1
|
van der Stouwe AMM, Riemersma NL, Knobbe TJ, Kremer D, Nolte S, Plasmeijer DB, Gomes‐Neto AW, Bakker SJL, Drost G, Elting JWJ. Tremor after solid organ transplantation: Results from the TransplantLines Biobank and Cohort Study. Eur J Neurol 2024; 31:e16412. [PMID: 39440518 PMCID: PMC11554862 DOI: 10.1111/ene.16412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/14/2024] [Accepted: 07/04/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND AND PURPOSE Tremor is a frequent complaint of solid organ transplant recipients. We report on the largest population investigated with clinical neurophysiological methods. Our aim is to objectively establish the tremor prevalence and syndrome in the largest population of solid organ transplant recipients. METHODS Tremor was measured in heart, kidney, liver, and lung recipients, using accelerometers during rest, postural, and weight-loaded conditions. The 95th percentile of healthy kidney donors' tremor amplitude was used as the cutoff to determine the presence of tremor in transplant recipients. Tremor frequency, frequency variability, and effect of loading were used to investigate enhanced physiological tremor as the likely tremor syndrome. Impact on activities of daily life was assessed, and correlations with tacrolimus blood levels were investigated. RESULTS Tremor was present in 52% of 246 transplant recipients, typically in postural positions. Mean tremor frequency was 6.1 (±2.0) Hz; mean tremor variability was 2.6 (±1.8) Hz. A frequency decrease upon loading was found in 83% of patients with tremor. Sixty-five percent of patients met formal clinical neurophysiological criteria for enhanced physiological tremor. Tremor-related impairment was present in 55% and correlated with tremor amplitude (ρ = 0.23, p ≤ 0.001). In a binominal regression analysis, tacrolimus blood levels were independently associated with tremor prevalence (p = 0.009). CONCLUSIONS More than half of solid organ transplant recipients experience a tremor that best fits the syndrome of enhanced physiological tremor. This is the first objective study on tremor that has established a better understanding of the neurophysiological mechanisms of tremor in a large population of solid organ transplant recipients.
Collapse
Affiliation(s)
- A. Madelein M. van der Stouwe
- Expertise Center Movement Disorders Groningen, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Department of Neurology, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Niels L. Riemersma
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Tim J. Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Svea Nolte
- Department of Neurology, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Danieke B. Plasmeijer
- Department of Neurology, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Antonio. W. Gomes‐Neto
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Gea Drost
- Department of Neurology, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Department of Neurosurgery, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| | - Jan Willem J. Elting
- Expertise Center Movement Disorders Groningen, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
- Department of Neurology, University Medical Center GroningenUniversity of GroningenGroningenthe Netherlands
| |
Collapse
|
2
|
Mohamed AAA, Walland P, Stevens J, van Londen M, Heerspink HJL, Gansevoort RT, van de Merbel NC. A validated LC-MS/MS method for the simultaneous quantification of iothalamate and hippuran in serum and urine for non-radioactive kidney function assessment. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1247:124329. [PMID: 39378524 DOI: 10.1016/j.jchromb.2024.124329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024]
Abstract
A novel liquid chromatography-tandem mass spectrometry method is described for the quantitative determination of the kidney function markers iothalamate and hippuran in human serum and urine. It is based on protein precipitation with methanol followed by dilution of the supernatant for serum and simple dilution for urine. The polar analytes are chromatographically separated by a 6.5-min gradient on a low-ligand density reversed-phase column; detection is performed by electrospray ionization tandem mass spectrometry in the positive ion mode against stable-isotope labeled internal standards. The results of a thorough method validation show that iothalamate and hippuran can be simultaneously quantified in the concentration ranges 0.500-30.0 ng/mL and 10.0-5000 ng/mL for serum and urine, respectively, with values for CV and absolute bias not exceeding 10 %, and with sufficient stability in all relevant matrices and solvents. The method was successfully applied for the analysis of serum and urine samples of multiple individuals who received both iothalamate and hippuran.
Collapse
Affiliation(s)
- Abdulfataah A A Mohamed
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Peter Walland
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, The Netherlands
| | - Jasper Stevens
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Marco van Londen
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Hiddo J L Heerspink
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Ron T Gansevoort
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Nico C van de Merbel
- ICON Bioanalytical Laboratories, Amerikaweg 18, 9407 TK Assen, The Netherlands; Department of Analytical Biochemistry, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
3
|
Lockwood MB, Sung C, Alvernaz SA, Lee JR, Chin JL, Nayebpour M, Bernabé BP, Tussing-Humphreys LM, Li H, Spaggiari M, Martinino A, Park CG, Chlipala GE, Doorenbos AZ, Green SJ. The Gut Microbiome and Symptom Burden After Kidney Transplantation: An Overview and Research Opportunities. Biol Res Nurs 2024; 26:636-656. [PMID: 38836469 DOI: 10.1177/10998004241256031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Many kidney transplant recipients continue to experience high symptom burden despite restoration of kidney function. High symptom burden is a significant driver of quality of life. In the post-transplant setting, high symptom burden has been linked to negative outcomes including medication non-adherence, allograft rejection, graft loss, and even mortality. Symbiotic bacteria (microbiota) in the human gastrointestinal tract critically interact with the immune, endocrine, and neurological systems to maintain homeostasis of the host. The gut microbiome has been proposed as an underlying mechanism mediating symptoms in several chronic medical conditions including irritable bowel syndrome, chronic fatigue syndrome, fibromyalgia, and psychoneurological disorders via the gut-brain-microbiota axis, a bidirectional signaling pathway between the enteric and central nervous system. Post-transplant exposure to antibiotics, antivirals, and immunosuppressant medications results in significant alterations in gut microbiota community composition and function, which in turn alter these commensal microorganisms' protective effects. This overview will discuss the current state of the science on the effects of the gut microbiome on symptom burden in kidney transplantation and future directions to guide this field of study.
Collapse
Affiliation(s)
- Mark B Lockwood
- Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Choa Sung
- Post-Doctoral Fellow, Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Suzanne A Alvernaz
- Graduate Student, Department of Biomedical Engineering, University of Illinois ChicagoColleges of Engineering and Medicine, Chicago, IL, USA
| | - John R Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jennifer L Chin
- Medical Student, Touro College of Osteopathic Medicine, Middletown, NY, USA
| | - Mehdi Nayebpour
- Virginia BioAnalytics LLC, Washington, District of Columbia, USA
| | - Beatriz Peñalver Bernabé
- Graduate Student, Department of Biomedical Engineering, University of Illinois ChicagoColleges of Engineering and Medicine, Chicago, IL, USA
| | - Lisa M Tussing-Humphreys
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Hongjin Li
- Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Mario Spaggiari
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Alessandro Martinino
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Chang G Park
- Department of Population Health Nursing Science, Office of Research Facilitation, University of Illinois Chicago, Chicago, IL, USA
| | - George E Chlipala
- Research Core Facility, Research Resources Center, University of Illinois Chicago, Chicago, IL, USA
| | - Ardith Z Doorenbos
- Department of Biobehavioral Nursing Science, University of Illinois ChicagoCollege of Nursing, Chicago, IL, USA
| | - Stefan J Green
- Department of Internal Medicine, Division of Infectious Diseases, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
4
|
Swarte JC, Zhang S, Nieuwenhuis LM, Gacesa R, Knobbe TJ, De Meijer VE, Damman K, Verschuuren EAM, Gan TC, Fu J, Zhernakova A, Harmsen HJM, Blokzijl H, Bakker SJL, Björk JR, Weersma RK. Multiple indicators of gut dysbiosis predict all-cause and cause-specific mortality in solid organ transplant recipients. Gut 2024; 73:1650-1661. [PMID: 38955400 DOI: 10.1136/gutjnl-2023-331441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/12/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVE Gut microbiome composition is associated with multiple diseases, but relatively little is known about its relationship with long-term outcome measures. While gut dysbiosis has been linked to mortality risk in the general population, the relationship with overall survival in specific diseases has not been extensively studied. In the current study, we present results from an in-depth analysis of the relationship between gut dysbiosis and all-cause and cause-specific mortality in the setting of solid organ transplant recipients (SOTR). DESIGN We analysed 1337 metagenomes derived from faecal samples of 766 kidney, 334 liver, 170 lung and 67 heart transplant recipients part of the TransplantLines Biobank and Cohort-a prospective cohort study including extensive phenotype data with 6.5 years of follow-up. To analyze gut dysbiosis, we included an additional 8208 metagenomes from the general population of the same geographical area (northern Netherlands). Multivariable Cox regression and a machine learning algorithm were used to analyse the association between multiple indicators of gut dysbiosis, including individual species abundances, and all-cause and cause-specific mortality. RESULTS We identified two patterns representing overall microbiome community variation that were associated with both all-cause and cause-specific mortality. The gut microbiome distance between each transplantation recipient to the average of the general population was associated with all-cause mortality and death from infection, malignancy and cardiovascular disease. A multivariable Cox regression on individual species abundances identified 23 bacterial species that were associated with all-cause mortality, and by applying a machine learning algorithm, we identified a balance (a type of log-ratio) consisting of 19 out of the 23 species that were associated with all-cause mortality. CONCLUSION Gut dysbiosis is consistently associated with mortality in SOTR. Our results support the observations that gut dysbiosis is associated with long-term survival. Since our data do not allow us to infer causality, more preclinical research is needed to understand mechanisms before we can determine whether gut microbiome-directed therapies may be designed to improve long-term outcomes.
Collapse
Affiliation(s)
- J Casper Swarte
- Gastroenterology and Hepatology, University Medical Centre, Groningen, Netherlands
| | - Shuyan Zhang
- Gastroenterology and Hepatology, University Medical Centre, Groningen, Netherlands
| | | | - Ranko Gacesa
- Gastroenterology and Hepatology, University Medical Centre, Groningen, Netherlands
- Department of Genetics, University of Groningen, University Medical Center, Groningen, Netherlands
| | - Tim J Knobbe
- University Medical Centre, Groningen, Netherlands
| | | | - Kevin Damman
- University Medical Centre, Groningen, Netherlands
| | | | - Tji C Gan
- University Medical Centre, Groningen, Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center, Groningen, Netherlands
- Department of Pediatrics, University Medical Center, Groningen, Netherlands
| | | | - Hermie J M Harmsen
- Medical Microbiology, University of Groningen, University Medical Center, Groningen, Netherlands
| | | | | | - Johannes R Björk
- Gastroenterology and Hepatology, University Medical Centre, Groningen, Netherlands
| | - Rinse K Weersma
- Gastroenterology and Hepatology, University Medical Centre, Groningen, Netherlands
| |
Collapse
|
5
|
Yepes-Calderón M, van der Veen Y, Martín Del Campo S F, Kremer D, Sotomayor CG, Knobbe TJ, Vos MJ, Corpeleijn E, de Borst MH, Bakker SJL. Vitamin C deficiency after kidney transplantation: a cohort and cross-sectional study of the TransplantLines biobank. Eur J Nutr 2024; 63:2357-2366. [PMID: 38811416 PMCID: PMC11377669 DOI: 10.1007/s00394-024-03426-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/02/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE Vitamin C deficiency is associated with excess mortality in kidney transplant recipients (KTR). We aim to evaluate plasma vitamin C status at different post-transplantation moments and assess the main characteristics associated with vitamin C deficiency in KTR. METHODS Plasma vitamin C was assessed in 598 KTR at 3-, 6-, 12-, 24-, and 60-months post-transplantation, 374 late KTR with a functioning graft ≥ 1 year, and 395 potential donors. Vitamin C deficiency was defined as plasma vitamin C ≤ 28 µmol/L. Diet was assessed by a 177-item food frequency questionnaire. Data on vitamin C-containing supplements use were extracted from patient records and verified with the patients. RESULTS Vitamin C deficiency ranged from 46% (6-months post-transplantation) to 30% (≥ 1 year post-transplantation). At all time points, KTR had lower plasma vitamin C than potential donors (30-41 µmol/L vs 58 µmol/L). In cross-sectional analyses of the 953 KTR at their first visit ≥ 12 months after transplantation (55 ± 14 years, 62% male, eGFR 55 ± 19 mL/min/1.73 m2), the characteristics with the strongest association with vitamin C deficiency were diabetes and smoking (OR 2.67 [95% CI 1.84-3.87] and OR 1.84 [95% CI 1.16-2.91], respectively). Dietary vitamin C intake and vitamin C supplementation were associated with lower odds (OR per 100 mg/day 0.38, 95% CI 0.24-0.61 and OR 0.21, 95% CI 0.09-0.44, respectively). CONCLUSION Vitamin C deficiency is frequent among KTR regardless of the time after transplantation, especially among those with diabetes and active smokers. The prevalence of vitamin C deficiency was lower among KTR with higher vitamin C intake, both dietary and supplemented. Further research is warranted to assess whether correcting this modifiable risk factor could improve survival in KTR.
Collapse
Affiliation(s)
- Manuela Yepes-Calderón
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands.
| | - Yvonne van der Veen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
| | - Fernando Martín Del Campo S
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
- Faculty of Medicine, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
| | - Camilo G Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
- Clinical Hospital University of Chile, Independencia, Santiago, Chile
- Institute of Biomedical Sciences, University of Chile, Independencia, Santiago, Chile
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
| | - Michel J Vos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin H de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Hanzeplein 1, Groningen, 9700 RB, The Netherlands
| |
Collapse
|
6
|
Kremer D, Salamah S, Groothof D, Post A, Knobbe TJ, Neto AWG, Peterson S, van Londen M, Yerramilli M, Berger SP, Alkaff FF, de Borst MH, Bakker SJL. Plasma symmetric dimethylarginine and kidney graft function in kidney transplant recipients. Nephrol Dial Transplant 2024; 39:1533-1536. [PMID: 38749569 DOI: 10.1093/ndt/gfae109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Indexed: 08/31/2024] Open
Affiliation(s)
- Daan Kremer
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - Sovia Salamah
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
- Department of Public Health and Preventive Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Dion Groothof
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - Adrian Post
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - Tim J Knobbe
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - António W Gomes Neto
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | | | - Marco van Londen
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | | | - Stefan P Berger
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - Firas F Alkaff
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Martin H de Borst
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| | - Stephan J L Bakker
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, The Netherlands
| |
Collapse
|
7
|
Weerd JCVD, Wegberg AMJV, Boer TS, Engelke UFH, Coene KLM, Wevers RA, Bakker SJL, Blaauw PD, Groen J, Spronsen FJV, Heiner-Fokkema MR. Impact of Phenylketonuria on the Serum Metabolome and Plasma Lipidome: A Study in Early-Treated Patients. Metabolites 2024; 14:479. [PMID: 39330486 PMCID: PMC11434371 DOI: 10.3390/metabo14090479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Data suggest that metabolites, other than blood phenylalanine (Phe), better and independently predict clinical outcomes in patients with phenylketonuria (PKU). METHODS To find new biomarkers, we compared the results of untargeted lipidomics and metabolomics in treated adult PKU patients to those of matched controls. Samples (lipidomics in EDTA-plasma (22 PKU and 22 controls) and metabolomics in serum (35 PKU and 20 controls)) were analyzed using ultra-high-performance liquid chromatography and high-resolution mass spectrometry. Data were subjected to multivariate (PCA, OPLS-DA) and univariate (Mann-Whitney U test, p < 0.05) analyses. RESULTS Levels of 33 (of 20,443) lipid features and 56 (of 5885) metabolite features differed statistically between PKU patients and controls. For lipidomics, findings include higher glycerolipids, glycerophospholipids, and sphingolipids species. Significantly lower values were found for sterols and glycerophospholipids species. Seven features had unknown identities. Total triglyceride content was higher. Higher Phe and Phe catabolites, tryptophan derivatives, pantothenic acid, and dipeptides were observed for metabolomics. Ornithine levels were lower. Twenty-six metabolite features were not annotated. CONCLUSIONS This study provides insight into the metabolic phenotype of PKU patients. Additional studies are required to establish whether the observed changes result from PKU itself, diet, and/or an unknown reason.
