological, Swampscott,

MA). Ab staining was then detected with the ImmPress

Reagent (Vector Laboratories) according to the manufac-

turer’s instructions. The VIP Substrate Kit (Vector Labo-

ratories) (generating a purple colour) was used as a

substrate. All sections were counter-stained with methyl-

green (Vector Laboratories).

Serum C3 ELISA

Serum C3 levels were determined by ELISA. Plates were

first coated with goat anti-mouse C3 (Cappel Laborato-

ries, Durham, NC). Serially diluted serum samples were

added followed by horseradish peroxidase (HRP)-goat

anti-mouse C3 (Cappel). Serum C3 levels from normal

C57BL/6 (n = 9), C3)/) (n = 9), C3+/) (n = 9) and

Crry)/) C3+/) (n = 9) mice were measured. To evaluate

whether systemic consumption of C3 occurred during

development of serum sickness, serum samples at the end

of the respective experiments were also measured. Results

are expressed as relative optical density (OD450) values

adjusted for standard wild-type mouse serum samples

included in all ELISA plates.

Western blotting

Western blotting was used to detect DAF in kidney tissue.

Frozen renal cortical tissue protein was obtained and

quantified as described previously.21 Equal amounts of

protein samples were reduced and separated by sodium

dodecyl sulphate–polyacrylamide gel electrophoresis

(SDS-PAGE) following electrophoretic transfer to a poly-

vinylidene difluoride membrane (Millipore, Bedford,

MA). Membranes were blocked with 5% non-fat milk for

1 hr at room temperature, and incubated with rabbit

anti-DAF Ab (Santa Cruz Biotechnology) followed by

HRP-anti-rabbit Ab (Cell Signaling Technology, Danvers,

MA). Renal tissue samples from DAF-deficient mice were

used as negative controls. Membranes were also probed

with anti-actin antibody (Sigma) to confirm equal loading

of samples.

Statistical analyses

All data are expressed as mean ± standard error of the

mean (SEM) and were analysed using MINITAB software

(Minitab Inc., State College, PA). Two-sample t-tests and

Kruskal–Wallis tests were used for parametric and non-

parametric data analysis of differences between two

groups. For comparisons for data obtained from different

groups and at different time-points, analysis of variance

(ANOVA) followed by Tukey’s pairwise comparisons was

used. The v2 test was used to analyse the difference of the

incidences between the two groups. Potential correlations

among variables were examined using the Pearson prod-

uct moment correlation coefficient.

Results

In normal C57BL/6 mice, repetitive active immunization

with heterologous horse spleen apoferritin leads to gener-

ation of chronic serum sickness and the progressive

glomerular deposition of immune complexes primarily

within mesangial regions. Because Crry is the primary

complement regulator in mesangial cells, we were inter-

ested in whether its absence would affect pathological

manifestations of glomerular disease. Thus, we induced

chronic serum sickness in Crry)/) C3+/) mice. As the

most appropriate controls for these animals, a group of

C3+/) mice was studied concurrently. Because of the time

dependence of disease manifestation, mice were studied

after either 3 or 5 weeks of disease.

As expected, C3+/) mice had IgG (presumably in

immune complexes) in mesangial regions after 3 weeks of

apoferritin immunization; although the amounts of IgG

were high, they were even higher after 5 weeks. C3 depo-

sition tracked with mesangial immunoglobulin deposits,

and increased similarly with time (Fig. 1). Despite the

apparent complement activation, none of the six mice at

� 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e897

Crry in IC glomerulonephritis

the 3 weeks and three of the eight mice at 5 weeks devel-

oped evidence of GN.

Crry)/) C3+/) mice developed mesangial deposition of

IgG after 3 and 5 weeks of apoferritin administration,

and this deposition did not differ from that found in

C3+/) mice (Fig. 1a). At 3 weeks, C3 deposition was

greater in Crry)/) C3+/) mice than in C3+/) mice

(P = 0�022), at which time six of nine Crry)/) C3+/)

mice developed histological features of GN (Fig. 2a)

(P = 0�01 versus C3+/) mice). After 5 weeks of immuni-

zation, C3 staining was further increased in Crry)/)

C3+/) mice (Fig. 1b), with six of nine Crry)/) C3+/)

mice developing GN (Fig. 2a). The conclusion that C3

deposition was relevant to the development of GN was

supported by the correlation between C3 staining and

GN scores (r = 0�558, P = 0�001). Consistent with the

lack of significant tubulointerstitial or glomerular capil-

lary wall/podocyte disease, there was no significant ele-

vation or difference in BUN and urinary albumin

values among the groups.

