1
|
Spector BL, Koseva BS, Sante D, Cheung WA, Alisch RS, Kats A, Bergmann P, Grundberg E, Wyckoff GJ, Willig LK. Total plasma cfDNA methylation in pediatric kidney transplant recipients provides insight into acute allograft rejection pathophysiology. Clin Immunol 2025; 275:110475. [PMID: 40107586 DOI: 10.1016/j.clim.2025.110475] [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: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Cell-free DNA (cfDNA) is a marker of organ injury and immune response. DNA methylation is an epigenetic regulator of gene expression. Here, we elucidate total plasma cfDNA methylation from kidney transplant recipients in presence versus absence of rejection. In doing so, we exploit cfDNA as a real-time biomarker to define molecular pathways of rejection. Twenty plasma cfDNA samples from pediatric kidney transplant recipients were collected at allograft biopsy. Differentially methylated cytosine residues (>20 % methylation difference, q-value <0.05) were identified in presence (N = 7) versus absence (N = 9) of acute rejection. Separate analyses were performed comparing borderline rejection (N = 4) to rejection and non-rejection. In rejection versus non-rejection, there were 1269 differentially methylated genes corresponding to 533 pathways. These numbers were 4-13× greater than comparisons against borderline samples. Enriched pathways between rejection and non-rejection samples were related to immune cell/inflammatory response, lipid metabolism, and tryptophan-kynurenine metabolism, suggesting differential methylation of these pathways contributes to rejection.
Collapse
Affiliation(s)
- Benjamin L Spector
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States; Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States.
| | - Boryana S Koseva
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Drinnan Sante
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City School of Pharmacy, Kansas City, MO, United States
| | - Warren A Cheung
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Alexander Kats
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO, United States
| | - Phillip Bergmann
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Elin Grundberg
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Gerald J Wyckoff
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City School of Pharmacy, Kansas City, MO, United States
| | - Laurel K Willig
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| |
Collapse
|
2
|
Liu Y, Fu Q, Yang M, Xu J, Zhou Z, Chen X, Zhang Y, Yuan H, Jia G, He S, Yang L, Zhao G. Downregulation of N6-methyladenosine (m6A) methylation of Sema4D mRNA contributes to Treg dysfunction and allograft rejection. Am J Transplant 2025:S1600-6135(25)00023-1. [PMID: 39848338 DOI: 10.1016/j.ajt.2025.01.017] [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: 09/11/2024] [Revised: 12/08/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
Regulatory T cells (Tregs) have been shown to be involved in the induction of transplantation tolerance in numerous models. Our previous work demonstrated that methyltransferase-like 14 (METTL14) loss impaired Treg function and hindered the establishment of transplantation tolerance. However, the underlying mechanisms remain unclear. In this study, we found that METTL14 knockdown in Tregs significantly impaired their regulatory function, leading to poor allograft function and accelerated transplant rejection. Using methylated RNA immunoprecipitation- and mRNA-sequencing approaches, we discovered that METTL14 deficiency fostered the expression of semaphorin 4D (Sema4D) mRNA, a key semaphorin family member with immunoregulatory activity. Methylation of target adenosines reduced Sema4D mRNA degradation, a process mediated by the METTL14-YTH N6-methyladenosine RNA binding protein 2 axis. Inhibition of Sema4D suppressed its interaction with its receptor, thereby preserving Treg immunoregulation capability and prolonging allograft survival through the p21-activated kinase-signal transducer and activator of transcription 5signaling pathway. Importantly, Sema4D expression in kidney transplant biopsies were negatively correlated with renal allograft survival. In summary, our findings suggest that METTL14 deficiency in Tregs leads to transplant rejection and reveal for the first time that Sema4D may serve as a potential therapeutic target to enhance Treg function in transplantation.
Collapse
Affiliation(s)
- Yanzhuo Liu
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Department of Oncology, Medical Research Center, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan Province, China
| | - Qiang Fu
- Organ Transplant Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Maozhu Yang
- Department of International Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Jianli Xu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Zili Zhou
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Xingmin Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yanling Zhang
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Hao Yuan
- The Seventh People's Hospital of Chengdu Affiliated Cancer Hospital of Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Shu He
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China.