Collapse
Affiliation(s)
- Jorine C van der Weerd
- Department of Laboratory Medicine, Laboratory of Metabolic Disease, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Annemiek M J van Wegberg
- Division of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Theo S Boer
- Department of Laboratory Medicine, Laboratory of Metabolic Disease, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Udo F H Engelke
- Department of Human Genetics, Translational Metabolic Laboratory (TML), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Karlien L M Coene
- Department of Human Genetics, Translational Metabolic Laboratory (TML), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Laboratory of Clinical Chemistry and Hematology, Máxima Medical Centre, 5504 DB Veldhoven, The Netherlands
| | - Ron A Wevers
- Department of Human Genetics, Translational Metabolic Laboratory (TML), Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Pim de Blaauw
- Department of Laboratory Medicine, Laboratory of Metabolic Disease, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Joost Groen
- Department of Laboratory Medicine, Laboratory of Metabolic Disease, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - Francjan J van Spronsen
- Division of Metabolic Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, Laboratory of Metabolic Disease, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, The Netherlands
| |
Collapse
|
8
|
Rodrigues FG, Bruins MSM, Vliegenthart R, Kremer D, Sotomayor CG, Nolte IM, Douwe J Mulder U, Navis GJ, Heilberg IP, Pol RA, Bakker SJL, de Borst MH, Te Velde-Keyzer CA. Phase angle and donor type are determinants of coronary artery calcification in stable kidney transplant recipients at twelve months after transplantation. Nutr Metab Cardiovasc Dis 2024; 34:1912-1921. [PMID: 38740537 DOI: 10.1016/j.numecd.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 04/03/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND AND AIM Coronary artery calcification (CAC) partially explains the excess cardiovascular morbidity and mortality after kidney transplantation. This study aimed to investigate determinants of CAC in stable kidney transplant recipients at 12 months post-transplantation. METHODS AND RESULTS CAC-score was quantified by the Agatston method using non-contrast enhanced computed tomography, and age- and sex-standardized CAC-percentiles were calculated. Univariable and multivariable multinomial logistic regression was performed to study potential determinants of CAC. The independent determinants were included in multivariable multinomial logistic regression adjusting for potential confounders. 203 KTRs (age 54.0 ± 14.7 years, 61.1% male) were included. Participants were categorized into four groups according to CAC percentiles (p = 0 [CAC-score = 0], n = 68; p ≥ 1%-p ≤ 50% [CAC score = 29.0 (4.0-166.0)], n = 31; p > 50 ≤ 75% [CAC score = 101.0 (23.8-348.3)], n = 26; and p>75% [CAC score = 581.0 (148.0-1652)], n = 83). Upon multivariable multinomial logistic regression, patients with a narrower phase angle and patients who had received a graft from a deceased donor had a higher risk of being in the >75th CAC-percentile. CONCLUSIONS This study identifies not only metabolic and transplant-related factors, but also phase angle, a composite marker of cell integrity, as an independent determinant of CAC at 12 months after kidney transplantation. This study offers new perspectives for future research into the value of bioelectrical impedance analysis in relation to vascular calcification in kidney transplant recipients.
Collapse
Affiliation(s)
- Fernanda G Rodrigues
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| | - Megan S M Bruins
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rozemarijn Vliegenthart
- Department of Radiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daan Kremer
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Camilo G Sotomayor
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ilja M Nolte
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Udo Douwe J Mulder
- Department of Internal Medicine, Division Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerjan J Navis
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ita Pfeferman Heilberg
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil; Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Robert A Pol
- Department of Vascular and Transplant Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Charlotte A Te Velde-Keyzer
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
9
|
Kremer D, Knobbe TJ, Vinke JSJ, Groothof D, Post A, Annema C, Abrahams AC, van Jaarsveld BC, de Borst MH, Berger SP, Bakker SJL, Eisenga MF. Iron deficiency, anemia, and patient-reported outcomes in kidney transplant recipients. Am J Transplant 2024; 24:1456-1466. [PMID: 38493925 DOI: 10.1016/j.ajt.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 03/19/2024]
Abstract
Kidney transplant recipients (KTRs) experience more fatigue, anxiety, and depressive symptoms and lower concentration and health-related quality of life (HRQoL) compared with the general population. Anemia is a potential cause that is well-recognized and treated. Iron deficiency, however, is often unrecognized, despite its potential detrimental effects related to and unrelated to anemia. We investigated the interplay of anemia, iron deficiency, and patient-reported outcomes in 814 outpatient KTRs (62% male, age 56 ± 13 years) enrolled in the TransplantLines Biobank and Cohort Study (Groningen, The Netherlands). In total, 28% had iron deficiency (ie, transferrin saturation < 20% and ferritin < 100 μg/L), and 29% had anemia (World Health Organization criteria). In linear regression analyses, iron deficiency, but not anemia, was associated with more fatigue, worse concentration, lower wellbeing, more anxiety, more depressive symptoms, and lower HRQoL, independent of age, sex, estimated glomerular filtration rate, anemia, and other potential confounders. In the fully adjusted logistic regression models, iron deficiency was associated with an estimated 53% higher risk of severe fatigue, a 100% higher risk of major depressive symptoms, and a 51% higher chance of being at risk for sick leave/work disability. Clinical trials are needed to investigate the effect of iron deficiency correction on patient-reported outcomes and HRQoL in KTRs.
Collapse
Affiliation(s)
- Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Joanna Sophia J Vinke
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Coby Annema
- Section of Nursing Science, Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Alferso C Abrahams
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Brigit C van Jaarsveld
- Department of Nephrology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Martin H de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
10
|
van Wegberg AMJ, van der Weerd JC, Engelke UFH, Coene KLM, Jahja R, Bakker SJL, Huijbregts SCJ, Wevers RA, Heiner-Fokkema MR, van Spronsen FJ. The clinical relevance of novel biomarkers as outcome parameter in adults with phenylketonuria. J Inherit Metab Dis 2024; 47:624-635. [PMID: 38556470 DOI: 10.1002/jimd.12732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
Recent studies in PKU patients identified alternative biomarkers in blood using untargeted metabolomics. To test the added clinical value of these novel biomarkers, targeted metabolomics of 11 PKU biomarkers (phenylalanine, glutamyl-phenylalanine, glutamyl-glutamyl-phenylalanine, N-lactoyl-phenylalanine, N-acetyl-phenylalanine, the dipeptides phenylalanyl-phenylalanine and phenylalanyl-leucine, phenylalanine-hexose conjugate, phenyllactate, phenylpyruvate, and phenylacetate) was performed in stored serum samples of the well-defined PKU patient-COBESO cohort and a healthy control group. Serum samples of 35 PKU adults and 20 healthy age- and sex-matched controls were analyzed using ultra-high performance liquid chromatography quadrupole time-of-flight mass spectrometry. Group differences were tested using the Mann-Whitney U test. Multiple linear regression analyses were performed with these biomarkers as predictors of (neuro-)cognitive functions working memory, sustained attention, inhibitory control, and mental health. Compared to healthy controls, phenylalanine, glutamyl-phenylalanine, N-lactoyl-phenylalanine, N-acetyl-phenylalanine, phenylalanine-hexose conjugate, phenyllactate, phenylpyruvate, and phenylacetate were significant elevated in PKU adults (p < 0.001). The remaining three were below limit of detection in PKU and controls. Both phenylalanine and N-lactoyl-phenylalanine were associated with DSM-VI Attention deficit/hyperactivity (R2 = 0.195, p = 0.039 and R2 = 0.335, p = 0.002, respectively) of the ASR questionnaire. In addition, N-lactoyl-phenylalanine showed significant associations with ASR DSM-VI avoidant personality (R2 = 0.265, p = 0.010), internalizing (R2 = 0.192, p = 0.046) and externalizing problems (R2 = 0.217, p = 0.029) of the ASR questionnaire and multiple aspects of the MS2D and FI tests, reflecting working memory with R2 between 0.178 (p = 0.048) and 0.204 (p = 0.033). Even though the strength of the models was not considered strong, N-lactoyl-phenylalanine outperformed phenylalanine in its association with working memory and mental health outcomes.
Collapse
Affiliation(s)
- A M J van Wegberg
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| | - J C van der Weerd
- Department of Laboratory Medicine, Laboratory of Metabolic Diseases, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - U F H Engelke
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - K L M Coene
- Laboratory of Clinical Chemistry and Hematology, Máxima Medical Centre, Veldhoven, The Netherlands
| | - R Jahja
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| | - S J L Bakker
- Department of Internal Medicine, University of Groningen, University Medical Center Groningen, The Netherlands
| | - S C J Huijbregts
- Department of Clinical Child and Adolescent Studies-Neurodevelopmental Disorders, Faculty of Social Sciences, Leiden University, Leiden, The Netherlands
| | - R A Wevers
- Department of Human Genetics, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M R Heiner-Fokkema
- Department of Laboratory Medicine, Laboratory of Metabolic Diseases, University of Groningen, University Medical Centre Groningen, The Netherlands
| | - F J van Spronsen
- Division of Metabolic Diseases, University of Groningen, University Medical Center Groningen, Beatrix Children's Hospital, The Netherlands
| |
Collapse
|
11
|
Zhang S, Swarte JC, Gacesa R, Knobbe TJ, Kremer D, Jansen BH, de Borst MH, Harmsen HJM, Erasmus ME, Verschuuren EAM, Bakker SJL, Gan CT, Weersma RK, Björk JR. The gut microbiome in end-stage lung disease and lung transplantation. mSystems 2024; 9:e0131223. [PMID: 38712927 PMCID: PMC11237811 DOI: 10.1128/msystems.01312-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/03/2024] [Indexed: 05/08/2024] Open
Abstract
Gut dysbiosis has been associated with impaired outcomes in liver and kidney transplant recipients, but the gut microbiome of lung transplant recipients has not been extensively explored. We assessed the gut microbiome in 64 fecal samples from end-stage lung disease patients before transplantation and 219 samples from lung transplant recipients after transplantation using metagenomic sequencing. To identify dysbiotic microbial signatures, we analyzed 243 fecal samples from age-, sex-, and BMI-matched healthy controls. By unsupervised clustering, we identified five groups of lung transplant recipients using different combinations of immunosuppressants and antibiotics and analyzed them in relation to the gut microbiome. Finally, we investigated the gut microbiome of lung transplant recipients in different chronic lung allograft dysfunction (CLAD) stages and longitudinal gut microbiome changes after transplantation. We found 108 species (58.1%) in end-stage lung disease patients and 139 species (74.7%) in lung transplant recipients that were differentially abundant compared with healthy controls, with several species exhibiting sharp longitudinal increases from before to after transplantation. Different combinations of immunosuppressants and antibiotics were associated with specific gut microbial signatures. We found that the gut microbiome of lung transplant recipients in CLAD stage 0 was more similar to healthy controls compared to those in CLAD stage 1. Finally, the gut microbial diversity of lung transplant recipients remained lower than the average gut microbial diversity of healthy controls up to more than 20 years post-transplantation. Gut dysbiosis, already present before lung transplantation was exacerbated following lung transplantation.IMPORTANCEThis study provides extensive insights into the gut microbiome of end-stage lung disease patients and lung transplant recipients, which warrants further investigation before the gut microbiome can be used for microbiome-targeted interventions that could improve the outcome of lung transplantation.
Collapse
Affiliation(s)
- Shuyan Zhang
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - J. Casper Swarte
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Ranko Gacesa
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tim J. Knobbe
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Bernadien H. Jansen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Martin H. de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - TransplantLines InvestigatorsAnnemaCobyBakkerStephan J. L.BergerStefan P.BlokzijlHansBodewesFrank A. J. A.de BoerMarieke T.DammanKevinde BorstMartin H.DiepstraArjanDijkstraGerardDouwesRianne M.DoorenbosCaecilia S. E.EisengaMichele F.ErasmusMichiel E.GanC. TjiHakEelkoHepkemaBouke G.KlontFrankKnobbeTim J.KremerDaanLeuveninkHenri G. D.LexmondWillem S.de MeijerVincent E.NiestersHubert G. M.Nieuwenhuis-MoekeGertrude J.van PeltL. JoostPolRobert A.PorteRobert J.RanchorAdelta V.SiebelinkMarion J.SlartRiemer J. H. J. A.SwarteJ. CasperTouwDaan J.van den HeuvelMarius C.van Leer-ButerCorettavan LondenMarcoVerschuurenErik A. M.VosMichel J.WeersmaRinse K.Gomes NetoAntonio W.SandersJan Stephan F.
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Medical Pulmonary Diseases, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Hermie J. M. Harmsen
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Michiel E. Erasmus
- Department of Cardiothoracic Surgery, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Erik A. M. Verschuuren
- Department of Medical Pulmonary Diseases, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - C. Tji Gan
- Department of Medical Pulmonary Diseases, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Rinse K. Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Johannes R. Björk
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
12
|
Post A, Groothof D, Kremer D, Knobbe TJ, Abma W, Koops CA, Tsikas D, Wallimann T, Dullaart RPF, Franssen CFM, Kema IP, Heiner-Fokkema MR, Bakker SJL. Creatine homeostasis and the kidney: comparison between kidney transplant recipients and healthy controls. Amino Acids 2024; 56:42. [PMID: 38869518 PMCID: PMC11176230 DOI: 10.1007/s00726-024-03401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Creatine is a natural nitrogenous organic acid that is integral to energy metabolism and crucial for proper cell functioning. The kidneys are involved in the first step of creatine production. With kidney transplantation being the gold-standard treatment for end-stage kidney disease, kidney transplant recipients (KTR) may be at risk of impaired creatine synthesis. We aimed to compare creatine homeostasis between KTR and controls. Plasma and urine concentrations of arginine, glycine, guanidinoacetate, creatine and creatinine were measured in 553 KTR and 168 healthy controls. Creatine intake was assessed using food frequency questionnaires. Iothalamate-measured GFR data were available in subsets of 157 KTR and 167 controls. KTR and controls had comparable body weight, height and creatine intake (all P > 0.05). However, the total creatine pool was 14% lower in KTR as compared to controls (651 ± 178 vs. 753 ± 239 mmol, P < 0.001). The endogenous creatine synthesis rate was 22% lower in KTR as compared to controls (7.8 ± 3.0 vs. 10.0 ± 4.1 mmol per day, P < 0.001). Despite lower GFR, the plasma guanidinoacetate and creatine concentrations were 21% and 41% lower in KTR as compared to controls (both P < 0.001). Urinary excretion of guanidinoacetate and creatine were 66% and 59% lower in KTR as compared to controls (both P < 0.001). In KTR, but not in controls, a higher measured GFR was associated with a higher endogenous creatine synthesis rate (std. beta: 0.21, 95% CI: 0.08; 0.33; P = 0.002), as well as a higher total creatine pool (std. beta: 0.22, 95% CI: 0.11; 0.33; P < 0.001). These associations were fully mediated (93% and 95%; P < 0.001) by urinary guanidinoacetate excretion which is consistent with production of the creatine precursor guanidinoacetate as rate-limiting factor. Our findings highlight that KTR have a disturbed creatine homeostasis as compared to controls. Given the direct relationship of measured GFR with endogenous creatine synthesis rate and the total creatine pool, creatine supplementation might be beneficial in KTR with low kidney function.Trial registration ID: NCT02811835.Trial registration URL: https://clinicaltrials.gov/ct2/show/NCT02811835 .
Collapse
Affiliation(s)
- Adrian Post
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands.
| | - Dion Groothof
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
| | - Daan Kremer
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
| | - Tim J Knobbe
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
| | - Willem Abma
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ, the Netherlands
| | - Christa A Koops
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ, the Netherlands
| | - Dimitrios Tsikas
- Institute of Toxicology, Core Unit Proteomics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | | | - Robin P F Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
| | - Casper F M Franssen
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ, the Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, 9713 GZ, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, The Netherlands
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, 9700 RB, the Netherlands
| |
Collapse
|
13
|
Trillos-Almanza MC, Chvatal-Medina M, Connelly MA, Moshage H, Bakker SJL, de Meijer VE, Blokzijl H, Dullaart RPF. Circulating Trimethylamine-N-Oxide Is Elevated in Liver Transplant Recipients. Int J Mol Sci 2024; 25:6031. [PMID: 38892218 PMCID: PMC11172608 DOI: 10.3390/ijms25116031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Liver transplant recipients (LTRs) have lower long-term survival rates compared with the general population. This underscores the necessity for developing biomarkers to assess post-transplantation mortality. Here we compared plasma trimethylamine-N-oxide (TMAO) levels with those in the general population, investigated its determinants, and interrogated its association with all-cause mortality in stable LTRs. Plasma TMAO was measured in 367 stable LTRs from the TransplantLines cohort (NCT03272841) and in 4837 participants from the population-based PREVEND cohort. TMAO levels were 35% higher in LTRs compared with PREVEND participants (4.3 vs. 3.2 µmol/L, p < 0.001). Specifically, TMAO was elevated in LTRs with metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and polycystic liver disease as underlying etiology (p < 0.001 for each). Among LTRs, TMAO levels were independently associated with eGFR (std. β = -0.43, p < 0.001) and iron supplementation (std. β = 0.13, p = 0.008), and were associated with mortality (29 deaths during 8.6 years follow-up; log-rank test p = 0.017; hazard ratio of highest vs. lowest tertile 4.14, p = 0.007). In conclusion, plasma TMAO is likely elevated in stable LTRs, with impaired eGFR and iron supplementation as potential contributory factors. Our preliminary findings raise the possibility that plasma TMAO could contribute to increased mortality risk in such patients, but this need to be validated through a series of rigorous and methodical studies.
