Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Common variants in glyoxalase I do not increase chronic pancreatitis risk

  • Tom Kaune ,

    Contributed equally to this work with: Tom Kaune, Marcus Hollenbach

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Software, Writing – original draft

    ‡ These authors are joint first authors on this work.

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Marcus Hollenbach ,

    Contributed equally to this work with: Tom Kaune, Marcus Hollenbach

    Roles Conceptualization, Funding acquisition, Project administration, Supervision, Visualization, Writing – original draft

    ‡ These authors are joint first authors on this work.

    Affiliation Medical Department II–Gastroenterology, Hepatology, Infectious Diseases, Pulmonology, University of Leipzig Medical Center, Leipzig, Germany

  • Bettina Keil,

    Roles Methodology

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Jian-Min Chen,

    Roles Resources, Writing – review & editing

    Affiliations Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Etablissement Français du Sang (EFS)–Bretagne, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France, Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France

  • Emmanuelle Masson,

    Roles Resources, Writing – review & editing

    Affiliations Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Etablissement Français du Sang (EFS)–Bretagne, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France, Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France

  • Carla Becker,

    Roles Methodology, Writing – review & editing

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Marko Damm,

    Roles Resources, Writing – review & editing

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Claudia Ruffert,

    Roles Data curation, Formal analysis, Methodology, Writing – review & editing

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Robert Grützmann,

    Roles Resources, Writing – review & editing

    Affiliation Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Chirurgische Klinik, Erlangen, Germany

  • Albrecht Hoffmeister,

    Roles Resources, Writing – review & editing

    Affiliation Medical Department II–Gastroenterology, Hepatology, Infectious Diseases, Pulmonology, University of Leipzig Medical Center, Leipzig, Germany

  • Rene H. M. te Morsche,

    Roles Resources, Writing – review & editing

    Affiliation Department of Gastroenterology and Hepatology, Radboud umc, Nijmegen, The Netherlands

  • Giulia Martina Cavestro,

    Roles Resources, Writing – review & editing

    Affiliation Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy

  • Raffaella Alessia Zuppardo,

    Roles Resources, Writing – review & editing

    Affiliation Gastroenterology and Gastrointestinal Endoscopy Unit, Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS Ospedale San Raffaele Scientific Institute, Milan, Italy

  • Adrian Saftoiu,

    Roles Resources, Writing – review & editing

    Affiliation Department of Internal Medicine and Gastroenterology, University of Medicine and Pharmacy, Craiova, Romania

  • Ewa Malecka-Panas,

    Roles Resources, Writing – review & editing

    Affiliation Department of Digestive Tract Diseases, Medical University of Łódź, Łódź, Poland

  • Stanislaw Głuszek,

    Roles Resources, Writing – review & editing

    Affiliation Faculty of Medicine and Health Sciences, Jan Kochanowski University, Kielce, Poland

  • Peter Bugert,

    Roles Resources, Writing – review & editing

    Affiliation Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service of Baden-Württemberg, Mannheim, Germany

  • Markus M. Lerch,

    Roles Resources, Writing – review & editing

    Affiliation Department of Medicine A, University Medicine Greifswald, Greifswald, Germany

  • Frank Ulrich Weiss,

    Roles Funding acquisition, Resources, Writing – original draft

    Affiliation Department of Medicine A, University Medicine Greifswald, Greifswald, Germany

  • Wen-Bin Zou,

    Roles Resources, Writing – review & editing

    Affiliation Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai Institute of Pancreatic Diseases, Shanghai, China

  • Zhuan Liao,

    Roles Resources, Writing – review & editing

    Affiliation Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai Institute of Pancreatic Diseases, Shanghai, China

  • Peter Hegyi,

    Roles Funding acquisition, Resources, Writing – review & editing

    Affiliations Institute for Translational Medicine and First Department of Internal Medicine, Medical School, University of Pécs, Pécs, Hungary, HAS-SZTE, Momentum Gastroenterology Multidisciplinary Research Group, Szeged, Hungary

  • Joost PH Drenth,

    Roles Resources, Writing – original draft

    Affiliation Department of Gastroenterology and Hepatology, Radboud umc, Nijmegen, The Netherlands

  • Jan Riedel,

    Roles Resources, Writing – review & editing

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Claude Férec,

    Roles Resources, Writing – review & editing

    Affiliations Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Etablissement Français du Sang (EFS)–Bretagne, Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale, Brest, France, Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU) Brest, Hôpital Morvan, Brest, France

  • Markus Scholz,

    Roles Methodology, Writing – original draft

    Affiliations Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany, LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany

