Skip to main content
medRxiv
  • Home
  • About
  • Submit
  • ALERTS / RSS
Advanced Search

Synergistic impact of three complement polymorphisms in the donor, not the recipient, on long-term kidney allograft survival

View ORCID ProfileFelix Poppelaars, View ORCID ProfileMariana Gaya da Costa, View ORCID ProfileBernardo Faria, View ORCID ProfileSiawosh K. Eskandari, View ORCID ProfileVojtech Petr, View ORCID ProfileV. Michael Holers, View ORCID ProfileMohamed R. Daha, View ORCID ProfileStefan P. Berger, View ORCID ProfileJeffrey Damman, View ORCID ProfileMarc A. Seelen, View ORCID ProfileJoshua M. Thurman
doi: https://doi.org/10.1101/2023.10.24.23297481
Felix Poppelaars
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
2Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Felix Poppelaars
  • For correspondence: f.poppelaars{at}umcg.nl
Mariana Gaya da Costa
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
3Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Mariana Gaya da Costa
Bernardo Faria
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
4Nephrology R&D Group, Institute for Research and Innovation in Health (i3S), São João University Hospital Center, University of Porto, Porto, Portugal
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Bernardo Faria
Siawosh K. Eskandari
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Siawosh K. Eskandari
Vojtech Petr
2Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
5Department of Nephrology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Vojtech Petr
V. Michael Holers
2Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for V. Michael Holers
Mohamed R. Daha
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
6Department of Nephrology, University of Leiden, Leiden University Medical Center, Leiden, The Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Mohamed R. Daha
Stefan P. Berger
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Stefan P. Berger
Jeffrey Damman
7Department of Pathology, Erasmus University Medical Center, Erasmus University, Rotterdam, the Netherlands.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Jeffrey Damman
Marc A. Seelen
1Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Marc A. Seelen
Joshua M. Thurman
2Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • ORCID record for Joshua M. Thurman
  • Abstract
  • Full Text
  • Info/History
  • Metrics
  • Supplementary material
  • Data/Code
  • Preview PDF
Loading

Abstract

Background Genetic analysis in transplantation offers potential for personalized medicine. Given the crucial role of the complement system in renal allograft injury, we investigated in kidney transplant pairs the impact of complement polymorphisms on long-term outcomes.

Methods In this observational cohort study, we analyzed polymorphisms in C3 (C3R102G), factor B (CFBR32Q), and factor H (CFHV62I) genes of 1,271 donor-recipient kidney transplant pairs and assessed their association with 15-year death-censored allograft survival.

Results Individually, only the presence of the CFB32Q variant in the donor and the combined presence in donor-recipient pairs were associated with better graft survival (P=0.027 and P=0.045, respectively). In the combined analysis, the C3R102G, CFBR32Q, and CFHV62I variants in the donor independently associated with the risk of graft loss (HR 1.32; 95%-CI, 1.08– 1.58; P=0.005). Thus, donor kidneys carrying the genetic variants that promote the highest complement activity exhibited the worst graft survival, whereas those with the genetic variants causing the lowest complement activity showed the best graft survival (15-year death-censored allograft survival: 48.8% vs 87.8%, P=0.001).

Conclusion Our study demonstrates that the combination of complement polymorphisms in the donor strongly associates with long-term allograft survival following kidney transplantation. These findings hold significance for therapeutic strategies involving complement inhibition in kidney transplantation.

1. Introduction

The complement system can be activated through the classical, lectin, and/or alternative pathway, all leading to the formation of C3-convertases that cleave C3 into C3b and C3a.1 The newly generated C3b, in combination with factor B, can create more C3-convertases, resulting in an amplification loop with exponential potential.2,3 This amplification loop is tightly regulated by factor H, which rapidly dissociates factor B from the C3-convertase and catalyzes the inactivation of C3b through factor I-mediated cleavage to the iC3b form.4 Understandably, activation of the complement cascade is recognized as a critical proponent of kidney allograft injury throughout various phases of transplantation; in deceased donors before transplantation, during organ perfusion, following reperfusion, and in the recipient in various settings such as antibody-mediated rejection.5–9 Pre-clinical studies have shown promising results with complement inhibition in kidney transplantation to improve allograft outcomes.10–14 Furthermore, ongoing clinical trials with complement-targeting therapeutics aim to assess their impact on long-term outcomes after kidney transplantation.7

In recent decades, numerous studies have linked variations in complement genes to a wide range of inflammatory and infectious diseases.15–17 Functional characterization of these complement variants has yielded valuable insights into the underlying pathogenic mechanisms of complement activation in these disorders.18 Using in vitro experiments, recent studies revealed that individual polymorphisms only cause small changes in the activity of the complement system. However, when these complement variants are combined, their collective impact becomes significant.19,20 Thus, to achieve a comprehensive genetic understanding of the complement system, the total repertoire of polymorphisms in complement genes should therefore be studied, as this system operates as a network of proteins.21 The total make-up of the inherited set of complement variants is called the complotype and is believed to determine one’s individual ability to activate and regulate the complement system.19,21 A complotype leading to amplified complement activity makes an individual susceptible to inflammation, while a complotype that dampens complement activity increases the individual risks of infection.20,21

Several studies have demonstrated the impact of complement polymorphisms on outcomes after kidney transplantation.22–27 Yet, the majority of these studies focused on individual complement polymorphisms or haplotypes of a single complement gene. The combination of complement polymorphisms is likely to have a larger impact on long-term outcomes than one particular variant.28 Given the crucial role of complement in the pathogenesis of transplant renal injury, we, therefore, hypothesized that the complotype could be a major determinant of long-term allograft survival.28 Furthermore, genetic analysis in transplantation offers a unique opportunity to understand the effects of donor versus recipient genotype as well as develop personalized strategies that can improve donor-recipient matching, advance individualized risk stratification and drive personalized medicine.

As proof of concept, we examined the combined effect of three common complement polymorphisms (C3R102G, CFBR32Q, and CFHV62I) in donor-recipient pairs on long-term allograft survival after kidney transplantation. These three polymorphisms represent the most extensively researched and comprehensively characterized genetic variants within the complement system.19,21 The risk variant C3102Gexhibits reduced binding to factor H compared with the reference C3102R variant, thereby amplifying complement activation.19 The protective CFB32Q variant, on the other hand, shows decreased potential to form C3- convertases and amplify complement activation compared to the reference CFB32R variant.29 Finally, the protective CFH62I variant demonstrates a higher capacity to bind C3b, catalyzing its inactivation and more effectively competing with factor B in C3-convertase formation compared to the reference CFH62V variant.30 The combination of these 3 genetic variants was selected based on prior research, which revealed that when the disease "risk" variants (C3102G, CFB32R, and CFH62V) were combined in a functional in vitro assay there was a sixfold higher complement activity compared than with the "protective" variants (C3102R, CFB32Q, and CFH62I).19 To address this hypothesis, we first analyzed the individual impact of complement variants in both the donor and recipient on 15-year death-censored allograft survival. Subsequently, we investigated the association of the combined presence of these variants with allograft survival.