Collapse
Affiliation(s)
- Maria Camila Trillos-Almanza
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - Mateo Chvatal-Medina
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | | | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - TransplantLines Investigators
- Groningen Institute for Organ Transplantation, University Medical Center Groningen, University of Groningen, 9700 AD Groningen, The Netherlands;
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| | - Vincent E. de Meijer
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.C.-M.); (H.M.); (H.B.)
| | - Robin P. F. Dullaart
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| |
Collapse
|
14
|
de Boer SE, Knobbe TJ, Kremer D, van Munster BC, Nieuwenhuijs-Moeke GJ, Pol RA, Bakker SJL, Berger SP, Sanders JSF. Kidney Transplantation Improves Health-Related Quality of Life in Older Recipients. Transpl Int 2024; 37:12071. [PMID: 38686099 PMCID: PMC11057459 DOI: 10.3389/ti.2024.12071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/12/2024] [Indexed: 05/02/2024]
Abstract
Kidney transplantation is the best treatment for kidney failure in older patients. However, little is known regarding changes in health-related quality of life (HRQoL) from before to after transplantation and determinants of HRQoL in older kidney transplant recipients (KTR). We studied both, using data of older (≥65 years) patients waitlisted for kidney transplantation and older KTR 1 year after transplantation from the TransplantLines Biobank and Cohort Study. HRQoL was assessed using the SF-36 questionnaire. We included 145 older waitlisted patients (68% male, age 70 ± 4 years) and 115 older KTR at 1 year after transplantation (73% male, age 70 ± 4 years). Both mental (48.5 ± 8.4 versus 51.2 ± 7.7, p = 0.009) and physical (47.4 ± 8.5 versus 52.1 ± 7.2, p < 0.001) HRQoL were higher among included KTR, compared to the waitlisted patients. In paired analyses among 46 patients with HRQoL-data both before and after transplantation, there was a trend towards increased mental HRQoL (49.1 ± 8.4 to 51.6 ± 7.5, p = 0.054), and significantly increased physical HRQoL (48.1 ± 8.0 to 52.4 ± 6.7, p = 0.001) after transplantation. Among all assessed factors, the number of patient-reported immunosuppressive drug-related side effects was most strongly negatively associated with both mental and physical HRQoL. In conclusion, HRQoL is significantly higher among older KTR after kidney transplantation compared to older waitlisted patients.
Collapse
Affiliation(s)
- Silke E. de Boer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Tim. J. Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Barbara C. van Munster
- Department of Internal Medicine, Division of Geriatrics, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Robert A. Pol
- Department of Surgery, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Stefan P. Berger
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| | - Jan Stephan F. Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen and University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Westenberg LB, Pol RA, van der Weijden J, de Borst MH, Bakker SJ, van Londen M. Central Body Fat Distribution and Kidney Function after Living Kidney Donation. Clin J Am Soc Nephrol 2024; 19:503-513. [PMID: 38190119 PMCID: PMC11020429 DOI: 10.2215/cjn.0000000000000403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/03/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND In most screening guidelines, high body mass index (BMI) is considered a contraindication for kidney donation. New insights suggest that central body fat distribution might provide greater power in assessing kidney risk. This study aimed to determine whether BMI and central body fat distribution measures are associated with long-term kidney function after donor nephrectomy. We hypothesized that higher BMI, waist circumference (WC), and waist-to-height ratio (WHtR) were associated with lower kidney function long term after donation. METHODS The study population consisted of living kidney donors. BMI, WC, and WHtR were measured during donor screening. The outcome postdonation kidney function was assessed using measured GFR (mGFR) (mGFR, 125 I-iothalamate infusion) at 3 months ( n =1042), 5 years ( n =556), and 10 years ( n =210) of follow-up. Primary multivariable linear regression analyses were performed with BMI and WC and secondary analyses with WHtR. Linear mixed models were performed to investigate change in postdonation eGFR. RESULTS The donor age was 52±11 years, and 48% were male. The mean BMI was 26.1±3.6 kg/m 2 , and WC was 91±11 cm. Higher predonation BMI was associated with lower mGFR throughout follow-up: -1.35 (95% confidence interval [CI], -1.95 to -0.80), -1.55 (95% CI, -2.50 to -0.65), and -2.35 (95% CI, -4.10 to -0.60) ml/min per m 2 per 5 kg/m 2 higher BMI at 3 months, 5, and 10 years after donation, respectively, adjusted for sex, age, and predonation GFR. For WC, differences in mGFR were -1.30 (95% CI, -1.70 to -0.90), -1.50 (95% CI, -2.20 to -0.80), and -1.70 (95% CI, -3.00 to -0.50) ml/min per m 2 per 10 cm higher WC at 3 months, 5, and 10 years after donation, respectively. In male donors, BMI and WC were significantly associated with a negative postdonation change in eGFR. CONCLUSIONS Higher BMI and WC were independently associated with lower GFR (long term) after living kidney donation.
Collapse
Affiliation(s)
- Lisa B. Westenberg
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Division of Transplant Surgery, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert A. Pol
- Division of Transplant Surgery, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Jessica van der Weijden
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin H. de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J.L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco van Londen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
16
|
van der Vaart A, Kremer D, Niekolaas T, Bakker SJL, van Dijk PR, de Borst MH. Time-updated Fibroblast Growth Factor 23 Is Predictive for Posttransplant Diabetes Mellitus in Kidney Transplant Recipients. J Endocr Soc 2024; 8:bvae055. [PMID: 38577264 PMCID: PMC10993900 DOI: 10.1210/jendso/bvae055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Indexed: 04/06/2024] Open
Abstract
Objective This work aimed to study whether fibroblast growth factor 23 (FGF23) is predictive for incident posttransplant diabetes mellitus (PTDM) in kidney transplant recipients (KTRs). Methods We repeatedly analyzed plasma C-terminal FGF23 concentrations in 170 KTRs enrolled in the TransplantLines Biobank and Cohort Study. Associations of time-updated plasma FGF23 with incident PTDM were studied by Cox regression. Results A total of 170 KTRs (46% female, aged 54.4 ± 12.4 years) with 540 FGF23 measurements were included. Plasma FGF23 concentrations at transplantation were 31.1 (0.76-2576) pmol/L. During a follow-up of 24 (12-24) months, 38 patients developed PTDM. The highest FGF23 tertile (compared to the lowest) was associated with an increased risk for PTDM (fully adjusted hazard ratio 20.9; 95% CI, 3.4-130.0; P < .001). Conclusion In KTRs without diabetes at baseline, the highest tertile of FGF23, compared to the lowest, is predictive for development of PTDM.
Collapse
Affiliation(s)
- Amarens van der Vaart
- Department of Internal Medicine, Divisions of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
- Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB Groningen, the Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Divisions of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Tessa Niekolaas
- Department of Internal Medicine, Divisions of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Divisions of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| | - Peter R van Dijk
- Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, 9700 RB Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Divisions of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB Groningen, the Netherlands
| |
Collapse
|
17
|
Klont F, Nijdam FB, Bakker SJL, Keski-Rahkonen P, Hopfgartner G, Investigators T. High-abundance peaks and peak clusters associate with pharmaceutical polymers and excipients in urinary untargeted clinical metabolomics data: exploration of their origin and possible impact on label-free quantification. Analyst 2024; 149:1061-1067. [PMID: 38251754 PMCID: PMC10866140 DOI: 10.1039/d3an01874a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 12/08/2023] [Indexed: 01/23/2024]
Abstract
Pharmaceutical polymers and excipients represent interesting but often overlooked chemical classes in clinical exposure and bioanalytical research. These chemicals may cause hypersensitivity reactions, they can be useful to confirm exposure to pharmaceuticals, and they may pose bioanalytical challenges, including ion suppression in liquid chromatography-mass spectrometry (LC-MS-)based workflows. In this work, we assessed these chemicals in light of a rather surprising finding presented in two previously published studies, namely that usage of cyclosporine A, an immunosuppressive drug which is known to be cleared through excretion in the bile, explained the largest amount of variance in principal component analysis of urinary LC-SWATH/MS small-molecule profiling data. Specifically, we examined the freely-accessible 24-hour urine metabolomics data of 570 kidney transplant recipients included in the TransplantLines Biobank and Cohort Study (NCT03272841). These data unveiled thousands of high-abundance polymer peaks in some samples, which were associated with the use of the macrogol (i.e., polyethylene glycol) 3350 oral laxative agent. In addition, we found multiple clusters of high-abundance peaks which were linked to the exposure to two pharmaceutical excipients, namely short-chain polyethylene glycol (molecular weight <1000 Da) and polyethoxylated castor oil (also known as Kolliphor® EL or Cremophor® EL). Respectively, these excipients are used in temazepam capsules and cyclosporine A capsules, and the latter provides a plausible explanation for the rather surprising finding that instigated our work. Moreover, such explanation and our findings in general put emphasis on taking into consideration these and other pharmaceutical polymers and excipients when exploring, processing, and interpreting clinical small-molecule profiling data.
Collapse
Affiliation(s)
- Frank Klont
- Unit of PharmacoTherapy, -Epidemiology & -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
- Life Sciences Mass Spectrometry, Department of Inorganic and Analytical Chemistry, University of Geneva, Quai Ernest Ansermet 24, 1211 Geneva, Switzerland
| | - Fleur B Nijdam
- Unit of PharmacoTherapy, -Epidemiology & -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC/WHO), Avenue Tony Garnier 25, 69007 Lyon, France
| | - Gérard Hopfgartner
- Life Sciences Mass Spectrometry, Department of Inorganic and Analytical Chemistry, University of Geneva, Quai Ernest Ansermet 24, 1211 Geneva, Switzerland
| | - TransplantLines Investigators
- Group of Authors on Behalf of the Transplant Lines Biobank and Cohort Study, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands
| |
Collapse
|
18
|
Rodrigues FG, Van Der Plas WY, Sotomayor CG, Van Der Vaart A, Kremer D, Pol RA, Kruijff S, Heilberg IP, Bakker SJL, De Borst MH. Pre-Transplant Hyperparathyroidism and Graft or Patient Outcomes After Kidney Transplantation. Transpl Int 2024; 37:11916. [PMID: 38384325 PMCID: PMC10880800 DOI: 10.3389/ti.2024.11916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 01/23/2024] [Indexed: 02/23/2024]
Abstract
The impact of pre-transplant parathyroid hormone (PTH) levels on early or long-term kidney function after kidney transplantation is subject of debate. We assessed whether severe hyperparathyroidism is associated with delayed graft function (DGF), death-censored graft failure (DCGF), or all-cause mortality. In this single-center cohort study, we studied the relationship between PTH and other parameters related to bone and mineral metabolism, including serum alkaline phosphatase (ALP) at time of transplantation with the subsequent risk of DGF, DCGF and all-cause mortality using multivariable logistic and Cox regression analyses. In 1,576 kidney transplant recipients (51.6 ± 14.0 years, 57.3% male), severe hyperparathyroidism characterized by pre-transplant PTH ≥771 pg/mL (>9 times the upper limit) was present in 121 patients. During 5.2 [0.2-30.0] years follow-up, 278 (15.7%) patients developed DGF, 150 (9.9%) DCGF and 432 (28.6%) died. A higher pre-transplant PTH was not associated with DGF (HR 1.06 [0.90-1.25]), DCGF (HR 0.98 [0.87-1.13]), or all-cause mortality (HR 1.02 [0.93-1.11]). Results were consistent in sensitivity analyses. The same applied to other parameters related to bone and mineral metabolism, including ALP. Severe pre-transplant hyperparathyroidism was not associated with an increased risk of DGF, DCGF or all-cause mortality, not supporting the need of correction before kidney transplantation to improve graft or patient survival.
Collapse
Affiliation(s)
- Fernanda Guedes Rodrigues
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Willemijn Y. Van Der Plas
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Camilo German Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amarens Van Der Vaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Robert A. Pol
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Schelto Kruijff
- Department of Surgery, Division of Surgical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Surgery, Martini Hospital Groningen, Groningen, Netherlands
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ita Pfeferman Heilberg
- Nutrition Post Graduation Program, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
- Nephrology Division, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | | | - Martin H. De Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
19
|
Wijma AG, Bongers BC, Annema C, Dekker R, Blokzijl H, van der Palen JA, De Meijer VE, Cuperus FJ, Klaase JM. 'Effects of a home-based bimodal lifestyle intervention in frail patients with end-stage liver disease awaiting orthotopic liver transplantation': study protocol of a non-randomised clinical trial. BMJ Open 2024; 14:e080430. [PMID: 38286689 PMCID: PMC10826538 DOI: 10.1136/bmjopen-2023-080430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/17/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Patients with end-stage liver disease awaiting orthotopic liver transplantation (OLT) are generally classified as frail due to disease-related malnutrition and a progressive decline in musculoskeletal and aerobic fitness, which is associated with poor pre-OLT, peri-OLT and post-OLT outcomes. However, frailty in these patients may be reversable with adequate exercise and nutritional interventions. METHODS AND ANALYSIS Non-randomised clinical trial evaluating the effect of a home-based bimodal lifestyle programme in unfit patients with a preoperative oxygen uptake (VO2) at the ventilatory anaerobic threshold ≤13 mL/kg/min and/or VO2 at peak exercise ≤18 mL/kg/min listed for OLT at the University Medical Center Groningen (UMCG). The programme is patient tailored and comprises high-intensity interval and endurance training, and functional exercises three times per week, combined with nutritional support. Patients will go through two training periods, each lasting 6 weeks.The primary outcome of this study is the impact of the programme on patients' aerobic fitness after the first study period. Secondary outcomes include aerobic capacity after the second study period, changes in sarcopenia, anthropometry, functional mobility, perceived quality of life and fatigue, incidence of hepatic encephalopathy and microbiome composition. Moreover, number and reasons of intercurrent hospitalisations during the study and postoperative outcomes up to 12 months post OLT will be recorded. Finally, feasibility of the programme will be assessed by monitoring the participation rate and reasons for non-participation, number and severity of adverse events, and dropout rate and reasons for dropout. ETHICS AND DISSEMINATION This study was approved by the Medical Research Ethics Committee of the UMCG (registration number NL83612.042.23, August 2023) and is registered in the Clinicaltrials.gov register (NCT05853484). Good Clinical Practice guidelines and the principles of the Declaration of Helsinki will be applied. Results of this study will be submitted for presentation at (inter)national congresses and publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT05853484.
Collapse
Affiliation(s)
- Allard G Wijma
- Department of Surgery, division of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bart C Bongers
- Department of Nutrition and Movement Sciences, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Department of Surgery, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Coby Annema
- Section of Nursing Science, Department of Health Sciences, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Rienk Dekker
- Department for Rehabilitation Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Job Am van der Palen
- Department of Epidemiology, Medisch spectrum Twente, Enschede, Netherlands
- Section Cognition, Data and Education, Faculty of Behavioural, Management and Social Sciences, University of Twente, Enschede, Netherlands
| | - Vincent E De Meijer
- Department of Surgery, division of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Frans Jc Cuperus
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joost M Klaase
- Department of Surgery, division of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
20
|
Knobbe TJ, Kremer D, Eisenga MF, van Londen M, Annema C, Bültmann U, Kema IP, Navis GJ, Berger SP, Bakker SJL. Sleep quality, fatigue, societal participation and health-related quality of life in kidney transplant recipients: a cross-sectional and longitudinal cohort study. Nephrol Dial Transplant 2023; 39:74-83. [PMID: 37418245 PMCID: PMC10730797 DOI: 10.1093/ndt/gfad148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND Fatigue and impaired health-related quality of life (HRQoL) are common among kidney transplant recipients (KTR). We hypothesized that both may partially be attributable to poor sleep. METHODS Cross-sectional and longitudinal data of KTR enrolled in the TransplantLines Biobank and Cohort Study were used. Sleep quality was assessed using the Pittsburgh Sleep Quality Index questionnaire. Individual strength (i.e. a composite of fatigue, concentration, motivation and physical activity), societal participation and HRQoL were assessed using validated questionnaires. RESULTS We included 872 KTR (39% female, age 56 ± 13 years) and 335 healthy controls. In total, 33% of male KTR and 49% of female KTR reported poor sleep quality, which was higher compared with male and female healthy controls (19% and 28%, respectively, P < .001 for both). In logistic regression analyses, female sex, anxiety, active smoking, low protein intake, physically inactive lifestyle, low plasma magnesium concentration, using calcineurin inhibitors, not using mTOR inhibitors and using benzodiazepine agonists were associated with poor sleep quality. In adjusted linear regression analyses, poor sleep was strongly and independently associated with lower individual strength [standardized β (st.β) = 0.59, 95% confidence interval (CI) 0.45 to 0.74, P < .001], poorer societal participation (frequency: st.β = -0.17, 95% CI -0.32 to -0.01, P = .04; restrictions: st.β = -0.36, 95% CI -0.51 to -0.21, P < .001; satisfaction: st.β = -0.44, 95% CI -0.59 to -0.28, P < .001) and lower HRQoL (physical: st.β = -0.53, 95% CI -0.68 to -0.38, P < .001; mental: st.β = -0.64, 95% CI -0.78 to -0.50, P < .001). The associations with poorer societal participation and lower HRQoL were strongly mediated by individual strength (P < .001 for all), yet the suggested direct effects of poor sleep quality on HRQoL remained significant (Pphysical = .03, Pmental = .002). Longitudinal data of 292 KTR showed that sleep quality improves after kidney transplantation in males (P < .001), but not in females (P = .9). CONCLUSIONS Poor sleep quality is common among KTR, and may be a potential target to improve fatigue, societal participation and HRQoL among KTR.