  • Holger Kirsten,

    Roles Formal analysis, Visualization, Writing – review & editing

    Affiliations Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany, LIFE- Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany

  • Andrea Tóth,

    Roles Data curation, Formal analysis, Investigation, Methodology, Software, Writing – review & editing

    Affiliation Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany

  • Maren Ewers,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Writing – review & editing

    Affiliation Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany

  • Heiko Witt,

    Roles Funding acquisition, Methodology, Resources, Writing – original draft

    Affiliation Else Kröner-Fresenius-Zentrum für Ernährungsmedizin (EKFZ), Paediatric Nutritional Medicine, Technische Universität München (TUM), Freising, Germany

  • Heidi Griesmann,

    Roles Conceptualization, Writing – original draft

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • Patrick Michl,

    Roles Conceptualization, Supervision, Writing – original draft

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  •  [ ... ],
  • Jonas Rosendahl

    Roles Conceptualization, Funding acquisition, Methodology, Software, Supervision, Validation, Visualization, Writing – original draft

    jonas.rosendahl@uk-halle.de

    Affiliation Department of Internal Medicine I, Martin Luther University, Halle, Germany

  • [ view all ]
  • [ view less ]

Abstract

Introduction

Chronic pancreatitis (CP) may be caused by oxidative stress. An important source of reactive oxygen species (ROS) is the methylglyoxal-derived formation of advanced glycation endproducts (AGE). Methylglyoxal is detoxified by Glyoxalase I (GLO1). A reduction in GLO1 activity results in increased ROS. Single nucleotide polymorphisms (SNPs) of GLO1 have been linked to various inflammatory diseases. Here, we analyzed whether common GLO1 variants are associated with alcoholic (ACP) and non-alcoholic CP (NACP).

Methods

Using melting curve analysis, we genotyped a screening cohort of 223 ACP, 218 NACP patients, and 328 controls for 11 tagging SNPs defined by the SNPinfo LD TAG SNP Selection tool and the functionally relevant variant rs4746. For selected variants the cohorts were extended to up to 1,441 patient samples.

Results

In the ACP cohort, comparison of genotypes for rs1937780 between patients and controls displayed an ambiguous result in the screening cohort (p = 0.08). However, in the extended cohort of 1,441 patients no statistically significant association was found for the comparison of genotypes (p = 0.11), nor in logistic regression analysis (p = 0.214, OR 1.072, 95% CI 0.961–1.196). In the NACP screening cohort SNPs rs937662, rs1699012, and rs4746 displayed an ambiguous result when patients were compared to controls in the recessive or dominant model (p = 0.08, 0.08, and 0.07, respectively). Again, these associations were not confirmed in the extended cohorts (rs937662, dominant model: p = 0.07, logistic regression: p = 0.07, OR 1.207, 95% CI 0.985–1.480) or in the replication cohorts for rs4746 (Germany, p = 0.42, OR 1.080, 95% CI 0.673–1.124; France, p = 0.19, OR 0.90, 95% CI 0.76–1.06; China, p = 0.24, OR 1.18, 95% CI 0.90–1.54) and rs1699012 (Germany, Munich; p = 0.279, OR 0.903, 95% CI 0.750–1.087).

Conclusions

Common GLO1 variants do not increase chronic pancreatitis risk.

Introduction

Chronic pancreatitis (CP) is a recurring inflammation of the pancreas with progressive fibrosis by tissue destruction that in some patients results in exocrine and endocrine pancreatic insufficiency [1]. Several studies have identified that the underlying pathomechanisms can range from premature intrapancreatic activation of proteases to local and systemic inflammatory processes, which are relevant for the initiation and progression of the disease [2]. Recently, it was demonstrated that oxidative stress (ROS) is involved in these inflammatory and fibrotic processes [35]. As advanced-glycation-end products (AGE) impact on ROS, they may contribute to CP development [6] as it was shown in acute pancreatitis [7].

“Dicarbonyl stress” indicates a cellular condition where α-oxoaldehyde metabolites accumulate, leading to an increased modification of protein and DNA which contribute to cellular dysfunction in ageing and disease. “Dicarbonyl stress” is mainly caused by methylglyoxal (MGO) that is formed as a by-product in glycolysis [8], ketone body metabolism and threonine catabolism [911]. MGO is highly reactive with nucleotides, phospholipids and proteins [12,13] with the result of a rapid formation of AGE. In addition, reducing sugars like glucose react with amino groups and trigger MGO formation and AGE generation in a non-enzymatic protein glycation within the Maillard reaction [6]. AGE themselves induce several detrimental processes on a cellular level and furthermore activate different signaling pathways via the RAGE receptor. Moreover, they induce ROS and have been associated with various disease entities [14].