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China.
| |
Collapse
|
3
|
Zhu J, Xiang X, Hu X, Li C, Song Z, Dong Z. miR-147 Represses NDUFA4, Inducing Mitochondrial Dysfunction and Tubular Damage in Cold Storage Kidney Transplantation. J Am Soc Nephrol 2023; 34:1381-1397. [PMID: 37211637 PMCID: PMC10400108 DOI: 10.1681/asn.0000000000000154] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/25/2023] [Indexed: 05/23/2023] Open
Abstract
SIGNIFICANCE STATEMENT Cold storage-associated transplantation (CST) injury occurs in renal transplant from deceased donors, the main organ source. The pathogenesis of CST injury remains poorly understood, and effective therapies are not available. This study has demonstrated an important role of microRNAs in CST injury and revealed the changes in microRNA expression profiles. Specifically, microRNA-147 (miR-147) is consistently elevated during CST injury in mice and in dysfunctional renal grafts in humans. Mechanistically, NDUFA4 (a key component of mitochondrial respiration complex) is identified as a direct target of miR-147. By repressing NDUFA4, miR-147 induces mitochondrial damage and renal tubular cell death. Blockade of miR-147 and overexpression of NDUFA4 reduce CST injury and improve graft function, unveiling miR-147 and NDUFA4 as new therapeutic targets in kidney transplantation. BACKGROUND Kidney injury due to cold storage-associated transplantation (CST) is a major factor determining the outcome of renal transplant, for which the role and regulation of microRNAs remain largely unclear. METHODS The kidneys of proximal tubule Dicer (an enzyme for microRNA biogenesis) knockout mice and their wild-type littermates were subjected to CST to determine the function of microRNAs. Small RNA sequencing then profiled microRNA expression in mouse kidneys after CST. Anti-microRNA-147 (miR-147) and miR-147 mimic were used to examine the role of miR-147 in CST injury in mouse and renal tubular cell models. RESULTS Knockout of Dicer from proximal tubules attenuated CST kidney injury in mice. RNA sequencing identified multiple microRNAs with differential expression in CST kidneys, among which miR-147 was induced consistently in mouse kidney transplants and in dysfunctional human kidney grafts. Anti-miR-147 protected against CST injury in mice and ameliorated mitochondrial dysfunction after ATP depletion injury in renal tubular cells in intro . Mechanistically, miR-147 was shown to target NDUFA4, a key component of the mitochondrial respiration complex. Silencing NDUFA4 aggravated renal tubular cell death, whereas overexpression of NDUFA4 prevented miR-147-induced cell death and mitochondrial dysfunction. Moreover, overexpression of NDUFA4 alleviated CST injury in mice. CONCLUSIONS microRNAs, as a class of molecules, are pathogenic in CST injury and graft dysfunction. Specifically, miR-147 induced during CST represses NDUFA4, leading to mitochondrial damage and renal tubular cell death. These results unveil miR-147 and NDUFA4 as new therapeutic targets in kidney transplantation.
Collapse
Affiliation(s)
- Jiefu Zhu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Xiaohong Xiang
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
- Department of Critical Care Medicine, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Xiaoru Hu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, China
| | - Chenglong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| | - Zhixia Song
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
- Department of Nephrology, Yichang Central People's Hospital, The First Clinical Medical College of Three Gorges University, Yichang, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia
| |
Collapse
|
4
|
do Nascimento Gonçalves N, Caldas HC, da Silva Florim GM, Sormani GM, Arantes LMRB, Sorroche BP, Baptista MASF, Fernandes-Charpiot IMM, Nascimento-Filho CHV, de Castilho RM, Abbud-Filho M. Distinct global DNA methylation and NF-κB expression profile of preimplantation biopsies from ideal and non-ideal kidneys. J Nephrol 2022; 35:1831-1840. [PMID: 35524842 DOI: 10.1007/s40620-022-01341-w] [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: 02/09/2022] [Accepted: 04/20/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Epigenetic mechanisms may affect the ideal and non-ideal kidneys selected for transplantation and their inflammatory gene expression profile differently and may contribute to poor clinical outcomes. OBJECTIVE Study the Global DNA methylation and the expression profiles of the DNA methyltransferases (DNMTs) and nuclear factor kappa B (NF-κB) in preimplantation kidney biopsies from ideal and non-ideal kidneys (expanded criteria donor (ECD) and with KDPI > 85%). METHODS In a sample consisting of 45 consecutive pre-implantation biopsies, global DNA methylation levels were detected by LINE-1 repeated elements using bisulfite pyrosequencing. DNMT gene expression was assessed by real-time quantitative polymerase chain reaction, and NF-κB protein expression by immunofluorescence. RESULTS ECD kidneys displayed increased methylation levels in LINE-1, and DNMT1 and DNMT3B expression was upregulated when comparing ECD to standard criteria donor kidneys. Similarly, kidneys with KDPI > 85% exhibited increased LINE-1 methylation and DNMT1 upregulation when compared to a KDPI ≤ 85%. NF-κB protein expression levels were greatly increased in both types of non-ideal kidneys compared to ideal kidneys. Moreover, hypermethylation of LINE-1 was associated with cold ischemia time > 20 h and ECD kidney classification. CONCLUSIONS This study shows that global DNA hypermethylation and high expression of NF-κB occurred in both types of non-ideal kidneys and were associated with prolonged cold ischemia time. Global DNA methylation can be a useful tool to assess non-ideal kidneys and hence, could be used to expand the pool of kidneys donors.