C3 IgG Merge

Crry –/–C3 +/–

Crry–/–C3+/–

(3 weeks, n = 9)

Crry–/–C3+/–

(5 weeks, n = 9)

(3 weeks, n = 6)

C3+/–

(5 weeks, n = 8)C3+/–

C3+/–

C3 C3IgG IgG

3 weeks 5 weeks

0

1

2

3

4

0·5

1·5

2·5

3·5

*

IF in

tens

ity

(a)

(b)

Figure 1. Immunopathological features in

complement receptor 1 (CR1)-related gene/

protein y (Crry)-deficient mice with serum

sickness. (a) Representative immunofluores-

cence (IF) photomicrographs show co-localiza-

tion of glomerular deposits containing mouse

C3 and immunoglobulin (IgG) in Crry)/) C3+/)

mice compared with control C3+/) mice. (b)

Semiquantitative data for IF intensity of glomer-

ular C3 and IgG staining are also shown. Magni-

fication, ·600. *P = 0�022 versus C3+/) mice.

e898 � 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904

L. Bao et al.

The scores for GN were based on increased numbers of

cells within glomeruli, without specifically determining

their origins. As these cells could have included intrinsic

glomerular cells and extrinsic blood-derived inflammatory

cells, we enumerated these cell types in glomeruli. There

was no evidence for intrinsic glomerular cell proliferation

or injury (i.e. PCNA+ or a-SMA+ cells, respectively) in

any of the C3+/) and Crry)/) C3+/) mice immunized with

apoferritin. Furthermore, there were few B or T lympho-

cytes or F4/80+ monocytic cells within glomeruli or the

tubulointerstitium in any of the groups of animals. In

contrast, there were significant numbers of neutrophils in

glomeruli (Fig. 2a); glomerular neutrophil numbers were

highly correlated with GN scores (r = 0�879, P = 0�000),

consistent with the premise that this was an exudative

GN model. At 3 weeks, glomerular neutrophil numbers

were significantly greater in Crry)/) C3+/) mice compared

with C3+/) mice (Fig. 2b). These data demonstrate that

the lack of complement regulation by absent mesangial

cell Crry allowed a greater influx of neutrophils in glome-

ruli in the presence of potentially inflammatory immune

complexes.

We were next interested in the systemic complement

status of the two groups of mice. It was previously

reported that Crry)/) C3+/) mice had a systemic C3 level

that was about 20% that of wild-type mice;10,11,22 we

extended these results by evaluating the C3+/) controls

used here. As shown in Fig. 3, C3+/) mice had C3 levels

approximately half those of wild-type controls. Crry)/)

C3+/) mice had even greater reductions in C3 levels to

approximately 20% and 30% those of wild-type and

C3+/) mice, respectively. Serum C3 levels were affected by

neither the gender of the mice nor the presence or

absence of apoferritin immunization.

In studies to determine whether an increase in systemic

C3 levels would affect development of GN in Crry)/)

C3+/) mice, we transplanted kidneys from Crry)/) C3+/)

mice into C3-sufficient wild-type animals. In our previous

studies of Crry-deficient kidneys transplanted into wild-

Serum sickness 3 weeks 5 weeks

PAS

Neutrophils+

a-SMA

GN = 0

GN = 1

GN = 2

GN = 0·5

0

1

2 *

3

4

5

Neu

trop

hils

/glo

mer

ulus

C3 +/– Crry–/–C3 +/–

C3+/– (n = 9) C3+/– (n = 9)Crry–/–C3+/– (n = 6) Crry–/–C3+/– (n = 8)

3 weeks

C3 +/– Crry –/–C3+/–

5 weeks

(a)

(b)

Figure 2. Complement receptor 1 (CR1)-

related gene/protein y (Crry) deficiency

exacerbates development of proliferative

glomerulonephritis (GN) and glomerular

inflammation with neutrophils. (a) Representa-

tive histology and double-stain immunohisto-

chemistry showing the development of GN and

neutrophil influx (stained brown), but not

a-smooth muscle actin (a-SMA)-positive mes-

angial cells (stained purple), in glomeruli in

different groups of mice. (b) Quantitative data

for glomerular neutrophils relative to GN score

in individual mice from each group are shown.

