Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Nov 28.
Published in final edited form as: Transpl Int. 2017 Nov 8;31(3):263–277. doi: 10.1111/tri.13084

Genetics of acute rejection after kidney transplantation

Casey R Dorr 1,2, William S Oetting 3, Pamala A Jacobson 3, Ajay K Israni 1,2,4
PMCID: PMC6260834  NIHMSID: NIHMS997663  PMID: 29030886

SUMMARY

Treatment of acute rejection (AR) following kidney transplantation has improved in recent years, but there are still limitations to successful outcomes. This review article covers literature in regard to recipient and donor genetics of AR kidney and secondarily of liver allografts. Many candidate gene and some genome-wide association studies (GWASs) have been conducted for AR in kidney transplantation. Genetic associations with AR in kidney and liver are mostly weak, and in most cases, the associations have not been reproducible. A limitation in the study of AR is the lack of sufficiently large populations that account for population stratification to study the AR phenotype which in this era occurs in <10% of transplants. Furthermore, the AR phenotype has been difficult to define and the definitions of classifications have evolved over time. Literature related to the pharmacogenomics of tacrolimus is robust and has been validated in many studies. Associations between gene expression and AR are emerging as markers of outcomes and AR classification. In the future, combinations of pretransplant genotype for AR risk prediction, genotype-based immune suppressant dosing, and pharmacogenomic markers to select AR maintenance or treatment and expression markers from biopsies may provide valuable clinical tools for guiding treatment.

Keywords: acute rejection, genetics, kidney, single nucleotide polymorphism, tacrolimus, transcriptomics, transplantation

Introduction

This article is a review of literature pertaining to the genetics of acute rejection (AR) in kidney, and to some extent liver, allograft recipients. There are three primary types of allograft rejection: hyperacute rejection that occurs minutes after the transplant, AR, which occurs days to months after transplant, and chronic rejection that occurs long after transplant. AR has further been classified as antibody-mediated rejection (ABMR), T cell-mediated rejection (TCMR), C4d-negative ABMR, and mixed; in this review, we focused on all forms of AR. The definition of the AR phenotype has changed over time as it is further understood. Mechanistically, these types of AR are quite different but have a similar endpoint of the allograft being rejected. Thus, the heterogeneity of the AR phenotype has made genetic association studies difficult to determine and validate. As there is continual updating of the classification and definition of AR, such as the Banff classifications [1,2], this review article does not restrict manuscripts reviewed to a single AR definition, but leaves the definition to the authors of the reviewed literature.

As with the definition of AR, methods used to study the genetics associated with AR have also changed over time. DNA and/or RNA is extracted from the blood or the biopsy and then genetically assessed for association with AR. These association tests have evolved from testing candidate single nucleotide polymorphisms (SNPs), to testing multiple candidate SNPs, then panels of SNPs on gene chips, to genome-wide SNP chips, and we are now identifying known and novel SNPs using whole-genome and targeted next-generation sequencing. The majority of these AR studies were case controls investigating recipients with or without AR. Although some of these studies account for population stratification, such as a recent GWAS [3], many of them did not. This article also reviews literature on the pharmacogenomics and transcriptomics of AR.

Single nucleotide polymorphisms associated with acute rejection

Although outcome after solid organ transplants has improved in recent years, success is still limited by AR, chronic rejection (chronic allograft nephropathy), and graft failure. AR has been repeatedly shown to be one of the strongest predictors of allograft survival, and therefore, there is great interest in finding genetic markers associated with AR and AR risk [4]. Throughout the last few decades, there have been multiple reports in which investigators have attempted to associate genetic variation with AR risk. The strongest association between genetic variants and transplant outcome has been with the major histocompatibility complex (MHC) antigens, also called human leukocyte antigens (HLA) [5]. Because of this strong association, transplant centers try to match as many HLA alleles between the donor and the recipient to lower the immunogenicity of the allograft and reduce the risk of AR. There have also been additional reports showing an association with AR of genetic variants in other genes. Most genetic association studies aim to find interesting genes and then attempt to understand the biological relevance for those genes to AR. It is difficult to use the genetic association studies to predict susceptibility for AR because of small effect size of individual SNPs and the heterogeneous nature of AR. These genes would include those in immune pathways, both innate and adaptive immune systems, stimulatory molecules, tissue repair pathways, and genes involved in systemic hypertension among others.

For many variants, the initial report for a given SNP presents with a statistically significant association with AR; however, validation for most of these SNPs has remained elusive [69]. We identified 76 variants in the literature which were associated with AR (Table 1). This list was compiled from an extensive literature search of numerous genetic variants that have been associated with AR in kidney and some liver transplant studies. Nearly all the reports in Table 1 are from candidate gene analysis of recipient genomic DNA. Few studies have been conducted that investigate the genetic interactions between donor and recipient beyond HLA matching. Given the small cohort size of most transplant studies, donor and recipient genetics have not been thoroughly studied in combination. Note that there are several limitations of the reports in the literature. As stated in the Introduction, there are variations in the definition of AR between studies and that definition has changed over time. Additionally, population stratification was not done in the analysis for many of the reviewed studies. In most of these reports, the statistical power to detect a true association was low. It is common for small cohorts to be used in the analysis (N < 300) or to not take into account the testing of multiple SNPs. Additionally, validation cohorts are typically absent in the studies, or subsequent studies failed to validate the initial positive reports. Validation could fail because of the heterogeneity of the AR phenotype. Each type of AR has different mechanisms, so each type would likely have different genetics and thus be difficult to validate unless large sample sizes of each type of AR are obtained. However, some of these SNPs in Table 1 have been associated with AR by multiple studies, such as ACE (rs4340), CCR2 (rs1799864), CCR5 (rs1799987), CD28C (rs3116496), CTLA4 (rs5742909, rs231775), F5 (rs6025) and IL10 (rs1800896, rs1800872). Most of these studies have focused on recipient SNPs, but there have been a few studies analyzing donor-related SNPs. At present, there are no candidate SNPs that have been unambiguously shown to be associated with AR, through the use of large (1000+) discovery and validation cohorts.

Table 1.

Genetic variants associated with acute rejection.