2. Materials and methods

2.1 Study design

Patients receiving a first single kidney transplantation at the University Medical Center Groningen (UMCG) between March 1993 and February 2008 were selected for this study.31–36 Subsequent, exclusion criteria consisted of perioperative technical complications (3 pairs excluded), lack of available DNA (65 pairs excluded), loss of follow-up (4 pairs excluded), re-transplantation at the time of recruitment (22 pairs excluded) and simultaneous transplantation of other organs (65 pairs excluded). In summary, we began with 1430 kidney transplant pairs, and after applying exclusion criteria, we included 1271 kidney transplant pairs in this study. The endpoint was 15-year death-censored graft failure, defined as the return to dialysis or re-transplantation.

2.2 DNA extraction and genotyping

Peripheral blood mononuclear cells from blood or splenocytes were obtained from the kidney donors and their respective transplant recipients. DNA was isolated using commercial kits. DNA samples of 1,271 donor-recipient kidney transplant pairs was analyzed for three common complement polymorphisms, namely the C3R102G variant (rs2230199 C>G, Arg102Gly), the CFHV62I variant (rs800292 G>A, Val62Ile), and the CFBR32Qvariant (rs641153 G>A, Arg32Gln). Genotyping of target single nucleotide polymorphism (SNP) was performed using the Illumina VeraCode GoldenGate Assay kit. Genotype clustering and calling were performed using BeadStudio Software. The overall genotype success rate was between 99.7 - 100%. Samples with a missing call rate were excluded from subsequent analyses, resulting in the exclusion of 12 donor-recipient pairs. Previously, we published the C3 SNPs in this cohort.26 The distribution of all assessed SNP was in Hardy-Weinberg equilibrium.

2.3 Statistics

Statistical analysis was performed using SPSS 26.0. The data were presented as mean ± standard deviation (SD) for parametric variables, median [IQR] for non-parametric variables, and percentage [n (%)] for nominal data. To compare multiple groups, One-way ANOVA or Kruskal-Wallis H-test were used for normally and non-normally distributed variables, respectively, while χ2-tests were employed for categorical variables. The associations between complement polymorphisms and death-censored graft survival were assessed using Kaplan-Meier analyses with log-rank tests. Cox proportional-hazards regression analyses were performed to further examine the associations with graft loss, with adjustments made for potential confounders. Additionally, multivariable Cox regressions with stepwise forward selection were performed incorporating all variables that showed significant associations with graft loss in the univariable analyses. All statistical tests were two-tailed and P<0.05 was considered significant for all analyses.

2.4 Ethics

The study protocol was approved by the medical ethics committee at the UMCG under file number METc 2014/077 and the study was performed in accordance with the principles of the Declaration of Helsinki.

3. Results

3.1 The frequency of C3, CFH, and CFB polymorphisms in donor-recipient transplant pairs

We first examined the distribution of the C3R102G, CFHV62I, and CFBR32Q polymorphisms. There were no significant differences in the genotypic frequencies between the recipients and donors, also when compared to the European cohort of the 1000 Genomes Project (Table 1). Within the donor population, the number of minor alleles for the three complement polymorphisms per individual ranged from 0 to 4. Among the donors, 28.5% had no minor alleles, 41.9% had one, 22.5% had two, 6.6% had three, and 0.6% had four minor alleles (Figure 1). Similar results were seen in the recipient population; the number of minor alleles per individual ranged from 0 to 5, with 26.8% having no minor alleles, 43.1% having one, 22.6% having two, 6.6% having three, 0.5% having four, and 0.5% having five minor alleles (Figure 1). These results confirm that the occurrence of multiple complement polymorphisms within a single individual is frequently seen.

Figure 1
  • Download figure
  • Open in new tab
Figure 1 The number of minor alleles of complement polymorphisms in donors and recipients

The number of minor alleles of common polymorphic variants in C3R102G (rs2230199), CFBR32Q (rs641153), and CFHV62I (rs800292) was counted for each donor and recipient.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 1: Genotypic frequencies of complement polymorphisms in donors and recipients

3.2 The association of single complement polymorphisms with allograft outcome

Next, we assessed the association between individual complement polymorphisms and late allograft loss. Kaplan-Meier analysis revealed significantly higher 15-year death-censored graft survival rate for donor kidneys carrying at least one ‘less activating’ CFB32Q variant when compared to donors who are homozygous for the reference CFB32R variant (Fig. 2A, P = 0.027). After complete follow-up, graft loss incidence was 16.7% for renal grafts with at least one ‘less activating’ CFB32Q variant and 28.1% for renal grafts carrying the reference CFB32R variant. Conversely, no significant difference in long-term graft survival was observed based on the C3R102Gand CFHV62Ipolymorphism in the donors (Supplementary Figure S1). Additionally, no significant difference in graft loss was seen for the C3R102G, CFHV62I, and CFBR32Q polymorphism in recipients (Supplementary Figure S2). In univariable analysis, the ‘less activating’ CFB32Q variant in the donor was associated with a lower risk of graft loss and a hazard ratio of 0.60 (95%-CI, 0.38–0.95; P = 0.03). Subsequently, we performed a multivariable Cox regression analysis with a stepwise forward selection procedure using all variables that were significantly associated with graft loss in the univariable analysis (Table 2). In the final model, the ‘less activating’ CFBR32Qpolymorphism in the donor, donor and recipient age, recipient blood type, and delayed graft function (DGF) were included. After adjustment, the ‘less activating’ CFB32Q variant in the donor remained significantly associated with a lower risk of graft loss (HR, 0.55; 95%-CI, 0.34 – 0.89; P = 0.015). Overall, the presence of the CFB32Q variant in the donor, which has reduced capacity to amplify complement activation, is linked to a lower risk of graft failure after kidney transplantation.