Collapse
Affiliation(s)
- Naiane do Nascimento Gonçalves
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | - Heloisa Cristina Caldas
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | - Greiciane Maria da Silva Florim
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | - Giovanna Mattiello Sormani
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | | | | | - Maria Alice Sperto Ferreira Baptista
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | - Ida Maria Maximina Fernandes-Charpiot
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil
| | | | - Rogério Moraes de Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Mario Abbud-Filho
- Department of Medicine, Faculdade de Medicina de São José do Rio Preto (FAMERP), Laboratório de Imunologia e Transplante Experimental (LITEX), Hospital de Base, Av. Brigadeiro Faria Lima, 5416, São Jose do Rio Preto, SP, 15090-000, Brazil.
| |
Collapse
|
5
|
Xiang X, Zhu J, Dong G, Dong Z. Epigenetic Regulation in Kidney Transplantation. Front Immunol 2022; 13:861498. [PMID: 35464484 PMCID: PMC9024296 DOI: 10.3389/fimmu.2022.861498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/17/2022] [Indexed: 12/29/2022] Open
Abstract
Kidney transplantation is a standard care for end stage renal disease, but it is also associated with a complex pathogenesis including ischemia-reperfusion injury, inflammation, and development of fibrosis. Over the past decade, accumulating evidence has suggested a role of epigenetic regulation in kidney transplantation, involving DNA methylation, histone modification, and various kinds of non-coding RNAs. Here, we analyze these recent studies supporting the role of epigenetic regulation in different pathological processes of kidney transplantation, i.e., ischemia-reperfusion injury, acute rejection, and chronic graft pathologies including renal interstitial fibrosis. Further investigation of epigenetic alterations, their pathological roles and underlying mechanisms in kidney transplantation may lead to new strategies for the discovery of novel diagnostic biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Xiaohong Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States.,Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiefu Zhu
- Center of Nephrology and Dialysis, Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| |
Collapse
|
6
|
Abstract
Exhaustion of T cells occurs in response to long-term exposure to self and foreign antigens. It limits T cell capacity to proliferate and produce cytokines, leading to an impaired ability to clear chronic infections or eradicate tumors. T-cell exhaustion is associated with a specific transcriptional, epigenetic, and metabolic program and characteristic cell surface markers' expression. Recent studies have begun to elucidate the role of T-cell exhaustion in transplant. Higher levels of exhausted T cells have been associated with better graft function in kidney transplant recipients. In contrast, reinvigorating exhausted T cells by immune checkpoint blockade therapies, while promoting tumor clearance, increases the risk of acute rejection. Lymphocyte depletion and high alloantigen load have been identified as major drivers of T-cell exhaustion. This could account, at least in part, for the reduced rates of acute rejection in organ transplant recipients induced with thymoglobulin and for the pro-tolerogenic effects of a large organ such as the liver. Among the drugs that are widely used for maintenance immunosuppression, calcineurin inhibitors have a contrasting inhibitory effect on exhaustion of T cells, while the influence of mTOR inhibitors is still unclear. Harnessing or encouraging the natural processes of exhaustion may provide a novel strategy to promote graft survival and transplantation tolerance.
Collapse
|
7
|
Cristoferi I, Giacon TA, Boer K, van Baardwijk M, Neri F, Campisi M, Kimenai HJAN, Clahsen-van Groningen MC, Pavanello S, Furian L, Minnee RC. The applications of DNA methylation as a biomarker in kidney transplantation: a systematic review. Clin Epigenetics 2022; 14:20. [PMID: 35130936 PMCID: PMC8822833 DOI: 10.1186/s13148-022-01241-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 01/27/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Although kidney transplantation improves patient survival and quality of life, long-term results are hampered by both immune- and non-immune-mediated complications. Current biomarkers of post-transplant complications, such as allograft rejection, chronic renal allograft dysfunction, and cutaneous squamous cell carcinoma, have a suboptimal predictive value. DNA methylation is an epigenetic modification that directly affects gene expression and plays an important role in processes such as ischemia/reperfusion injury, fibrosis, and alloreactive immune response. Novel techniques can quickly assess the DNA methylation status of multiple loci in different cell types, allowing a deep and interesting study of cells' activity and function. Therefore, DNA methylation has the potential to become an important biomarker for prediction and monitoring in kidney transplantation. PURPOSE OF THE STUDY The aim of this study was to evaluate the role of DNA methylation as a potential biomarker of graft survival and complications development in kidney transplantation. MATERIAL AND METHODS: A systematic review of several databases has been conducted. The Newcastle-Ottawa scale and the Jadad scale have been used to assess the risk of bias for observational and randomized studies, respectively. RESULTS Twenty articles reporting on DNA methylation as a biomarker for kidney transplantation were included, all using DNA methylation for prediction and monitoring. DNA methylation pattern alterations in cells isolated from different tissues, such as kidney biopsies, urine, and blood, have been associated with ischemia-reperfusion injury and chronic renal allograft dysfunction. These alterations occurred in different and specific loci. DNA methylation status has also proved to be important for immune response modulation, having a crucial role in regulatory T cell definition and activity. Research also focused on a better understanding of the role of this epigenetic modification assessment for regulatory T cells isolation and expansion for future tolerance induction-oriented therapies. CONCLUSIONS Studies included in this review are heterogeneous in study design, biological samples, and outcome. More coordinated investigations are needed to affirm DNA methylation as a clinically relevant biomarker important for prevention, monitoring, and intervention.