Collapse
Affiliation(s)
- Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco van Londen
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Coby Annema
- Department of Health Sciences, Section of Nursing Science, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ute Bültmann
- Department of Health Sciences, Community and Occupational Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ido P Kema
- Department of Laboratory Medicine Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerjan J Navis
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
21
|
Doorenbos CSE, Jonker J, Hao J, Gore EJ, Kremer D, Knobbe TJ, de Joode AAE, Sanders JSF, Thaunat O, Niesters HGM, Van Leer-Buter CC, Bakker SJL. Smoking, Alcohol Intake and Torque Teno Virus in Stable Kidney Transplant Recipients. Viruses 2023; 15:2387. [PMID: 38140628 PMCID: PMC10748022 DOI: 10.3390/v15122387] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Torque Teno Virus (TTV) is a non-pathogenic virus that is highly prevalent among kidney transplant recipients (KTRs). Its circulating load is associated with an immunological status in KTR and is considered a promising tool for guiding immunosuppression. To allow for optimal guidance, it is important to identify other determinants of TTV load. We aimed to investigate the potential association of smoking and alcohol intake with TTV load. For this cross-sectional study, serum TTV load was measured using PCR in stable kidney transplant recipients at ≥1 year after transplantation, and smoking status and alcohol intake were assessed through questionnaires and measurements of urinary cotinine and ethyl glucuronide. A total of 666 KTRs were included (57% male). A total of 549 KTR (82%) had a detectable TTV load (3.1 ± 1.5 log10 copies/mL). In KTR with a detectable TTV load, cyclosporin and tacrolimus use were positively associated with TTV load (St. β = 0.46, p < 0.001 and St. β = 0.66, p < 0.001, respectively), independently of adjustment for potential confounders. Current smoking and alcohol intake of >20 g/day were negatively associated with TTV load (St. β = -0.40, p = 0.004 and St. β = -0.33, p = 0.009, respectively), independently of each other and of adjustment for age, sex, kidney function, time since transplantation and calcineurin inhibitor use. This strong association of smoking and alcohol intake with TTV suggests a need to account for the smoking status and alcohol intake when applying TTV guided immunosuppression in KTR.
Collapse
Affiliation(s)
- Caecilia S. E. Doorenbos
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Jip Jonker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Jiasi Hao
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Edmund J. Gore
- Department of Medical Microbiology, Division of Clinical Virology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Tim J. Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Anoek A. E. de Joode
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Jan Stephan F. Sanders
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| | - Olivier Thaunat
- Department of Transplantation Nephrology and Clinical Immunology Hospices Civils de Lyon, Claude Bernard Lyon I University, INSERM Unit 1111, 69003 Lyon, France
| | - Hubert G. M. Niesters
- Department of Medical Microbiology, Division of Clinical Virology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Coretta C. Van Leer-Buter
- Department of Medical Microbiology, Division of Clinical Virology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands; (J.J.)
| |
Collapse
|
22
|
Swarte JC, Knobbe TJ, Björk JR, Gacesa R, Nieuwenhuis LM, Zhang S, Vila AV, Kremer D, Douwes RM, Post A, Quint EE, Pol RA, Jansen BH, de Borst MH, de Meijer VE, Blokzijl H, Berger SP, Festen EAM, Zhernakova A, Fu J, Harmsen HJM, Bakker SJL, Weersma RK. Health-related quality of life is linked to the gut microbiome in kidney transplant recipients. Nat Commun 2023; 14:7968. [PMID: 38042820 PMCID: PMC10693618 DOI: 10.1038/s41467-023-43431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 11/08/2023] [Indexed: 12/04/2023] Open
Abstract
Kidney transplant recipients (KTR) have impaired health-related quality of life (HRQoL) and suffer from intestinal dysbiosis. Increasing evidence shows that gut health and HRQoL are tightly related in the general population. Here, we investigate the association between the gut microbiome and HRQoL in KTR, using metagenomic sequencing data from fecal samples collected from 507 KTR. Multiple bacterial species are associated with lower HRQoL, many of which have previously been associated with adverse health conditions. Gut microbiome distance to the general population is highest among KTR with an impaired physical HRQoL (R = -0.20, P = 2.3 × 10-65) and mental HRQoL (R = -0.14, P = 1.3 × 10-3). Physical and mental HRQoL explain a significant part of variance in the gut microbiome (R2 = 0.58%, FDR = 5.43 × 10-4 and R2 = 0.37%, FDR = 1.38 × 10-3, respectively). Additionally, multiple metabolic and neuroactive pathways (gut brain modules) are associated with lower HRQoL. While the observational design of our study does not allow us to analyze causality, we provide a comprehensive overview of the associations between the gut microbiome and HRQoL while controlling for confounders.
Collapse
Affiliation(s)
- J Casper Swarte
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Johannes R Björk
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ranko Gacesa
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Lianne M Nieuwenhuis
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Shuyan Zhang
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rianne M Douwes
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Adrian Post
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Evelien E Quint
- Department of Surgery, division of Transplantation Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robert A Pol
- Department of Surgery, division of Transplantation Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bernadien H Jansen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent E de Meijer
- Department of Surgery, section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology and Infection prevention, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
23
|
Swaab TDA, Quint EE, Westenberg LB, Zorgdrager M, Segev DL, McAdams‐DeMarco MA, Bakker SJL, Viddeleer AR, Pol RA. Validity of computed tomography defined body composition as a prognostic factor for functional outcome after kidney transplantation. J Cachexia Sarcopenia Muscle 2023; 14:2532-2539. [PMID: 37731200 PMCID: PMC10751408 DOI: 10.1002/jcsm.13316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 06/08/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND The prevalence of sarcopenia is markedly higher in kidney transplant candidates than in the general population. It is a syndrome characterized by progressive and generalized loss of skeletal muscle mass and strength, which increases the risk of adverse postoperative outcomes. METHODS We studied the impact of computed tomography defined preoperative sarcopenia, defined as a skeletal muscle index below age and gender specific cut-off values, on postoperative physical functional outcomes (grip strength, 4-m walking test, timed up and go, and sit to stand) at 6 months follow up. RESULTS A total of 107 patients transplanted between 2015 and 2019 were included in this single-centre study. Mean age was 60.3 (±13.1), and 68.2% of patients were male. Ten patients (9.4%) were identified as sarcopenic. Sarcopenic patients were younger (55.6 (±15.1) vs. 60.8 (±12.9) years), more likely to be female (60.0% vs. 28.9%), and had an increased dialysis vintage (19 [2.5-32.8] vs. 9 [0.0-21.0] months) in comparison with their non-sarcopenic counterparts. In univariate analysis, they had a significantly lower body mass index and skeletal muscle area (P ≤ 0.001). In multivariate regression analysis, skeletal muscle index was significantly associated with grip strength (β = 0.690, R2 = 0.232) and timed up and go performance (β = -0.070, R2 = 0.154). CONCLUSIONS We identified a significant association between sarcopenia existing pre-transplantation and poorer 6 months post-transplantation physical functioning with respect to hand grip strength and timed up and go tests in kidney transplant recipients. These results could be used to preoperatively identify patients with an increased risk of poor postoperative physical functional outcome, allowing for preoperative interventions to mitigate these risks.
Collapse
Affiliation(s)
- Tim D. A. Swaab
- Department of Surgery, Division of Transplantation Surgery, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Evelien E. Quint
- Department of Surgery, Division of Transplantation Surgery, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Lisa B. Westenberg
- Department of Surgery, Division of Transplantation Surgery, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Marcel Zorgdrager
- Department of Radiology, Medical Imaging CenterUniversity Medical Center GroningenGroningenThe Netherlands
| | - Dorry L. Segev
- Department of SurgeryNYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | - Stephan J. L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| | - Alain R. Viddeleer
- Department of Radiology, Medical Imaging CenterUniversity Medical Center GroningenGroningenThe Netherlands
| | - Robert A. Pol
- Department of Surgery, Division of Transplantation Surgery, University Medical Center GroningenUniversity of GroningenGroningenThe Netherlands
| |
Collapse
|
24
|
Post A, Garcia E, Shalaurova I, Matyus SP, González-Delgado JM, Doorenbos CSE, van der Veen Y, Shah SH, Kraus WE, Kremer D, Knobbe TJ, Bakker SJL, Dullaart RPF, Connelly MA. Creatine and creatinine quantified using nuclear magnetic resonance: A method validation study and clinical associations between circulating creatine and fatigue in kidney transplant recipients. Clin Chim Acta 2023; 548:117517. [PMID: 37591418 DOI: 10.1016/j.cca.2023.117517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND A potential contributor to fatigue in kidney transplant recipients (KTR) may be impaired creatine homeostasis. We developed and validated a high-throughput NMR assay allowing for simultaneous measurement of circulating creatine and creatinine, and determined plasma creatine and estimated intramuscular creatine concentrations in KTRs, delineated their determinants and explored their associations with self-reported fatigue. METHODS An NMR assay was developed and validated for measurement of circulating creatinine and creatine concentrations. Plasma creatine and creatinine concentrations were measured in 618 KTR. Fatigue was assessed using the checklist individual strength. Associations of creatine parameters with fatigue was assessed using linear mixed effect models. RESULTS The NMR-based assay had good sensitivity, precision and demonstrated linearity across a large range of values. Among KTR, the mean age was 56 ± 13 years, 62% were men and eGFR was 54 ± 18 ml/min/1.73 m2. Plasma creatine concentration was 27 [19-39] µmol/L. Estimated intramuscular creatine concentration was 27 ± 7 mmol/kg. Higher plasma creatine concentration and higher estimated intramuscular creatine concentration were independently associated with a lower total fatigue score and less motivation problems. CONCLUSION An NMR method for measurement of circulating creatine and creatinine which offers the potential for accurate and efficient quantification was developed. The found associations suggest that improving creatine status may play a beneficial role in mitigating fatigue.
Collapse
Affiliation(s)
- Adrian Post
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | | | | | | | | | - Caecilia S E Doorenbos
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | - Yvonne van der Veen
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | - Svati H Shah
- Division of Cardiology, Department of Medicine and Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA; Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - William E Kraus
- Division of Cardiology, Department of Medicine and Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Daan Kremer
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | - Tim J Knobbe
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | - Stephan J L Bakker
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | - Robin P F Dullaart
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands.
| | | |
Collapse
|
25
|
Denhaerynck K, Dobbels F, Košťálová B, De Geest S. Psychometric Properties of the BAASIS: A Meta-analysis of Individual Participant Data. Transplantation 2023; 107:1795-1809. [PMID: 36949037 PMCID: PMC10358438 DOI: 10.1097/tp.0000000000004574] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 11/16/2022] [Accepted: 12/11/2022] [Indexed: 03/24/2023]
Abstract
BACKGROUND Nonadherence to immunosuppressives, a risk factor for poor posttransplant outcomes, can be assessed by self-report using the Basel Assessment of Adherence to Immunosuppressive Medications Scale (BAASIS). Available in written and interview versions, and previously validated on content, the BAASIS is widely used in research and clinical practice. The aim of this study was to investigate its psychometric properties. METHODS Using a literature search and our BAASIS database, this meta-analysis identified completed studies in adult transplant recipients whose data were usable to examine the BAASIS' reliability and 3 validity aspects: (1) relationships with other variables (electronic monitoring, other self-report scales, tacrolimus blood-level variability, collateral report, depressive symptoms, psycho-behavioral constructs, and interventions); (2) response processes; and (3) internal structure. Testing used random-effects logistic regressions. RESULTS Our sample included 12 109 graft recipients from 26 studies. Of these 26, a total of 20 provided individual participant data. Evidence of the BAASIS' stability over time supports its reliability. Validity testing of relationships with other variables showed that BAASIS-assessed nonadherence was significantly associated with the selected variables: electronically monitored nonadherence ( P < 0.03), other self- and collaterally-reported nonadherence ( P < 0.001), higher variability in tacrolimus concentrations ( P = 0.02), higher barriers ( P < 0.001), lower self-efficacy ( P < 0.001), lower intention ( P < 0.001), and higher worries ( P = 0.02). Nonadherence also decreased after regimen change interventions ( P = 0.03). Response process evaluation indicated good readability and slightly higher nonadherence with the written version. Structurally, items on taking and timing shared variability. CONCLUSIONS The BAASIS shows good validity and reliability as a self-report instrument to assess medication nonadherence in transplantation.
Collapse
Affiliation(s)
- Kris Denhaerynck
- Institute of Nursing Science, Department of Public Health, University of Basel, Switzerland
| | - Fabienne Dobbels
- Academic Centre for Nursing and Midwifery, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| | - Barbora Košťálová
- Department of Social and Clinical Pharmacy, Faculty of Pharmacy in Hradec Králové, Charles University, Prague, Czech Republic
| | - Sabina De Geest
- Institute of Nursing Science, Department of Public Health, University of Basel, Switzerland
- Academic Centre for Nursing and Midwifery, Department of Public Health and Primary Care, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Quint EE, Haanstra AJ, van der Veen Y, Maring H, Berger SP, Ranchor A, Bakker SJL, Finnema E, Pol RA, Annema C. PREhabilitation of CAndidates for REnal Transplantation (PreCareTx) study: protocol for a hybrid type I, mixed method, randomised controlled trial. BMJ Open 2023; 13:e072805. [PMID: 37500274 PMCID: PMC10387712 DOI: 10.1136/bmjopen-2023-072805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
INTRODUCTION Kidney transplant candidates (KTCs) need to be in optimal physical and psychological condition prior to surgery. However, KTCs often experience compromised functional capacity which can be characterised as frailty. Prehabilitation, the enhancement of a person's functional capacity, may be an effective intervention to improve the health status of KTCs. The PREhabilitation of CAndidates for REnal Transplantation (PreCareTx) study aims to examine the effectiveness of a multimodal prehabilitation programme on the health status of KTCs, and to explore the potential of implementation of prehabilitation in daily clinical practice. METHODS AND ANALYSIS This study uses a single centre, effectiveness-implementation hybrid type I study design, comprised of a randomised controlled trial and a mixed-methods study. Adult patients who are currently on the transplant waiting list or are waitlisted during the study period, at a university medical centre in The Netherlands, will be randomly assigned to either prehabilitation (n=64) or care as usual (n=64) groups. The prehabilitation group will undergo a 12-week home-based, tailored prehabilitation programme consisting of physical and/or nutritional and/or psychosocial interventions depending on the participant's deficits. This programme will be followed by a 12-week maintenance programme in order to enhance the incorporation of the interventions into daily life. The primary endpoint of this study is a change in frailty status as a proxy for health status. Secondary endpoints include changes in physical fitness, nutritional status, psychological well-being, quality of life and clinical outcomes. Tertiary endpoints include the safety, feasibility and acceptability of the prehabilitation programme, and the barriers and facilitators for further implementation. ETHICS AND DISSEMINATION Medical ethical approval was granted by the Medical Ethics Committee Groningen, Netherlands (M22.421). Written informed consent will be obtained from all participants. The results will be disseminated at international conferences and in peer-reviewed journals. TRIAL REGISTRATION NUMBER ClinicalTrials.gov, NCT05489432.