There were significantly more glomerular neu-

trophils in Crry)/) C3+/) mice compared with

control C3+/) mice. *P = 0�024 versus C3+/)

mice. PAS, periodic acid Schiff.

0

0·05

0·1

0·15

*

*

0·2

0·25

Ser

um C

3 (O

D45

0)

Serum sickness (week) 0 0 3 5 0 3 5

WT

(n = 9) (n = 23) (n = 26)C3+/– Crry–/–3+/–

Figure 3. C3 levels are significantly reduced in complement receptor

1 (CR1)-related gene/protein y (Crry))/) C3+/) mice. C3 serum levels

were measured by enzyme-linked immunosorbent assay (ELISA) in

wild-type (WT), C3+/) and Crry)/) C3+/) mice. C3 levels from mice

of different genders and with or without serum sickness were com-

parable. *P < 0�01 versus the other two groups.

� 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e899

Crry in IC glomerulonephritis

type hosts, we observed a marked tubulointerstitial

inflammation which led to failure of the majority of kid-

neys within 3 weeks of transplantation.8 Notably, in these

kidneys, there was no evident glomerular pathology in

spite of the absence of Crry. Thus, we studied kidneys

after 1 and 2 weeks of chronic serum sickness, with the

hypothesis that their exposure to roughly fivefold higher

circulating C3 levels would represent a further challenge

to mesangial cells lacking Crry, and lead to severe GN.

This reasonable hypothesis turned out to be incorrect, as

none of the Crry)/) C3+/) kidneys developed GN after 1

or 2 weeks of apoferritin immunization (n = 3 each),

although tubular complement activation and damage were

evident (Fig. 4a). Consistent with our previous studies,

there were also no glomerular disease features in trans-

planted kidneys in control animals receiving saline

(n = 3). The native wild-type kidneys left in situ were also

analysed as controls for the transplanted Crry)/) C3+/)

kidneys. After 1 or 2 weeks of immunization, native wild-

type and transplanted Crry)/) C3+/) kidneys had compa-

rable glomerular IgG and C3 deposition (Fig. 4b),

although this deposition was at lesser extend compared

with that found at 3 weeks, as described above. As in

transplanted Crry)/) C3+/) kidneys, none of the native

wild-type kidneys developed GN after 1 or 2 weeks of

immunization.

Control (n = 3) 1 week (n = 3) 2 weeks (n = 3)

C3

IgG

PAS

0

1

2

3

0·5

1·5

2·5

3·5

IF In

tens

ity

C3 IgG

1 week

C3 IgG

2 weeks

Transplanted Crry–/–C3+/– kidney

Native WT kidney

(a)

(b)

Figure 4. Normalizing serum C3 levels induced tubular complement activation and damage but did not increase susceptibility to serum sickness-

induced glomerulonephritis (GN) in complement receptor 1 (CR1)-related gene/protein y (Crry))/) C3+/) mouse kidneys transplanted into wild-

type (WT) mice. (a) Prominent tubular deposits of C3 were observed in Crry)/) C3+/) mouse kidneys (white arrows) transplanted into wild-type

mice, which led to inflammation and tubular damage (black arrows). After 1 or 2 weeks of apoferritin immunization, none of the Crry)/) C3+/)

mouse kidneys developed proliferative GN. (b) Semiquantitative data showing immunofluorescence (IF) staining intensity for C3 and immuno-

globulin G (IgG) in glomeruli from transplanted Crry)/) C3+/) kidneys as well as the native wild-type kidneys. PAS, periodic acid Schiff.

e900 � 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904

L. Bao et al.

As described previously, Crry)/) C3+/) mouse kidneys

transplanted into wild-type mice had spontaneous com-

plement activation in the tubulointerstitium and consid-

erable tubular injury and inflammatory cell infiltration

(Fig. 4a); a composite ‘injury’ score for these variables in

the nine transplanted kidneys (2–3 weeks post-transplant)

was 1�2 ± 0�1. This was lower than we had previously

found in Crry)/) C3)/) kidneys transplanted into wild-

type mice (e.g. 1�7 ± 0�4 within 1 week after trans-

plantation). Although these data sets are not directly

comparable, it is possible that the presence of C3 in

Crry)/) C3+/) mice may have conferred relative resistance

to complement-mediated kidney injury when transplanted

into C3-sufficient hosts.