Figure 2
  • Download figure
  • Open in new tab
Figure 2 Kaplan–Meier curves of renal allograft survival according to the CFBR32Q variant

(A) Cumulative death-censored survival of kidney allografts based on the presence of the rs641153 complement factor B gene polymorphism (CFBR32Q) in allograft donors. Due to the low prevalence of donors homozygous for the CFB32Q variant, we merged heterozygote and homozygote donors into a single group. Donor kidneys carrying at least one CFB32Q variant (blue line – CFB32QR+CFB32QQ) were compared to donors who are homozygous for the reference CFB32R variant (black line - CFB32RR). (B) Cumulative death-censored survival of kidney allografts based on the donor-recipient paired genotypes of the CFB32Q variant, comparing (i) pairs with both the donor and recipient being homozygous for the reference CFB32R variant (black line), (ii) pairs where only the recipient carries the CFB32Q variant (light blue line), (iii) pairs where only the donor carries the CFB32Q variant (blue line), and (iv) pairs where both the donor and recipient carry the CFB32Q variant (dark blue line). Data represent death-censored survival curves and P-values were calculated using log-rank tests.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 2: Associations of the CFB polymorphism in the donor with graft loss

Next, we explored whether the combined genotype of donor-recipient pairs for the CFBR32Q polymorphism exerted a more significant influence on graft survival than the donor genotype alone. Based on the CFB32Q variant, donor-recipient pairs were divided into four groups (Fig. 2B). Recipients carrying the ‘less activating’ CFB32Q variant that received a graft with the ‘less activating’ CFB32Q variant demonstrated the most favorable outcome. Kaplan-Meier survival analyses revealed a significant difference in 15-year death-censored graft survival between donor-recipient pairs lacking the ‘less activating’ CFB32Q variant and those where both the donor and recipient carried the ‘less activating’ CFB32Q variant (graft loss: 28.3% versus 5.3%, P = 0.045). The ‘less activating’ CFB32Q variant in the donor appeared to exert a more substantial impact on graft survival compared to its presence in the recipient (Fig. 2B). Collectively, these findings confirm that complement variants influence the risk of graft loss, and their functional consequence substantiates the role of complement in kidney transplant injury.

3.3 The association of the complotype with allograft outcome

Finally, we investigated the impact of the combined presence of the C3R102G, CFHV62I, and CFBR32Qpolymorphisms on long-term allograft survival after kidney transplantation. Donor-recipient pairs were classified into 5 groups based on the combination of the three polymorphisms, referred to as the complotype. Analyses were performed separately for the complotype of the donor and the recipient. The groups consisted of the (i) high activity complotype, consisting of the gain-of-function C3102G variant and the reference variants for the other two polymorphisms (C3102G/CFB32R/CFH62V), the (ii) normal activity complotype, consisting of the reference variants for all three polymorphisms (C3102R/CFB32R/CFH62V), the (iii) dampened complotype, consisting of either the gain-of-function CFH62I or the loss-of-function CFB32Qvariant with reference variants for the other two polymorphisms (C3102R/CFB32R/CFH62I or C3102R/CFB32Q/CFH62V), the (iv) mixed Complotype, consisting of the gain-of-function C3102G variant together with the gain-of-function CFH62I and/or the loss-of-function CFB32Q, and the (v) low activity complotype, consisting of both the gain-of-function CFH62Iand the loss-of-function CFB32Q variant together with the reference C3102R variant (C3102R/CFB32Q/CFH62I).

Kaplan-Meier survival analysis revealed a significant difference in graft survival based on the donor’s complotype (Fig. 3; P = 0.024). Donor kidneys carrying the variant set that promotes the highest complement activity (high activity complotype – C3102G/CFB32R/CFH62V) had the shortest graft survival with a mean time to graft loss of 9.3 years, whereas donor kidneys with the variant set causing the lowest complement activity (low activity complotype – C3102R/CFB32Q/CFH62I) had the longest graft survival with a mean time to graft loss of 14.0 years (P = 0.001). After complete follow-up, the incidence of graft loss was 51.2% for donor kidneys with a high activity complotype, and 12.2% for donor kidneys with a low activity complotype. In univariable analysis, the donor’s complotype (as an ordinal variable) was significantly associated with 15-year death-censored graft survival (HR, 1.30; 95%-CI, 1.09 – 1.56; P = 0.005). When comparing donors with a low activity complotype to those with a high activity complotype, the HR for graft loss was 5.82 (95%-CI, 1.90 – 17.79; P = 0.002). In contrast, no association was observed between the recipient’s complotype and graft loss (Supplementary Figure S3, P = 0.99).

Figure 3
  • Download figure
  • Open in new tab
Figure 3 Kaplan–Meier curves of renal allograft survival according to the donor complotype

Cumulative death-censored graft survival of kidney transplants based on the combined presence of the C3R102G, CFHV62I, and CFBR32Q polymorphisms in allograft donors. The comparisons include: (i) high activity complotype (red line), which consists of the gain-of-function C3102Gvariant and the reference variants for the other two polymorphisms (C3102G/CFB32R/CFH62V); (ii) normal activity complotype (black line), consisting of the reference variants for all three polymorphisms (C3102R/CFB32R/CFH62V); (iii) dampened complotype (light blue line), consisting of either the gain-of-function CFH62I or the loss-of-function CFB32Q variant with reference variants for the other two polymorphisms (C3102R/CFB32R/CFH62I or C3102R/CFB32Q/CFH62V); (iv) mixed Complotype (yellow line), consisting of the gain-of-function C3102G variant together with the gain-of-function CFH62Iand/or the loss-of-function CFB32Q; and (v) low activity complotype (green line), consisting of both the gain-of-function CFH62I and the loss-of-function CFB32Q variant together with the reference C3102R variant (C3102R/CFB32Q/CFH62I). The data are represented through death-censored survival curves, and p-values were calculated using log-rank tests.

We conducted further investigations to examine potential differences in clinical characteristics among donor-recipient pairs with different donor complotypes. The only significant variation observed was Sirolimus use among the groups (Table 3, P = 0.04). Nevertheless, when adjusting for Sirolimus use, the donor complotype remained significantly associated with graft survival in Cox regression analysis (HR, 1.30; 95%-CI, 1.08 – 1.56; P = 0.005). To ensure an accurate interpretation of death-censored graft survival, we also analyzed patient survival to exclude this as a potential confounding variable. All-cause mortality was comparable among the donor complotype groups (Table 3, P = 0.84). Multivariable analysis was then conducted with stepwise adjustments for relevant clinical variables (Table 4), including donor characteristics (model 2), recipient characteristics (model 3), and transplant variables (model 4). In these analyses, the donor complotype retained its significant association with graft loss, irrespective of adjusting for other clinical variables. Lastly, we performed a multivariate analysis with a stepwise forward selection procedure (Table 5). In the final model, donor complotype, donor and recipient age, recipient blood type, and DGF were included. Following adjustment, the donor complotype (as an ordinal variable) exhibited an association with graft loss, with a hazard ratio of 1.31 (95%-CI, 1.08 – 1.58; P = 0.005) using the low activity complotype as a reference. In summary, these analyses unveil the synergistic impact of the C3, CFH, and CFB polymorphisms in the donor on graft loss following kidney transplantation, and this association remains significant even when accounting for other determinants and potential confounders.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 3: Baseline characteristics of transplant pairs based on the donor complotype.
View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 4: Associations of the complotype in the donor with graft loss
View this table:
  • View inline
  • View popup
  • Download powerpoint
Table 5: Competitive analysis of the associations of clinical factors with graft loss

4. Discussion

Developing a comprehensive understanding of the immunological mechanisms underlying graft loss is crucial to create tailored treatment strategies in kidney transplantation.37,38 Studies of human genetics offer a complementary model to investigate mechanisms for the purpose of target validation.39,40 Moreover, therapeutic targets that are substantiated by human genetic evidence in disease association studies have a twofold higher probability of resulting in approved clinical drugs.41,42 The main finding of the current study is that disease-associated complement variants are common in both kidney transplant recipients and their donors, while these complement polymorphisms individually only have a modest impact on graft loss risk, when combined their collective effects significantly influence graft survival after kidney transplantation. Furthermore, our data support the notion that locally produced complement, originating from the donor kidney, plays a more prominent role in graft failure following kidney transplantation than complement generated by the recipient.