Collapse
Affiliation(s)
- Evelien E Quint
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Avril J Haanstra
- Department of Health Sciences, Section of Nursing Science, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yvonne van der Veen
- Department of Health Sciences, Section of Nursing Science, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Internal Medicine, Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Heleen Maring
- Department of Health Sciences, Section of Nursing Science, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Physical Therapy, University Medical Center Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Internal Medicine, Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Adelita Ranchor
- Department of Health Sciences, Section of Health Psychology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Internal Medicine, Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Evelyn Finnema
- Department of Health Sciences, Section of Nursing Science, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert A Pol
- Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Coby Annema
- Department of Health Sciences, Section of Nursing Science, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
27
|
Vinke JSJ, Ziengs AL, Buunk AM, van Sonderen L, Gomes-Neto AW, Berger SP, Bakker SJL, Eisenga MF, Spikman JM, De Borst MH. Iron deficiency and cognitive functioning in kidney transplant recipients: findings of the TransplantLines biobank and cohort study. Nephrol Dial Transplant 2023; 38:1719-1728. [PMID: 36662046 PMCID: PMC10310504 DOI: 10.1093/ndt/gfad013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Neurocognitive impairment is common in kidney transplant recipients (KTRs). Adequate brain functioning requires energy and neurotransmitter activity, for which iron is essential. We aimed to investigate iron deficiency (ID) as a potentially modifiable risk factor for cognitive impairment in KTRs. METHODS We analyzed stable KTRs participating in the TransplantLines Biobank and Cohort study. Participants underwent neuropsychological tests for memory, mental speed, and attention and executive functioning. ID was defined as ferritin <100 µg/mL or 100-299 µg/mL with transferrin saturation (TSAT) ≤20%. Associations between iron status and norm scores of neurocognitive outcomes, corrected for age, sex and education, were assessed using multivariable linear regression analyses adjusted for potential confounders including hemoglobin. RESULTS We included 166 KTRs [median (IQR) age 57 (45-65) years, 59% male, estimated glomerular filtration rate 51±18 mL/min/1.73 m2]. Time since transplantation was 5.8 (1.0-12.0) years. Prevalence of ID was 65%. ID was independently associated with lower scores for mental speed (std.β = -0.19, P = .02) and attention and executive functioning (std.β = -0.19, P = .02), and tended to be associated with worse memory (std.β = -0.16, P = .07). Lower plasma ferritin levels were associated with worse memory (std.β = 0.23, P = .007), mental speed (std.β = 0.34, P < .001), and attention and executive functioning (std.β = 0.30, P = .001). Lower TSAT was associated with worse memory (std.β = 0.19, P = .04) and mental speed (std.β = 0.27, P = .003), and tended to be associated with worse attention and executive functioning (std.β = 0.16, P = .08). CONCLUSIONS Iron-deficient KTRs performed worse on neurocognitive tasks measuring memory, mental speed, and attention and executive functioning. These findings set the stage for prospective studies addressing whether ID correction restores cognitive function after kidney transplantation.
Collapse
Affiliation(s)
- Joanna Sophia J Vinke
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Aaltje L Ziengs
- Department of Neuropsychology, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne M Buunk
- Department of Neuropsychology, University Medical Center Groningen, Groningen, The Netherlands
| | - Lisanne van Sonderen
- Department of Neuropsychology, University Medical Center Groningen, Groningen, The Netherlands
| | - Antonio W Gomes-Neto
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - TransplantLines Investigators
- Groningen Transplant Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacoba M Spikman
- Department of Neuropsychology, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin H De Borst
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
28
|
Yepes-Calderón M, Kremer D, Post A, Sotomayor CG, Seidel U, Huebbe P, Knobbe TJ, Lüersen K, Eisenga MF, Corpeleijn E, de Borst MH, Navis GJ, Rimbach G, Bakker SJ. Urinary Copper Excretion Is Associated with Long-Term Graft Failure in Kidney Transplant Recipients. Am J Nephrol 2023; 54:425-433. [PMID: 37231776 PMCID: PMC10687917 DOI: 10.1159/000531147] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023]
Abstract
INTRODUCTION In chronic kidney disease, proteinuria increases urinary copper excretion, inducing oxidative tubular damage and worsening kidney function. We investigated whether this phenomenon occurred in kidney transplant recipients (KTRs). In addition, we studied the associations of urinary copper excretion with the biomarker of oxidative tubular damage urinary liver-type fatty-acid binding protein (u-LFABP) and death-censored graft failure. METHODS This prospective cohort study was performed in the Netherlands between 2008 and 2017, including outpatient KTR with a functioning graft for longer than 1 year, who were extensively phenotyped at baseline. Twenty-four-hour urinary copper excretion was measured by inductively coupled plasma mass spectrometry. Multivariable linear and Cox regression analyses were performed. RESULTS In 693 KTR (57% men, 53 ± 13 years, estimated glomerular filtration rate [eGFR] 52 ± 20 mL/min/1.73 m2), baseline median urinary copper excretion was 23.6 (interquartile range 11.3-15.9) µg/24 h. Urinary protein excretion was positively associated with urinary copper excretion (standardized β = 0.39, p < 0.001), and urinary copper excretion was positively associated with u-LFABP (standardized β = 0.29, p < 0.001). During a median follow-up of 8 years, 109 (16%) KTR developed graft failure. KTR with relatively high copper excretion were at higher risk of long-term graft failure (hazard ratio [HR]: 1.57, 95% confidence interval [CI]: 1.32-1.86 per log2, p < 0.001), independent of multiple potential confounders like eGFR, urinary protein excretion, and time after transplantation. A dose-response relationship was observed over increasing tertiles of copper excretion (HR: 5.03, 95% CI: 2.75-9.19, tertile 3 vs. 1, p < 0.001). u-LFABP was a significant mediator of this association (74% of indirect effect, p < 0.001). CONCLUSION In KTR, urinary protein excretion is positively correlated with urinary copper excretion. In turn, higher urinary copper excretion is associated with an independent increased risk of kidney graft failure, with a substantial mediating effect through oxidative tubular damage. Further studies are warranted to investigate whether copper excretion-targeted interventions could improve kidney graft survival.
Collapse
Affiliation(s)
- Manuela Yepes-Calderón
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Camilo G. Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
- Clinical Hospital University of Chile, Independencia, Chile
| | - Ulrike Seidel
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Tim J. Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Kai Lüersen
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin H. de Borst
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan J. Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Stephan J.L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Trillos-Almanza MC, Wessel H, Martínez-Aguilar M, van den Berg EH, Douwes RM, Moshage H, Connelly MA, Bakker SJL, de Meijer VE, Dullaart RPF, Blokzijl H. Branched Chain Amino Acids Are Associated with Physical Performance in Patients with End-Stage Liver Disease. Biomolecules 2023; 13:biom13050824. [PMID: 37238694 DOI: 10.3390/biom13050824] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Decreased circulating branched chain amino acids (BCAA) represent a prominent change in amino acid profiles in patients with end-stage liver disease (ESLD). These alterations are considered to contribute to sarcopenia and hepatic encephalopathy and may relate to poor prognosis. Here, we cross-sectionally analyzed the association between plasma BCAA levels and the severity of ESLD and muscle function in participants of the liver transplant subgroup of TransplantLines, enrolled between January 2017 and January 2020. Plasma BCAA levels were measured by nuclear magnetic resonance spectroscopy. Physical performance was analyzed with a hand grip strength test, 4 m walking test, sit-to-stand test, timed up and go test, standing balance test and clinical frailty scale. We included 92 patients (65% men). The Child Pugh Turcotte classification was significantly higher in the lowest sex-stratified BCAA tertile compared to the highest tertile (p = 0.015). The times for the sit-to-stand (r = -0.352, p < 0.05) and timed up and go tests (r = -0.472, p < 0.01) were inversely correlated with total BCAA levels. In conclusion, lower circulating BCAA are associated with the severity of liver disease and impaired muscle function. This suggests that BCAA may represent a useful prognostic marker in the staging of liver disease severity.
Collapse
Affiliation(s)
- Maria Camila Trillos-Almanza
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Hanna Wessel
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Magnolia Martínez-Aguilar
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Eline H van den Berg
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Rianne M Douwes
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Han Moshage
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | | | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Vincent E de Meijer
- Department of Surgery, Division of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Internal Medicine, Division of Endocrinology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700RB Groningen, The Netherlands
| |
Collapse
|
30
|
de Mul N, Verlaan D, Ruurda JP, van Grevenstein WMU, Hagendoorn J, de Borst GJ, Vriens MR, de Bree R, Zweemer RP, Vogely C, Haitsma Mulier JLG, Vernooij LM, Reitsma JB, de Zoete MR, Top J, Kluijtmans JAJ, Hoefer IE, Noordzij P, Rettig T, Marsman M, de Smet AMGA, Derde L, van Waes J, Rijsdijk M, Schellekens WJM, Bonten MJM, Slooter AJC, Cremer OL. Cohort profile of PLUTO: a perioperative biobank focusing on prediction and early diagnosis of postoperative complications. BMJ Open 2023; 13:e068970. [PMID: 37076142 PMCID: PMC10124280 DOI: 10.1136/bmjopen-2022-068970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
PURPOSE Although elective surgery is generally safe, some procedures remain associated with an increased risk of complications. Improved preoperative risk stratification and earlier recognition of these complications may ameliorate postoperative recovery and improve long-term outcomes. The perioperative longitudinal study of complications and long-term outcomes (PLUTO) cohort aims to establish a comprehensive biorepository that will facilitate research in this field. In this profile paper, we will discuss its design rationale and opportunities for future studies. PARTICIPANTS Patients undergoing elective intermediate to high-risk non-cardiac surgery are eligible for enrolment. For the first seven postoperative days, participants are subjected to daily bedside visits by dedicated observers, who adjudicate clinical events and perform non-invasive physiological measurements (including handheld spirometry and single-channel electroencephalography). Blood samples and microbiome specimens are collected at preselected time points. Primary study outcomes are the postoperative occurrence of nosocomial infections, major adverse cardiac events, pulmonary complications, acute kidney injury and delirium/acute encephalopathy. Secondary outcomes include mortality and quality of life, as well as the long-term occurrence of psychopathology, cognitive dysfunction and chronic pain. FINDINGS TO DATE Enrolment of the first participant occurred early 2020. During the inception phase of the project (first 2 years), 431 patients were eligible of whom 297 patients consented to participate (69%). Observed event rate was 42% overall, with the most frequent complication being infection. FUTURE PLANS The main purpose of the PLUTO biorepository is to provide a framework for research in the field of perioperative medicine and anaesthesiology, by storing high-quality clinical data and biomaterials for future studies. In addition, PLUTO aims to establish a logistical platform for conducting embedded clinical trials. TRIAL REGISTRATION NUMBER NCT05331118.
Collapse
Affiliation(s)
- Nikki de Mul
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
- Julius Center, Department of Epidemiology, Program of Infectious Diseases, UMC Utrecht, Utrecht, The Netherlands
| | - Diede Verlaan
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
- Julius Center, Department of Epidemiology, Program of Infectious Diseases, UMC Utrecht, Utrecht, The Netherlands
| | - Jelle P Ruurda
- Department of Surgical Oncology, Upper Gastro-Intestinal Surgery, UMC Utrecht, Utrecht, The Netherlands
| | | | - Jeroen Hagendoorn
- Department of Surgical Oncology, Hepatobilliary and Pancreatic Surgery, UMC Utrecht, Utrecht, The Netherlands
| | - Gert-Jan de Borst
- Department of Vascular Surgery, UMC Utrecht, Utrecht, The Netherlands
| | - Menno R Vriens
- Department of Endocrine and Surgical Oncology, Cancer Center, UMC Utrecht, Utrecht, The Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, UMC Utrecht, Utrecht, The Netherlands
| | - Ronald P Zweemer
- Department of Gynaecological Oncology, UMC Utrecht, Utrecht, The Netherlands
| | - Charles Vogely
- Department of Orthopaedic Surgery, UMC Utrecht, Utrecht, The Netherlands
| | - Jelle L G Haitsma Mulier
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
- Julius Center, Department of Epidemiology, Program of Infectious Diseases, UMC Utrecht, Utrecht, The Netherlands
| | - Lisette M Vernooij
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
- Department of Anaesthesiology and Intensive Care, Antonius Ziekenhuis Nieuwegein, Nieuwegein, The Netherlands
| | - Johannes B Reitsma
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Marcel R de Zoete
- Department of Medical Microbiology, UMC Utrecht, Utrecht, The Netherlands
| | - Janetta Top
- Department of Medical Microbiology, UMC Utrecht, Utrecht, The Netherlands
| | - Jan A J Kluijtmans
- Department of Medical Microbiology, UMC Utrecht, Utrecht, The Netherlands
| | - Imo E Hoefer
- Central Diagnostic Laboratory, Universitair Medisch Centrum, Utrecht, The Netherlands
| | - Peter Noordzij
- Department of Anaesthesiology and Intensive Care, Antonius Ziekenhuis Nieuwegein, Nieuwegein, The Netherlands
| | - Thijs Rettig
- Department of Anesthesiology, Intensive Care and Pain Medicine, Amphia Hospital site Molengracht, Breda, The Netherlands
| | - Marije Marsman
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
| | | | - Lennie Derde
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Judith van Waes
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
| | - Mienke Rijsdijk
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
| | - Willem Jan M Schellekens
- Department of Anaesthesiology, UMC Utrecht, Utrecht, The Netherlands
- Department of Anaesthesiology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Marc J M Bonten
- Julius Center, Department of Epidemiology, Program of Infectious Diseases, UMC Utrecht, Utrecht, The Netherlands
| | - Arjen J C Slooter
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Olaf L Cremer
- Department of Intensive Care Medicine, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Westenberg LB, Zorgdrager M, Swaab TDA, van Londen M, Bakker SJL, Leuvenink HGD, Viddeleer AR, Pol RA. Reference values for low muscle mass and myosteatosis using tomographic muscle measurements in living kidney donors. Sci Rep 2023; 13:5835. [PMID: 37037940 PMCID: PMC10086018 DOI: 10.1038/s41598-023-33041-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/06/2023] [Indexed: 04/12/2023] Open
Abstract
Low muscle mass and myosteatosis are associated with poor clinical outcomes. Computed tomography (CT) imaging is an objective method for muscle mass and quality assessment; however consensus on cut-off values is lacking. This study assessed age-, sex-, and body mass index (BMI)-specific reference values of skeletal muscle parameters and correlated muscle mass with 24-h urinary creatinine excretion (24-h UCE). In total, 960 healthy subjects were included in this study. Muscle mass and quality were determined using axial CT slices at the vertebral level L3. The muscle area was indexed for height (skeletal muscle index [SMI]). The mean age was 53 ± 11 years, and 50% were male. The SMI reference values for low muscle mass in males were 38.8 cm2/m2 (20-29 years), 39.2 (30-39 years), 39.9 (40-49 years), 39.0 (50-59 years), 37.0 (60-69 years), and 36.8 (70-79 years). For females, these reference values were 37.5 cm2/m2 (20-29 years), 35.5 (30-39 years), 32.8 (40-49 years), 33.2 (50-59 years), 31.2 (60-69 years), and 31.5 (70-79 years). 24-h UCE and SMI were significantly correlated (r = 0.54, p < 0.001) without bias between the two methods of assessing muscle mass. This study provides age-, sex-, and BMI-specific reference values for skeletal muscle parameters that will support clinical decision making.
Collapse
Affiliation(s)
- Lisa B Westenberg
- Division of Transplant Surgery, Department of Surgery, University Medical Centre Groningen, University of Groningen, PO Box 30 001, 9700 RB, Groningen, The Netherlands.
| | - Marcel Zorgdrager
- Department of Radiology, Medical Imaging Center, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim D A Swaab
- Department of Radiology, Medical Imaging Center, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Marco van Londen
- Division of Nephrology, Department of Internal Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Henri G D Leuvenink
- Division of Transplant Surgery, Department of Surgery, University Medical Centre Groningen, University of Groningen, PO Box 30 001, 9700 RB, Groningen, The Netherlands
| | - Alain R Viddeleer
- Department of Radiology, Medical Imaging Center, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Robert A Pol
- Division of Transplant Surgery, Department of Surgery, University Medical Centre Groningen, University of Groningen, PO Box 30 001, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
32
|
Zijp TR, Knobbe TJ, van Hateren K, Roggeveld J, Blokzijl H, Tji Gan C, Jl Bakker S, Jongedijk EM, Investigators T, Touw DJ. Expeditious quantification of plasma tacrolimus with liquid chromatography tandem mass spectrometry in solid organ transplantation. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1222:123709. [PMID: 37060814 DOI: 10.1016/j.jchromb.2023.123709] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/17/2023]
Abstract
Traditionally, tacrolimus is assessed in whole blood samples, but this is suboptimal from the perspective that erythrocyte-bound tacrolimus is not a good representative of the active fraction. In this work, a straightforward and rapid method was developed for determination of plasma tacrolimus in solid organ transplant recipients, using liquid chromatography tandem mass spectrometry (LC-MS/MS) with heated electrospray ionisation. Sample preparation was performed through protein precipitation of 200 µl plasma with 500 µl stable isotopically labelled tacrolimus I.S. in methanol, where 20 µl was injected on the LC-MS/MS system. Separation was done using a chromatographic gradient on a C18 column (50 × 2.1 mm, 2.6 µm). The method was linear in the concentration range 0.05-5.00 µg/L, with within-run and between-run precision in the range 2-6 % and a run time of 1.5 min. Furthermore, the method was validated for selectivity, sensitivity, carry-over, accuracy and precision, process efficiency, recovery, matrix effect, and stability following EMA and FDA guidelines. Clinical validation was performed in 2333 samples from 1325 solid organ transplant recipients using tacrolimus (liver n = 312, kidney n = 1714, and lung n = 307), which had median plasma tacrolimus trough concentrations of 0.10 µg/L, 0.15 µg/L and 0.23 µg/L, respectively. This method is suitable for measurement of tacrolimus in plasma and will facilitate ongoing observational and prospective studies on the relationship of plasma tacrolimus concentrations with clinical outcomes.