Gene Name SNP rs# Protein change Nucleotide change Organ Study n, #AR, # test P-value, OR (95% Cl) [ref]
ABCB1 ATP-binding cassette, subfamily B, member 1 rs2032582 p.Ser893Alap.Ser893Thr c.2677T>Ac.2677T>G K N = 232, 64, 21 P = 0.003, 3.16 (1.50–6.67) [92]
ACE Angiotensin l-converting enzyme rs4340 Intronic 288 bp in/del K N = 206, 19, 4 P < 0.05, 5.34 (1.27–22.42) [93] 1.
K N = Meta-analysis   62 (1.14–2.29) [94]
AIF Allograft inflammatory factor-1 rs2269475 p.Arg15Trp c.43C>T K N = 458, 118, 3 P = 0.05, 0.61 (0.39–0.97) [95]
AT1R Angiotensin type 1 receptor rs5186 3′ UTR c.*86A>C K N = 206, 19, 4 P < 0.05, 8.34 (2.43–28.69) [93]
CCDC67 Coiled-coil domain containing 67 rs10765602 5′ of gene g.93048165G>T K N = 778, 503, GWAS P = 0.02 1.98 (1.21–3.25) [12]
CCL2 C-C motif chemokine ligand 2 rs1024611 5′ of gene c.−2582A>G K N = 167, 27, 2 P = 0.022, 2.6 (1.12–6.01) [96]
CCL5 C-C motif chemokine ligand 5 rs2107538 5′ of gene C.−471 G>A K N = 261, 92, 3 P = 0.035, None, [97]
CCR2 C-C motif chemokine receptor 2 rs1799864 p.Val64Ile c.190G>A K N = 163, 39, 5 P = 0.014, 0.30 (0.12–0.78)
K N = 85, 33, 2   [98]P = 0.003, None [99]
CCR5 C-C motif chemokine receptor 5 rs1799987 Intronic c.−301+246A>G K N = 163, 39, 5 P = 0.016, 0.37 (0.16–0.85)
K N = 243, 46, 2   [98]P = 0.029, 2.76 (1.11–6.90)
K N = 85, 33, 2   [100]P = 0.003, None [99]
CD28 CD28 molecule rs3116496 Intronic c.243+17T>C K N = 270, 70, 1 P = 0.026, 1.93 (1.10–3.39)
K   [101]meta-analysis [102]
CD46 CD46 molecule rs2796267 5′ of gene c.−652G>A K N = 334, 68, 1 P = 0.012, 0.47 (0.26–0.84) [103]
CD86 CD86 molecule rs1 129055 p.Alal 98Thr c.592G>A K, L N = Meta-analysis Meta-analysis [102]
CTLA4 Cytotoxic T-lymphocyte-associated protein 4 rs733618 5′ of gene c.−1722T>C K N = 167, 45, 5 P = 0.002, 0.411 (0.24–0.72) [104]
CTLA4 Cytotoxic T-lymphocyte-associated protein 4 rs5742909 5′ of gene c.−319C>T K N = 131, 34, 5 P = 0.015, 3.45 (1.18–10.1)
K N = Meta-analysis   [105]P = 0.014, 0.805
  (0.68–0.96) [106]
CTLA4 Cytotoxic T-lymphocyte-associated protein 4 rs231775 p.Thr17Ala c.49A>G K N = 190, 36, 7 P = 0.0374, 2.78 (1.07–7.19)
K N = 63, 17, 12   [107]P = 0.043, 5.5 (0.962–31.44)
K N = Meta-analysis   [108]Meta-analysis [109]P = 0.028, 0.30 [104]
K N = 72, 29, 5
CTLA4 Cytotoxic T-lymphocyte-associated protein 4 rs3087243 3′ of gene c.*1421G>A K N = 72, 29, 5 P = 0.035, 4.51 [110]
CXCL8 C-X-C motif chemokine ligand 8 rs4073 5′ of gene c.−352A>T K N = 296, 61, 2 P = 0.032, 2.7 (1.09–6.69) [111]
CYP2E1 Cytochrome P450 2ΕΊ rs251 5641 p.Phe421=p. c.1263C>Gc. K N = 347, 63, 3 P = 0.003, 2.55 (1.37–4.75) [112]
  Phe421Leu 1263C>T
CYP3A5 Cytochrome P450 3A5 rs776746 Intronic c.219–237G/A K N = 136, 19, 3 P = 0.01, 10.1 (1.21–83.3) [113]
EPHX2 Epoxy hydrolase 2 rs 1042032 3′ UTR c.*35A>G K N = 259, 31, 3 P = 0.015, 6.34 (1.35–29.90) [114]
F2 Coagulation factor II, thrombin 1799963 3′ UTR c.*97G>A K N = 165, 39, 3 P = 0.03, 12.2 (1.3–118.7) [115]
F5 Coagulation factor V rs6025 p.Arg534Gln c.1601 G>A K N = 394, 192, 1 P < 0.02, 3.83 (1.38–10.59)
N = 165, 39, 3   [116]P = 0.01, 5.7 (1.6–20.2) [115]
F0XP3 Forkhead box P3 rs2232365 5′ UTR c.−22–902A>G K N = 292, 56, 6 P = 0.001, 3.10 (1.58–6.08) [117]
F0XP3 Forkhead box P3 rs3761548 5′ UTR c.−23+2882A>C K N = 292, 56, 6 P < 0.001, 3.08 (1.67–5.660 [117]
FCGR2A Fc fragment of IgG receptor IIA rs1801274 p.Hls166Arg c.500A>G K N = 99, 53, 1 P < 0.05, None [118]
HLA-DOA Major histocompatibility complex, class II DO alpha rs9296068 5′ of gene g.4470122T>G L N = 122, 60, 2 P = 0.001, 2.55 (1.44–4.61) [119]
HLA-DOA Major histocompatibility complex, class II DO alpha rs2395304 5′ of gene g.4464339C>T L N = 122, 60, 2 P = 0.008, 2.34 (1.20–4.65) [119]
HLAG Major histocompatibility complex, class I, G rs66554220 14 bp in/del L N = 146, 57, 2 P =< 0.001, 17.657 (3.73–83.67)
HLAG Major histocompatibility complex, class I, G rs1063320 3′ UTR c.*233C>G L N = 146, 57, 2 P =< 0.001, 6.80 (2.58–17.91) [120]
ICAM1 Intercellular adhesion molecule-1 rs5498 p.Lys469Glu c.1405A>G K N = 42, 11, 2 P = 0.013, 0.23 (None) [121]
IL1B Interleukin 1 beta rs1143634 p.Phe105= c.315C>T K N = 100, 18, 12 P = 0.045, 3.11 (1.02–9.44) [122]
IL2 Interleukin 2 rs2069762 5′ of gene c.−385T>G K N = 63, 20, 1 P < 0.05, None, [123]
IL2RB Interleukin 2 receptor subunit beta rs228942 p.Asp391Glu c.1173C>A K N = 337, 61, 4 P = 0.0096 2.11 (1.19–3.74) [124]
IL2RB Interleukin 2 receptor subunit beta rs228953 p.Gly250= c.750C>T K N = 337, 61, 4 P = 0.029 1.58 (1.04–2.38) [124]
IL3 Interleukin 3 rs181781 5′ of gene c.−1285G>A K N = 330, 60, 3 P = 0.041 0.55 (0.31–0.98) [125]
IL3 Interleukin 3 rs2073506 5′ of gene c.−1662C>A K N = 330, 60, 3 P = 0.003 0.32 (0.15–0.68) [125]
IL3 Interleukin 3 rs40401 p.Pro27Ser c.79C>T K N = 330, 60, 3 P = 0.014 2.18 (1.17–4.05) [125]
IL4 Interleukin 4 rs2243250 5′ of gene c.−589C>T K N = 120, 14, 4 P = 0.02, donor None, [126]
IL4R Interleukin 4 receptor rs1801275 p.Gln576Arg c.1727A>G K N = 344, 62, 5 P = 0.019, 0.44 (0.21–0.91) [127]
IL6 Interleukin 6 rs1800796 5′ of gene c.−636G>C L N = 335, 40, 1 P = 0.033, 0.45 (0.21–0.98) [128]
IL6 Interleukin 6 rs1800795 5′ of gene c.−237C>G K N = 145, 77, 20 P = 0.0002 donor [129]
IL10 Interleukin 10 rs1800896 5′ of gene c.−1117A>G K N = 291, 50, 8 P = 0.016*, 1.9 (1.1–3.1)
K N = 95, 21‒, 2   [130]P = 0.045, 2.8 (1.17–6.69)[131]
IL10 Interleukin 10 rs1800871 5′ of gene c.−854T>C K N = 291, 50, 8 P = 0.016*, 1.9 (1.1–3.1) [130]
IL10 Interleukin ΊΟ rs1800872 5′ of gene c.−627A>C K N = 232, 64, 21 P = 0.007, 4.71 (1.52–14.55)
K N = 291, 50, 8   [92]P = 0.016*, 1.9 (1.1–3.1) [130]
IL18 Interleukin 18 rs187238 5′ of gene c.−368G>C K N = 226, 37, 1 P = 0.015, 3.65 (1.24–10.79) [132]
IMPDH1 Inoslne 5-prlme-monophosphate dehydrogenase 1 rs2278293 Intronic c.579+119G>A K N = 191, 29, 11 P = 0.008, 0.34 (0.1 5–0.76) [133]
IMPDH1 Inoslne 5-prlme-monophosphate dehydrogenase 1 rs2278294 Intronic c.580–106G>A K N = 191, 29, 11 P = 0.02, 0.40 (0.18–0.89) [133]
IMPDH2 Inoslne 5-prlme-monophosphate dehydrogenase 2 rs11706052 Intronic c.819+10T>C K N = 232, 64, 21 P = 0.006, 3.39 (1.42–8.09) [92]
INFG lnterferon-γ rs2430561 Intronic c.115–483A>T K N = 118, 20, 4 P-None, 2.6 (1.6–6.0) [134]
IRF5 Interferon regulatory factor 5 rs3757385 5′ of gene C.−811T>G L N = 289, 39, 3 P = 0.042, 2.34 (1.07–5.10) [135]
ITGB3 Integrin subunit beta 3 rs5918 p.Leu59Pro c.176T>C K N = 119, 52, 1 P = 0.04, 2.75 (1.01–7.93) [136]
MBL2 Mannose-binding lectin Haplotypers 5′ of genep. c.−290C>Gc. K N = 710, 144, 11 P = 0.01 2.05 (1.16–3.64) [137]
  7096206rs Arg52Cyp. 154C>Tc.
  5030737rs Gly54Aspp. 161G>Ac.
  1800450rs Gly57Glu 170G>A
  1800451
MTHFR Methylenetetrahydrofolate rs1801133 p.Ala222Val c.788C>T K N = 165, 39, 3 P = 0.04, 4.3 (1.4–12.9) [8,115]
K N = 585, 98, 19
NFKB1 Nuclear factor kappa B subunit 1 rs28362491 5′ of gene c.−798_−795delATTG K N = 292, 56, 6 P = 0.001, 2.61 (1.50–4.53) [117]
NFKBIA NFKB Inhibitor alpha rs696 3′ UTR c.*126G>A K N = 292, 56, 6 P = 0.007, 1.85 (1.18–2.91) [117]
PDCD1 Programmed cell death 1 rs2227982 p.Ala215Val c.644C>T K N = 122, 60, 4 P = 0.04, 0.30 (0.09–1.02) [138]
PTGS2 Prostaglandln-endoperoxlde synthase 2 rs689466 5′ of gene c.−1329A>G K N = 458, 118, 3 P = 0.01 0.59 (0.38–0.91) [95]
PTPN22 Protein tyrosine phosphatase nonreceptor 22 rs2476601 p.Arg620Trp c.1858C>T L N = 345, 180, 6 P = 0.022, 2.33 (1.13–4.79) [139]
PTPRO Protein tyrosine phosphatase receptor-type O rs7976329 Intronic c.76–34269T>C K N = 778, 503, GWAS P = 0.01, 1.61 (0.96–2.70) [12]
STAT4 Signal transducer and activator of transcription 4 rs7574865 Intronic c.274–23582A>C K N = 453, 134, 1 P = 0.003, 0.542 (0.36–0.82 [140]
TGFB Transforming growth factor-β1 rs1800470 p.Pro10Leu c.29C>T K N = 291, 50, 8 P = 0.043, 1.8 (1.0–3.0)
K N = 164, 7   [130]P = 0.02, 4.0 (None)
K N = 359, 165, 6   [141]P = 0.000, 0.146 (0.076–0.281 [142]
TGFB Transforming growth factor-β1 rs1800471 p.Arg25Pro c.74G>C K N = 291, 50, 8 P = 0.043, 1.8 (1.0–3.0)
K N = 118, 20, 4   [130]P = None, 0.09 (0.02–0.49)
K N = 164, 13, 7   [134]P = 0.02, 4.0 (None) [141]
TGFBR2 Transforming growth factor-β receptor II rs2228048 p.Asn414= c.1242C>T K N = 347, 63, 3 P = 0.020 [143]
TLR3 Toll-like receptor 3 rs3775291 p.Leu412Phe c.1234C>G L N = 100, 37, 10 P = 0.017, 0.1 (0.01–0.86) [144]
TLR4 Toll-like receptor 4 rs4986790 p.Asp299Gly c.896A>G N = 238, 57, 2 P = 0.01, 0.41 (0.30–0.83) [145]
TLR4 Toll-like receptor 4 rs10759932 5′ of gene c.−1847T>C N = 216, 42, 6 P = 0.001, 0.25 (0.11–0.57) [146]
TNF Tumor necrosis factor rs1800629 5′ of gene c.−488G>A K N = 232, 64, 21 P = 0.029, 2.18 (1.08–4.41)
K N = 100, 18, 12   [92]P = 0.03, 6.277 (1.18–33.2)
K N = 291, 50, 8   [122]P = 0.003, 5.0 (3.0–8.3)
K N = 95, 21, 2   [130]P = 0.005, 3.92 (1.61–9.57)
K N = 164, 13, 7   [131]P = 0.009, 6.1 (None)
N = 129, 57, 2   [141]P < 0.05, 2.53 (1.19–5.37)
N = 623, 1   [147]P = 0.001, 4.05 (1.76–9.28) [148]
TP53 Tumor protein gene rs1625895 Intronic c.672+62A>G K N = 100, 31, 1 P = 0.009, 0.36 (0.16–0.78) [149]
UGT1A9 Uridine diphosphate glucuronosyltransferase 1A9 rs17868320 5′ of gene c.−2152C>T K N = 100, 18, 3 P = 0.07, 3.62 (0.90–14.5) [150]
UGT1A9 Uridine diphosphate glucuronosyltransferase 1A9 rs6714486 5′ of gene c.−275T>A K N = 100, 18, 3 P = 0.05, 4.40 (1.05–18.4) [150]
UGT2B7 Uridine diphosphate glucuronosyltransferase 2B7 rs7439366 p.Tyr268His c.802T>C K N = 235, 38, 1 P < 0.046, 2.5 (1.00–6.41) [151]
VEGFA Vascular endothelial growth factor A rs699947 5′ of gene c.−2055A>C K N = 173, 65, 4 P = 0.005, 4.1 (1.5–11.3) [152]
VEGFA Vascular endothelial growth factor A rs1570360 5′ of gene c.−614A>G K N = 173, 65, 4 P = 0.001, 6.8 (1.8–25.0) [152]
MIR146A MicroRNA 146a rs2910164 n.60C>G K N = 350 P = 0.04, 2.63 (1.04–6.62) [153]
MIR196A2 MicroRNA 196a-2 rs11614913 n.78C>T K N = 350 P = 0.027, 2.86 (1.12–7.27) [153]
MIR499A MicroRNA 499a rs3746444 n.73A>G K N = 350 P = 0.027, 3.31 (1.14–9.58) [153]