One plausible explanation for why Crry)/) C3+/) mouse

kidneys transplanted into wild-type hosts were relatively

resistant to both GN and tubulointerstitial disease was

compensation by another complement regulator. As DAF

is the other cell-bound C3 regulator in mice, we examined

its protein abundance in the various kidneys. However,

DAF protein quantities were not increased in Crry-

deficient kidneys from unmanipulated animals (Fig. 5),

nor in the setting of chronic serum sickness, in which

there was probably generation of complement activation

products capable of up-regulating DAF expression.23

Thus, a compensatory increase in cellular complement

regulation was an unlikely explanation for these results.

Discussion

Complement activation has long been considered as

pathogenic in many different forms of immune complex-

mediated GN diseases, including lupus nephritis,

membranoproliferative GN, postinfectious GN and mem-

branous nephropathy.24 Consistent with such a premise,

inhibition of complement activation by the provision of

exogenous soluble Crry, either transgenically expressed or

administered as a recombinant protein, has been shown

to be protective in murine lupus nephritis 3,4,21 and nephro-

toxic serum nephritis.5,6 These findings are consistent

with the premise that complement activation in these dis-

ease models is sufficiently robust to overcome intrinsic

complement regulation. By providing exogenous comple-

ment regulators, the balance is brought back towards suc-

cessful restriction of complement activation.

In serum sickness, various glomerular pathologies can

result which depend on the species/strain being studied,

as well as the immunization schemes. Here we used the

protocol as originally described by Stilmant, Couser and

Cotran,15 and subsequently modified by Iskandar, Eman-

cipator and colleagues,16 of active immunization of mice

with apoferritin. In this study, we were interested in

mesangial cell Crry, given the significant deposition of

immune complexes (ICs) within the mesangium.

Crry deficiency is embryonic lethal in mice, unless there

is maternal C3 deficiency.7 Here we crossed Crry)/) C3)/)

females with Crry)/) C3+/) males, thereby generating

Crry)/) C3+/) mice with absent Crry but a single wild-

type C3 allele. These mice became susceptible to develop-

ing GN early in the chronic serum sickness model,

illustrating the relevance of mesangial cell Crry in protect-

ing the C57BL/6 mouse from immune complex-mediated

complement activation and its downstream effects.

To examine whether the combined inherited and

acquired hypocomplementaemia in Crry)/) C3+/) mice

affected glomerular disease features in these studies, we

transplanted Crry)/) C3+/) mouse kidneys into syngeneic

C3-sufficient wild-type mice and then induced serum sick-

ness. Under these circumstances, glomeruli with immune

complex deposition were exposed to normal circulating C3

levels. In our previous work, in which Crry)/) C3)/)

mouse kidneys were transplanted into wild-type hosts,

Crry-deficient kidneys rapidly developed pathology in the

tubulointerstitial compartment, ultimately leading to organ

failure.8 Thus, like the placenta, the tubulointerstitial

compartment relies upon Crry as its critical complement

regulator. Under these conditions, spontaneous comple-

ment activation did not occur in Crry-deficient glomeruli,

and there was no evident glomerular pathology. Because of

the spontaneous complement activation occurring in the

kidney outside the glomerulus, it was necessary to use an

abbreviated disease model. Nonetheless, given the approxi-

mately fivefold increase in circulating C3 levels in the

recipient animals, this experimental design did allow us to

evaluate whether normal C3 levels but absent glomerular

Crry conferred upon transplanted Crry)/) C3+/) kidneys a

greater degree of susceptibility to this immune complex-

mediated GN. As anticipated following renal transplanta-

tion, Crry)/) C3+/) mouse kidneys developed complement

activation and injury within the tubulointerstitial compart-

ment. In contrast, they appeared resistant to developing

immune complex-mediated GN; an explanation for this

somewhat unexpected result is not obvious. The renal

transplantation protocol itself does not prevent develop-

ment of immune complex-mediated GN in this model, as

evidenced by past studies of wild-type kidneys transplanted

into complement factor H Cfh)/) mice.25 Clearly, the

DAF

Intensity

a-actin

ratio 0·94 0·92 0·90 0·88 0·84 0·86 0·88 0·88

WT C3+/– Crry–/–C3+/–

Figure 5. Renal expression of decay-accelerating factor (DAF) was

unaffected by the absence of complement receptor 1 (CR1)-related

gene/protein y (Crry). Western blot analysis showed that renal DAF

expression was comparable in wild-type (WT), C3+/) and Crry)/)

C3+/) mice. Shown are representative immunoblots from at least

three different experiments.