Collapse
Affiliation(s)
- Iacopo Cristoferi
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands.
| | - Tommaso Antonio Giacon
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
- Environmental and Respiratory Physiology Laboratory, Department of Biomedical Sciences, Padua University, Via Marzolo 3, 35131, Padua, Italy
- Institute of Anaesthesia and Intensive Care, Department of Medicine - DIMED, Padua University Hospital, Via Cesare Battisti 267, 35128, Padua, Italy
| | - Karin Boer
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Division of Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, The Netherlands
| | - Myrthe van Baardwijk
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Flavia Neri
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
| | - Manuela Campisi
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
| | - Hendrikus J A N Kimenai
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Marian C Clahsen-van Groningen
- Department of Pathology and Clinical Bioinformatics, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Sofia Pavanello
- Occupational Medicine, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Padua University, Via Giustiniani 2, 35128, Padua, Italy
| | - Lucrezia Furian
- Kidney and Pancreas Transplantation Unit, Department of Surgical, Oncological and Gastroenterological Sciences, Padua University Hospital, Via Giustiniani 2, 35128, Padua, Italy
| | - Robert C Minnee
- Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
- Erasmus MC Transplant Institute, Erasmus MC, University Medical Center Rotterdam, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| |
Collapse
|
8
|
At the dawn: cell-free DNA fragmentomics and gene regulation. Br J Cancer 2021; 126:379-390. [PMID: 34815523 PMCID: PMC8810841 DOI: 10.1038/s41416-021-01635-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Epigenetic mechanisms play instrumental roles in gene regulation during embryonic development and disease progression. However, it is challenging to non-invasively monitor the dynamics of epigenomes and related gene regulation at inaccessible human tissues, such as tumours, fetuses and transplanted organs. Circulating cell-free DNA (cfDNA) in peripheral blood provides a promising opportunity to non-invasively monitor the genomes from these inaccessible tissues. The fragmentation patterns of plasma cfDNA are unevenly distributed in the genome and reflect the in vivo gene-regulation status across multiple molecular layers, such as nucleosome positioning and gene expression. In this review, we revisited the computational and experimental approaches that have been recently developed to measure the cfDNA fragmentomics across different resolutions comprehensively. Moreover, cfDNA in peripheral blood is released following cell death, after apoptosis or necrosis, mainly from haematopoietic cells in healthy people and diseased tissues in patients. Several cfDNA-fragmentomics approaches showed the potential to identify the tissues-of-origin in cfDNA from cancer patients and healthy individuals. Overall, these studies paved the road for cfDNA fragmentomics to non-invasively monitor the in vivo gene-regulatory dynamics in both peripheral immune cells and diseased tissues.
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW DNA methylation is involved in gene transcription and as such important for cellular function. Here, the literature on DNA methylation in relation to acute rejection is summarized with a focus on the potential clinical utility of DNA methylation for monitoring transplant rejection. RECENT FINDINGS The tight transcriptional control of DNA methylation in immune cell function, e.g. demethylation in regulatory T-cell-specific genes for stable immunosuppressive capacities, suggests an important role for DNA methylation variations in the antidonor-directed immune response. Until today, differentially methylated DNA in immune cells, however, has not been described at the moment of allograft rejection. The ability to locus-specific modify DNA methylation could facilitate the generation of stable cells for cellular therapy purposes. The unique cell-specific characteristics of DNA methylation provide the opportunity to identify its cellular origin. Examining methylation of cell-free DNA in blood or urine may serve as a 'liquid biopsy' enabling minimally invasive detection of allograft rejection. SUMMARY Actual research publications on DNA methylation in relation to allograft rejection are scarce, which makes it challenging to determine its potential clinical value. Extensive research is needed to investigate the value of DNA methylation in early recognition, diagnosis, and/or successful treatment of allograft rejection.