L, liver transplant; K, kidney transplant.

All SNPs In the table are In recipients, unless otherwise noted.

“Study n” is the number of individuals used in that candidate gene study to determine the P-value.

“# test” is the number of polymorphisms tested in the reported analysis.

#

AR is the number of acute rejection events in the study population.

*

Slgnlflcant P-value based on an IL-10 haplotype containing three promoter variants.

Significant P-value based on haplotype containing the codon 10 and codon 25 TGFB variants.

0 and 1 acute rejection events were combined In the no rejection category for analysis.

The choice of candidate SNPs for investigation is typically limited to those that have come to the attention of the investigator, either through previous reports, or were hypothesized to be important variants which functionally impact a protein in a biological pathway thought to impact the occurrence of AR. A GWAS allows for multiple variants spanning the entire genome to be investigated. Therefore, the GWAS provides a more robust platform for discovery than single SNP analysis. A limitation to GWAS analysis is the high expectation of statistical significance (typically a P-value threshold of 108) due to multiple testing of up to 1 000 000 individual SNPs or more. Statistical power can also be limited by the typically small influence of an individual SNP on the outcome. Large cohorts are required to overcome these obstacles to identifying a statistically significant variant. At this time, very few GWASs for AR have been attempted.

In a candidate SNP analysis of 2724 SNPs and 990 kidney allograft recipients, no statistically significant SNPs were found to be associated with AR [10]. Multivariate models also failed to identify with any statistically significant SNPs, after accounting for a false discovery rate (FDR) of 10%, although many of the SNPs with the lowest P-values play a role in signaling pathways involved in T-cell activation. Additionally, some SNPs were associated with the severity of AR based on the i (inflammation) and t (tubulitis) scores on kidney allograft biopsy.

A larger-scale GWAS of 1528 European American renal allograft recipients and 450 130 SNPs again did not identify any statistically significant SNPs associated with AR [11]. SNPs with smallest P-values were rs146480420, within the epithelial cell adhesion molecule (EPCAM) gene, and rs59677415, near LOC102467213, both with a P value of 9e–7. The top 83 SNPs with smallest P-values were in the canonical pathways thought to impact AR such as NFkB activation, IL-8 signaling, and leukocyte extravasation signaling.

In a GWAS of European renal allograft recipients, the investigators used a pooled DNA approach consisting of 275 cases, 503 controls, and 1109 SNPs that were analyzed using a 50-kb sliding window to identify at least five contiguous SNPs for association with AR [12]. The pooled DNA approach reduces the number of tests needed which reduces cost. This method involves mixing equimolar amounts of DNA from each individual in the case or control groups and hybridizing the pools on a DNA-Bead Chip array [12], assaying, and then using statistical association tests for the genotypes between the case and control pools. DNA pooling approaches for association studies have been described elsewhere [1315]. For validation, 313 cases and 531 controls were used to validate 14 SNPs. Two SNPs were identified as significantly associated with AR, one in PTPRO, coding for a receptor-type tyrosine kinase essential for B-cell receptor signaling and the second in the ciliary gene CCDC67 [12]. Validation studies in other cohorts will be needed to confirm these two SNPs association with AR of kidney allografts.

For the aforementioned studies, only two SNPs were identified that were significantly associated with AR although there were also several SNPs just below the threshold of significance and were within pathways thought to impact AR. Reasons for these limited results are most likely due to the heterogeneity of AR where multiple pathways (and variants) may be associated with inducing or maintaining AR. This results in decreased statistical power and the ability to detect statistical significance. Additionally, it was shown that transplant center effects can greatly impact results and this may impact statistical associations when recipients from different transplant centers are combined into a single cohort [10]. This transplant center effect may also impact validation studies of candidate SNPs where independent cohorts are used to validate an initially reported variant. Furthermore, other clinical factors and genetic differences between populations can effect associations of genetics and AR, but are not always controlled for in studies. Thus, larger-scale, multicenter studies accounting for population stratification and clinical factors may lead to better identification of SNPs with significant association with AR.

At present, efforts are under way to expand the size of cohorts to be studied. The International Genetics & Translational Research in Transplantation Network (iGeneTRAiN) consortium [16,17] includes investigators from multiple studies to create a large cohort of different types of allograft recipients to overcome both the problem of multiple testing and the small impact of these SNPs. This group is currently conducting meta-analyses of AR from multiple cohorts where a GWAS has been performed. The primary advantage of meta-analysis is that studies can be combined to increase sample sizes and increase statistical power [17]. A disadvantage of combining GWAS across many clinical sites for such meta-analysis requires the centers to agree on specific endpoints and a common definition of AR. Additionally, the genetic data should be collected by similar methods and use of similar gene chips. If different GWAS chips are used by participating centers, genotypes can be imputed to a common variant set, although this requires additional bioinformatics steps prior to analysis. The iGeneTRAiN consortium has used imputed genotypes and common definitions of AR to conduct a robust meta-analysis of specific types of transplant and AR such as kidney.

More recent investigations are using DNA sequencing to identify rare SNPs which will be necessary to identify the genetic variances associated with AR which has not being identified through the analysis of common high allele frequency (AF) SNPs. These methods may lead to better understanding of the genetics of AR and possibly better therapy for the patients with AR.

Pharmacogenomics and acute rejection

To reduce the risk of AR, research has focused on optimizing immune suppression drugs following transplantation. Tacrolimus (TAC) is the primary immune suppressant used in >90% of kidney transplants to prevent AR and has differing efficacy in different populations, in large part due to the diverse rates of TAC metabolism, and has been the predominate focus of pharmacogenomics research in transplantation [18]. SNPs in the CYP3A5 and CYP3A4 drug metabolism genes, as well as TAC transporters, can influence the metabolic rates of TAC. Some of these SNPs, which impact the pharmacokinetics of TAC, lead to higher or lower dose requirements to achieve the therapeutic target. There is also evidence that high metabolic rates of immune suppressants in the blood, such as TAC [19] or cyclosporine [20], are associated with higher risk of AR. TAC and cyclosporine are transported by ABCB1 and metabolized by the CYP3A4 and CYP3A5 enzymes [21]. SNPs in the genes that express these, and related, enzymes have been the focus of most investigations of TAC trough blood concentrations and have been extensively reviewed elsewhere [2224]. The CYP3A5*3 SNP (rs776746) is the most commonly studied SNP and significantly decreases TAC metabolism [3,25] and the dose needed to reach therapeutic TAC blood concentrations. The CYP3A5*3 SNP has inconsistently been associated with rejection [26,27]. Further studies have shown that combinations of SNPs affect TAC metabolism [28]. In a study of mainly Caucasian subjects, the effects of ABCB1 SNPs on TAC concentrations were strongly accentuated by CYP3A4 and CYP3A5 genotype [29] although this has been inconsistently shown. Some studies have shown that the *28 SNP in the cytochrome P450 oxidoreductase gene (POR) is associated with increased metabolic activity of CYP3A4 and CYP3A5 [30]. Subjects with rapid TAC-metabolizing SNPs such as CYP3A5*1 and POR*28T when compared with slow-metabolizing CYP3A5*3 and CYP3A4*22 SNPs have dose requirements two to three times greater and a significantly longer time to reach therapeutic trough blood concentrations. However, no differences were observed in AR by SNP genotype [31]. Another study suggested that SNPs in the POR, ABCB1 and CYP3A5 genes should all be considered when dosing TAC [32]. Optimal dosing of immune suppressants may lead to lower rates of AR.