Evidence of the polymorphic nature of the complement system dates back to the 1960s, with the identification of two distinct C3 variants.43 At the protein level, the C3R102G polymorphism was classified as a slow variant (C3S – C3102R) and a fast variant (C3F – C3102G), based on the speed of C3 to migrate through a gel-electrophoresis system.43 Associations of the C3102G variant with different diseases has been well established, and the molecular basis for these disease associations was recently elucidated.19 Similarly, in the 1970s, the CFBR32Q variant was also first described at the protein level.44 Later on, it was shown that factor H was highly polymorphic as well.45 The relationship between the C3R102G polymorphism and outcome after kidney transplantation has been explored in multiple studies with varying results.22,26,46,47 To our knowledge, the current study is the first to examine the individual impact of the CFBR32Q and CFHV62Ipolymorphisms in both the donor and recipient on long-term graft survival after kidney transplantation. We observed a 34% risk reduction for graft loss in donor kidneys carrying at least one CFB32Q variant compared to kidneys homozygous for the reference CFB32R variant. Fittingly, the CFB32Q variant exhibits lower efficiency in amplifying complement activation. Notably, donor-recipient pairs both harboring the CFB32Q variant had the most favorable long-term outcome, revealing significant interactions between the complement systems of the donor and recipient, with a prominent role for donor-derived complement. In addition, our data strongly suggest that factor B presents an attractive therapeutic target in transplantation, further supported by preclinical models demonstrating the efficacy of factor B inhibitors in kidney transplantation.11,48,49 Altogether, these findings contribute to the growing body of evidence highlighting the pivotal role of the complement system in kidney transplant survival, underscoring the potential effectiveness of complement inhibitors to improve these outcomes.5,6

The concept of the complotype, defined as the complete inherited set of complement genes, was first introduced by Harris et al.21 Evidence supporting this concept was provided by in vitro experiments that simultaneously examined the combined effects of multiple polymorphic variants on complement activity. These studies revealed that individual complement gene variants had a relatively small impact, but when combined, their effects were additive and resulted in significantly higher complement turnover.19,20 Subsequently, Paun et al. provided the first in vivo evidence by showing an association between the combination of the C3102G, CFBL9H, CFBR32Q, and CFHV62I variants with higher systemic complement activation as well as with age-related macular degeneration (AMD).50 Presently, we found a dose-response relationship of a complotype composed of the C3R102G, CFBR32Q, and CFHV62Ivariants in the donor with long-term allograft survival. Furthermore, when comparing donor kidneys carrying the three "risk" variants (C3102G/CFB32R/CFH62V) to those carrying the three "protective" variants (C3102R/CFB32Q/CFH62I), the risk of graft loss was increased by approximately 400%. In contrast, the individual ’risk’ variants in the donor raised graft loss risk only by 51% for CFB32R, 33% for C3102G, and 17% for CFH62V. Mező et al. attempted to analyze the same combination in kidney transplantation, but their study focused solely on recipient genotypes, dividing them into two groups and assessing shorter graft survival. Nevertheless, graft survival was worse in recipients harboring at least one C3102G, CFB32R, and CFH62V variant.51 Another study demonstrated that the combined presence of two polymorphisms in the membrane-bound complement regulators CD46 and CD59 was associated with a favorable outcome after kidney transplantation, but the functional consequences of these variants are not well described, making their results harder to interpret.27 In addition to the additive effects of complement polymorphisms shown here, we have previously demonstrated the possibility of genetic compensation by different complement variants with opposing effects.35 Overall, these data present compelling evidence that instead of focusing solely on individual complement polymorphisms or complement genes for graft loss risk assessment, we should analyze the repertoire of common complement variants.

Clinical trials testing the efficacy of therapeutic complement inhibition in kidney transplantation have so far provided mixed results.7 Collectively, treatment of kidney transplant recipients with an anti-C5 monoclonal antibody did not result in major outcome improvements.52,53 Clinical trials involving C1-inhibitor in sensitized recipients yielded disappointing results as well.54 Yet, the perioperative administration of C1-inhibitor in kidney transplantation showed promising effects in a recent placebo-controlled randomized trial.55 The findings of our study offer valuable insights regarding the use of complement inhibitors in kidney transplantation, specifically on the site of action and patient selection. Importantly, we found that the complotype of the donor graft, rather than that of the recipient, was associated with the risk of graft loss. In accordance, Pratt et al. were to first to reveal the key role of donor-derived complement in allograft survival following experimental kidney transplantation in a murine model.56 Since then, accumulating data has further established the critical significance of locally produced complement in impacting outcomes after kidney transplantation.26,57–62 Moreover, the kidney significantly contributes to the circulating levels of complement.63,64 Altogether, this suggests that complement-targeted therapy in kidney transplantation should focus on the donor graft as the site of action. Immunosuppression in transplantation forms a delicate balance between the risks of graft rejection and infection, and while complement therapeutics may be beneficial in some patients, the harms may outweigh the benefits in others. Analyzing the complotype of donor-recipient pairs can identify those who are more susceptible to complement-mediated transplant kidney injury and may benefit from complement inhibition while avoiding over-immunosuppression in those with a low activity complotype, which would increase infectious risk without significantly improving graft outcomes. Further research is first needed to enable more comprehensive complotype testing that includes the analysis of functional polymorphisms of all complement genes. In addition, while the complotype sets an individual’s intrinsic complement activity, it does not provide information on activation. Ideally, the genetic analysis should thus be combined with additional tools such as plasma complement measurements and molecular imaging.65,66 Altogether, this could enable an individualized approach to help select individuals that would benefit from complement inhibition and help determine the timing, dosing, duration, and type of complement blocker needed.

This study has some limitations. While the association found in this study is expected to be causal, our study is prospective and observational in nature, which means we cannot definitively prove causality with our results. Additionally, due to the lack of available plasma samples, we could not measure complement activation in our cohort. However, it is worth noting that we deliberately selected the three most widely studied and functionally characterized complement polymorphisms, which helps strengthen the validity of our findings. The current study did not have sufficient statistical power to conduct donor-recipient complotype combinations or subgroup analyses (e.g., by sex, donor type, or recipient cause of renal failure) to explore the potential influence of these factors on the observed association. On the contrary, our study has several notable strengths, including the large size of the transplantation cohort, the combined analysis of the donor and recipient genotypes, the lengthy and thorough follow-up period, and the utilization of a robust and clinically significant endpoint (graft loss).