Collapse
|
10
|
Lai X, Zheng X, Mathew JM, Gallon L, Leventhal JR, Zhang ZJ. Tackling Chronic Kidney Transplant Rejection: Challenges and Promises. Front Immunol 2021; 12:661643. [PMID: 34093552 PMCID: PMC8173220 DOI: 10.3389/fimmu.2021.661643] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/27/2021] [Indexed: 01/09/2023] Open
Abstract
Despite advances in post-transplant management, the long-term survival rate of kidney grafts and patients has not improved as approximately forty percent of transplants fails within ten years after transplantation. Both immunologic and non-immunologic factors contribute to late allograft loss. Chronic kidney transplant rejection (CKTR) is often clinically silent yet progressive allogeneic immune process that leads to cumulative graft injury, deterioration of graft function. Chronic active T cell mediated rejection (TCMR) and chronic active antibody-mediated rejection (ABMR) are classified as two principal subtypes of CKTR. While significant improvements have been made towards a better understanding of cellular and molecular mechanisms and diagnostic classifications of CKTR, lack of early detection, differential diagnosis and effective therapies continue to pose major challenges for long-term management. Recent development of high throughput cellular and molecular biotechnologies has allowed rapid development of new biomarkers associated with chronic renal injury, which not only provide insight into pathogenesis of chronic rejection but also allow for early detection. In parallel, several novel therapeutic strategies have emerged which may hold great promise for improvement of long-term graft and patient survival. With a brief overview of current understanding of pathogenesis, standard diagnosis and challenges in the context of CKTR, this mini-review aims to provide updates and insights into the latest development of promising novel biomarkers for diagnosis and novel therapeutic interventions to prevent and treat CKTR.
Collapse
Affiliation(s)
- Xingqiang Lai
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Organ Transplant Center, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xin Zheng
- Department of Urology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - James M. Mathew
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Lorenzo Gallon
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Medicine, Nephrology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph R. Leventhal
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Zheng Jenny Zhang
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
11
|
Vasco M, Benincasa G, Fiorito C, Faenza M, De Rosa P, Maiello C, Santangelo M, Vennarecci G, Napoli C. Clinical epigenetics and acute/chronic rejection in solid organ transplantation: An update. Transplant Rev (Orlando) 2021; 35:100609. [PMID: 33706201 DOI: 10.1016/j.trre.2021.100609] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
The lack of a precise stratification algorithm for predicting patients at high risk of graft rejection challenges the current solid organ transplantation (SOT) clinical setting. In fact, the established biomarkers for transplantation outcomes are unable to accurately predict the onset time and severity of graft rejection (acute or chronic) as well as the individual response to immunosuppressive drugs. Thus, identifying novel molecular pathways underlying early immunological responses which can damage transplant integrity is needed to reach precision medicine and personalized therapy of SOT. Direct epigenetic-sensitive mechanisms, mainly DNA methylation and histone modifications, may play a relevant role for immune activation and long-term effects (e.g., activation of fibrotic processes) which may be translated in new non-invasive biomarkers and drug targets. In particular, the measure of DNA methylation by using the blood-based "epigenetic clock" system may be an added value to the donor eligibility criteria providing an estimation of the heart biological age as well as a predictive biomarkers. Besides, monitoring of DNA methylation changes may aid to predict acute vs chronic graft damage in kidney transplantation (KT) patients. For example, hypermethylation of genes belonging to the Notch and Wnt pathways showed a higher predictive value for chronic injury occurring at 12 months post-KT with respect to established clinical parameters. Detecting higher circulating cell-free DNA (cfDNA) fragments carrying hepatocyte-specific unmethylated loci in the inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4), insulin like growth factor 2 receptor (IGF2R), and vitronectin (VTN) genes may be useful to predict acute graft injury after liver transplantation (LT) in serum samples. Furthermore, hypomethylation in the forkhead box P3 (FOXP3) gene may serve as a marker of infiltrating natural Treg percentage in the graft providing the ability to predict acute rejection events after heart transplantation (HTx). We aim to update on the possible clinical relevance of DNA methylation changes regulating immune-related pathways underlying acute or chronic graft rejection in KT, LT, and HTx which might be useful to prevent, monitor, and treat solid organ rejection at personalized level.