Collapse
Affiliation(s)
- Tanja R Zijp
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, the Netherlands
| | - Tim J Knobbe
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, the Netherlands
| | - Kai van Hateren
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, the Netherlands
| | - Jan Roggeveld
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, the Netherlands
| | - Hans Blokzijl
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, the Netherlands
| | - C Tji Gan
- University of Groningen, University Medical Center Groningen, Department of Pulmonary Diseases and Tuberculosis, Groningen, the Netherlands
| | - Stephan Jl Bakker
- University of Groningen, University Medical Center Groningen, Department of Internal Medicine, Division of Nephrology, Groningen, the Netherlands
| | - Erwin M Jongedijk
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, the Netherlands
| | - TransplantLines Investigators
- University Medical Center Groningen Transplant Center, University Medical Center Groningen, Groningen, the Netherlands
| | - Daan J Touw
- University of Groningen, University Medical Center Groningen, Department of Clinical Pharmacy and Pharmacology, Groningen, the Netherlands; University of Groningen, Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, Groningen, the Netherlands.
| |
Collapse
|
33
|
Kremer D, Alkaff FF, Post A, Knobbe TJ, Tepel M, Thaunat O, Berger SP, van den Born J, Genovese F, Karsdal MA, Rasmussen DGK, Bakker SJL. Plasma endotrophin, reflecting tissue fibrosis, is associated with graft failure and mortality in KTRs: results from two prospective cohort studies. Nephrol Dial Transplant 2023; 38:1041-1052. [PMID: 36535643 PMCID: PMC10064980 DOI: 10.1093/ndt/gfac332] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Fibrosis is a suggested cause of graft failure and mortality among kidney transplant recipients (KTRs). Accumulating evidence suggests that collagen type VI is tightly linked to fibrosis and may be a marker of systemic fibrosis and ageing. We studied whether plasma endotrophin, a pro-collagen type VI fragment, is associated with graft failure and mortality among KTRs. METHODS In cohort A (57% male, age 53 ± 13 years), we measured plasma endotrophin in 690 prevalent KTRs ≥1 year after transplantation. The non-overlapping cohort B included 500 incident KTRs with serial endotrophin measurements before and after kidney transplantation to assess trajectories and intra-individual variation of endotrophin. RESULTS In cohort A, endotrophin was higher in KTRs compared with healthy controls. Concentrations were positively associated with female sex, diabetes, cardiovascular disease, markers of inflammation and kidney injury. Importantly, endotrophin was associated with graft failure {hazard ratio [HR] per doubling 1.87 [95% confidence interval (CI) 1.07-3.28]} and mortality [HR per doubling 2.59 (95% CI 1.73-3.87)] independent of potential confounders. Data from cohort B showed that endotrophin concentrations strongly decrease after transplantation and remain stable during post-transplantation follow-up [intra-individual coefficient of variation 5.0% (95% CI 3.7-7.6)]. CONCLUSIONS Plasma endotrophin is strongly associated with graft failure and mortality among KTRs. These findings suggest a key role of abnormal extracellular matrix turnover and fibrosis in graft and patient prognosis among KTRs and highlight the need for (interventional) studies targeting the profibrotic state of KTRs. The intra-individual stability after transplantation indicates potential use of endotrophin as a biomarker and outcome measure of fibrosis. TRIAL REGISTRATION ClinicalTrials.gov NCT02811835.
Collapse
Affiliation(s)
- Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Firas F Alkaff
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin Tepel
- Odense University Hospital, Department of Nephrology, Odense, Denmark
- Institute of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, Odense, Denmark
| | - Olivier Thaunat
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Transplantation, Néphrologie et Immunologie Clinique, Lyon, France
| | - Stefan P Berger
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
34
|
Riemersma NL, Kremer D, Knobbe TJ, Gan CT, Nolte S, Gomes-Neto AW, Blokzijl H, de Meijer VE, Damman K, Eisenga MF, Drost G, Elting JWJ, Touw DJ, Berger SP, Bakker SJL, van der Stouwe AMM. Tremor, Daily Functioning, and Health-Related Quality of Life in Solid Organ Transplant Recipients. Transpl Int 2023; 36:10951. [PMID: 37008718 PMCID: PMC10062599 DOI: 10.3389/ti.2023.10951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/23/2023] [Indexed: 04/04/2023]
Abstract
Solid organ transplant recipients (SOTR) frequently report tremor. Data concerning tremor-related impairment and its potential impact on health-related quality of life (HRQoL) are lacking. This cross-sectional study assesses impact of tremor on activities of daily living and HRQoL using validated questionnaires among SOTR enrolled in the TransplantLines Biobank and Cohort Study. We included 689 SOTR (38.5% female, mean [±SD] age 58 [±14] years) at median [interquartile range] 3 [1-9] years after transplantation, of which 287 (41.7%) reported mild or severe tremor. In multinomial logistic regression analyses, whole blood tacrolimus trough concentration was an independent determinant of mild tremor (OR per µg/L increase: 1.11, 95% CI: 1.02 to 1.21, p = 0.019). Furthermore, in linear regression analyses, severe tremor was strongly and independently associated with lower physical and mental HRQoL (β = -16.10, 95% CI: -22.23 to -9.98, p < 0.001 and β = -12.68, 95% CI: -18.23 to -7.14, p < 0.001 resp.). SOTR frequently report tremor-related impairment of activities of daily living. Tacrolimus trough concentrations appeared as a main determinant of tremor among SOTR. The strong and independent association of tremor-related impairment with lower HRQoL warrants further studies into the effects of tacrolimus on tremor. Clinical Trial Registration: ClinicalTrials.gov, Identifier NCT03272841.
Collapse
Affiliation(s)
- Niels L. Riemersma
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Tim J. Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - C. Tji Gan
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Pulmonary Diseases and Lung Transplantation, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Svea Nolte
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - António W. Gomes-Neto
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Vincent E. de Meijer
- Department of Hepato-Pancreato-Biliary Surgery and Liver Transplantation, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Kevin Damman
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Michele F. Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Gea Drost
- Department of Neurology and Clinical Neurophysiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jan Willem J. Elting
- Department of Neurology and Clinical Neurophysiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Daan J. Touw
- Department of Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, Netherlands
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Stephan J. L. Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - A. M. Madelein van der Stouwe
- Department of Neurology and Clinical Neurophysiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Expertise Center Movement Disorders Groningen, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
35
|
Knobbe TJ, Kremer D, Douwes RM, Eisenga MF, Gomes-Neto AW, Annema C, Swarte JC, Klont F, Navis G, Berger SP, Bakker SJL, Bodewes FAJA, de Boer MT, Damman K, de Borst MH, Diepstra A, Dijkstra G, Doorenbos CSE, Erasmus ME, Gan CT, Hak E, Hepkema BG, Leuvenink HGD, Lexmond WS, de Meijer VE, Niesters HGM, Pelt LJV, Pol RA, Porte RJ, Ranchor AV, Sanders JSF, Siebelink MJ, Slart RJHJA, Touw DJ, van den Heuvel MC, van Leer-Buter C, van Londen M, Verschuuren EAM, Vos MJ, Weersma RK. Proton Pump Inhibitor Use, Fatigue, and Health-Related Quality of Life in Kidney Transplant Recipients: Results From the TransplantLines Biobank and Cohort Study. Am J Kidney Dis 2023:S0272-6386(23)00532-2. [PMID: 36801431 DOI: 10.1053/j.ajkd.2022.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/21/2022] [Indexed: 02/19/2023]
Abstract
RATIONALE & OBJECTIVE Prior studies report that the use of proton pump inhibitors (PPIs) can adversely affect gut microbiota and gastrointestinal uptake of micronutrients, in particular iron and magnesium, and are used frequently by kidney transplant recipients. Altered gut microbiota, iron deficiency, and magnesium deficiency have been implicated in the pathogenesis of chronic fatigue. Therefore, we hypothesized that PPI use may be an important and underappreciated cause of fatigue and reduced health-related quality of life (HRQoL) in this population. STUDY DESIGN Cross-sectional study. SETTING & PARTICIPANTS Kidney transplant recipients (≥1 year after transplantation) enrolled in the TransplantLines Biobank and Cohort Study. EXPOSURE PPI use, PPI type, PPI dosage, and duration of PPI use. OUTCOME Fatigue and HRQoL, assessed using the validated Checklist Individual Strength 20 Revised questionnaire and Short Form-36 questionnaire. ANALYTICAL APPROACH Logistic and linear regression. RESULTS We included 937 kidney transplant recipients (mean age 56±13 years, 39% female) at a median of 3 (1-10) years after transplantation. PPI use was associated with fatigue severity (regression coefficient 4.02, 95% CI, 2.18 to 5.85, P<0.001), a higher risk of severe fatigue (OR 2.05, 95% CI, 1.48 to 2.84, P<0.001), lower physical HRQoL (regression coefficient-8.54, 95% CI, -11.54 to-5.54, P<0.001), and lower mental HRQoL (regression coefficient-4.66, 95% CI, -7.15 to-2.17, P<0.001). These associations were independent of potential confounders including age, time since transplantation, history of upper gastrointestinal disease, antiplatelet therapy, and the total number of medications. They were present among all individually assessed PPI types and were dose dependent. Duration of PPI exposure was only associated with fatigue severity. LIMITATIONS Residual confounding and inability to assess causal relationships. CONCLUSIONS PPI use is independently associated with fatigue and lower HRQoL among kidney transplant recipients. PPI use might be an easily accessible target for alleviating fatigue and improving HRQoL among kidney transplant recipients. Further studies examining the effect of PPI exposure in this population are warranted.
Collapse
Affiliation(s)
- Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen.
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Rianne M Douwes
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Michele F Eisenga
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - António W Gomes-Neto
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Coby Annema
- Department of Health Sciences, Section of Nursing Research, University Medical Center Groningen
| | - J Casper Swarte
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Frank Klont
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen; Unit of PharmacoTherapy, -Epidemiology and -Economics, Groningen Research Institute of Pharmacy, University of Groningen, Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Stefan P Berger
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
van den Berg EH, Flores-Guerrero JL, Garcia E, Connelly MA, de Meijer VE, Bakker SJL, Blokzijl H, Dullaart RPF. High plasma levels of betaine, a trimethylamine N-Oxide-related metabolite, are associated with the severity of cirrhosis. Liver Int 2023; 43:424-433. [PMID: 35585781 PMCID: PMC10084422 DOI: 10.1111/liv.15310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/09/2022] [Accepted: 05/16/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND AND AIMS The gut microbiome-related metabolites betaine and trimethylamine N-oxide (TMAO) affect major health issues. In cirrhosis, betaine metabolism may be diminished because of impaired hepatic betaine homocysteine methyltransferase activity, whereas TMAO generation from trimethylamine may be altered because of impaired hepatic flavin monooxygenase expression. Here, we determined plasma betaine and TMAO levels in patients with end-stage liver disease and assessed their relationships with liver disease severity. METHODS Plasma betaine and TMAO concentrations were measured by nuclear magnetic resonance spectroscopy in 129 cirrhotic patients (TransplantLines cohort study; NCT03272841) and compared with levels from 4837 participants of the PREVEND cohort study. Disease severity was assessed by Child-Pugh-Turcotte (CPT) classification and Model for End-stage Liver Disease (MELD) score. RESULTS Plasma betaine was on average 60% higher (p < .001), whereas TMAO was not significantly lower in cirrhotic patients vs. PREVEND population (p = .44). After liver transplantation (n = 13), betaine decreased (p = .017; p = .36 vs. PREVEND population), whereas TMAO levels tended to increase (p = .085) to higher levels than in the PREVEND population (p = .003). Betaine levels were positively associated with the CPT stage and MELD score (both p < .001). The association with the MELD score remained in the fully adjusted analysis (p < .001). The association of TMAO with the MELD score did not reach significance (p = .11). Neither betaine nor TMAO levels were associated with mortality on the waiting list for liver transplantation (adjusted p = .78 and p = .44, respectively). CONCLUSION Plasma betaine levels are elevated in cirrhotic patients in parallel with disease severity and decrease after liver transplantation.
Collapse
Affiliation(s)
- Eline H van den Berg
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jose L Flores-Guerrero
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Erwin Garcia
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, North Carolina, USA
| | - Vincent E de Meijer
- Department of Hepatopancreatobiliary Surgery and Liver Transplantation, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | - Stephan J L Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
37
|
Eelderink C, Kremer D, Riphagen IJ, Knobbe TJ, Schurgers LJ, Pasch A, Mulder DJ, Corpeleijn E, Navis G, Bakker SJL, de Borst MH, Te Velde-Keyzer CA. Effect of vitamin K supplementation on serum calcification propensity and arterial stiffness in vitamin K-deficient kidney transplant recipients: A double-blind, randomized, placebo-controlled clinical trial. Am J Transplant 2023; 23:520-530. [PMID: 36695702 DOI: 10.1016/j.ajt.2022.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/21/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023]
Abstract
Vitamin K deficiency is common among kidney transplant recipients (KTRs) and likely contributes to progressive vascular calcification and stiffness. In this single-center, randomized, double-blind, placebo-controlled trial, we aimed to investigate the effects of vitamin K supplementation on the primary end point, serum calcification propensity (calciprotein particle maturation time, T50), and secondary end points arterial stiffness (pulse wave velocity [PWV]) and vitamin K status in 40 vitamin K-deficient KTRs (plasma dephosphorylated uncarboxylated matrix Gla protein [dp-ucMGP] ≥500 pmol/L). Participants (35% female; age, 57 ± 13 years) were randomized 1:1 to vitamin K2 (menaquinone-7, 360 μg/day) or placebo for 12 weeks. Vitamin K supplementation had no effect on calcification propensity (change in T50 vs baseline +2.3 ± 27.4 minutes) compared with placebo (+0.8 ± 34.4 minutes; Pbetween group = .88) but prevented progression of PWV (change vs baseline -0.06 ± 0.26 m/s) compared with placebo (+0.27 ± 0.43 m/s; Pbetween group = .010). Vitamin K supplementation strongly improved vitamin K status (change in dp-ucMGP vs baseline -385 [-631 to -269] pmol/L) compared with placebo (+39 [-188 to +183] pmol/L; Pbetween group < .001), although most patients remained vitamin K-deficient. In conclusion, vitamin K supplementation did not alter serum calcification propensity but prevented progression of arterial stiffness, suggesting that vitamin K has vascular effects independent of calciprotein particles. These results set the stage for longer-term intervention studies with vitamin K supplementation in KTRs. TRIAL REGISTRY: EU Clinical Trials Register (EudraCT Number: 2019-004906-88) and the Dutch Trial Register (NTR number: NL7687).
Collapse
Affiliation(s)
- Coby Eelderink
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands.
| | - Ineke J Riphagen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Leon J Schurgers
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, Maastricht, the Netherlands
| | - Andreas Pasch
- Calciscon AG, Biel, Switzerland; Department of Physiology and Pathophysiology, Johannes Kepler University Linz, Linz, Austria
| | - D J Mulder
- Department of Internal Medicine, Division of Vascular Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Martin H de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| | - Charlotte A Te Velde-Keyzer
- Department of Internal Medicine, Division of Nephrology, University of Groningen and University Medical Center, Groningen, Groningen, the Netherlands
| |
Collapse
|
38
|
van Vliet IM, Post A, Kremer D, Boslooper‐Meulenbelt K, van der Veen Y, de Jong MF, Pol RA, Jager‐Wittenaar H, Navis GJ, Bakker SJ. Muscle mass, muscle strength and mortality in kidney transplant recipients: results of the TransplantLines Biobank and Cohort Study. J Cachexia Sarcopenia Muscle 2022; 13:2932-2943. [PMID: 36891995 PMCID: PMC9745460 DOI: 10.1002/jcsm.13070] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/10/2022] [Accepted: 07/23/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Survival of kidney transplant recipients (KTR) is low compared with the general population. Low muscle mass and muscle strength may contribute to lower survival, but practical measures of muscle status suitable for routine care have not been evaluated for their association with long-term survival and their relation with each other in a large cohort of KTR. METHODS Data of outpatient KTR ≥ 1 year post-transplantation, included in the TransplantLines Biobank and Cohort Study (ClinicalTrials.gov Identifier: NCT03272841), were used. Muscle mass was determined as appendicular skeletal muscle mass indexed for height2 (ASMI) through bio-electrical impedance analysis (BIA), and by 24-h urinary creatinine excretion rate indexed for height2 (CERI). Muscle strength was determined by hand grip strength indexed for height2 (HGSI). Secondary analyses were performed using parameters not indexed for height2. Cox proportional hazards models were used to investigate the associations between muscle mass and muscle strength and all-cause mortality, both in univariable and multivariable models with adjustment for potential confounders, including age, sex, body mass index (BMI), estimated glomerular filtration rate (eGFR) and proteinuria. RESULTS We included 741 KTR (62% male, age 55 ± 13 years, BMI 27.3 ± 4.6 kg/m2), of which 62 (8%) died during a median [interquartile range] follow-up of 3.0 [2.3-5.7] years. Compared with patients who survived, patients who died had similar ASMI (7.0 ± 1.0 vs. 7.0 ± 1.0 kg/m2; P = 0.57), lower CERI (4.2 ± 1.1 vs. 3.5 ± 0.9 mmol/24 h/m2; P < 0.001) and lower HGSI (12.6 ± 3.3 vs. 10.4 ± 2.8 kg/m2; P < 0.001). We observed no association between ASMI and all-cause mortality (HR 0.93 per SD increase; 95% confidence interval [CI] [0.72, 1.19]; P = 0.54), whereas CERI and HGSI were significantly associated with mortality, independent of potential confounders (HR 0.57 per SD increase; 95% CI [0.44, 0.81]; P = 0.002 and HR 0.47 per SD increase; 95% CI [0.33, 0.68]; P < 0.001, respectively), and associations of CERI and HGSI with mortality remained independent of each other (HR 0.68 per SD increase; 95% CI [0.47, 0.98]; P = 0.04 and HR 0.53 per SD increase; 95% CI [0.36, 0.76]; P = 0.001, respectively). Similar associations were found for unindexed parameters. CONCLUSIONS Higher muscle mass assessed by creatinine excretion rate and higher muscle strength assessed by hand grip strength are complementary in their association with lower risk of all-cause mortality in KTR. Muscle mass assessed by BIA is not associated with mortality. Routine assessment using both 24-h urine samples and hand grip strength is recommended, to potentially target interdisciplinary interventions for KTR at risk for poor survival to improve muscle status.