In conclusion, the current study has demonstrated that a complotype composed of C3R102G, CFBR32Q, and CFHV62I variants in the donor is strongly associated with long-term allograft survival following kidney transplantation. These findings are relevant in the context of therapeutic strategies for complement inhibition in kidney transplantation. The introduction of genomic and other molecular profiling techniques presents an unparalleled opportunity to implement precision pharmacotherapy in transplantation with the aim of enhancing patient outcomes. By utilizing genotype-based patient stratification, we can potentially identify donor-recipient pairs with a genetic predisposition to complement-mediated transplant kidney injury, rendering them excellent candidates for therapeutic complement inhibition in kidney transplantation. Furthermore, future studies should explore whether other complement polymorphisms in recipients can counteract the observed effects of the C3R102G, CFBR32Q, and CFHV62Ivariants in the donors.

Funding

FP was supported by a Veni grant of the Netherlands Organization for Scientific Research (NWO), project no. 09150162010171.

Statement of competing financial interests

FP owns or owned stock in Apellis Pharmaceuticals, Chemocentryx, InflaRx, Iveric Bio, as well as Omeros and has been involved as a consultant for Invizius and Alnylam Pharmaceuticals. JMT and VMH are consultants for Q32 Bio, Inc., a company developing complement inhibitors. Both also hold stock and may receive royalty income from Q32 Bio, Inc. The remaining authors of this paper declare that they have no competing interests.

Data availability statement

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

Author contributions

J.D. and M.A.S. formulated the overarching research aim and designed the study; F.P., and J.D. performed experiments and collected data; F.P. analyzed and interpreted the data; M.G.C., B.F., S.K.E., V.P., V.M.H., M.R.D., S.P.B., J.D., M.A.S., and J.M.T., critically reviewed the manuscript and provided commentary; F.P. wrote the first draft of the manuscript and revised it for publication; All authors read and approved the final manuscript.

Acknowledgements

The authors thank the members of the REGaTTA cohort (REnal GeneTics TrAnsplantation; University Medical Center Groningen, University of Groningen, Groningen, the Netherlands): S.J.L. Bakker, J. van den Born, M.H. de Borst, H. van Goor, J.L. Hillebrands, B.G. Hepkema, G.J. Navis and H. Snieder.

Abbreviations

AMD
Age-related macular degeneration
C3
Complement component 3 gene
CFB
Complement Factor B gene
CFH
Complement Factor H gene
DGF
Delayed graft function
HLA
Human leukocyte antigen
SNP
Single-nucleotide polymorphism