Collapse
Affiliation(s)
- Maria Vasco
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Carmela Fiorito
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mario Faenza
- Multidisciplinary Department of Medical Surgical and Dental Sciences-Plastic Surgery Unit, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paride De Rosa
- General Surgery and Transplantation Unit, "San Giovanni di Dio e Ruggi D'Aragona" University Hospital, Scuola Medica Salernitana, Salerno, Italy
| | - Ciro Maiello
- Department of Cardiovascular Surgery and Transplants, Monaldi Hospital, Azienda dei Colli, Naples, Italy
| | - Michele Santangelo
- General Surgery and Kidney Transplantation Unit, "Federico II" University Hospital, Naples, Italy
| | - Giovanni Vennarecci
- Division of General Surgery and Liver Transplantation, AO Cardarelli, Naples, Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, University of Campania "Luigi Vanvitelli", Naples, Italy; Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
12
|
Nobakht E, Jagadeesan M, Paul R, Bromberg J, Dadgar S. Precision Medicine in Kidney Transplantation: Just Hype or a Realistic Hope? Transplant Direct 2021; 7:e650. [PMID: 33437865 PMCID: PMC7793397 DOI: 10.1097/txd.0000000000001102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Desirable outcomes including rejection- and infection-free kidney transplantation are not guaranteed despite current strategies for immunosuppression and using prophylactic antimicrobial medications. Graft survival depends on factors beyond human leukocyte antigen matching such as the level of immunosuppression, infections, and management of other comorbidities. Risk stratification of transplant patients based on predisposing genetic modifiers and applying precision pharmacotherapy may help improving the transplant outcomes. Unlike certain fields such as oncology in which consistent attempts are being carried out to move away from the "error and trial approach," transplant medicine is lagging behind in implementing personalized immunosuppressive therapy. The need for maintaining a precarious balance between underimmunosuppression and overimmunosuppression coupled with adverse effects of medications calls for a gene-based guidance for precision pharmacotherapy in transplantation. Technologic advances in molecular genetics have led to increased accessibility of genetic tests at a reduced cost and have set the stage for widespread use of gene-based therapies in clinical care. Evidence-based guidelines available for precision pharmacotherapy have been proposed, including guidelines from Clinical Pharmacogenetics Implementation Consortium, the Pharmacogenomics Knowledge Base National Institute of General Medical Sciences of the National Institutes of Health, and the US Food and Drug Administration. In this review, we discuss the implications of pharmacogenetics and potential role for genetic variants-based risk stratification in kidney transplantation. A single score that provides overall genetic risk, a polygenic risk score, can be achieved by combining of allograft rejection/loss-associated variants carried by an individual and integrated into practice after clinical validation.
Collapse
Affiliation(s)
- Ehsan Nobakht
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University School of Medicine, Washington, DC
| | - Muralidharan Jagadeesan
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University School of Medicine, Washington, DC
| | - Rohan Paul
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University School of Medicine, Washington, DC
| | - Jonathan Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD
| | - Sherry Dadgar
- Division of Renal Diseases and Hypertension, Department of Medicine, George Washington University School of Medicine, Washington, DC
- Personalized Medicine Care Diagnostics Laboratory (PMCDx), Inc., Germantown, MD
| |
Collapse
|
13
|
Wu J, Zhang F, Zhang J, Sun Z, Wang W. Advances of miRNAs in kidney graft injury. Transplant Rev (Orlando) 2020; 35:100591. [PMID: 33309915 DOI: 10.1016/j.trre.2020.100591] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/14/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Kidney transplantation is the preferred treatment for patients with end-stage renal disease. However, various types of kidney graft injury after transplantation are still key factors that affect the survival of the kidney graft. Therefore, exploring the underlying mechanisms involved is very important. Current diagnostic measures for kidney graft injury (including needle biopsy, blood creatinine, eGFR, etc.) have many limiting factors such as invasiveness, insufficient sensitivity and specificity, so they cannot provide timely and effective information to clinicians. As for kidney grafts that have occurred injury, the traditional treatment has a little efficacy and many side effects. Therefore, there is an urgent need for developing new biomarkers and targeted treatment for kidney graft injury. Recently, studies have found that miRNAs are involved in the regulation of the progression of kidney graft injury. At the same time, it has high stability in blood, urine, and other body fluids, so it is suggested to have the potential as a biomarker and therapeutic target for kidney graft injury. Here, we reviewed the miRNAs involved in the pathophysiology of kidney graft injury such as ischemia/reperfusion injury, acute rejection, drug-induced nephrotoxicity, chronic allograft dysfunction, BK virus infection, and the latest advances of miRNAs as biomarkers and therapeutic targets of kidney graft injury, then summarized the specific data of miRNAs expression level in kidney graft injury, which aims to provide a reference for subsequent basic research and clinical transformation.