Collapse
Affiliation(s)
- Iris M.Y. van Vliet
- Department of DieteticsUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Adrian Post
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Karin Boslooper‐Meulenbelt
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Yvonne van der Veen
- Department of DieteticsUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Margriet F.C. de Jong
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Robert A. Pol
- Department of SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | | | - Harriët Jager‐Wittenaar
- Department of Oral and Maxillofacial SurgeryUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Research Group Healthy Ageing, Health Care and NursingHanze University of Applied SciencesGroningenThe Netherlands
| | - Gerjan J. Navis
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Stephan J.L. Bakker
- Department of Internal Medicine, Division of NephrologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
39
|
Visser K, van der Horn HJ, Bourgonje AR, Jacobs B, de Borst MH, Vos PE, Bulthuis MLC, van Goor H, van der Naalt J. Acute serum free thiols: a potentially modifiable biomarker of oxidative stress following traumatic brain injury. J Neurol 2022; 269:5883-5892. [PMID: 35776194 PMCID: PMC9553822 DOI: 10.1007/s00415-022-11240-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/16/2022] [Accepted: 06/17/2022] [Indexed: 11/01/2022]
Abstract
Serum concentrations of free thiols (key components of the extracellular antioxidant machinery) reflect the overall redox status of the human body. The objective of this exploratory study was to determine the concentrations of serum free thiols in the acute phase after traumatic brain injury (TBI) and their association with long-term outcome. In this observational cohort study, patients with TBI of various severity were included from a biobank of prospectively enrolled TBI patients. Further eligibility criteria included an available blood sample and head computed tomography data, obtained within 24 h of injury, as well as a functional outcome assessment (Glasgow Outcome Scale Extended (GOSE)) at 6 months post-injury. Serum free thiol concentrations were markedly lower in patients with TBI (n = 77) compared to healthy controls (n = 55) (mean ± standard deviation; 210.3 ± 63.3 vs. 301.8 ± 23.9 μM, P < 0.001) indicating increased oxidative stress. Concentrations of serum free thiols were higher in patients with complete functional recovery (GOSE = 8) than in patients with incomplete recovery (GOSE < 8) (median [interquartile range]; 235.7 [205.1-271.9] vs. 205.2 [173-226.7] μM, P = 0.016), suggesting that patients with good recovery experience less oxidative stress in the acute phase after TBI or have better redox function. Acute TBI is accompanied by a markedly lower concentration of serum free thiols compared to healthy controls indicating that serum free thiols may be a novel biomarker of TBI. Future studies are warranted to validate our findings and explore the clinical applicability and prognostic capability of this candidate-biomarker.
Collapse
Affiliation(s)
- Koen Visser
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Harm Jan van der Horn
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Arno R. Bourgonje
- Department of Gastroenterology and Hepatology, University of Groninger, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Bram Jacobs
- Department of Neurology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Martin H. de Borst
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Pieter E. Vos
- Department of Neurology, Slingeland Hospital, 7009 BL Doetinchem, The Netherlands
| | - Marian L. C. Bulthuis
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - Joukje van der Naalt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
40
|
Knobbe TJ, Kremer D, Abma FI, Annema C, Berger SP, Navis GJ, van der Mei SF, Bültmann U, Visser A, Bakker SJ. Employment Status and Work Functioning among Kidney Transplant Recipients. Clin J Am Soc Nephrol 2022; 17:1506-1514. [PMID: 36162849 PMCID: PMC9528259 DOI: 10.2215/cjn.05560522] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/18/2022] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND OBJECTIVES To date, employment figures of kidney transplant recipients in Europe are inconsistent. Additionally, little is known about work functioning of employed kidney transplant recipients and work functioning trajectories before and after transplantation. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Data from the ongoing TransplantLines Biobank and Cohort study and from community-dwelling employed adults were used. Health-related work functioning of kidney transplant recipients was assessed with the Work Role Functioning Questionnaire 2.0 and compared with potential kidney donors and community-dwelling employed adults. RESULTS We included 668 kidney transplant recipients of working age (59% men, age 51±11 years) at median 3 (interquartile range, 2-10) years after transplantation, 246 potential kidney donors of working age (43% men, age 53±9 years), and 553 community-dwelling employed adults (70% men, age 45±11 years). The proportion of employed kidney transplant recipients was lower compared with potential kidney donors (56% versus 79%). If employed, the work functioning score of kidney transplant recipients was slightly lower compared with employed potential kidney donors yet higher compared with community-dwelling employed adults (medians 91 [interquartile range, 76-98], 94 [interquartile range, 85-99], and 88 [interquartile range, 79-95], respectively). Backward linear regression analyses revealed that lower educational level, having a kidney from a deceased donor, presence of tingling or numbness of hands or feet, presence of concentration/memory problems, presence of anxiety, and presence of severe fatigue were independently associated with lower work functioning among kidney transplant recipients. Additional subgroup analyses showed that work functioning scores were lower before transplantation than at 12 months after transplantation (83 [interquartile range, 66-93] versus 92 [interquartile range, 88-98], respectively; P=0.002). CONCLUSIONS Stable employed kidney transplant recipients report to function well at work. In addition, this study shows that self-reported work functioning is higher after successful kidney transplantation compared with before transplantation. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER TransplantLines Biobank and Cohort study, NCT03272841 PODCAST: This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2022_09_26_CJN05560522.mp3.
Collapse
Affiliation(s)
- Tim J. Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Femke I. Abma
- Department of Health Sciences, Community and Occupational Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Coby Annema
- Department of Health Sciences, Section of Nursing Science, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P. Berger
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerjan J. Navis
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sijrike F. van der Mei
- Department of Health Sciences, Applied Health Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ute Bültmann
- Department of Health Sciences, Community and Occupational Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Annemieke Visser
- Department of Health Sciences, Applied Health Research, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J.L. Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- University Medical Center Groningen Transplant Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
41
|
Knobbe TJ, Kremer D, Eisenga MF, Corpeleijn E, Annema C, Spikman JM, Navis G, Berger SP, Bakker SJL. Hand dexterity, daily functioning and health-related quality of life in kidney transplant recipients. Sci Rep 2022; 12:16208. [PMID: 36171358 PMCID: PMC9519570 DOI: 10.1038/s41598-022-19952-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Impaired interplay between sensory and motor function may be an important, often overlooked cause of the decreased daily functioning and impaired health-related quality of life (HRQoL) of kidney transplant recipients (KTR). We assessed this interplay using a hand dexterity test, and investigated its potential associations with daily functioning and HRQoL among KTR enrolled at the TransplantLines Biobank and Cohort Study. A total of 309 KTR (58% male, mean age 56 ± 13 years) at median 4 [IQR: 1-11] years after transplantation were included. Impaired hand dexterity, as defined by a test performance slower than the 95th percentile of an age- and sex-specific reference population, was observed in 71 (23%) KTR. Worse hand dexterity was independently associated with worse performance on almost all measures of physical capacity, activities of daily living and societal participation. Finally, hand dexterity was independently associated with physical HRQoL (standardized beta - 0.22, 95%CI - 0.34 to - 0.09, P < 0.001). In conclusion, impaired interplay between sensory and motor function, as assessed by hand dexterity, is prevalent among KTR. In addition, poor hand dexterity was associated with impaired daily functioning and limited physical HRQoL. Impaired interplay between sensory and motor function may be therefore an important, hitherto overlooked, phenomenon in KTR.
Collapse
Affiliation(s)
- Tim J Knobbe
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands.
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Hanzeplein 1, P.O. Box 30.001, 9713 GZ, Groningen, The Netherlands.
| | - Daan Kremer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Eva Corpeleijn
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Coby Annema
- Department of Health Sciences, Section of Nursing Science, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Joke M Spikman
- Department of Neurology, Division of Neuropsychology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
42
|
Swarte JC, Li Y, Hu S, Björk JR, Gacesa R, Vich Vila A, Douwes RM, Collij V, Kurilshikov A, Post A, Klaassen MAY, Eisenga MF, Gomes-Neto AW, Kremer D, Jansen BH, Knobbe TJ, Berger SP, Sanders JSF, Heiner-Fokkema MR, Porte RJ, Cuperus FJC, de Meijer VE, Wijmenga C, Festen EAM, Zhernakova A, Fu J, Harmsen HJM, Blokzijl H, Bakker SJL, Weersma RK. Gut microbiome dysbiosis is associated with increased mortality after solid organ transplantation. Sci Transl Med 2022; 14:eabn7566. [PMID: 36044594 DOI: 10.1126/scitranslmed.abn7566] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Organ transplantation is a life-saving treatment for patients with end-stage disease, but survival rates after transplantation vary considerably. There is now increasing evidence that the gut microbiome is linked to the survival of patients undergoing hematopoietic cell transplant, yet little is known about the role of the gut microbiome in solid organ transplantation. We analyzed 1370 fecal samples from 415 liver and 672 renal transplant recipients using shotgun metagenomic sequencing to assess microbial taxonomy, metabolic pathways, antibiotic resistance genes, and virulence factors. To quantify taxonomic and metabolic dysbiosis, we also analyzed 1183 age-, sex-, and body mass index-matched controls from the same population. In addition, a subset of 78 renal transplant recipients was followed longitudinally from pretransplantation to 24 months after transplantation. Our data showed that both liver and kidney transplant recipients suffered from gut dysbiosis, including lower microbial diversity, increased abundance of unhealthy microbial species, decreased abundance of important metabolic pathways, and increased prevalence and diversity of antibiotic resistance genes and virulence factors. These changes were found to persist up to 20 years after transplantation. Last, we demonstrated that the use of immunosuppressive drugs was associated with the observed dysbiosis and that the extent of dysbiosis was associated with increased mortality after transplantation. This study represents a step toward potential microbiome-targeted interventions that might influence the outcomes of recipients of solid organ transplantation.
Collapse
Affiliation(s)
- J Casper Swarte
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Yanni Li
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Shixian Hu
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Johannes R Björk
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Ranko Gacesa
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Arnau Vich Vila
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Rianne M Douwes
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Valerie Collij
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Adrian Post
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Marjolein A Y Klaassen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - António W Gomes-Neto
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Bernadien H Jansen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Stefan P Berger
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Jan-Stephan F Sanders
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - M Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Robert J Porte
- Department of Surgery, Section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Frans J C Cuperus
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Vincent E de Meijer
- Department of Surgery, Section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Cisca Wijmenga
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Eleonora A M Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands.,Department of Pediatrics, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9713 GZ, Groningen, Netherlands
| |
Collapse
|
43
|
Vinke JSJ, Wouters HJCM, Stam SP, Douwes RM, Post A, Gomes-Neto AW, van der Klauw MM, Berger SP, Bakker SJL, De Borst MH, Eisenga MF. Decreased haemoglobin levels are associated with lower muscle mass and strength in kidney transplant recipients. J Cachexia Sarcopenia Muscle 2022; 13:2044-2053. [PMID: 35666066 PMCID: PMC9397498 DOI: 10.1002/jcsm.12999] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 02/18/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Post-transplant anaemia and reduced muscle mass and strength are highly prevalent in kidney transplant recipients (KTRs). Decreased haemoglobin levels, a marker of anaemia, could adversely affect muscle mass and strength through multiple mechanisms, among others, through diminished tissue oxygenation. We aimed to investigate the association between haemoglobin levels with muscle mass and strength in KTRs. METHODS We included stable KTRs from the TransplantLines Biobank and Cohort study with a functional graft ≥1 year post-transplantation. Muscle mass was assessed using 24 h urinary creatinine excretion rate (CER) and bioelectrical impedance analysis (BIA). Muscle strength was assessed with a handgrip strength test using a dynamometer and, in a subgroup (n = 290), with the five-times sit-to-stand (FTSTS) test. We used multivariable linear and logistic regression analyses to investigate the associations of haemoglobin levels with muscle mass and strength. RESULTS In 871 included KTRs [median age 58 (interquartile range (IQR), 48-66)] years; 60% men; eGFR 51 ± 18 mL/min/1.73 m2 ) who were 3.5 (1.0-10.2) years post-transplantation, the mean serum haemoglobin level was 13.9 ± 1.8 g/dL in men and 12.8 ± 1.5 g/dL in women. Lower haemoglobin levels were independently associated with a lower CER (std. β = 0.07, P = 0.01), BIA-derived skeletal muscle mass (std. β = 0.22, P < 0.001), handgrip strength (std. β = 0.15, P < 0.001), and worse FTSTS test scores (std. β = -0.17, P = 0.02). KTRs in the lowest age-specific and sex-specific quartile of haemoglobin levels had an increased risk of being in the worst age-specific and sex-specific quartile of CER (fully adjusted OR, 2.09; 95% CI 1.15-3.77; P = 0.02), handgrip strength (fully adjusted OR, 3.30; 95% CI 1.95-5.59; P < 0.001), and FTSTS test score (fully adjusted OR, 7.21; 95% CI 2.59-20.05; P < 0.001). CONCLUSIONS Low haemoglobin levels are strongly associated with decreased muscle mass and strength in KTRs. Future investigation will need to investigate whether maintaining higher haemoglobin levels may improve muscle mass and strength in KTRs.
Collapse
Affiliation(s)
- Joanna Sophia J Vinke
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Hanneke J C M Wouters
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Suzanne P Stam
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Rianne M Douwes
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Adrian Post
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Antonio W Gomes-Neto
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Melanie M van der Klauw
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stefan P Berger
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | -
- Groningen Transplant Center, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Martin H De Borst
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
44
|
Li Y, Nieuwenhuis LM, Keating BJ, Festen EA, de Meijer VE. The Impact of Donor and Recipient Genetic Variation on Outcomes After Solid Organ Transplantation: A Scoping Review and Future Perspectives. Transplantation 2022; 106:1548-1557. [PMID: 34974452 PMCID: PMC9311456 DOI: 10.1097/tp.0000000000004042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/16/2021] [Accepted: 11/25/2021] [Indexed: 11/25/2022]
Abstract
At the outset of solid organ transplantation, genetic variation between donors and recipients was recognized as a major player in mechanisms such as allograft tolerance and rejection. Genome-wide association studies have been very successful in identifying novel variant-trait associations, but have been difficult to perform in the field of solid organ transplantation due to complex covariates, era effects, and poor statistical power for detecting donor-recipient interactions. To overcome a lack of statistical power, consortia such as the International Genetics and Translational Research in Transplantation Network have been established. Studies have focused on the consequences of genetic dissimilarities between donors and recipients and have reported associations between polymorphisms in candidate genes or their regulatory regions with transplantation outcomes. However, knowledge on the exact influence of genetic variation is limited due to a lack of comprehensive characterization and harmonization of recipients' or donors' phenotypes and validation using an experimental approach. Causal research in genetics has evolved from agnostic discovery in genome-wide association studies to functional annotation and clarification of underlying molecular mechanisms in translational studies. In this overview, we summarize how the recent advances and progresses in the field of genetics and genomics have improved the understanding of outcomes after solid organ transplantation.