References

  1. 1.↵
    Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: A key system for immune surveillance and homeostasis. Nat Immunol. 2010;11(9):785–797. doi:10.1038/ni.1923
    OpenUrlCrossRefPubMedWeb of Science
  2. 2.↵
    Harboe M, Garred P, Borgen MS, Stahl GL, Roos A, Mollnes TE. Design of a complement mannose-binding lectin pathway-specific activation system applicable at low serum dilutions. Clin Exp Immunol. 2006;144(3):512–520. doi:10.1111/j.1365-2249.2006.03072.x
    OpenUrlCrossRefPubMedWeb of Science
  3. 3.↵
    Harboe M, Ulvund G, Vien L, Fung M, Mollnes TE. The quantitative role of alternative pathway amplification in classical pathway induced terminal complement activation. Clin Exp Immunol. 2004;138(3):439–446. doi:10.1111/j.1365-2249.2004.02627.x
    OpenUrlCrossRefPubMedWeb of Science
  4. 4.↵
    Poppelaars F, Goicoechea de Jorge E, Jongerius I, et al. A Family Affair: Addressing the Challenges of Factor H and the Related Proteins. Front Immunol. 2021;12. doi:10.3389/fimmu.2021.660194
    OpenUrlCrossRef
  5. 5.↵
    Jager NM, Poppelaars F, Daha MR, Seelen MA. Complement in renal transplantation: The road to translation. Mol Immunol. 2017. doi:10.1016/j.molimm.2017.05.014
    OpenUrlCrossRef
  6. 6.↵
    Poppelaars F, Seelen MA. Complement-mediated inflammation and injury in brain dead organ donors. Mol Immunol. 2016. doi:10.1016/j.molimm.2016.11.004
    OpenUrlCrossRef
  7. 7.↵
    Golshayan D, Schwotzer N, Fakhouri F, Zuber J. Targeting the Complement Pathway in Kidney Transplantation. J Am Soc Nephrol. July 2023. doi:10.1681/ASN.0000000000000192
    OpenUrlCrossRef
  8. 8.
    Damman J, Daha MR, van Son WJ, Leuvenink HG, Ploeg RJ, Seelen MA. Crosstalk between complement and Toll-like receptor activation in relation to donor brain death and renal ischemia-reperfusion injury. Am J Transplant. 2011;11(4):660–669. doi:10.1111/j.1600-6143.2011.03475.x
    OpenUrlCrossRefPubMed
  9. 9.↵
    Bromberg JS, Heeger PS, Li XC. Evolving paradigms that determine the fate of an allograft: Minireview. Am J Transplant. 2010;10(5):1143–1148. doi:10.1111/j.1600-6143.2010.03033.x
    OpenUrlCrossRefPubMed
  10. 10.↵
    Poppelaars F, Jager NM, Kotimaa J, et al. C1-inhibitor Treatment Decreases Renal Injury in an Established Brain-dead Rat Model. Transplantation. 2017. doi:10.1097/TP.0000000000001895
    OpenUrlCrossRef
  11. 11.↵
    Jager NM, van Zanden JE, Subías M, et al. Blocking Complement Factor B Activation Reduces Renal Injury and Inflammation in a Rat Brain Death Model. Front Immunol. 2019;10:2528. doi:10.3389/FIMMU.2019.02528/BIBTEX
    OpenUrlCrossRef
  12. 12.
    Poppelaars F, van Werkhoven MB, Kotimaa J, et al. Critical role for complement receptor C5aR2 in the pathogenesis of renal ischemia-reperfusion injury. FASEB J. April 2017:fj.201601218R. doi:10.1096/fj.201601218R
    OpenUrlCrossRefPubMed
  13. 13.
    Schmitz R, Fitch ZW, Schroder PM, et al. C3 complement inhibition prevents antibody-mediated rejection and prolongs renal allograft survival in sensitized non-human primates. Nat Commun. 2021;12(1). doi:10.1038/s41467-021-25745-7
    OpenUrlCrossRef
  14. 14.↵
    Castellano G, Melchiorre R, Loverre A, et al. Therapeutic targeting of classical and lectin pathways of complement protects from ischemia-reperfusion-induced renal damage. Am J Pathol. 2010;176(4):1648–1659. doi:10.2353/ajpath.2010.090276
    OpenUrlCrossRefPubMedWeb of Science
  15. 15.↵
    Lemaire M, Noone D, Lapeyraque AL, Licht C, Frémeaux-Bacchi V. Inherited Kidney Complement Diseases. Clin J Am Soc Nephrol. 2021;16(6):942–956. doi:10.2215/CJN.11830720
    OpenUrlAbstract/FREE Full Text
  16. 16.
    Goicoechea de Jorge E, López Lera A, Bayarri-Olmos R, Yebenes H, Lopez-Trascasa M, Rodríguez de Córdoba S. Common and rare genetic variants of complement components in human disease. Mol Immunol. 2018;102:42–57. doi:10.1016/j.molimm.2018.06.011
    OpenUrlCrossRefPubMed
  17. 17.↵
    Rodríguez de Córdoba S. Genetic variability shapes the alternative pathway complement activity and predisposition to complement-related diseases. Immunol Rev. 2023;313(1):71–90. doi:10.1111/imr.13131
    OpenUrlCrossRef
  18. 18.↵
    Grumach AS, Kirschfink M. Are complement deficiencies really rare? Overview on prevalence, clinical importance and modern diagnostic approach. Mol Immunol. 2014;61(2):110–117. doi:10.1016/j.molimm.2014.06.030
    OpenUrlCrossRef
  19. 19.↵
    Heurich M, Martinez-Barricarte R, Francis NJ, et al. Common polymorphisms in C3, factor B, and factor H collaborate to determine systemic complement activity and disease risk. Proc Natl Acad Sci. 2011;108(21):8761–8766. doi:10.1073/pnas.1019338108
    OpenUrlAbstract/FREE Full Text
  20. 20.↵
    Lay E, Nutland S, Smith JE, et al. Complotype affects the extent of down-regulation by Factor I of the C3b feedback cycle in vitro. Clin Exp Immunol. 2015;181(2):314–322. doi:10.1111/cei.12437
    OpenUrlCrossRef
  21. 21.↵
    Harris CL, Heurich M, Rodriguez de Cordoba S, Morgan BP. The complotype: dictating risk for inflammation and infection. Trends Immunol. 2012;33(10):513–521. doi:10.1016/j.it.2012.06.001
    OpenUrlCrossRefPubMedWeb of Science
  22. 22.↵
    Brown KM, Kondeatis E, Vaughan RW, et al. Influence of donor C3 allotype on late renal-transplantation outcome. N Engl J Med. 2006;354(19):2014–2023. doi:10.1056/NEJMoa052825
    OpenUrlCrossRefPubMedWeb of Science
  23. 23.
    Bay JT, Schejbel L, Madsen HO, Sørensen SS, Hansen JM, Garred P. Low C4 gene copy numbers are associated with superior graft survival in patients transplanted with a deceased donor kidney. Kidney Int. 2013;84(3):562–569. doi:10.1038/ki.2013.195
    OpenUrlCrossRefPubMed
  24. 24.
    Bay JT, Sørensen SS, Hansen JM, Madsen HO, Garred P. Low mannose-binding lectin serum levels are associated with reduced kidney graft survival. Kidney Int. 2013;83(2):264–271. doi:10.1038/ki.2012.373
    OpenUrlCrossRefPubMed
  25. 25.
    Eikmans M, de Canck I, van der Pol P, et al. The Functional Polymorphism Ala258Ser in the Innate Receptor Gene Ficolin-2 in the Donor Predicts Improved Renal Transplant Outcome. Transplant J. 2012;94(5):478–485. doi:10.1097/TP.0b013e31825c5967
    OpenUrlCrossRefPubMed
  26. 26.↵
    Damman J, Daha MR, Leuvenink HG, et al. Association of complement C3 gene variants with renal transplant outcome of deceased cardiac dead donor kidneys. Am J Transplant. 2012;12(3):660–668. doi:10.1111/j.1600-6143.2011.03880.x
    OpenUrlCrossRefPubMed
  27. 27.↵
    Michielsen LA, van Zuilen AD, Kardol-Hoefnagel T, Verhaar MC, Otten HG. Association between promoter polymorphisms in CD46 and CD59 in kidney donors and transplant outcome. Front Immunol. 2018;9(MAY):352548. doi:10.3389/fimmu.2018.00972
    OpenUrlCrossRef
  28. 28.↵
    Michielsen LA, van Zuilen AD, Muskens IS, Verhaar MC, Otten HG. Complement Polymorphisms in Kidney Transplantation: Critical in Graft Rejection? Am J Transplant. 2017;17(8):2000–2007. doi:10.1111/ajt.14199
    OpenUrlCrossRef
  29. 29.↵
    Montes T, Tortajada A, Morgan BP, De Córdoba SR, Harris CL. Functional basis of protection against age-related macular degeneration conferred by a common polymorphism in complement factor B. Proc Natl Acad Sci U S A. 2009;106(11):4366–4371. doi:10.1073/PNAS.0812584106
    OpenUrlAbstract/FREE Full Text
  30. 30.↵
    Tortajada A, Montes T, Martínez-Barricarte R, Morgan BP, Harris CL, de Córdoba SR. The disease-protective complement factor H allotypic variant Ile62 shows increased binding affinity for C3b and enhanced cofactor activity. Hum Mol Genet. 2009;18(18):3452–3461. doi:10.1093/HMG/DDP289
    OpenUrlCrossRefPubMedWeb of Science
  31. 31.↵
    Eskandari SK, Gaya da Costa M, Faria B, et al. An interleukin 6-based genetic risk score strengthened with interleukin 10 polymorphisms associated with long-term kidney allograft outcomes. Am J Transplant. 2022;22(August):45–57. doi:10.1111/ajt.17212
    OpenUrlCrossRef
  32. 32.
    Poppelaars F, Gaya da Costa M, Eskandari SK, Damman J, Seelen MA. Donor genetic variants in interleukin-6 and interleukin-6 receptor associate with biopsy-proven rejection following kidney transplantation. Sci Rep. 2021;11(1):2021.04.17.21255669. doi:10.1038/s41598-021-95714-z
    OpenUrlCrossRef
  33. 33.
    Poppelaars F, Gaya Da Costa M, Faria B, Eskandari SK, Damman J, Seelen MA. A functional TGFB1 polymorphism in the donor associates with long-term graft survival after kidney transplantation. Clin Kidney J. 2022;15(2):278–286. doi:10.1093/ckj/sfab175
    OpenUrlCrossRef
  34. 34.