Collapse
Affiliation(s)
- Jiyue Wu
- Institute of Urology, Beijing Chaoyang Hospital, Capital Medical Unversity, China
| | - Feilong Zhang
- Institute of Urology, Beijing Chaoyang Hospital, Capital Medical Unversity, China
| | - Jiandong Zhang
- Institute of Urology, Beijing Chaoyang Hospital, Capital Medical Unversity, China
| | - Zejia Sun
- Institute of Urology, Beijing Chaoyang Hospital, Capital Medical Unversity, China
| | - Wei Wang
- Institute of Urology, Beijing Chaoyang Hospital, Capital Medical Unversity, China.
| |
Collapse
|
14
|
Freitas RCC, Bortolin RH, Genvigir FDV, Bonezi V, Hirata TDC, Felipe CR, Tedesco-Silva H, Medina-Pestana JO, Cerda A, Doi SQ, Hirata MH, Hirata RDC. Differentially expressed urinary exo-miRs and clinical outcomes in kidney recipients on short-term tacrolimus therapy: a pilot study. Epigenomics 2020; 12:2019-2034. [PMID: 33275448 DOI: 10.2217/epi-2020-0160] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Aim: To analyze the expression of urinary exosome-derived miRNAs (exo-miRs) in kidney recipients on tacrolimus-based therapy. Patients & methods: Clinical and drug monitoring data were recorded from 23 kidney recipients. Expression of 93 exo-miRs was measured by quantitative PCR array and mRNA targets were explored. Results: 16 exo-miRs were differentially expressed, including marked upregulation of miR-155-5p, and downregulation of miR-223-3p and miR-1228-3p. Expression of miR-155-5p and miR-223-3p correlated with tacrolimus dose (p < 0.05), miR-223-3p with serum creatinine (p < 0.05), and miR-223-3p and miR-1228-3p with blood leukocytes (p < 0.05). 12 miRNAs have predicted targets involved in cell proliferation, apoptosis, stress response, PIK3/AKT/mTOR and TGF-β signaling pathways. Conclusion: Differentially expressed urinary exo-miRs may be useful markers to monitor tacrolimus therapy and graft function in kidney transplantation.
Collapse
Affiliation(s)
- Renata Caroline Costa Freitas
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Raul Hernandes Bortolin
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Fabiana Dalla Vecchia Genvigir
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Vivian Bonezi
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Claudia Rosso Felipe
- Nephrology Division, Hospital do Rim, Federal University of Sao Paulo, Sao Paulo 04038-002, Brazil
| | - Helio Tedesco-Silva
- Nephrology Division, Hospital do Rim, Federal University of Sao Paulo, Sao Paulo 04038-002, Brazil
| | | | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, BIOREN, Universidad de La Frontera, Temuco 4810296, Chile
| | - Sonia Quateli Doi
- School of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
| | - Mario Hiroyuki Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
15
|
Bonezi V, Genvigir FDV, Salgado PDC, Felipe CR, Tedesco-Silva H, Medina-Pestana JO, Cerda A, Doi SQ, Hirata MH, Hirata RDC. Differential expression of genes related to calcineurin and mTOR signaling and regulatory miRNAs in peripheral blood from kidney recipients under tacrolimus-based therapy. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1051. [PMID: 33145270 PMCID: PMC7575939 DOI: 10.21037/atm-20-1757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Genetic and epigenetics factors have been implicated in drug response, graft function and rejection in solid organ transplantation. Differential expression of genes involved in calcineurin and mTOR signaling pathway and regulatory miRNAs was analyzed in the peripheral blood of kidney recipient cohort (n=36) under tacrolimus-based therapy. Methods PPP3CA, PPP3CB, MTOR, FKBP1A, FKBP1B and FKBP5 mRNA expression and polymorphisms in PPP3CA and MTOR were analyzed by qPCR. Expression of miRNAs targeting PPP3CA (miR-30a, miR-145), PPP3CB (miR-10b), MTOR (miR-99a, miR-100), and FKBP1A (miR-103a) was measured by qPCR array. Results PPP3CA and MTOR mRNA levels were reduced in the first three months of treatment compared to pre-transplant (P<0.05). PPP3CB, FKBP1A, FKBP1B, and FKBP5 expression was not changed. In the 3rd month of treatment, the expression of miR-99a, which targets MTOR, increased compared to pre-transplant (P<0.05). PPP3CA c.249G>A (GG genotype) and MTOR c.2997C>T (TT genotype) were associated with reduced expression of PPP3CA mRNA and MTOR, respectively. FKBP1B mRNA levels were higher in patients with acute rejection (P=0.026). Conclusions The expression of PPP3CA, MTOR and miR-99a in the peripheral blood of renal recipients is influenced by tacrolimus-based therapy and by PPP3CA and MTOR variants. These molecules can be potential biomarkers for pharmacotherapy monitoring.