� 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e901

Crry in IC glomerulonephritis

transplanted kidneys are exposed to systemic complement

(and do not rely upon intrinsic C3 synthesis), as shown by

marked complement activation in the tubulointerstitium

in the current and previous studies.8 It may be that the

Crry)/) C3+/) mouse (and kidney) is able to compensate

for the lack of Crry from conception until the transplant

procedure. This theory is supported by the lower amounts

of C3 activation and tubulointerstitial damage that were

evident in Crry)/) C3+/) kidneys relative to Crry)/) C3)/)

kidneys in our previous studies.8

DAF is another complement regulator expressed in

normal mouse kidneys,26 with a similar main function to

Crry, namely regulation of C3 activation. Although it only

dissociates the C3 (and C5) convertase, and does not

facilitate complement factor I-mediated C3b inactivation,

it is an effective classical pathway C3 convertase inhibi-

tor,27 which is relevant to the current studies. Hence, we

considered whether a compensatory up-regulation of DAF

would compensate for the absence of Crry in these stud-

ies. However, there was no evidence for an increase in

DAF total protein, making this compensation an unlikely

explanation for the relative resistance of Crry)/) C3+/)

kidneys to immune complex-mediated GN (or spontane-

ous complement activation in the tubulointerstitium).

The different distributions of DAF and Crry in normal

rodent kidneys may also explain the lack of compensation

by DAF when Crry is absent and indicate their distinct

functions under different pathological circumstances. Crry

is normally present in the mesangium,13,14 while DAF is

localized in rodent glomerular capillary walls,26 as a result

in particular of its presence on podocytes.28

In the absence of systemic Cfh or renal Crry, unre-

stricted complement activation occurs in glomeruli 12 and

the tubulointerstitium,8 respectively. In immune complex-

mediated disease, Cfh is clearly relevant to limit further

glomerular complement activation, and prevent inflam-

matory GN.29 In the present studies, we show that glo-

merular Crry does have a role in limiting complement

activation by immune complexes within the mesangial

region. A third member of this group in mice is DAF. As

is true in humans, DAF may be relatively dispensable

compared with Cfh and Crry (or MCP in humans), inso-

far as there is no evidence for spontaneous complement

activation in animals lacking DAF. Yet, it does appear to

have a role in limiting Ab-directed complement activa-

tion. This has been shown by studies of nephrotoxic

serum nephritis in DAF)/) Crry)/) C3+/) mice (i.e. defi-

cient in both DAF and Crry) and our studies with

immune complex-mediated GN in DAF)/) mice.22,30 In

the former setting of Ab-directed complement activation

in the glomerular capillary wall, mice have exacerbated

proteinuria, yet comparable C3 deposition and glomerular

pathology between DAF)/) Crry)/) C3+/) and control

DAF)/) Crry+/) C3+/) mice.22 The finding that there is

little overlap in the sites of complement activation clearly

shows that, in addition to differences in specific comple-

ment regulatory functions, the distributions of the C3

regulators have substantial effects on where complement

can be activated and on the resultant disease expression

within the kidney. This concept extends further to differ-

ent disease models, in which different sites of comple-

ment activation occur which can be limited locally by the

presence of specific complement regulators.31

In this study, the histopathological expression of me-

sangial proliferative GN could be attributed almost exclu-

sively to an influx of neutrophils into glomeruli. This was

most predominant in Crry)/) C3+/) mice, and occurred

early in the disease course; however, it was also seen in a

few C3+/) mice after 5 weeks of disease. Such a neutro-

phil predominance was also observed in the inflammation

occurring in the Crry-deficient embryo.7 It is worth not-

ing that the Schlondorff group previously reported that

macrophages were the major infiltrating cell type in a

similar model induced in wild-type mice on the BALB/c

background, although neutrophils were not specifically

examined.32–34 Thus, even in the same apoferritin-

induced chronic serum sickness disease model in mice,

there can be considerable differences in phenotypic out-

come which depend on many factors, including the fun-

damental genetic background and the site and tempo of

complement activation.