For protection on the cellular level MGO is detoxified by the Glyoxalase system. Glyoxalase I (GLO1) catalyzes the conversion of α-oxo-aldehydes such as MGO and L-glutathione (GSH) to form the corresponding hemithioacethal S-D-lactoylglutathione [15]. In the next step, hydroxyacyl glutathione hydrolase (GLO2) converts S-D-Lactoylglutathione to D-lactate and GSH. Herein, GLO1 is the rate-limiting enzyme in this series of reactions [16].

Thus far, GLO1 single nucleotide polymorphisms (SNPs) were associated with distinct inflammatory diseases. The rs4746 (p.Ala111Glu) variant displayed a decrease of GLO1 enzymatic activity for the A-allele in lymphoblastoid cells of the brain [17,18]. In addition, rs4746 has been linked to diabetes [19], atherosclerosis [20], chronic renal failure [21], vascular diseases [22,23], neuropsychiatric disorders [24,25], and different cancer types [2629]. Moreover, rs1130534 (c.372A>T, p.Gly124 = ) and rs1049346 were correlated with lower enzyme activity, but did not associate with vascular complications in diabetes mellitus [30].

In conclusion, GLO1 variants with a diminished GLO1 activity cause increased MGO levels and consecutive ROS generation. Therefore, we reasoned that GLO1 SNPs contribute to the development of CP and investigated whether genetic variants in GLO1 are associated with alcoholic CP (ACP) or non-alcoholic CP (NACP).

Material and methods

Patients and controls

The study was approved by the medical ethical review committee of the Martin-Luther-University of Halle-Wittenberg (Medical ethical committee, University Halle-Wittenberg, Medical Faculty, Bearbeitungsnummer 2015–106, date: 22.01.2016, title: “Erforschung molekulargenetischer Ursachen von Pankreaserkrankungen”). All patients gave written informed consent. The diagnosis of CP was based on two or more of the following findings: history of recurrent acute pancreatitis or recurrent or persisting abdominal pain typical for CP, pancreatic calcifications and/or pancreatic ductal irregularities indicated by computed tomography imaging, magnetic resonance imaging, endoscopic retrograde pancreaticography or (endo)sonography of the pancreas and/or the diagnosis of exocrine pancreatic insufficiency [31].

ACP was diagnosed in patients with a history of chronic alcohol intake (> 80 g per day for males or >60 g per day for females) for more than 2 years. NACP was diagnosed in the absence of known precipitating factors as alcohol consumption and/or smoking. Patients with a positive family history were included in the NACP group. The data on past ethanol consumption and the clinical presentation were based on research records and/or physician's history and/or completion of a detailed questionnaire by the patient.

Patients and Controls were recruited throughout Germany and in the European centres in The Netherlands, Romania, Poland, Italy, and Hungary. Controls were blood donors and healthy volunteers as described in our former publications [32].

In the screening cohort we investigated 223 ACP, 218 NACP patients, and 328 controls for 12 common GLO1 SNPs with a minor allele frequency of a least 5%. Variants with nominal significance according to uncorrected p-values (rs1699012, rs937662, rs4746, and rs1937780) were analyzed in further subjects (extended cohorts). In addition, we screened European ACP cohorts for rs1937780 and an additional German, French, and Chinese NACP cohort for rs4746 and an independent German NACP cohort for rs1699012 (see flow chart in Fig 1). For a detailed description of the screening cohort and the extended cohorts see Table 1.

thumbnail
Fig 1. Flowchart of patients analyzed in this study.

Note that the extended cohorts comprise the screening cohort and novel patients for all German patients (SNPs rs937662, rs1937780, and rs4746). For rs1699012 the numbers summarize the extended with the replication cohort from Munich. The European cohorts (rs1937780) and the German, French, and Chinese replication cohorts for rs4746 are additional patients.

https://doi.org/10.1371/journal.pone.0222927.g001

thumbnail
Table 1. Description of the cohorts included in the analysis.

https://doi.org/10.1371/journal.pone.0222927.t001

Selection of tagging SNPs in the GLO1 locus

We selected 11 tagging SNPs in the GLO1 locus using the SNPinfo LD TAG SNP Selection tool (LD map Figure A in S1 File). For this purpose we used an LD threshold of 0.8, a minimum of one SNP tagged, a minimum of 5 valid genotypes to calculate LD in populations with European ancestry (CEU) and extended the region of interest by 10.000 bp in the 5’-region and the 3’-region. In addition, we analyzed SNP rs4746 that was described to be functionally relevant [17,18]. The other functionally relevant SNPs rs1049346 and rs1130534 were tagged by rs1621788 and rs13212218, respectively. Details on the selected SNPs are summarized in Table A in S1 File.