The CYP3A5 SNPs important in TAC metabolism differ significantly by race. SNP frequencies in many genes have been found to vary between different world populations [33] and may cause variability in response to drugs including TAC [34], which is similar to what was discovered about cyclosporine in the 1990s [20]. The CYP3A5*3 loss-of-function (LoF) SNP causes alternative RNA splicing [35,36] resulting in mRNA decay [37] and loss of TAC metabolic activity [38]. The CYP3A5 *1 (functional) and *3 (LoF) SNPs have been validated by in vitro experiments using CRISPR/Cas9 genetic engineering of the CYP3A5 gene in cell lines and TAC metabolism assays [38]. This *3 LoF SNP has an AF of >90% in Caucasians but only around 14% in sub-Saharan Africans [39]. African Americans also carry the CYP3A5 *6 and *7 SNPs with AFs = 10–22% [40], which also result in low CYP3A5 enzyme activity but are extremely rare in Caucasians. Therefore, nearly all Caucasians will have low capacity for TAC metabolism. However, TAC metabolism in people of African descent is highly unpredictable as some will have low, intermediate, or high metabolic rates of TAC. A recent GWAS showed definitively that TAC trough concentrations were tightly associated with CYP3A5*3, *6 and *7 SNPs in African Americans [3] and a dosing model for the prediction of TAC dose using these genotypes has been developed to facilitate clinical implementation [41]. In addition, the Clinical Pharmacogenomic Implementation Consortium (CPIC) has published guidelines for genotype-guided TAC dosing [42]. Genotype-based dosing of TAC has a potential to improve management of TAC which could lead to better outcomes and reduction in AR rates.

The association between AR and intrapatient variability in TAC concentration has been the subject of many recent papers. Causes of intrapatient TAC variability in trough concentrations are multifactorial including genetics, diarrhea, drug–drug interactions, nonadherence, or generic TAC substitution [43]. High intrapatient variability in TAC blood concentrations was associated with increased risk of AR [44]. TAC intrapatient variability was significantly associated with kidney AR, but the TAC intrapatient variability was not explained by the CYP3A5*3 genotype alone [45], suggesting that other genetic variants and clinical factors may be important. Other studies have shown that high TAC intrapatient variability in blood concentrations is associated with poorer short- and long-term outcomes and biopsy histology post-transplant [4649]. Studies have shown that high TAC intrapatient trough variability led to increased rates of late rejection and graft loss in pediatric kidney allografts [50,51]. Also, the once-daily TAC formulation, despite potential of improving adherence, did not reduce the high TAC intrapatient trough variability compared with the twice-daily dosing [52]. As many of the immune suppressants used in transplantation are impacted by metabolic and transport genes, and also target similar immunological and pharmacogenetic pathways, proper dosing is important. Dosing that reduces intrapatient variability in immune suppressant blood concentrations may lead to improved outcomes. Currently, there are no trials of genotype-based dosing to reduce intrapatient trough TAC variability. Genotype-based TAC dosing trials are needed, especially for the African Americans who are prone to under-dosing, in large part due to higher frequencies of SNPs associated with higher rates of TAC metabolism.

In the future, we envision implementing genotype-based dosing models for TAC that account for genetic and clinical factors. The goal of dosing models would be to reach therapeutic TAC concentration early and to maintain the optimal troughs following transplantation. One such dosing equation determined that the CYP3A5*1 genotype and four clinical factors were important for TAC clearance [53]. Another study developed a TAC dosing model for African American kidney transplant recipients [41]. These studies were limited to common genotypes and future dosing models will need to include low-frequency variants identified from next-generation sequencing studies and possibly include epigenetics and more clinical factors. We foresee genetic-based TAC dosing as a method that can improve transplant outcomes and possibly reduce rates of AR through optimized immune suppression.

Future directions in transcriptomics

Beyond genotype-based dosing and SNPs that have been associated with AR, variation in gene expression has also been evaluated to predict the risk of AR. Gene expression can also be used as a method to better define subtypes of AR. Global gene expression, known as transcriptomics, can be used to investigate the total RNA expressed in a tissue at a given time by microarray or RNA sequencing (RNAseq). These studies use markers in the blood of transplant patients or in the biopsy with the objective of understanding whether gene expression profiles can be used to predict transplant outcomes or how to better treat episodes of AR. Studies conducted primarily by the laboratories of Drs. Philip Halloran [5477] and Minnie Sarwal [7883] report differences in gene expression profiles in cells taken from of an extra kidney biopsy core, or blood, can predict the risk of TCMR and ABMR. Studies show improvement in TCMR and better risk stratification for allograft loss among recipients with ABMR [77]. Unfortunately, obtaining this extra biopsy core is not always feasible in the clinical setting, limiting clinical implementation. Most transcriptomic studies in transplantation have used microarray chips that detect only known gene segments that are customized on a chip. In contrast, newer technology such as RNAseq can determine the presence of RNA expression from the entire gene, partial gene segments, and some isoforms of genes and also can quantify the level of gene expression. RNAseq is becoming the preferred method to detect global gene expression and could be useful in AR association studies to determine AR pathology, classification, risk, and possibly AR therapy.

Transcriptomic studies in the blood of renal allograft patients have been used to predict risk for AR. Our team published an RNA profile, over time, using RNAseq from a cohort of immunosuppressed kidney allograft recipients that did not undergo AR that can be used as a baseline transcriptomic RNAseq signature [84]. Similar studies have been performed with microarray to develop a signature for renal transplant tolerance [85,86]. Other studies have used gene profiling from blood [87] using microarray to study gene expression profiles of AR. Molecular markers in blood could also be used to detect AR and AR risk such as specific mRNAs, proteins, free DNA, or metabolites [2]. It has also been reported that donor-derived cell free DNA in the blood can be used as a marker for active AR of kidney allografts [88]. Furthermore, the kSORT assay has been developed to test expression of specific genes in blood for AR risk [89,90]. Although gene expression testing of renal allograft function, and risk for AR, in blood is minimally invasive, further studies have been conducted to understand AR in renal allograft biopsy tissue where rejection actively occurs.

Gene expression analysis of biopsies can be used, along with histology and Banff scores, to better define, classify, and detect the types of AR. A recent publication following the XIII Banff meeting has begun the discussion of how to use transcriptomics as a method to classify AR [2]. To this point, a number of studies aimed to better understand AR using transcriptomics in biopsy tissue. One study showed that expression analysis of dysregulated gene pathways in renal allograft biopsies with interstitial fibrosis and tubular atrophy, but no histological evidence of inflammation, has been shown to be a sensitive method to detect future graft loss [91]. When histology of biopsies is ambiguous, pathologists do not always agree on the diagnosis; thus, molecular analysis can be useful. Using microarray results from 403 kidney transplant biopsies, a microarray probe set was used to determine TCMR scores [74]. The TCMR scores, in the study, had an accuracy rate of 89% in concordance with histology which includes TCMR and mixed; the primary disagreements were when histology was ambiguous (“borderline”) [74]. In a study of 403 biopsies, of the 45 biopsies that were diagnosed as ABMR by gene expression, 39 were diagnosed as ABMR by histology and donor-specific antibodies; the ABMR gene expression score strongly predicted graft loss [75]. These gene expression diagnostic studies were further validated by an international prospective study (INTERCOM) investigating 300 new biopsies from six centers [62,63]. The INTERCOM study determined that the ABMR scores agreed with conventional assessment at an accuracy rate of 85% [62]. The INTERCOM study also determined that gene expression TCMR diagnosis accuracy rate was 87% compared with histological diagnosis that included both TCMR and mixed rejection [63]. However, 77 of the 300 biopsies were further reclassified with 16 histologically TCMR and molecularly non-TCMR, 15 histologic non-TCMR were molecularly TCMR, while all 46 “borderline” biopsies by histology were reclassified with 8 TCMR or 38 non-TCMR. Thus, gene expression diagnosis of biopsies can be used to further classify ambiguous, borderline, and mixed rejection. Taken together, transcriptomics has potential to aid in the molecular classification and definition of specific types of AR that could be used in concordance with standard histological and clinical AR diagnostics.

In summary, this literature review shows numerous genetic variants that have been associated with AR in kidney transplantation. Many of these genetic variants’ associations with AR require larger studies, meta-analysis, and subsequent validation. Furthermore, there are large amounts of intrapatient variability in TAC concentrations in the blood of the kidney recipients that have been linked to AR. Dosing equations that incorporate genetic variants and clinical factors to personalize the TAC dose may be able to reduce the intrapatient variability in blood concentrations and achieve stable and therapeutic concentrations effectively. Additionally, understanding the RNA expression signatures in kidney transplant recipients may lead to better diagnosis, classification, and treatment of AR, based on gene expression. Thus, we think genetics will continue to play an important role in the future of diagnosing, defining, and treating AR in kidney transplant recipients.

Acknowledgments

Funding

This was funded with grant support from United States National Institute for Allergy and Infectious Diseases Grant AI U19 AI070119.

Footnotes

Conflict of Interest

The authors declare no conflicts of interest.