Given the fact that extensive complement activation

occurred in the glomerular mesangial area, even in the

mice with intact Crry, one would assume that excessive

production of anaphylatoxins C3a and C5a played a

significant role in the glomerular recruitment of neutro-

phils. The seven membrane-spanning G protein-coupled

receptors for C3a and C5a are both expressed on neu-

trophils and monocytes,35,36 and C5a is chemotactic to

neutrophils.37 More importantly, a large body of evi-

dence supports the functional significance of C3aR and

C5aR signalling in neutrophil recruitment in many dif-

ferent experimental disease models. Blockade of C3aR

signalling with a specific antagonist significantly reduced

neutrophil influx in a murine focal cerebral ischemia

model,38 while preventing C5aR activation with a spe-

cific antagonist or neutralizing Ab significantly inhibited

neutrophil recruitment in rodent models of zymosan-

induced hyper-nociception,39 monarticular arthritis,40

neurodegeneration,41 immune-complex peritonitis42 and

renal ischemia-reperfusion.43 In our hands, we observed

a significant reduction of neutrophil infiltration in the

murine lupus model using C3aR and C5aR antago-

nists.44,45 Considering that C3a and C5a have broad

effects in both myeloid and lymphoid cells,46 and, as

mentioned earlier, variants of the very same chronic

serum sickness model (i.e. in wild-type BALB/c) generate

a monocytic inflammatory GN,32–34 the fundamental

basis for the neutrophil predominance remains to be

determined in the current model.

e902 � 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904

L. Bao et al.

In summary, here we provide evidence that glomerular

Crry limits experimental immune complex-mediated pro-

liferative GN. The proliferative nature of this disease can

be attributed to neutrophil infiltration into glomeruli,

and is progressive over time. These results add to our

understanding of the important role of effective local

complement regulation within the kidney, in this case to

limit complement activation brought about by tissue-

deposited immune complexes. These data support the

conclusion that complement inhibition represents a viable

approach to the treatment of the commonly occurring

clinical condition of immune complex-associated GN.

Acknowledgements

Grant support: National Institutes of Health grant

R01DK041873.

Disclosures

All authors declare no financial conflict of interest.

References

1 Walport MJ. Advances in immunology: complement (first of

two parts). N Engl J Med 2001; 344:1058–66.

2 Kim Y-U, Kinoshita T, Molina H, Hourcade D, Seya T, Wagner

LM, Holers VM. Mouse complement regulatory protein Crry/p65

uses the specific mechanisms of both human decay-accelerating

factor and membrane cofactor protein. J Exp Med 1995; 181:151–9.

3 Bao L, Haas M, Kraus DM, Hack BK, Rakstang JK, Holers VM,

Quigg RJ. Administration of a soluble recombinant complement

C3 inhibitor protects against renal disease in MRL/lpr mice.

J Am Soc Nephrol 2003; 14:670–9.

4 Bao L, Haas M, Boackle SA et al. Transgenic expression of a solu-

ble complement inhibitor protects against renal disease and pro-

motes survival in MRL/lpr mice. J Immunol 2002; 168:3601–7.

5 Quigg RJ, Kozono Y, Berthiaume D et al. Blockade of antibody-

induced glomerulonephritis with Crry-Ig, a soluble murine com-

plement inhibitor. J Immunol 1998; 160:4553–60.

6 Quigg RJ, He C, Lim A, Berthiaume D, Alexander JJ, Kraus D,

Holers VM. Transgenic mice overexpressing the complement

inhibitor Crry as a soluble protein are protected from antibody-

induced glomerular injury. J Exp Med 1998; 188:1321–31.

7 Xu C, Mao D, Holers VM, Palanca B, Cheng AM, Molina H. A

critical role for murine complement regulator Crry in fetomater-

nal tolerance. Science 2000; 287:498–501.

8 Bao L, Wang Y, Chang A et al. Unrestricted C3 activation

occurs in Crry-deficient kidneys and rapidly leads to chronic

renal failure. J Am Soc Nephrol 2007; 18:811–22.