DNA extraction and SNP genotyping

DNA was isolated from EDTA blood using a commercial system (QIAamp Blood DNA Mini Kit; Qiagen, Hilden, Germany). Polymerase chain reaction (PCR) was conducted using OneTaq® 2X Master Mix (NEB) with 200 μM dNTPs, 1.8 mM MgCl2 and 0.1 μM forward primer as well as 0.1 μM reverse primer (0.4 μM forward primer for rs3778443 and rs17544798; 0.2 μM reverse primer for rs4746) in a total volume of 25 μl. Cycle conditions were an initial denaturation at 95°C for 5 minutes followed by 45 cycles of 20 seconds denaturation at 95°C, 40 seconds annealing (Table B in S1 File), 90 seconds primer extension at 72°C followed by final extension for 5 minutes at 72°C in an automated thermal cycler. Primers and probes (Table B in S1 File) were synthesized by TIB Molbiol (Berlin, Germany). Genotyping was performed using the LightCycler480® system (Roche Diagnostics).

Probe oligomers were diluted in H2O to a concentration of 200 nM. For genotyping we used the PCR products from standard PCR (see above) with 50 nM (final) of probe oligomers followed by melting curve analysis with the following protocol: 95°C for 60 seconds, 40°C for 60 seconds, continuous increase to 70°C with various ramp rates (0.29°C/s rs12198212 and rs1621788, 0.14°C/s rs17544798, rs132212218 and rs937662, 0.19°C/s for the other seven SNPs). Call rates for all SNPs were > 95%. For quality control 2.6% of all samples were genotyped in duplicates blinded to the investigator. Resulting concordance rate was 99.7%.

Statistical analysis

Quality of SNP genotypes was assessed by study-wise call rate and exact test for Hardy-Weinberg disequilibrium (HWE) in patients and controls. We compared the results for genotype frequencies of the different polymorphisms between patient cohorts and control populations with the Chi-square test and logistic binary regression. All other models (dominant, recessive, allele frequencies) were computed by using two-tailed Fisher's Exact test. P-values were calculated using IBM SPSS Statistics 25 and GraphPad Prism 5. A p-value of less than 0.05 was considered to be significant. When a significant or nominal significant association was found, at least an extended or one replication cohort was screened.

Results

No study-wide significant association of GLO1 SNPs with ACP

We identified no significant association in logistic regression analysis and no significant difference in the genotype distribution in the ACP screening cohort compared to controls (Table 2 and Table C in S1 File). As for rs937662 the HWE was nominal significant in our controls (p = 0.047) and for both rs937662 and rs1699012 nominal significant results were observed in the NACP screening cohort in the dominant or recessive model (p = 0.08, respectively) we extended our analysis in the ACP cohort to 430 and 447 patients. Here, we found no association for both SNPs and no deviation from HWE for rs937662 in the controls (p = 0.38; controls) (Table 3). Furthermore, genotype data of rs1937780 displayed a borderline significance in the screening cohort (p = 0.08). Again, we extended our investigated groups and analysed rs1937780 in a further German ACP cohort and additional European ACP patients from The Netherlands, Hungary, Italy, Romania and Poland. In the German cohort (n = 872) compared to controls (n = 1,474) the association was statistically significant (p = 0.003) as the genotype distribution between ACP patients and controls differed (GG: 40.0% vs. 33.1%; GA 45.9% vs. 51.6%; AA 14.1% vs. 15.3%) (Table D in S1 File). Of note, there was a deviation from HWE in the controls of the extended German cohort (p = 0.01). However, we did not find a significant association in logistic regression analysis (p = 0.134, odds ratio (OR) OR 1.117, 95% confidence interval (95% CI) 0.966–1.290) for this cohort. Otherwise, in the overall cohort of European ACP patients and controls we found no significant association in logistic regression analysis either (p = 0.214, OR 1.075, 95% CI 0.961–1.196) (Table E in S1 File). This result was confirmed for the corresponding genotype data and none of the five replication cohorts demonstrated a statistically significant association in the different genetic models used for computations (Table 4 and Tables D and E in S1 File). Except for the named SNPs and SNP rs2736655 (ACP patients p = 0.026), all SNPs corresponded to the HWE.

thumbnail
Table 2. Binary logistic regression results of the GLO1 single nucleotide polymorphisms in patients with alcoholic and non-alcoholic chronic pancreatitis with covariate sex.

https://doi.org/10.1371/journal.pone.0222927.t002

thumbnail
Table 3. Data of the analysed GLO1 SNPs in patients with alcoholic chronic pancreatitis (ACP) and controls.