REFERENCES

  • 1.Solez K, Colvin RB, Racusen LC, et al. Banff 07 classification of renal allograft pathology: updates and future directions. Am J Transplant 2008; 8: 753. [DOI] [PubMed] [Google Scholar]
  • 2.Loupy A, Haas M, Solez K, et al. The Banff 2015 kidney meeting report: current challenges in rejection classification and prospects for adopting molecular pathology. Am J Transplant 2017; 17: 28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Oetting WS, Schladt DP, Guan W, et al. Genomewide association study of tacrolimus concentrations in African American kidney transplant recipients identifies multiple CYP3A5 alleles. Am J Transplant 2016; 16: 574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Matas AJ, Gillingham KJ, Payne WD, Najarian JS. The impact of an acute rejection episode on long-term renal allograft survival (t1/2). Transplantation 1994; 57: 857. [DOI] [PubMed] [Google Scholar]
  • 5.Sheldon S, Hasleton PS, Yonan NA, et al. Rejection in heart transplantation strongly correlates with HLA-DR antigen mismatch. Transplantation 1994; 58: 719. [PubMed] [Google Scholar]
  • 6.Goldfarb-Rumyantzev AS, Naiman N. Genetic prediction of renal transplant outcome. Curr Opin Nephrol Hypertens 2008; 17: 573. [DOI] [PubMed] [Google Scholar]
  • 7.Oetting WS, Schladt DP, Leduc RE, et al. Validation of single nucleotide polymorphisms associated with acute rejection in kidney transplant recipients using a large multi-center cohort. Transpl Int 2011; 24: 1231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Oetting WS, Zhu Y, Brott MJ, Matas AJ, Cordner GK, Pan W. Validation of genetic variants associated with early acute rejection in kidney allograft transplantation. Clin Transplant 2012; 26: 418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Chen Z, Bouamar R, Van Schaik RH, et al. Genetic polymorphisms in IL-2, IL-10, TGF-beta1, and IL-2RB and acute rejection in renal transplant patients. Clin Transplant 2014; 28: 649. [DOI] [PubMed] [Google Scholar]
  • 10.Israni A, Leduc R, Holmes J, et al. Single-nucleotide polymorphisms, acute rejection, and severity of tubulitis in kidney transplantation, accounting for center-to-center variation. Transplantation 2010; 90: 1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Israni A, Dorr C, Miller M, et al. Single nucleotide polymorphisms (SNPs) Associated with Acute Rejection in European American Kidney Transplant Recipients: a Genome Wide Association Study (GWAS). Am J Transplant 2015; 15 (suppl 3). http://atcmeetingabstracts.com/abstract/single-nucleotide-polymor phisms-snpsassociated-with-acute-rejection-in-european-american-kidney-transplant-recipients-agenome-wide-association-study-gwas/. Accessed October 23, 2017. [Google Scholar]
  • 12.Ghisdal L, Baron C, Lebranchu Y, et al. Genome-wide association study of acute renal graft rejection. Am J Transplant 2017; 17: 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sham P, Bader JS, Craig I, O’Donovan M, Owen M. DNA pooling: a tool for large-scale association studies. Nat Rev Genet 2002; 3: 862. [DOI] [PubMed] [Google Scholar]
  • 14.Norton N, Williams NM, O’Donovan MC, Owen MJ. DNA pooling as a tool for large-scale association studies in complex traits. Ann Med 2004; 36: 146. [DOI] [PubMed] [Google Scholar]
  • 15.Davis OS, Plomin R, Schalkwyk LC. The SNPMaP package for R: a framework for genome-wide association using DNA pooling on microarrays. Bioinformatics 2009; 25: 281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Li YR, van Setten J, Verma SS, et al. Concept and design of a genome-wide association genotyping array tailored for transplantation-specific studies. Genome Med 2015; 7: 90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.International Genetics & Translational Research in Transplantation Network (iGeneTRAiN). Design and implementation of the international genetics and translational research in transplantation network. Transplantation 2015; 99: 2401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Matas AJ, Smith JM, Skeans MA, et al. OPTN/SRTR 2013 annual data report: kidney. Am J Transplant 2015; 15 (Suppl. 2): 1. [DOI] [PubMed] [Google Scholar]
  • 19.Egeland EJ, Robertsen I, Hermann M, et al. High tacrolimus clearance is a risk factor for acute rejection in the early phase after renal transplantation. Transplantation 2017; 101: e273. [DOI] [PubMed] [Google Scholar]
  • 20.Lindholm A, Welsh M, Rutzky L, Kahan BD. The adverse impact of high cyclosporine. Clearance rates on the incidences of acute rejection and graft loss. Transplantation 1993; 55: 985. [DOI] [PubMed] [Google Scholar]
  • 21.Barbarino JM, Staatz CE, Venkataramanan R, Klein TE, Altman RB. PharmGKB summary: cyclosporine and tacrolimus pathways. Pharmacogenet Genomics 2013; 23: 563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Tang JT, Andrews LM, van Gelder T, et al. Pharmacogenetic aspects of the use of tacrolimus in renal transplantation: recent developments and ethnic considerations. Expert Opin Drug Metab Toxicol 2016; 12: 555. [DOI] [PubMed] [Google Scholar]
  • 23.Hesselink DA, Bouamar R, Elens L, van Schaik RH, van Gelder T. The role of pharmacogenetics in the disposition of and response to tacrolimus in solid organ transplantation. Clin Pharmacokinet 2014; 53: 123. [DOI] [PubMed] [Google Scholar]
  • 24.van Gelder T, van Schaik RH Hesselink DA. Pharmacogenetics and immunosuppressive drugs in solid organ transplantation. Nat Rev Nephrol 2014; 10: 725. [DOI] [PubMed] [Google Scholar]
  • 25.Rojas L, Neumann I, Herrero MJ, et al. Effect of CYP3A5*3 on kidney transplant recipients treated with tacrolimus: a systematic review and meta-analysis of observational studies. Pharmacogenomics J 2015; 15: 38. [DOI] [PubMed] [Google Scholar]
  • 26.Yaowakulpatana K, Vadcharavivad S, Ingsathit A, et al. Impact of CYP3A5 polymorphism on trough concentrations and outcomes of tacrolimus minimization during the early period after kidney transplantation. Eur J Clin Pharmacol 2016; 72: 277. [DOI] [PubMed] [Google Scholar]
  • 27.Flahault A, Anglicheau D, Loriot MA, Thervet E, Pallet N. Clinical impact of the CYP3A5 6986A>G allelic variant on kidney transplantation outcomes. Pharmacogenomics 2017; 18: 165. [DOI] [PubMed] [Google Scholar]
  • 28.Pulk RA, Schladt DS, Oetting WS, et al. Multigene predictors of tacrolimus exposure in kidney transplant recipients. Pharmacogenomics 2015; 16: 841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Vanhove T, Annaert P, Lambrechts D, Kuypers DR. Effect of ABCB1 diplotype on tacrolimus disposition in renal recipients depends on CYP3A5 and CYP3A4 genotype. Pharmacogenomics J 2016; doi: 10.1038/tpj.2016.49. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • 30.Lunde I, Bremer S, Midtvedt K, et al. The influence of CYP3A, PPARA, and POR genetic variants on the pharmacokinetics of tacrolimus and cyclosporine in renal transplant recipients. Eur J Clin Pharmacol 2014; 70: 685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kuypers DR, de Loor H, Naesens M, Coopmans T, de Jonge H. Combined effects of CYP3A5*1, POR*28, and CYP3A4*22 single nucleotide polymorphisms on early concentration-controlled tacrolimus exposure in de-novo renal recipients. Pharmacogenet Genomics 2014; 24: 597. [DOI] [PubMed] [Google Scholar]
  • 32.Almeida-Paulo GN, Dapia GI, Lubomirov R, et al. Weight of ABCB1 and POR genes on oral tacrolimus exposure in CYP3A5 nonexpressor pediatric patients with stable kidney transplant. Pharmacogenomics J 2017; doi: 10.1038/tpj.2016.93. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 33.Zhou Y, Ingelman-Sundberg M, Lauschke VM. Worldwide distribution of cytochrome P450 alleles: a meta-analysis of population-scale sequencing projects. Clin Pharmacol Ther 2017; 102: 688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Yasuda SU, Zhang L, Huang SM. The role of ethnicity in variability in response to drugs: focus on clinical pharmacology studies. Clin Pharmacol Ther 2008; 84: 417. [DOI] [PubMed] [Google Scholar]
  • 35.Busi F, Cresteil T. Phenotyping-genotyping of alternatively spliced genes in one step: study of CYP3A5*3 polymorphism. Pharmacogenet Genomics 2005; 15: 433. [DOI] [PubMed] [Google Scholar]
  • 36.Kuehl P, Zhang J, Lin Y, et al. Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 2001; 27: 383. [DOI] [PubMed] [Google Scholar]
  • 37.Busi F, Cresteil T. CYP3A5 mRNA degradation by nonsense-mediated mRNA decay. Mol Pharmacol 2005; 68: 808. [DOI] [PubMed] [Google Scholar]