9 Mao D, Wu X, Deppong C, Friend LD, Dolecki G, Nelson DM,

Molina H. Negligible role of antibodies and C5 in pregnancy loss

associated exclusively with C3-dependent mechanisms through

complement alternative pathway. Immunity 2003; 19:813–22.

10 Wu X, Spitzer D, Mao D, Peng SL, Molina H, Atkinson JP.

Membrane protein Crry maintains homeostasis of the comple-

ment system. J Immunol 2008; 181:2732–40.

11 Ruseva MM, Hughes TR, Donev RM et al. Crry deficiency in

complement sufficient mice: C3 consumption occurs without

associated renal injury. Mol Immunol 2009; 46:803–11.

12 Pickering MC, Cook HT, Warren J, Bygrave AE, Moss J,

Walport MJ, Botto M. Uncontrolled C3 activation causes mem-

branoproliferative glomerulonephritis in mice deficient in com-

plement factor H. Nat Genet 2002; 31:424–8.

13 Li B, Sallee C, Dehoff M, Foley S, Molina H, Holers VM. Mouse

Crry/p65: characterization of monoclonal antibodies and the tis-

sue distribution of a functional homologue of human MCP and

DAF. J Immunol 1993; 151:4295–305.

14 Thurman JM, Ljubanovic D, Royer PA et al. Altered renal tubu-

lar expression of the complement inhibitor Crry permits com-

plement activation after ischemia/reperfusion. J Clin Invest 2006;

116:357–68.

15 Stilmant MM, Couser WG, Cotran RS. Experimental glomerulo-

nephritis in the mouse associated with mesangial deposition of

autologous ferritin immune complexes. Lab Invest 1975; 32:746–

56.

16 Iskandar SS, Gifford DR, Emancipator SN. Immune complex

acute necrotizing glomerulonephritis with progression to dif-

fuse glomerulosclerosis. A murine model. Lab Invest 1988;

59:772–9.

17 Cunningham PN, Wang Y, Guo R, He G, Quigg RJ. Role of

Toll-like receptor 4 in endotoxin-induced acute renal failure.

J Immunol 2004; 172:2629–35.

18 Quigg RJ, Lim A, Haas M, Alexander JJ, He C, Carroll MC.

Immune complex glomerulonephritis in C4- and C3-deficient

mice. Kidney Int 1998; 53:320–30.

19 Johnson RJ, Iida H, Alpers CE et al. Expression of smooth mus-

cle cell phenotype by rat mesangial cells in immune complex

nephritis: a-smooth muscle actin is a marker of mesangial cell

proliferation. J Clin Invest 1991; 87:847–58.

20 Ichimura K, Kurihara H, Sakai T. Involvement of mesangial cells

expressing alpha-smooth muscle actin during restorative glomer-

ular remodeling in Thy-1.1 nephritis. J Histochem Cytochem

2006; 54:1291–301.

21 Bao L, Zhou J, Holers VM, Quigg RJ. Excessive matrix accumu-

lation in the kidneys of MRL/lpr lupus mice is dependent on

complement activation. J Am Soc Nephrol 2003; 14:2516–25.

22 Miwa T, Zhou L, Tudoran R et al. DAF/Crry double deficiency

in mice exacerbates nephrotoxic serum-induced proteinuria

despite markedly reduced systemic complement activity. Mol

Immunol 2007; 44:139–46.

23 Cosio FG, Rovin BH. Complement induction of decay accelerat-

ing factor on human mesangial cells: signal transduction path-

ways. J Am Soc Nephrol 1993; 4:598.

24 Quigg RJ. Complement and autoimmune glomerular diseases.

Curr Dir Autoimmun 2004; 7:165–80.

25 Alexander JJ, Wang Y, Chang A et al. Mouse podocyte comple-

ment factor H: the functional analog to human complement

receptor 1. J Am Soc Nephrol 2007; 18:1157–66.

26 Lin F, Fukuoka Y, Spicer A et al. Tissue distribution of products

of the mouse decay-accelerating factor (DAF) genes. Exploitation

of a Daf1 knock-out mouse and site-specific monoclonal anti-

bodies. Immunology 2001; 104:215–25.

27 Lublin DM, Atkinson JP. Decay-accelerating factor: biochemis-

try, molecular biology, and function. Annu Rev Immunol 1989;

7:35–58.