For the calculations different genetic models were used.

https://doi.org/10.1371/journal.pone.0222927.t003

thumbnail
Table 4. Data of the European alcoholic chronic pancreatitis (ACP) replication cohorts for rs1937780 in comparison to controls.

Calculations were performed with different genetic models.

https://doi.org/10.1371/journal.pone.0222927.t004

No study-wide significant association of GLO1 SNPs with NACP

In the screening cohort the SNPs rs937662, rs1699012, and rs4746 displayed a nominal significant difference in the recessive or dominant model (p = 0.08, 0.08, and 0.07, respectively) (Table 5). Otherwise, no differences in the genotype distributions were observed (Table F in S1 File). Furthermore, we detected a significant association for SNP rs1699012 (p = 0.009, OR 0.763, 95% CI 0.623–0.933) and rs4746 (p = 0.008, OR 1.342, 95% CI 1.080–1.669) in logistic regression analysis (Table 2). To elucidate a potential association, we extended the German NACP cohort and found no statistically significant association of the genotype distribution for rs937662 (p = 0.15), whereas rs1699012 and rs4746 still displayed a significant association compared to controls (p = 0.02 and p = 0.008, respectively) (Table G in S1 File). Therefore, we investigated rs4746 in an independent German (n = 450, patients; n = 581, controls), French (n = 767, patients; n = 1,049, controls), and Chinese (n = 928, patients; n = 1,008, controls) NACP cohort. Here, no association was found in all three replication cohorts (Table 6; for genotype distribution see Table G in S1 File). Finally, for SNP rs1699012 we observed no significant association (p = 0.279, OR 0.903, 95% CI 0.750–1.087) in an independent German NACP cohort of 427 patients in logistic regression analysis (for different genetic models see Table 6). For the rare SNP rs3778443 we observed a significant HWE (p = 0.002).

thumbnail
Table 5. Data of the analysed GLO1 SNPs in patients with non-alcoholic chronic pancreatitis (NACP) and controls.

For the calculations different genetic models were used.

https://doi.org/10.1371/journal.pone.0222927.t005

thumbnail
Table 6. Data of the replication cohorts of GLO1 SNPs rs937662, rs1699012, rs4746 in patients with non-alcoholic chronic pancreatitis (NACP) and controls.

For computations different genetic models were used.

https://doi.org/10.1371/journal.pone.0222927.t006

Discussion

There is a biological plausibility that GLO1 is relevant in inflammatory processes and as such for the development of CP, although conflicting results have been reported in other diseases thus far. In this work, we investigated a potential genetic association of GLO1 variants with ACP and NACP. We failed to identify an association between CP and one functionally relevant variant and 11 tagging SNPs covering the GLO1 locus.

In our German ACP patients (total cohort) rs1937780 genotypes differed significantly between patients and controls (p = 0.003) and significance was also observed in the dominant model. Contrary, we found neither an association nor a comparable trend in the distinct European cohorts. The same variant has recently been investigated in pancreatic cancer patients and an association was absent [33]. As functional consequences of this variant are unknown and overall genetic data are statistically not significant, it is unlikely that rs1937780 plays a prominent role in CP development.

In the NACP cohort the three variants rs937662, rs1699012, and rs4746 showed borderline significant results in distinct analysis models that, however, were not confirmed in the extended or replication cohorts. For the variants rs937662 and rs1699012 no prior clinical relevance has been reported and therefore an association with CP is again unlikely. Contrary, rs4746 was associated with a wide spectrum of disorders ranging from diabetes [19], atherosclerosis [20], chronic renal failure [21], vascular diseases [22,34], neuropsychiatric disorders [24,25], and even to cancer [2629]. In our work the borderline significance of the screening cohort was disproved in three large NACP cohorts from Germany, France, and China. As such, although, a functional relevance for this variant has been reported, our data show no association with CP.

We investigated cohorts with a reasonable number of patients and extended these cohorts whenever statistically or nominal significant results were obtained. Therefore, a prominent disease association of GLO1 variants with CP can be ruled out with high certainty. Nevertheless, our approach is not capable of identifying rare associating variants. We therefore analyzed whole exome sequencing data from an ongoing project but did not identify rare variants enriched in our NACP patients (unpublished data). Nonetheless, our study is limited in its restriction to a Caucasian (German) cohort as only one SNP was analyzed in an Asian replication cohort and therefore we may have missed specific associations in other ethnicities.