  • 38.Dorr CR, Remmel RP, Muthusamy A, et al. CRISPR/Cas9 genetic modification of CYP3A5 *3 in HuH-7 human hepatocyte cell line leads to cell lines with increased midazolam and tacrolimus metabolism. Drug Metab Dispos 2017; 45: 957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Suarez-Kurtz G, Vargens DD, Santoro AB, et al. Global pharmacogenomics: distribution of CYP3A5 polymorphisms and phenotypes in the Brazilian population. PLoS One 2014; 9: e83472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Roy JN, Lajoie J, Zijenah LS, et al. CYP3A5 genetic polymorphisms in different ethnic populations. Drug Metab Dispos 2005; 33: 884. [DOI] [PubMed] [Google Scholar]
  • 41.Sanghavi K, Brundage RC, Miller MB, et al. Genotype-guided tacrolimus dosing in African-American kidney transplant recipients. Pharmacogenomics J 2017; 17: 61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Birdwell KA, Decker B, Barbarino JM, et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines for CYP3A5 genotype and tacrolimus dosing. Clin Pharmacol Ther 2015; 98: 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Shuker N, van Gelder T, Hesselink DA. Intra-patient variability in tacrolimus exposure: causes, consequences for clinical management. Transplant Rev (Orlando) 2015; 29: 78. [DOI] [PubMed] [Google Scholar]
  • 44.Huang CT, Shu KH, Ho HC, Wu MJ. Higher variability of tacrolimus trough level increases risk of acute rejection in kidney transplant recipients. Transplant Proc 2016; 48: 1978. [DOI] [PubMed] [Google Scholar]
  • 45.Ro H, Min SI, Yang J, et al. Impact of tacrolimus intraindividual variability and CYP3A5 genetic polymorphism on acute rejection in kidney transplantation. Ther Drug Monit 2012; 34: 680. [DOI] [PubMed] [Google Scholar]
  • 46.Whalen HR, Glen JA, Harkins V, et al. High intrapatient tacrolimus variability is associated with worse outcomes in renal transplantation using a low-dose tacrolimus immunosuppressive regime. Transplantation 2017; 101: 430. [DOI] [PubMed] [Google Scholar]
  • 47.Shuker N, Shuker L, van Rosmalen J, et al. A high intrapatient variability in tacrolimus exposure is associated with poor long-term outcome of kidney transplantation. Transpl Int 2016; 29: 1158. [DOI] [PubMed] [Google Scholar]
  • 48.Borra LC, Roodnat JI, Kal JA, Mathot RA, Weimar W, van Gelder T. High within-patient variability in the clearance of tacrolimus is a risk factor for poor long-term outcome after kidney transplantation. Nephrol Dial Transplant 2010; 25: 2757. [DOI] [PubMed] [Google Scholar]
  • 49.Vanhove T, Vermeulen T, Annaert P, Lerut E, Kuypers DR. High intrapatient variability of tacrolimus concentrations predicts accelerated progression of chronic histologic lesions in renal recipients. Am J Transplant 2016; 16: 2954. [DOI] [PubMed] [Google Scholar]
  • 50.Prytula AA, Bouts AH, Mathot RA, et al. Intra-patient variability in tacrolimus trough concentrations and renal function decline in pediatric renal transplant recipients. Pediatr Transplant 2012; 16: 613. [DOI] [PubMed] [Google Scholar]
  • 51.Pollock-Barziv SM, Finkelstein Y, Manlhiot C, et al. Variability in tacrolimus blood levels increases the risk of late rejection and graft loss after solid organ transplantation in older children. Pediatr Transplant 2010; 14: 968. [DOI] [PubMed] [Google Scholar]
  • 52.Shuker N, Cadogan M, van Gelder T, et al. Conversion from twice-daily to once-daily tacrolimus does not reduce intrapatient variability in tacrolimus exposure. Ther Drug Monit 2015; 37: 262. [DOI] [PubMed] [Google Scholar]
  • 53.Passey C, Birnbaum AK, Brundage RC, Oetting WS, Israni AK, Jacobson PA. Dosing equation for tacrolimus using genetic variants and clinical factors. Br J Clin Pharmacol 2011; 72: 948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bunnag S, Einecke G, Reeve J, et al. Molecular correlates of renal function in kidney transplant biopsies. J Am Soc Nephrol 2009; 20: 1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.de Freitas DG, Sellares J, Mengel M, et al. The nature of biopsies with “borderline rejection” and prospects for eliminating this category. Am J Transplant 2012; 12: 191. [DOI] [PubMed] [Google Scholar]
  • 56.Einecke G, Reeve J, Sis B, et al. A molecular classifier for predicting future graft loss in late kidney transplant biopsies. J Clin Invest 2010; 120: 1862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Famulski KS, de Freitas DG, Kreepala C, et al. Molecular phenotypes of acute kidney injury in kidney transplants. J Am Soc Nephrol 2012; 23: 948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Famulski KS, Einecke G, Sis B, et al. Defining the canonical form of T-cell-mediated rejection in human kidney transplants. Am J Transplant 2010; 10: 810. [DOI] [PubMed] [Google Scholar]
  • 59.Famulski KS, Reeve J, de Freitas DG, Kreepala C, Chang J, Halloran PF. Kidney transplants with progressing chronic diseases express high levels of acute kidney injury transcripts. Am J Transplant 2013; 13: 634. [DOI] [PubMed] [Google Scholar]
  • 60.Halloran PF, de Freitas DG, Einecke G, et al. An integrated view of molecular changes, histopathology and outcomes in kidney transplants. Am J Transplant 2010; 10: 2223. [DOI] [PubMed] [Google Scholar]
  • 61.Halloran PF, de Freitas DG, Einecke G, et al. The molecular phenotype of kidney transplants. Am J Transplant 2010; 10: 2215. [DOI] [PubMed] [Google Scholar]
  • 62.Halloran PF, Pereira AB, Chang J, et al. Microarray diagnosis of antibody-mediated rejection in kidney transplant biopsies: an international prospective study (INTERCOM). Am J Transplant 2013; 13: 2865. [DOI] [PubMed] [Google Scholar]
  • 63.Halloran PF, Pereira AB, Chang J, et al. Potential impact of microarray diagnosis of T cell-mediated rejection in kidney transplants: the INTERCOM study. Am J Transplant 2013; 13: 2352. [DOI] [PubMed] [Google Scholar]
  • 64.Hidalgo LG, Sis B, Sellares J, et al. NK cell transcripts and NK cells in kidney biopsies from patients with donor specific antibodies: evidence for NK cell involvement in antibody-mediated rejection. Am J Transplant 2010; 10: 1812. [DOI] [PubMed] [Google Scholar]
  • 65.Kreepala C, Famulski KS, Chang J, Halloran PF. Comparing molecular assessment of implantation biopsies with histologic and demographic risk assessment. Am J Transplant 2013; 13: 415. [DOI] [PubMed] [Google Scholar]
  • 66.Melk A, Mansfield ES, Hsieh S- C, et al. Transcriptional analysis of the molecular basis of human kidney aging using cDNA microarray profiling. Kidney Int 2005; 68: 2667. [DOI] [PubMed] [Google Scholar]
  • 67.Mengel M, Campbell P, Gebel H, et al. Precision diagnostics in transplantation: from bench to bedside. Am J Transplant 2013; 13: 562. [DOI] [PubMed] [Google Scholar]
  • 68.Mengel M, Chang J, Kayser D, et al. The molecular phenotype of 6-week protocol biopsies from human renal allografts: reflections of prior injury but not future course. Am J Transplant 2011; 11: 708. [DOI] [PubMed] [Google Scholar]
  • 69.Mengel M, Reeve J, Bunnag S, et al. Scoring total inflammation is superior to the current Banff inflammation score in predicting outcome and the degree of molecular disturbance in renal allografts. Am J Transplant 2009; 9: 1859. [DOI] [PubMed] [Google Scholar]
  • 70.Mengel M, Reeve J, Bunnag S, et al. Molecular correlates of scarring in kidney transplants: the emergence of mast cell transcripts. Am J Transplant 2009; 9: 169. [DOI] [PubMed] [Google Scholar]
  • 71.Mengel M, Sis B, Kim D, et al. The molecular phenotype of heart transplant biopsies: relationship to histopathological and clinical variables. Am J Transplant 2010; 10: 2105. [DOI] [PubMed] [Google Scholar]
  • 72.Merani S, Famulski KS, Ramassar V, Shapiro AJ, Halloran PF. Characterization of the transcriptome in isolated and transplanted mouse pancreatic islets: associations with engraftment and dysfunction. Islets 2012; 4: 158. [DOI] [PubMed] [Google Scholar]
  • 73.Reeve J, Einecke G, Mengel M, et al. Diagnosing rejection in renal transplants: a comparison of molecular- and histopathology-based approaches. Am J Transplant 2009; 9: 1802. [DOI] [PubMed] [Google Scholar]
  • 74.Reeve J, Sellares J, Mengel M, et al. Molecular diagnosis of T cell-mediated rejection in human kidney transplant biopsies. Am J Transplant 2013; 13: 645. [DOI] [PubMed] [Google Scholar]
  • 75.Sellares J, Reeve J, Loupy A, et al. Molecular diagnosis of antibody-mediated rejection in human kidney transplants. Am J Transplant 2013; 13: 971. [DOI] [PubMed] [Google Scholar]
  • 76.Sis B, Halloran PF. Endothelial transcripts uncover a previously unknown phenotype: C4d-negative antibody-mediated rejection. Curr Opin Organ Transplant 2010; 15: 42. [DOI] [PubMed] [Google Scholar]