    Poppelaars F, Costa MG da, Faria B, Eskandari SK, Seelen MA, Damman J. Tumor necrosis factor-α gene polymorphism is associated with short-and long-term kidney allograft outcomes. medRxiv. July 2021:2021.07.28.21261294. doi:10.1101/2021.07.28.21261294
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    Poppelaars F, Eskandari SK, Damman J, et al. Carboxypeptidase B2 gene polymorphisms in the donor associate with kidney allograft loss. medRxiv. 2023. doi:10.1101/2023.05.08.23289675
    OpenUrlAbstract/FREE Full Text
  36. 36.↵
    Poppelaars F, Eskandari SK, Damman J, Seelen MA, Faria B, da Costa MG. A non-muscle myosin heavy chain 9 genetic variant is associated with graft failure following kidney transplantation. medRxiv. January 2022:2022.03.29.22272996. doi:10.1101/2022.03.29.22272996
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    Ferrari P, Weimar W, Johnson RJ, Lim WH, Tinckam KJ. Kidney paired donation: principles, protocols and programs. Nephrol Dial Transplant Off Publ Eur Dial Transpl Assoc - Eur Ren Assoc. 2015;30(8):1276–1285. doi:10.1093/ndt/gfu309
    OpenUrlCrossRefPubMed
  38. 38.↵
    Faitot F, Michard B, Artzner T. Organ allocation in the age of the algorithm: avoiding futile transplantation - utility in allocation. Curr Opin Organ Transplant. 2020;25(3):305–309. doi:10.1097/MOT.0000000000000752
    OpenUrlCrossRef
  39. 39.↵
    Yang JYC, Sarwal MM. Transplant genetics and genomics. Nat Rev Genet. 2017;18(5):309-326. doi:10.1038/nrg.2017.12
    OpenUrlCrossRef
  40. 40.↵
    Zanoni F, Kiryluk K. Genetic background and transplantation outcomes: Insights from genome-wide association studies. Curr Opin Organ Transplant. 2020;25(1):35–41. doi:10.1097/MOT.0000000000000718
    OpenUrlCrossRef
  41. 41.↵
    Nelson MR, Tipney H, Painter JL, et al. The support of human genetic evidence for approved drug indications. Nat Genet. 2015;47(8):856–860. doi:10.1038/ng.3314
    OpenUrlCrossRefPubMed
  42. 42.↵
    King EA, Wade Davis J, Degner JF. Are drug targets with genetic support twice as likely to be approved? Revised estimates of the impact of genetic support for drug mechanisms on the probability of drug approval. PLoS Genet. 2019;15(12):e1008489. doi:10.1371/journal.pgen.1008489
    OpenUrlCrossRefPubMed
  43. 43.↵
    Wieme RJ, Demeulenaere L. Genetically determined electrophoretic variant of the human complement component C′;3. Nature. 1967;214(5092):1042–1043. doi:10.1038/2141042a0
    OpenUrlCrossRefPubMed
  44. 44.↵
    Alper CA, Boenisch T, Watson L. Genetic polymorphism in human glycine-rich beta-glycoprotein. J Exp Med. 1972;135(1):68–80. doi:10.1084/jem.135.1.68
    OpenUrlAbstract
  45. 45.↵
    Rodríguez de Córdoba S, Rubinstein P. Genetic polymorphism of human factor H (beta 1H). J Immunol. 1984;132(4):1906–1908. doi:10.4049/jimmunol.132.4.1906
    OpenUrlAbstract
  46. 46.↵
    Varagunam M, Yaqoob MM, Döhler B, Opelz G. C3 polymorphisms and allograft outcome in renal transplantation. N Engl J Med. 2009;360(9):874–880. doi:10.1056/NEJMoa0801861
    OpenUrlCrossRefPubMedWeb of Science
  47. 47.↵
    Bazyar N, Azarpira N, Khatami SR, Galehdari H, Salahi H. Complement C3 gene polymorphism in renal transplantation (an Iranian experience). Gene. 2012;498(2):254–258. doi:10.1016/j.gene.2012.01.062
    OpenUrlCrossRef
  48. 48.↵
    Thurman JM, Royer PA, Ljubanovic D, et al. Treatment with an inhibitory monoclonal antibody to mouse factor B protects mice from induction of apoptosis and renal ischemia/reperfusion injury. J Am Soc Nephrol. 2006;17(3):707–715. doi:10.1681/ASN.2005070698
    OpenUrlAbstract/FREE Full Text
  49. 49.↵
    Casiraghi F, Azzollini N, Todeschini M, et al. Complement Alternative Pathway Deficiency in Recipients Protects Kidney Allograft From Ischemia/Reperfusion Injury and Alloreactive T Cell Response. Am J Transplant. 2017;17(9):2312–2325. doi:10.1111/ajt.14262
    OpenUrlCrossRef
  50. 50.↵
    Paun CC, Lechanteur YTE, Groenewoud JMM, et al. A Novel Complotype Combination Associates with Age-Related Macular Degeneration and High Complement Activation Levels in vivo. Sci Rep. 2016;6. doi:10.1038/srep26568
    OpenUrlCrossRef
  51. 51.↵
    Mező B, Reindl-Schwaighofer R, Eskandary F, et al. High-activity Classical and Alternative Complement Pathway Genotypes - Association with Donor-specific Antibody-triggered Injury and Renal Allograft Survival. Transplant Direct. 2020;6(3). doi:10.1097/TXD.0000000000000978
    OpenUrlCrossRef
  52. 52.↵
    Schröppel B, Akalin E, Baweja M, et al. Peritransplant eculizumab does not prevent delayed graft function in deceased donor kidney transplant recipients: Results of two randomized controlled pilot trials. Am J Transplant. 2020;20(2):564–572. doi:10.1111/AJT.15580
    OpenUrlCrossRef
  53. 53.↵
    Marks WH, Mamode N, Montgomery RA, et al. Safety and efficacy of eculizumab in the prevention of antibody-mediated rejection in living-donor kidney transplant recipients requiring desensitization therapy: A randomized trial. Am J Transplant. 2019;19(10):2876–2888. doi:10.1111/AJT.15364
    OpenUrlCrossRef
  54. 54.↵
    Vo AA, Zeevi A, Choi J, et al. A phase I/II placebo-controlled trial of C1-inhibitor for prevention of antibody-mediated rejection in HLA sensitized patients. Transplantation. 2015;99(2):299–308. doi:10.1097/TP.0000000000000592
    OpenUrlCrossRefPubMed
  55. 55.↵
    Huang E, Vo A, Choi J, et al. Three-Year Outcomes of a Randomized, Double-Blind, Placebo-Controlled Study Assessing Safety and Efficacy of C1 Esterase Inhibitor for Prevention of Delayed Graft Function in Deceased Donor Kidney Transplant Recipients. Clin J Am Soc Nephrol. 2020;15(1):109–116. doi:10.2215/CJN.04840419
    OpenUrlAbstract/FREE Full Text
  56. 56.↵
    Pratt JR, Basheer SA, Sacks SH. Local synthesis of complement component C3 regulates acute renal transplant rejection. Nat Med. 2002;8(6):582–587. doi:10.1038/nm0602-582
    OpenUrlCrossRefPubMedWeb of Science
  57. 57.↵
    Damman J, Nijboer WN, Schuurs TA, et al. Local renal complement C3 induction by donor brain death is associated with reduced renal allograft function after transplantation. Nephrol Dial Transplant. 2011;26(7):2345–2354. doi:10.1093/ndt/gfq717
    OpenUrlCrossRefPubMed
  58. 58.
    Peng Q, Li K, Anderson K, et al. Local production and activation of complement up-regulates the allostimulatory function of dendritic cells through C3a-C3aR interaction. Blood. 2008;111(4):2452–2461. doi:10.1182/BLOOD-2007-06-095018
    OpenUrlAbstract/FREE Full Text
  59. 59.
    Pratt JR, Jones ME, Dong J, et al. Nontransgenic Hyperexpression of a Complement Regulator in Donor Kidney Modulates Transplant Ischemia/Reperfusion Damage, Acute Rejection, and Chronic Nephropathy. Am J Pathol. 2003;163(4):1457. doi:10.1016/S0002-9440(10)63503-1
    OpenUrlCrossRefPubMedWeb of Science
  60. 60.
    Damman J, Bloks VW, Daha MR, et al. Hypoxia and Complement-and-Coagulation Pathways in the Deceased Organ Donor as the Major Target for Intervention to Improve Renal Allograft Outcome. Transplantation. 2014;99(6):1. doi:10.1097/TP.0000000000000500
    OpenUrlCrossRef
  61. 61.
    Farrar CA, Zhou W, Lin T, Sacks SH. Local extravascular pool of C3 is a determinant of postischemic acute renal failure. FASEB J. 2006;20(2):217–226. doi:10.1096/FJ.05-4747COM
    OpenUrlCrossRefPubMedWeb of Science
  62. 62.↵
    Sheerm NS, Risley P, Abe K, et al. Synthesis of complement protein C3 in the kidney is an important mediator of local tissue injury. FASEB J. 2008;22(4):1065–1072. doi:10.1096/FJ.07-8719COM
    OpenUrlCrossRefPubMed
  63. 63.↵
    Naughton MA, Botto M, Carter MJ, Alexander GJ, Goldman JM, Walport MJ. Extrahepatic secreted complement C3 contributes to circulating C3 levels in humans. J Immunol. 1996;156(8):3051–3056. doi:10.4049/jimmunol.156.8.3051
    OpenUrlAbstract/FREE Full Text
  64. 64.↵
    Tang S, Zhou W, Sheerin NS, Vaughan RW, Sacks SH. Contribution of renal secreted complement C3 to the circulating pool in humans. J Immunol. 1999;162(7):4336–4341. http://www.ncbi.nlm.nih.gov/pubmed/10201966. Accessed August 29, 2017.
    OpenUrlAbstract/FREE Full Text
  65. 65.↵
    Renner B, Poppelaars F, Laskowski J, et al. Noninvasive Detection of iC3b/C3d Deposits in the Kidney Using a Novel Bioluminescent Imaging Probe. J Am Soc Nephrol. 2023;Publish Ah(7). doi:10.1681/asn.0000000000000129
    OpenUrlCrossRef
  66. 66.↵
    Banerjee P, Veuskens B, de Jorge EG, et al. Evaluating the clinical utility of measuring levels of factor H and the related proteins. Mol Immunol. 2022;151:166–182. doi:10.1016/j.molimm.2022.08.010
    OpenUrlCrossRef
Back to top
PreviousNext
Posted October 25, 2023.
Download PDF