In summary, we performed a comprehensive investigation of GLO1 variants and did not demonstrate a prominent role for CP development in alcoholic and the non-alcoholic etiologies of the disease.

Supporting information

S1 File.

Figure A. Linkage disequilibrium figure of the GLO1 locus generated by the SNPinfo LD TAG SNP Selection tool.

To generate the linkage disequilibrium (LD) figure, the SNPinfo LD TAG SNP Selection tool (https://snpinfo.niehs.nih.gov/snpinfo/snptag.html) with the following parameters was applied: LD threshold of 0.8; a minimum of one SNP tagged; a minimum of 5 valid genotypes to calculate LD in populations with European ancestry (CEU); integrated region with 10.000 bp in the 5’-region and the 3’-region of GLO1. Abbreviations: SNP, single nucleotide polymorphism; LD, linkage disequilibrium; CEU, Northern Europeans from Utah. For the following SNPs we used tagging SNPs in our study: rs10484854 was tagged by rs12198212; rs1781735 by rs1621788; rs6458064 by rs937662; and rs9394523 by rs13212218. As demonstrated in the figure the tagging SNPs represented the depicted haplotypes. In Table A in S1 File the information on the SNPs selected according to the published literature and by SNPinfo is summarized.

Table A. Overview of screened GLO1 SNPs that were identified by SNPinfo or by a literature research.

We used the SNPinfo LD TAG SNP Selection tool to identify SNPs in the GLO1 locus that cover the haplotypes of the gene. As several variants have been reported in the literature, we included these using tagging SNPs, where possible. The corresponding literature for the screened SNPs is indicated in brackets. For rs4746 several studies reported associations and functional data are available in addition. * These SNPs have been tagged by the screened SNP rs1616723, rs9380765, rs13212218, and rs1621788 respectively.

Table B. Polymerase chain reaction (PCR) primers and probes for melting curve analysis of all GLO1 SNPs. Abbreviations: fw, forward; rv, reverse; XI, internal dye modified base; LC610, 5´- LightCycler Red 610; LC640 (sensor probe), LightCycler Red 640 (sensor probe); FL, 3'-Fluorescein labelling (anchor probe); PH, 3´-phosphate.

Table C. Genotype data of the analysed GLO1 SNPs in patients with alcoholic chronic pancreatitis (ACP) and controls.

Note: For rs937662 and rs1699012 the extended German cohorts are shown. Calculations were performed using the Chi-square test (two-sided). Abbreviations: Contr. = controls, Pat. = patients.

Table D. Genotype data of the GLO1 SNP rs1937780 in patients with alcoholic chronic pancreatitis in European cohorts including Germany. Note: The German cohort comprises the screening cohort and additional samples. Calculations were performed using the Chi-square test (two-sided). Abbreviations: Contr. = controls, Pat. = patients.

Table E. Results of logistic regression with covariate gender for the GLO1 SNP rs1937780 in patients with alcoholic chronic pancreatitis in European cohorts including Germany.

Note: The German cohort comprises the screening cohort and additional samples. Calculations were performed using logistic regression. Abbreviations: OR, Odds ratio; CI, confidence interval; ACP, alcoholic chronic pancreatitis.

Table F. Genotype data of the analysed GLO1 SNPs in German patients with non-alcoholic chronic pancreatitis (NACP) and controls.

Calculations were performed using the Chi-square test (two-sided). Abbreviations: Contr. = controls, Pat. = patients.

Table G. Genotype data of the analysed GLO1 SNPs rs937662, rs1699012, rs4746 in the extended NACP cohorts.

Note: For rs937662, rs1699012, and rs4746 the extended German cohorts comprise the screening cohort and additional German samples. Calculations were performed using the Chi-square test (two-sided). Abbreviations: Contr. = controls, Pat. = patients. ‖ NACP replication cohort and controls from Germany. $ Independent NACP replication cohort from Munich. § NACP cohort and controls from France. € NACP cohort and controls from China.

https://doi.org/10.1371/journal.pone.0222927.s001

(DOCX)