  • 77.Loupy A, Lefaucheur C, Vernerey D, et al. Molecular microscope strategy to improve risk stratification in early antibody-mediated kidney allograft rejection. J Am Soc Nephrol 2014; 25: 2267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Weintraub LA, Sarwal MM. Microarrays: a monitoring tool for transplant patients? Transplant Int 2006; 19: 775. [DOI] [PubMed] [Google Scholar]
  • 79.Shah S, Sarwal MM. Microarrays: interrogating the transplant transcriptosome. Clin Transpl 2004: 261. [PubMed] [Google Scholar]
  • 80.Sarwal M, Chua M- S, Kambham N, et al. Molecular heterogeneity in acute renal allograft rejection identified by DNA microarray profiling. N Engl J Med 2003; 349: 125. [DOI] [PubMed] [Google Scholar]
  • 81.Mansfield ES, Sarwal MM. Arraying the orchestration of allograft pathology. Am J Transplant 2004; 4: 853. [DOI] [PubMed] [Google Scholar]
  • 82.Chua M- S, Mansfield E, Sarwal M. Applications of microarrays to renal transplantation: progress and possibilities. Front Biosci 2003; 8: s913. [DOI] [PubMed] [Google Scholar]
  • 83.Chua M- S, Barry C, Chen X, Salvatierra O, Sarwal MM. Molecular profiling of anemia in acute renal allograft rejection using DNA microarrays. Am J Transplant 2003; 3: 17. [DOI] [PubMed] [Google Scholar]
  • 84.Dorr C, Wu B, Guan W, et al. Differentially expressed gene transcripts using RNA sequencing from the blood of immunosuppressed kidney allograft recipients. PLoS One 2015; 10: e0125045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Sagoo P, Perucha E, Sawitzki B, et al. Development of a cross-platform biomarker signature to detect renal transplant tolerance in humans. J Clin Invest 2010; 120: 1848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Newell KA, Asare A, Kirk AD, et al. Identification of a B cell signature associated with renal transplant tolerance in humans. J Clin Invest 2010; 120: 1836. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Kurian SM, Williams AN, Gelbart T, et al. Molecular classifiers for acute kidney transplant rejection in peripheral blood by whole genome gene expression profiling. Am J Transplant 2014; 14: 1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Bloom RD, Bromberg JS, Poggio ED, et al. Cell-free DNA and active rejection in kidney allografts. J Am Soc Nephrol 2017; 28: 2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Roedder S, Sigdel T, Salomonis N, et al. The kSORT assay to detect renal transplant patients at high risk for acute rejection: results of the multicenter AART study. PLoS Med 2014; 11: e1001759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Crespo E, Roedder S, Sigdel T, et al. Molecular and functional noninvasive immune monitoring in the ESCAPE study for prediction of subclinical renal allograft rejection. Transplantation 2017; 101: 1400. [DOI] [PubMed] [Google Scholar]
  • 91.Modena BD, Kurian SM, Gaber LW, et al. Gene expression in biopsies of acute rejection and interstitial fibrosis/ tubular atrophy reveals highly shared mechanisms that correlate with worse long-term outcomes. Am J Transplant 2016; 16: 1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Grinyo J, Vanrenterghem Y, Nashan B, et al. Association of four DNA polymorphisms with acute rejection after kidney transplantation. Transpl Int 2008; 21: 879. [DOI] [PubMed] [Google Scholar]
  • 93.Zhang G, Wang H, Wang F, et al. Gene polymorphisms of the renin-angiotensin-aldosterone system and angiotensin II type 1-receptor activating antibodies in renal rejection. Tohoku J Exp Med 2007; 213: 203. [DOI] [PubMed] [Google Scholar]
  • 94.Huang Z, Wu B, Tao J, et al. Association between angiotensin I-converting enzyme insertion/deletion polymorphism and prognosis of kidney transplantation: a meta-analysis. PLoS One 2015; 10: e0127320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Vu D, Tellez-Corrales E, Shah T, Hutchinson I, Min DI. Influence of Cyclooxygenase-2 (COX-2) gene promoter-1195 and allograft inflammatory factor-1 (AIF-1) polymorphisms on allograft outcome in Hispanic kidney transplant recipients. Hum Immunol 2013; 74: 1386. [DOI] [PubMed] [Google Scholar]
  • 96.Kang SW, Park SJ, Kim YW, et al. Association of MCP-1 and CCR2 polymorphisms with the risk of late acute rejection after renal transplantation in Korean patients. Int J Immunogenet 2008; 35: 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kruger B, Boger CA, Obed A, et al. RANTES/CCL5 polymorphisms as a risk factor for recurrent acute rejection. Clin Transplant 2007; 21: 385. [DOI] [PubMed] [Google Scholar]
  • 98.Abdi R, Tran TB, Sahagun-Ruiz A, et al. Chemokine receptor polymorphism and risk of acute rejection in human renal transplantation. J Am Soc Nephrol 2002; 13: 754. [DOI] [PubMed] [Google Scholar]
  • 99.Yigit B, Bozkurt N, Berber I, Titiz I, Isbir T. Analysis of CC chemokine receptor 5 and 2 polymorphisms and renal transplant survival. Cell Biochem Funct 2007; 25: 423. [DOI] [PubMed] [Google Scholar]
  • 100.Cha RH, Yang SH, Kim HS, et al. Genetic interactions between the donor and the recipient for susceptibility to acute rejection in kidney transplantation: polymorphisms of CCR5. Nephrol Dial Transplant 2009; 24: 2919. [DOI] [PubMed] [Google Scholar]
  • 101.Pawlik A, Dabrowska-Zamojcin E, Dziedziejko V, Safranow K, Domanski L. Association between IVS3+ 17T/C CD28 gene polymorphism and the acute kidney allograft rejection. Transpl Immunol 2014; 30: 84. [DOI] [PubMed] [Google Scholar]
  • 102.Han FF, Fan H, Wang ZH, et al. Association between co-stimulatory molecule gene polymorphism and acute rejection of allograft. Transpl Immunol 2014; 31: 81. [DOI] [PubMed] [Google Scholar]
  • 103.Park MS, Kim SK, Lee TW, et al. A promoter polymorphism in the CD46 complement regulatory protein gene is associated with acute renal allograft rejection. Transplant Proc 2016; 48: 809. [DOI] [PubMed] [Google Scholar]
  • 104.Gao JW, Guo YF, Fan Y, et al. Polymorphisms in cytotoxic T lymphocyte associated antigen-4 influence the rate of acute rejection after renal transplantation in 167 Chinese recipients. Transpl Immunol 2012; 26: 207. [DOI] [PubMed] [Google Scholar]
  • 105.Ruhi C, Sallakci N, Yegin O, Suleymanlar G, Ersoy FF. The influence of CTLA-4 single nucleotide polymorphisms on acute kidney allograft rejection in Turkish patients. Clin Transplant 2015; 29: 612. [DOI] [PubMed] [Google Scholar]
  • 106.Duan Z, Zhang Y, Pan F, et al. Association between CTLA4 gene polymorphisms and acute rejection of kidney transplantation: a meta-analysis. J Nephrol 2012; 25: 996. [DOI] [PubMed] [Google Scholar]
  • 107.Misra MK, Kapoor R, Pandey SK, Sharma RK, Agrawal S. Association of CTLA-4 gene polymorphism with end-stage renal disease and renal allograft outcome. J Interferon Cytokine Res 2014; 34: 148. [DOI] [PubMed] [Google Scholar]
  • 108.Gendzekhadze K, Rivas-Vetencourt P, Montano RF. Risk of adverse posttransplant events after kidney allograft transplantation as predicted by CTLA-4+ 49 and TNF-alpha -308 single nucleotide polymorphisms: a preliminary study. Transpl Immunol 2006; 16: 194. [DOI] [PubMed] [Google Scholar]
  • 109.Gao JW, Zhou ZH, Guo SC, Guo YF, Guo F. A deeper understanding of the association between CTLA4+ 49A/G and acute rejection in renal transplantation: an updated meta-analysis. Ren Fail 2015; 37: 165. [DOI] [PubMed] [Google Scholar]
  • 110.Canossi A, Aureli A, Delreno F, et al. Influence of cytotoxic T-lymphocyte antigen-4 polymorphisms on acute rejection onset of cadaveric renal transplants. Transplant Proc 2013; 45: 2645. [DOI] [PubMed] [Google Scholar]
  • 111.Singh R, Kesarwani P, Ahirwar DK, Kapoor R, Mittal RD. Interleukin 8– 251T>A and Interferon gamma +874A>T polymorphism: potential predictors of allograft outcome in renal transplant recipients from north India. Transpl Immunol 2009; 21: 13. [DOI] [PubMed] [Google Scholar]
  • 112.Kim SK, Park HJ, Seok H, et al. Association studies of cytochrome P450, family 2, subfamily E, polypeptide 1 (CYP2E1) gene polymorphisms with acute rejection in kidney transplantation recipients. Clin Transplant 2014; 28: 707. [DOI] [PubMed] [Google Scholar]
  • 113.Quteineh L, Verstuyft C, Furlan V, et al. Influence of CYP3A5 genetic polymorphism on tacrolimus daily dose requirements and acute rejection in renal graft recipients. Basic Clin Pharmacol Toxicol 2008; 103: 546. [DOI] [PubMed] [Google Scholar]
  • 114.Gervasini G, Garcia-Cerrada M, Coto E, et al. A 3’-UTR polymorphism in soluble epoxide hydrolase gene is associated with acute rejection in renal transplant recipients. PLoS One 2015; 10: e0133563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Heidenreich S, Junker R, Wolters H, et al. Outcome of kidney transplantation in patients with inherited thrombophilia: data of a prospective study. J Am Soc Nephrol 2003; 14: 234. [DOI] [PubMed] [Google Scholar]