� 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904 e903

Crry in IC glomerulonephritis

28 Bao L, Spiller OB, St JP et al. Decay-accelerating factor expres-

sion in the rat kidney is restricted to the apical surface of podo-

cytes. Kidney Int 2002; 62:2010–21.

29 Alexander JJ, Pickering MC, Haas M, Osawe I, Quigg RJ. Com-

plement factor H limits immune complex deposition and pre-

vents inflammation and scarring in glomeruli of mice with

chronic serum sickness. J Am Soc Nephrol 2005; 16:52–7.

30 Bao L, Haas M, Minto AW, Quigg RJ. Decay-accelerating factor

but not CD59 limits experimental immune-complex glomerulo-

nephritis. Lab Invest 2007; 87:357–64.

31 Quigg RJ. Complement and the kidney. J Immunol 2003;

171:3319–24.

32 Anders HJ, Frink M, Linde Y et al. CC chemokine ligand

5/RANTES chemokine antagonists aggravate glomerulonephritis

despite reduction of glomerular leukocyte infiltration. J Immunol

2003; 170:5658–66.

33 Anders HJ, Vielhauer V, Kretzler M et al. Chemokine and

chemokine receptor expression during initiation and resolution

of immune complex glomerulonephritis. J Am Soc Nephrol 2001;

12:919–31.

34 Anders HJ, Vielhauer V, Schlondorff D. Chemokines and

chemokine receptors are involved in the resolution or progres-

sion of renal disease. Kidney Int 2003; 63:401–15.

35 Martin U, Bock D, Arseniev L et al. The human C3a receptor is

expressed on neutrophils and monocytes, but not on B or T

lymphocytes. J Exp Med 1997; 186:199–207.

36 Chenoweth DE, Hugli TE. Demonstration of specific C5a recep-

tor on intact human polymorphonuclear leukocytes. Proc Natl

Acad Sci U S A 1978; 75:3943–3947.

37 Snyderman R, Pike MC. Chemoattractant receptors on phagocy-

tic cells. Annu Rev Immunol 1984; 2:257–281.

38 Ducruet AF, Hassid BG, Mack WJ et al. C3a receptor modula-

tion of granulocyte infiltration after murine focal cerebral ische-

mia is reperfusion dependent. J Cereb Blood Flow Metab 2008;

28:1048–58.

39 Ting E, Guerrero AT, Cunha TM et al. Role of complement C5a

in mechanical inflammatory hypernociception: potential use of

C5a receptor antagonists to control inflammatory pain. Br

J Pharmacol 2008; 153:1043–53.

40 Woodruff TM, Strachan AJ, Dryburgh N, Shiels IA, Reid RC,

Fairlie DP, Taylor SM. Antiarthritic activity of an orally active

C5a receptor antagonist against antigen-induced monarticular

arthritis in the rat. Arthritis Rheum 2002; 46:2476–85.

41 Woodruff TM, Crane JW, Proctor LM et al. Therapeutic activity

of C5a receptor antagonists in a rat model of neurodegeneration.

FASEB J 2006; 20:1407–17.

42 Godau J, Heller T, Hawlisch H et al. C5a initiates the inflamma-

tory cascade in immune complex peritonitis. J Immunol 2004;

173:3437–45.

43 De Vries B, Kohl J, Leclercq WK, Wolfs TG, Van Bijnen AA,

Heeringa P, Buurman WA. Complement factor C5a mediates

renal ischemia-reperfusion injury independent from neutrophils.

J Immunol 2003; 170:3883–9.

44 Bao L, Osawe I, Puri T, Lambris JD, Haas M, Quigg RJ. C5a

promotes development of experimental lupus nephritis which

can be blocked with a specific receptor antagonist. Eur J Immu-

nol 2005; 35:3012–20.

45 Bao L, Osawe I, Haas M, Quigg RJ. Signaling through up-regu-

lated C3a receptor is key to the development of experimental

lupus nephritis. J Immunol 2005; 175:1947–1955.

46 Monk PN, Scola AM, Madala P, Fairlie DP. Function, structure

and therapeutic potential of complement C5a receptors. Br

J Pharmacol 2007; 152:429–48.

e904 � 2009 Blackwell Publishing Ltd, Immunology, 128, e895–e904

L. Bao et al.