References

  1. 1. Majumder S, Chari ST. Chronic pancreatitis. Lancet. 2016;387: 1957–1966 pmid:26948434
  2. 2. Mayerle J, Sendler M, Hegyi E, Beyer G, Lerch MM, Sahin-Toth M. Genetics, Cell Biology, and Pathophysiology of Pancreatitis. Gastroenterology. 2019;156: 1951–1968 pmid:30660731
  3. 3. Liu F, Shi Y, Zhang XQ, Xu XF, Chen Y, Zhang H. [The role of oxidative inflammatory cascade on pancreatic fibrosis progression in mice induced by DBTC plus ethanol]. Zhongguo Ying Yong Sheng Li Xue Za Zhi. 2015;31: 477–480 pmid:26827547
  4. 4. Yu JH, Kim H. Oxidative stress and inflammatory signaling in cerulein pancreatitis. World J Gastroenterol. 2014;20: 17324–17329 pmid:25516643
  5. 5. Verlaan M, Roelofs HM, van-Schaik A, Wanten GJ, Jansen JB, Peters WH, et al. Assessment of oxidative stress in chronic pancreatitis patients. World J Gastroenterol. 2006;12: 5705–5710 pmid:17007026
  6. 6. Singh R, Barden A, Mori T, Beilin L. Advanced glycation end-products: a review. Diabetologia. 2001;44: 129–146 pmid:11270668
  7. 7. Maciejczyk M, Skutnik-Radziszewska A, Zieniewska I, Matczuk J, Domel E, Waszkiel D, et al. Antioxidant Defense, Oxidative Modification, and Salivary Gland Function in an Early Phase of Cerulein Pancreatitis. Oxid Med Cell Longev. 2019;2019: 8403578 pmid:30984340
  8. 8. Ohmori S, Mori M, Shiraha K, Kawase M. Biosynthesis and degradation of methylglyoxal in animals. Prog Clin Biol Res. 1989;290: 397–412 pmid:2498903
  9. 9. Ray S, Ray M. Formation of methylglyoxal from aminoacetone by amine oxidase from goat plasma. J Biol Chem. 1983;258: 3461–3462 pmid:6833209
  10. 10. Casazza JP, Felver ME, Veech RL. The metabolism of acetone in rat. J Biol Chem. 1984;259: 231–236 pmid:6706932
  11. 11. Phillips SA, Thornalley PJ. The formation of methylglyoxal from triose phosphates. Investigation using a specific assay for methylglyoxal. Eur J Biochem. 1993;212: 101–105 pmid:8444148
  12. 12. Vaca CE, Fang JL, Conradi M, Hou SM. Development of a 32P-postlabelling method for the analysis of 2'-deoxyguanosine-3'-monophosphate and DNA adducts of methylglyoxal. Carcinogenesis. 1994;15: 1887–1894 pmid:7923582
  13. 13. Lo TW, Westwood ME, McLellan AC, Selwood T, Thornalley PJ. Binding and modification of proteins by methylglyoxal under physiological conditions. A kinetic and mechanistic study with N alpha-acetylarginine, N alpha-acetylcysteine, and N alpha-acetyllysine, and bovine serum albumin. J Biol Chem. 1994;269: 32299–32305 pmid:7798230
  14. 14. Hollenbach M. The Role of Glyoxalase-I (Glo-I), Advanced Glycation Endproducts (AGEs), and Their Receptor (RAGE) in Chronic Liver Disease and Hepatocellular Carcinoma (HCC). Int J Mol Sci. 2017;18:
  15. 15. Thornalley PJ. The glyoxalase system in health and disease. Mol Aspects Med. 1993;14: 287–371 pmid:8277832
  16. 16. Mannervik B. Glyoxalase I. In: Enzymatic Basis of Detoxification. Academic Press, New York. 1980;2: 263–293
  17. 17. Junaid MA, Kowal D, Barua M, Pullarkat PS, Sklower BS, Pullarkat RK. Proteomic studies identified a single nucleotide polymorphism in glyoxalase I as autism susceptibility factor. Am J Med Genet A. 2004;131: 11–17 pmid:15386471
  18. 18. Barua M, Jenkins EC, Chen W, Kuizon S, Pullarkat RK, Junaid MA. Glyoxalase I polymorphism rs2736654 causing the Ala111Glu substitution modulates enzyme activity—implications for autism. Autism Res. 2011;4: 262–270 pmid:21491613
  19. 19. Groener JB, Reismann P, Fleming T, Kalscheuer H, Lehnhoff D, Hamann A, et al. C332C genotype of glyoxalase 1 and its association with late diabetic complications. Exp Clin Endocrinol Diabetes. 2013;121: 436–439 pmid:23775136