  • 116.Hocher B, Slowinski T, Hauser I, et al. Association of factor V Leiden mutation with delayed graft function, acute rejection episodes and long-term graft dysfunction in kidney transplant recipients. Thromb Haemost 2002; 87: 194. [PubMed] [Google Scholar]
  • 117.Misra MK, Mishra A, Pandey SK, Kapoor R, Sharma RK, Agrawal S. Association of functional genetic variants of transcription factor Forkhead Box P3 and Nuclear Factor-kappaB with end-stage renal disease and renal allograft outcome. Gene 2016; 581: 57. [DOI] [PubMed] [Google Scholar]
  • 118.Yuan FF, Watson N, Sullivan JS, et al. Association of Fc gamma receptor IIA polymorphisms with acute renal-allograft rejection. Transplantation 2004; 78: 766. [DOI] [PubMed] [Google Scholar]
  • 119.Ningappa M, Ashokkumar C, Higgs BW, et al. Enhanced B cell alloantigen presentation and its epigenetic dysregulation in liver transplant rejection. Am J Transplant 2016; 16: 497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Thude H, Janssen M, Sterneck M, Nashan B, Koch M. 14-bp ins/del polymorphism and +3142C>G SNP of the HLA-G gene have a significant impact on acute rejection after liver transplantation. Hum Immunol 2016; 77: 1159. [DOI] [PubMed] [Google Scholar]
  • 121.Tajik N, Salari F, Ghods AJ, Hajilooi M, Radjabzadeh MF, Mousavi T. Association between recipient ICAM-1 K469 allele and renal allograft acute rejection. Int J Immunogenet 2008; 35: 9. [DOI] [PubMed] [Google Scholar]
  • 122.Manchanda PK, Mittal RD. Analysis of cytokine gene polymorphisms in recipient’s matched with living donors on acute rejection after renal transplantation. Mol Cell Biochem 2008; 311: 57. [DOI] [PubMed] [Google Scholar]
  • 123.Morgun A, Shulzhenko N, Rampim GF, et al. Interleukin-2 gene polymorphism is associated with renal but not cardiac transplant outcome. Transplant Proc 2003; 35: 1344. [DOI] [PubMed] [Google Scholar]
  • 124.Park SJ, Yoon YC, Kang SW, et al. Impact of IL2 and IL2RB genetic polymorphisms in kidney transplantation. Transplant Proc 2011; 43: 2383. [DOI] [PubMed] [Google Scholar]
  • 125.Lee DY, Song SB, Moon JY, et al. Association between interleukin-3 gene polymorphism and acute rejection after kidney transplantation. Transplant Proc 2010; 42: 4501. [DOI] [PubMed] [Google Scholar]
  • 126.Poole KL, Gibbs PJ, Evans PR, Sadek SA, Howell WM. Influence of patient and donor cytokine genotypes on renal allograft rejection: evidence from a single centre study. Transpl Immunol 2001; 8: 259. [DOI] [PubMed] [Google Scholar]
  • 127.Lee HJ, Kim TH, Kang SW, et al. Association interleukin-4 and interleukin4 receptor gene polymorphism and acute rejection and graft dysfunction after kidney transplantation. Transplant Proc 2016; 48: 813. [DOI] [PubMed] [Google Scholar]
  • 128.Yao J, Feng XW, Yu XB, et al. Recipient IL-6–572C/G genotype is associated with reduced incidence of acute rejection following liver transplantation. J Int Med Res 2013; 41: 356. [DOI] [PubMed] [Google Scholar]
  • 129.Marshall SE, McLaren AJ, McKinney EF, et al. Donor cytokine genotype influences the development of acute rejection after renal transplantation. Transplantation 2001; 71: 469. [DOI] [PubMed] [Google Scholar]
  • 130.Alakulppi NS, Kyllonen LE, Jantti VT, et al. Cytokine gene polymorphisms and risks of acute rejection and delayed graft function after kidney transplantation. Transplantation 2004; 78: 1422. [DOI] [PubMed] [Google Scholar]
  • 131.Sankaran D, Asderakis A, Ashraf S, et al. Cytokine gene polymorphisms predict acute graft rejection following renal transplantation. Kidney Int 1999; 56: 281. [DOI] [PubMed] [Google Scholar]
  • 132.Kim CD, Ryu HM, Choi JY, et al. Association of G-137C IL-18 promoter polymorphism with acute allograft rejection in renal transplant recipients. Transplantation 2008; 86: 1610. [DOI] [PubMed] [Google Scholar]
  • 133.Wang J, Yang JW, Zeevi A, et al. IMPDH1 gene polymorphisms and association with acute rejection in renal transplant patients. Clin Pharmacol Ther 2008; 83: 711. [DOI] [PubMed] [Google Scholar]
  • 134.Tinckam K, Rush D, Hutchinson I, et al. The relative importance of cytokine gene polymorphisms in the development of early and late acute rejection and sixmonth renal allograft pathology. Transplantation 2005; 79: 836. [DOI] [PubMed] [Google Scholar]
  • 135.Yu X, Wei B, Dai Y, et al. Genetic polymorphism of interferon regulatory factor 5 (IRF5) correlates with allograft acute rejection of liver transplantation. PLoS One 2014; 9: e94426. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 136.Salido E, Martin B, Barrios Y, et al. The PlA2 polymorphism of the platelet glycoprotein IIIA gene as a risk factor for acute renal allograft rejection. J Am Soc Nephrol 1999; 10: 2599. [DOI] [PubMed] [Google Scholar]
  • 137.Golshayan D, Wojtowicz A, Bibert S, et al. Polymorphisms in the lectin pathway of complement activation influence the incidence of acute rejection and graft outcome after kidney transplantation. Kidney Int 2016; 89: 927. [DOI] [PubMed] [Google Scholar]
  • 138.Zolfaghari L, Solgi G, Nafar M, et al. Association of programmed cell death 1 and programmed cell death 1 ligand gene polymorphisms with delayed graft function and acute rejection in kidney allograft recipients. Iran J Kidney Dis 2015; 9: 138. [PubMed] [Google Scholar]
  • 139.Dullin R, Koch M, Sterneck M, Nashan B, Thude H. Association between a gain-of-function variant of PTPN22 and rejection in liver transplantation. Transplantation 2015; 99: 431. [DOI] [PubMed] [Google Scholar]
  • 140.Yang H, Zhou Q, Chen ZM, Chen WQ, Wang MM, Chen JH. Polymorphisms in STAT4 increase the risk of acute renal allograft rejection in the Chinese population. Transpl Immunol 2011; 24: 216. [DOI] [PubMed] [Google Scholar]
  • 141.Park JY, Park MH, Park H, Ha J, Kim SJ, Ahn C. TNF-alpha and TGF-beta1 gene polymorphisms and renal allograft rejection in Koreans. Tissue Antigens 2004; 64: 660. [DOI] [PubMed] [Google Scholar]
  • 142.Zhang XX, Bian RJ, Wang J, Zhang QY. Relationship between cytokine gene polymorphisms and acute rejection following liver transplantation. Genet Mol Res 2016; 15: gmr.15027599. [DOI] [PubMed] [Google Scholar]
  • 143.Kim YH, Kim TH, Kang SW, et al. Association between a TGFBR2 gene polymorphism (rs2228048, Asn389Asn) and acute rejection in Korean kidney transplantation recipients. Immunol Invest 2013; 42: 285. [DOI] [PubMed] [Google Scholar]
  • 144.Citores MJ, Banos I, Noblejas A, Rosado S, Castejon R, Cuervas-Mons V. Toll-like receptor 3 L412F polymorphism may protect against acute graft rejection in adult patients undergoing liver transplantation for hepatitis C-related cirrhosis. Transplant Proc 2011; 43: 2224. [DOI] [PubMed] [Google Scholar]
  • 145.Ducloux D, Deschamps M, Yannaraki M, et al. Relevance of Toll-like receptor-4 polymorphisms in renal transplantation. Kidney Int 2005; 67: 2454. [DOI] [PubMed] [Google Scholar]
  • 146.Hwang YH, Ro H, Choi I, et al. Impact of polymorphisms of TLR4/ CD14 and TLR3 on acute rejection in kidney transplantation. Transplantation 2009; 88: 699. [DOI] [PubMed] [Google Scholar]
  • 147.Pawlik A, Domanski L, Rozanski J, et al. IL-2 and TNF-alpha promoter polymorphisms in patients with acute kidney graft rejection. Transplant Proc 2005; 37: 2041. [DOI] [PubMed] [Google Scholar]
  • 148.Sanchez-Fructuoso AI, Perez-Flores I, Valero R, et al. The polymorphism 308G/A of tumor necrosis factor-alpha gene modulates the effect of immunosuppressive treatment in first kidney transplant subjects who suffer an acute rejection. J Immunol Res 2016; 2016: 2197595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Azarpira N, Kazemi K, Darai M. Influence of p53 (rs1625895) polymorphism in kidney transplant recipients. Saudi J Kidney Dis Transpl 2014; 25: 1160. [DOI] [PubMed] [Google Scholar]
  • 150.Pazik J, Oldak M, Dabrowski M, et al. Association of UDP-glucuronosyltransferase 1A9 (UGT1A9) gene polymorphism with kidney allograft function. Ann Transplant 2011; 16: 69. [DOI] [PubMed] [Google Scholar]
  • 151.Pazik J, Oldak M, Lewandowski Z, et al. Uridine diphosphate glucuronosyltransferase 2B7 variant p.His268Tyr as a predictor of kidney allograft early acute rejection. Transplant Proc 2013; 45: 1516. [DOI] [PubMed] [Google Scholar]
  • 152.Shahbazi M, Fryer AA, Pravica V, et al. Vascular endothelial growth factor gene polymorphisms are associated with acute renal allograft rejection. J Am Soc Nephrol 2002; 13: 260. [DOI] [PubMed] [Google Scholar]
  • 153.Misra MK, Pandey SK, Kapoor R, Sharma RK, Agrawal S. Genetic variants of MicroRNA-related genes in susceptibility and prognosis of end-stage renal disease and renal allograft outcome among north Indians. Pharmacogenet Genomics 2014; 24: 442. [DOI] [PubMed] [Google Scholar]

RESOURCES