Collapse
Affiliation(s)
- Yanni Li
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Lianne M. Nieuwenhuis
- Department of Surgery, section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Brendan J. Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Eleonora A.M. Festen
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent E. de Meijer
- Department of Surgery, section of Hepatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
45
|
Sotomayor CG, Giubergia F, Groothof D, Ferreccio C, Nolte IM, Navis GJ, Gomes-Neto AW, Kremer D, Knobbe TJ, Eisenga MF, Rodrigo R, Touw DJ, Bakker SJL. Plasma Lead Concentration and Risk of Late Kidney Allograft Failure: Findings From the TransplantLines Biobank and Cohort Studies. Am J Kidney Dis 2022; 80:87-97.e1. [PMID: 34871698 DOI: 10.1053/j.ajkd.2021.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 10/15/2021] [Indexed: 11/11/2022]
Abstract
RATIONALE & OBJECTIVE Heavy metals are known to induce kidney damage, and recent studies have linked minor exposures to cadmium and arsenic with increased risk of kidney allograft failure, yet the potential association of lead with late graft failure in kidney transplant recipients (KTRs) remains unknown. STUDY DESIGN Prospective cohort study in The Netherlands. SETTING & PARTICIPANTS We studied outpatient KTRs (n = 670) with a functioning graft for ≥1 year recruited at a university setting (2008-2011) and followed for a median of 4.9 (interquartile range, 3.4-5.5) years. Additionally, patients with chronic kidney disease (n = 46) enrolled in the ongoing TransplantLines Cohort and Biobank Study (2016-2017, ClinicalTrials.gov identifier NCT03272841) were studied at admission for transplant and at 3, 6, 12, and 24 months after transplant. EXPOSURE Plasma lead concentration was log2-transformed to estimate the association with outcomes per doubling of plasma lead concentration and also considered categorically as tertiles of lead distribution. OUTCOME Kidney graft failure (restart of dialysis or repeat transplant) with the competing event of death with a functioning graft. ANALYTICAL APPROACH Multivariable-adjusted cause-specific hazards models in which follow-up of KTRs who died with a functioning graft was censored. RESULTS Median baseline plasma lead concentration was 0.31 (interquartile range, 0.22-0.45) μg/L among all KTRs. During follow-up, 78 (12%) KTRs experienced graft failure. Higher plasma lead concentration was associated with increased risk of graft failure (hazard ratio, 1.59 [95% CI, 1.14-2.21] per doubling; P = 0.006) independent of age, sex, transplant characteristics, estimated glomerular filtration rate, proteinuria, smoking status, alcohol intake, and plasma concentrations of cadmium and arsenic. These findings remained materially unchanged after additional adjustment for dietary intake and were consistent with those of analyses examining lead categorically. In serial measurements, plasma lead concentration was significantly higher at admission for transplant than at 3 months after transplant (P = 0.001), after which it remained stable over 2 years of follow-up (P = 0.2). LIMITATIONS Observational study design. CONCLUSIONS Pretransplant plasma lead concentrations, which decrease after transplant, are associated with increased risk of late kidney allograft failure. These findings warrant further studies to evaluate whether preventive or therapeutic interventions to decrease plasma lead concentration may represent novel risk-management strategies to decrease the rate of kidney allograft failure.
Collapse
Affiliation(s)
- Camilo G Sotomayor
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands; Program of Integrative Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile; Radiology Department, Clinical Hospital University of Chile, University of Chile, Santiago, Chile.
| | - Flavia Giubergia
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Dion Groothof
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Catterina Ferreccio
- Advanced Center for Chronic Diseases, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ilja M Nolte
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerjan J Navis
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Antonio W Gomes-Neto
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Daan Kremer
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Tim J Knobbe
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Michele F Eisenga
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Daan J Touw
- Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Stephan J L Bakker
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | |
Collapse
|
46
|
Stam SP, Vulto A, Vos MJ, Kerstens MN, Rutgers A, Kema I, Touw DJ, Bakker SJ, van Beek AP. Rationale and design of the CORE (COrticosteroids REvised) study: protocol. BMJ Open 2022; 12:e061678. [PMID: 35473729 PMCID: PMC9045047 DOI: 10.1136/bmjopen-2022-061678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Corticosteroids are an important pillar in many anti-inflammatory and immunosuppressive treatment regimens and are available in natural and synthetic forms, which are considered equipotent if clinical bioequivalence data are used. Current clinical bioequivalence data are however based on animal studies or studies with subjective endpoints. Furthermore, advancement in steroid physiology with regard to metabolism, intracellular handling and receptor activation have not yet been incorporated. Therefore, this study aims to re-examine the clinical bioequivalence and dose effects of the most widely used synthetic corticosteroids, prednisolone and dexamethasone. METHODS AND ANALYSIS In this double-blind, randomised cross-over clinical trial, 24 healthy male and female volunteers aged 18-75 years, will be included. All volunteers will randomly receive either first a daily dose of 7.5 mg prednisolone for 1 week, immediately followed by a daily dose of 30 mg prednisolone for 1 week, or first a presumed clinical bioequivalent dose of 1.125 mg dexamethasone per day, immediately followed by 4.5 mg of dexamethasone per day for 1 week. After a wash-out period of 4-8 weeks, the other treatment will be applied. The primary study endpoint is the difference in free cortisol excretion in 24 hours urine. Secondary endpoints will include differences in immunological parameters, blood pressure and metabolic measurements. ETHICS AND DISSEMINATION This study has been approved by the Medical Ethics Committee of the University Medical Center Groningen (METC 2020.398). The results of this study will be submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER ClinicalTrials.gov (Identifier: NCT04733144), and in the Dutch trial registry (NL9138).
Collapse
Affiliation(s)
- Suzanne P Stam
- Internal Medicine, Division of Nephrology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Annet Vulto
- Internal Medicine, Division of Endocrinology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Michel J Vos
- Laboratory Medicine, University Medical Centre Groningen, Groningen, The Netherlands
| | - Michiel N Kerstens
- Internal Medicine, Division of Endocrinology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Abraham Rutgers
- Rheumatology and Clinical Immunology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ido Kema
- Laboratory Medicine, University Medical Centre Groningen, Groningen, The Netherlands
| | - Daan J Touw
- Clincal Pharmacy and Pharmacology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stephan Jl Bakker
- Internal Medicine, Division of Nephrology, University Medical Centre Groningen, Groningen, The Netherlands
| | - André P van Beek
- Internal Medicine, Division of Endocrinology, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
47
|
Tiller G, Lammerts RGM, Karijosemito JJ, Alkaff FF, Diepstra A, Pol RA, Meter-Arkema AH, Seelen MA, van den Heuvel MC, Hepkema BG, Daha MR, van den Born J, Berger SP. Weak Expression of Terminal Complement in Active Antibody-Mediated Rejection of the Kidney. Front Immunol 2022; 13:845301. [PMID: 35493506 PMCID: PMC9044906 DOI: 10.3389/fimmu.2022.845301] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe role of the complement system in antibody-mediated rejection (ABMR) is insufficiently understood. We aimed to investigate the role of local and systemic complement activation in active (aABMR). We quantified complement activation markers, C3, C3d, and C5b-9 in plasma of aABMR, and acute T-cell mediated rejection (aTCMR), and non-rejection kidney transplant recipients. Intra-renal complement markers were analyzed as C4d, C3d, C5b-9, and CD59 deposition. We examined in vitro complement activation and CD59 expression on renal endothelial cells upon incubation with human leukocyte antigen antibodies.MethodsWe included 50 kidney transplant recipients, who we histopathologically classified as aABMR (n=17), aTCMR (n=18), and non-rejection patients (n=15).ResultsComplement activation in plasma did not differ across groups. C3d and C4d deposition were discriminative for aABMR diagnosis. Particularly, C3d deposition was stronger in glomerular (P<0,01), and peritubular capillaries (P<0,05) comparing aABMR to aTCMR rejection and non-rejection biopsies. In contrast to C3d, C5b-9 was only mildly expressed across all groups. For C5b-9, no significant difference between aABMR and non-rejection biopsies regarding peritubular and glomerular C5b-9 deposition was evident. We replicated these findings in vitro using renal endothelial cells and found complement pathway activation with C4d and C3d, but without terminal C5b-9 deposition. Complement regulator CD59 was variably present in biopsies and constitutively expressed on renal endothelial cells in vitro.ConclusionOur results indicate that terminal complement might only play a minor role in late aABMR, possibly indicating the need to re-evaluate the applicability of terminal complement inhibitors as treatment for aABMR.
Collapse
Affiliation(s)
- Gesa Tiller
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Rosa G. M. Lammerts
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jessy J. Karijosemito
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Firas F. Alkaff
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Arjan Diepstra
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Robert A. Pol
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anita H. Meter-Arkema
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Marc. A. Seelen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Marius C. van den Heuvel
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bouke G. Hepkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mohamed R. Daha
- Department of Nephrology, University of Leiden, Leiden, Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Stefan P. Berger,
| |
Collapse
|
48
|
de Jong IEM, Overi D, Carpino G, Gouw ASH, van den Heuvel MC, van Kempen LC, Mancone C, Onori P, Cardinale V, Casadei L, Alvaro D, Porte RJ, Gaudio E. Persistent biliary hypoxia and lack of regeneration are key mechanisms in the pathogenesis of posttransplant nonanastomotic strictures. Hepatology 2022; 75:814-830. [PMID: 34543480 PMCID: PMC9300015 DOI: 10.1002/hep.32166] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 08/30/2021] [Accepted: 09/16/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND AIMS Nonanastomotic biliary strictures (NAS) are a major cause of morbidity after orthotopic liver transplantation (OLT). Although ischemic injury of peribiliary glands (PBGs) and peribiliary vascular plexus during OLT has been associated with the later development of NAS, the exact underlying mechanisms remain unclear. We hypothesized that bile ducts of patients with NAS suffer from ongoing biliary hypoxia and lack of regeneration from PBG stem/progenitor cells. APPROACH AND RESULTS Forty-two patients, requiring retransplantation for either NAS (n = 18), hepatic artery thrombosis (HAT; n = 13), or nonbiliary graft failure (controls; n = 11), were included in this study. Histomorphological analysis of perihilar bile ducts was performed to assess differences in markers of cell proliferation and differentiation in PBGs, microvascular density (MVD), and hypoxia. In addition, isolated human biliary tree stem cells (hBTSCs) were used to examine exo-metabolomics during in vitro differentiation toward mature cholangiocytes. Bile ducts of patients with NAS or HAT had significantly reduced indices of PBG mass, cellular proliferation and differentiation (mucus production, secretin receptor expression, and primary cilia), reduced MVD, and increased PBG apoptosis and hypoxia marker expression, compared to controls. Metabolomics of hBTSCs during in vitro differentiation toward cholangiocytes revealed a switch from a glycolytic to oxidative metabolism, indicating the need for oxygen. CONCLUSIONS NAS are characterized by a microscopic phenotype of chronic biliary hypoxia attributed to loss of microvasculature, resulting in reduced proliferation and differentiation of PBG stem/progenitor cells into mature cholangiocytes. These findings suggest that persistent biliary hypoxia is a key mechanism underlying the development of NAS after OLT.
Collapse
Affiliation(s)
- Iris E M de Jong
- Surgical Research LaboratoryDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands.,Section of Hepatobiliary Surgery and Liver TransplantationDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Diletta Overi
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic SciencesSapienza University of RomeRomeItaly
| | - Guido Carpino
- Division of Health SciencesDepartment of Movement, Human and Health SciencesUniversity of Rome "Foro Italico"RomeItaly
| | - Annette S H Gouw
- Department of PathologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Marius C van den Heuvel
- Department of PathologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Léon C van Kempen
- Department of PathologyUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Carmine Mancone
- Department of Molecular MedicineSapienza University of RomeRomeItaly
| | - Paolo Onori
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic SciencesSapienza University of RomeRomeItaly
| | - Vincenzo Cardinale
- Department of Medico-Surgical Sciences and BiotechnologiesPolo Pontino, Sapienza University of RomeRomeItaly
| | - Luca Casadei
- Department of ChemistrySapienza University of RomeRomeItaly
| | - Domenico Alvaro
- Department of Translational and Precision MedicineSapienza University of RomeRomeItaly
| | - Robert J Porte
- Section of Hepatobiliary Surgery and Liver TransplantationDepartment of SurgeryUniversity of GroningenUniversity Medical Center GroningenGroningenThe Netherlands
| | - Eugenio Gaudio
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic SciencesSapienza University of RomeRomeItaly
| |
Collapse
|
49
|
Pretransplant endotrophin predicts delayed graft function after kidney transplantation. Sci Rep 2022; 12:4079. [PMID: 35260630 PMCID: PMC8904626 DOI: 10.1038/s41598-022-07645-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/15/2022] [Indexed: 12/27/2022] Open
Abstract
Delayed graft function after kidney transplantation is common and increases morbidity and health care costs. There is evidence that endotrophin, a specific fragment of pro-collagen type VI, promotes the inflammatory response in kidney diseases. We tested the hypothesis that pretransplant endotrophin in kidney transplant recipients may be associated with the risk of delayed graft function. Pretransplant plasma endotrophin was assessed using an enzyme-linked immunosorbent assay in three independent cohorts with 806 kidney transplant recipients. The primary outcome was delayed graft function, i.e., the necessity of at least one dialysis session within one-week posttransplant. In the discovery cohort median pretransplant plasma endotrophin was higher in 32 recipients (12%) who showed delayed graft function when compared to 225 recipients without delayed graft function (58.4 ng/mL [IQR 33.4-69.0]; N = 32; vs. 39.5 ng/mL [IQR 30.6-54.5]; N = 225; P = 0.009). Multivariable logistic regression, fully adjusted for confounders showed, that pretransplant plasma endotrophin as a continuous variable was independently associated with delayed graft function in both validation cohorts, odds ratio 2.09 [95% CI 1.30-3.36] and 2.06 [95% CI 1.43-2.97]. Pretransplant plasma endotrophin, a potentially modifiable factor, was independently associated with increased risk of delayed graft function and may be a new avenue for therapeutic interventions.
Collapse
|
50
|
van den Berg EH, Flores-Guerrero JL, Gruppen EG, Garcia E, Connelly MA, de Meijer VE, Bakker SJL, Blokzijl H, Dullaart RPF. Profoundly Disturbed Lipoproteins in Cirrhotic Patients: Role of Lipoprotein-Z, a Hepatotoxic LDL-like Lipoprotein. J Clin Med 2022; 11:jcm11051223. [PMID: 35268313 PMCID: PMC8910943 DOI: 10.3390/jcm11051223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 12/17/2022] Open
Abstract
Detailed information regarding lipoprotein concentrations and subfractions in cirrhotic patients before and after orthotopic liver transplantation (OLT) is lacking. Lipoprotein-Z (LP-Z) is a recently characterised abnormal, hepatotoxic free cholesterol-rich low-density lipoprotein (LDL)-like lipoprotein. We determined the lipoprotein profiles, including LP-Z, in cirrhotic patients and OLT recipients and assessed the prognostic significance of LP-Z on the OLT waiting list. We performed analyses in cirrhotic transplant candidates and non-cirrhotic OLT recipients. A population-based cohort was used as reference. The setting was a University hospital. Lipoprotein particle concentrations and subfractions were measured by nuclear magnetic resonance spectroscopy. In the cirrhotic patients (N = 130), most measures of triglyceride-rich lipoproteins (TRL), LDL, and high-density lipoproteins (HDL) were much lower compared to the OLT recipients (N = 372) and controls (N = 6027) (p < 0.01). In the OLT recipients, many lipoprotein variables were modestly lower, but HDL-cholesterol, triglycerides, and TRL and HDL size were greater vs. the control population. LP-Z was measurable in 40 cirrhotic patients and 3 OLT recipients (30.8% vs. 0.8%, p < 0.001). The cirrhotic patients with measurable LP-Z levels had profoundly lower HDL-cholesterol and particle concentrations (p < 0.001), and worse Child Pugh Turcotte classifications and MELD scores. The presence of LP-Z (adjusted for age, sex, and MELD score) predicted worse survival in cirrhotic patients (HR per 1 LnSD increment: 1.11, 95%CI 1.03−1.19, p = 0.003). In conclusion, cirrhotic patients have considerably lower plasma concentrations of all major lipoprotein classes with changes in lipoprotein subfraction distribution. After OLT, these lipoprotein abnormalities are in part reversed. LP-Z is associated with cirrhosis. Its presence may translate in disturbed HDL metabolism and worse survival.
Collapse
Affiliation(s)
- Eline H. van den Berg
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
- Correspondence: ; Tel.: +31-50-3610426
| | - Jose L. Flores-Guerrero
- Department of Nephrology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (J.L.F.-G.); (E.G.G.); (S.J.L.B.)
| | - Eke G. Gruppen
- Department of Nephrology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (J.L.F.-G.); (E.G.G.); (S.J.L.B.)
| | - Erwin Garcia
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, NC 27560, USA; (E.G.); (M.A.C.)
| | - Margery A. Connelly
- Laboratory Corporation of America Holdings (Labcorp), Morrisville, NC 27560, USA; (E.G.); (M.A.C.)
| | - Vincent E. de Meijer
- Department of Hepatopancreatobiliary Surgery and Liver Transplantation, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| | - Stephan J. L. Bakker
- Department of Nephrology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands; (J.L.F.-G.); (E.G.G.); (S.J.L.B.)
| | - Hans Blokzijl
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| | - Robin P. F. Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, 9700 RB Groningen, The Netherlands;
| |
Collapse
|