  20. 20. Wu JC, Li XH, Wang JB, Tang JF, Wang YF, Peng YD. Glyoxalase I and aldose reductase gene polymorphisms and susceptibility to carotid atherosclerosis in type 2 diabetes. Genet Test Mol Biomarkers. 2011;15: 273–279 pmid:21294693
  21. 21. Kalousova M, Germanova A, Jachymova M, Mestek O, Tesar V, Zima T. A419C (E111A) polymorphism of the glyoxalase I gene and vascular complications in chronic hemodialysis patients. Ann N Y Acad Sci. 2008;1126: 268–271 pmid:18079478
  22. 22. Gale CP, Futers TS, Summers LK. Common polymorphisms in the glyoxalase-1 gene and their association with pro-thrombotic factors. Diab Vasc Dis Res. 2004;1: 34–39 pmid:16305054
  23. 23. Rinaldi C, Bramanti P, Fama A, Scimone C, Donato L, Antognelli C, et al. GLYOXALASE I A111E, PARAOXONASE 1 Q192R AND L55M POLYMORPHISMS IN ITALIAN PATIENTS WITH SPORADIC CEREBRAL CAVERNOUS MALFORMATIONS: A PILOT STUDY. J Biol Regul Homeost Agents. 2015;29: 493–500 pmid:26122242
  24. 24. Williams R, Lim JE, Harr B, Wing C, Walters R, Distler MG, et al. A common and unstable copy number variant is associated with differences in Glo1 expression and anxiety-like behavior. PLoS One. 2009;4: e4649 pmid:19266052
  25. 25. Sidoti A, Antognelli C, Rinaldi C, D'Angelo R, Dattola V, Girlanda P, et al. Glyoxalase I A111E, paraoxonase 1 Q192R and L55M polymorphisms: susceptibility factors of multiple sclerosis? Mult Scler. 2007;13: 446–453 pmid:17463067
  26. 26. Antognelli C, Mezzasoma L, Mearini E, Talesa VN. Glyoxalase 1-419C>A variant is associated with oxidative stress: implications in prostate cancer progression. PLoS One. 2013;8: e74014 pmid:24040147
  27. 27. Krechler T, Jachymova M, Mestek O, Zak A, Zima T, Kalousova M. Soluble receptor for advanced glycation end-products (sRAGE) and polymorphisms of RAGE and glyoxalase I genes in patients with pancreas cancer. Clin Biochem. 2010;43: 882–886 pmid:20398646
  28. 28. Antognelli C, Del BC, Ludovini V, Gori S, Talesa VN, Crino L, et al. CYP17, GSTP1, PON1 and GLO1 gene polymorphisms as risk factors for breast cancer: an Italian case-control study. BMC Cancer. 2009;9: 115 pmid:19379515
  29. 29. Chocholaty M, Jachymova M, Schmidt M, Havlova K, Krepelova A, Zima T, et al. Polymorphisms of the receptor for advanced glycation end-products and glyoxalase I in patients with renal cancer. Tumour Biol. 2015;36: 2121–2126 pmid:25407489
  30. 30. Peculis R, Konrade I, Skapare E, Fridmanis D, Nikitina-Zake L, Lejnieks A, et al. Identification of glyoxalase 1 polymorphisms associated with enzyme activity. Gene. 2013;515: 140–143 pmid:23201419
  31. 31. Lohr JM, Dominguez-Munoz E, Rosendahl J, Besselink M, Mayerle J, Lerch MM, et al. United European Gastroenterology evidence-based guidelines for the diagnosis and therapy of chronic pancreatitis (HaPanEU). United European Gastroenterol J. 2017;5: 153–199 pmid:28344786
  32. 32. Derikx MH, Kovacs P, Scholz M, Masson E, Chen JM, Ruffert C, et al. Polymorphisms at PRSS1-PRSS2 and CLDN2-MORC4 loci associate with alcoholic and non-alcoholic chronic pancreatitis in a European replication study. Gut. 2015;64: 1426–1433 pmid:25253127
  33. 33. Duan Z, Chen G, Chen L, Stolzenberg-Solomon R, Weinstein SJ, Mannisto S, et al. Determinants of concentrations of N(epsilon)-carboxymethyl-lysine and soluble receptor for advanced glycation end products and their associations with risk of pancreatic cancer. Int J Mol Epidemiol Genet. 2014;5: 152–163 pmid:25379135
  34. 34. Rinaldi C, Bramanti P, Fama A, Scimone C, Donato L, Antognelli C, et al. GLYOXALASE I A111E, PARAOXONASE 1 Q192R AND L55M POLYMORPHISMS IN ITALIAN PATIENTS WITH SPORADIC CEREBRAL CAVERNOUS MALFORMATIONS: A PILOT STUDY. J Biol Regul Homeost Agents. 2015;29: 493–500 pmid:26122242