Supplementary Material

Data/Code
Email

Thank you for your interest in spreading the word about medRxiv.

NOTE: Your email address is requested solely to identify you as the sender of this article.

Enter multiple addresses on separate lines or separate them with commas.
Synergistic impact of three complement polymorphisms in the donor, not the recipient, on long-term kidney allograft survival
(Your Name) has forwarded a page to you from medRxiv
(Your Name) thought you would like to see this page from the medRxiv website.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Share
Synergistic impact of three complement polymorphisms in the donor, not the recipient, on long-term kidney allograft survival
Felix Poppelaars, Mariana Gaya da Costa, Bernardo Faria, Siawosh K. Eskandari, Vojtech Petr, V. Michael Holers, Mohamed R. Daha, Stefan P. Berger, Jeffrey Damman, Marc A. Seelen, Joshua M. Thurman
medRxiv 2023.10.24.23297481; doi: https://doi.org/10.1101/2023.10.24.23297481
Twitter logo Facebook logo LinkedIn logo Mendeley logo
Citation Tools
Synergistic impact of three complement polymorphisms in the donor, not the recipient, on long-term kidney allograft survival
Felix Poppelaars, Mariana Gaya da Costa, Bernardo Faria, Siawosh K. Eskandari, Vojtech Petr, V. Michael Holers, Mohamed R. Daha, Stefan P. Berger, Jeffrey Damman, Marc A. Seelen, Joshua M. Thurman
medRxiv 2023.10.24.23297481; doi: https://doi.org/10.1101/2023.10.24.23297481

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Subject Area

  • Transplantation
Subject Areas
All Articles
  • Addiction Medicine (349)
  • Allergy and Immunology (668)
  • Allergy and Immunology (668)
  • Anesthesia (181)
  • Cardiovascular Medicine (2648)
  • Dentistry and Oral Medicine (316)
  • Dermatology (223)
  • Emergency Medicine (399)
  • Endocrinology (including Diabetes Mellitus and Metabolic Disease) (942)
  • Epidemiology (12228)
  • Forensic Medicine (10)
  • Gastroenterology (759)
  • Genetic and Genomic Medicine (4103)
  • Geriatric Medicine (387)
  • Health Economics (680)
  • Health Informatics (2657)
  • Health Policy (1005)
  • Health Systems and Quality Improvement (985)
  • Hematology (363)
  • HIV/AIDS (851)
  • Infectious Diseases (except HIV/AIDS) (13695)
  • Intensive Care and Critical Care Medicine (797)
  • Medical Education (399)
  • Medical Ethics (109)
  • Nephrology (436)
  • Neurology (3882)
  • Nursing (209)
  • Nutrition (577)
  • Obstetrics and Gynecology (739)
  • Occupational and Environmental Health (695)
  • Oncology (2030)
  • Ophthalmology (585)
  • Orthopedics (240)
  • Otolaryngology (306)
  • Pain Medicine (250)
  • Palliative Medicine (75)
  • Pathology (473)
  • Pediatrics (1115)
  • Pharmacology and Therapeutics (466)
  • Primary Care Research (452)
  • Psychiatry and Clinical Psychology (3432)
  • Public and Global Health (6527)
  • Radiology and Imaging (1403)
  • Rehabilitation Medicine and Physical Therapy (814)
  • Respiratory Medicine (871)
  • Rheumatology (409)
  • Sexual and Reproductive Health (410)
  • Sports Medicine (342)
  • Surgery (448)
  • Toxicology (53)
  • Transplantation (185)
  • Urology (165)