Collapse
Affiliation(s)
- Vivian Bonezi
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Fabiana Dalla Vecchia Genvigir
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Patrícia de Cássia Salgado
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudia Rosso Felipe
- Nephrology Division, Hospital do Rim, Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Helio Tedesco-Silva
- Nephrology Division, Hospital do Rim, Federal University of Sao Paulo, Sao Paulo, Brazil
| | | | - Alvaro Cerda
- Department of Basic Sciences, Center of Excellence in Translational Medicine, BIOREN, Universidad de La Frontera, Temuco, Chile
| | - Sonia Quateli Doi
- School of Medicine, Uniformed Services University, Bethesda, MD, USA
| | - Mario Hiroyuki Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
16
|
Zhu C, Xiang W, Li B, Wang Y, Feng S, Wang C, Chen Y, Xie W, Qu L, Huang H, Annunziata F, Nunna S, Krepelova A, Mohammad M. Rasa S, Neri F, Chen J, Jiang H. DNA methylation modulates allograft survival and acute rejection after renal transplantation by regulating the mTOR pathway. Am J Transplant 2020. [PMCID: PMC7891393 DOI: 10.1111/ajt.16183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
17
|
Li J, Qiu Y, Li L, Wang J, Cheuk YC, Sang R, Jia Y, Wang J, Zhang Y, Rong R. Histone Methylation Inhibitor DZNep Ameliorated the Renal Ischemia-Reperfusion Injury via Inhibiting TIM-1 Mediated T Cell Activation. Front Med (Lausanne) 2020; 7:305. [PMID: 32754604 PMCID: PMC7365856 DOI: 10.3389/fmed.2020.00305] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Renal ischemia-reperfusion injury (IRI) after renal transplantation often leads to the loss of kidney graft function. However, there is still a lack of efficient regimens to prevent or alleviate renal IRI. Our study focused on the renoprotective effect of 3-Deazaneplanocin A (DZNep), which is a histone methylation inhibitor. We found that DZNep significantly alleviated renal IRI by suppressing nuclear factor kappa-B (NF-κB), thus inhibiting the expression of inflammatory factors in renal tubular epithelial cells in vivo or in vitro. After treatment with DZNep, T cell activation was impaired in the spleen and kidney, which correlated with the downregulated expression of T-cell immunoglobulin mucin (TIM)-1 on T cells and TIM-4 in macrophages. In addition, pretreatment with DZNep was not sufficient to protect the kidney, while administration of DZNep from before to after surgery significantly ameliorated IRI. Our findings suggest that DZNep can be a novel strategy for preventing renal IRI following kidney transplantation.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yue Qiu
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Department of Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Long Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiyan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Ruirui Sang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yichen Jia
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Jina Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| | - Yi Zhang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai, China.,Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Organ Transplantation, Shanghai, China
| |
Collapse
|
18
|
Regulation of Endothelial-to-Mesenchymal Transition by MicroRNAs in Chronic Allograft Dysfunction. Transplantation 2019; 103:e64-e73. [PMID: 30907855 DOI: 10.1097/tp.0000000000002589] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrosis is a universal finding in chronic allograft dysfunction, and it is characterized by an accumulation of extracellular matrix. The precise source of the myofibroblasts responsible for matrix deposition is not understood, and pharmacological strategies for prevention or treatment of fibrosis remain limited. One source of myofibroblasts in fibrosis is an endothelial-to-mesenchymal transition (EndMT), a process first described in heart development and involving endothelial cells undergoing a phenotypic change to become more like mesenchymal cells. Recently, lineage tracing of endothelial cells in mouse models allowed studies of EndMT in vivo and reported 27% to 35% of myofibroblasts involved in cardiac fibrosis and 16% of isolated fibroblasts in bleomycin-induced pulmonary fibrosis to be of endothelial origin. Over the past decade, mature microRNAs (miRNAs) have increasingly been described as key regulators of biological processes through repression or degradation of targeted mRNA. The stability and abundance of miRNAs in body fluids make them attractive as potential biomarkers, and progress is being made in developing miRNA targeted therapeutics. In this review, we will discuss the evidence of miRNA regulation of EndMT from in vitro and in vivo studies and the potential relevance of this to heart, lung, and kidney allograft dysfunction.
Collapse
|
19
|
Unveiling the Role of DNA Methylation in Kidney Transplantation: Novel Perspectives toward Biomarker Identification. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1602539. [PMID: 30766879 PMCID: PMC6350635 DOI: 10.1155/2019/1602539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/30/2018] [Indexed: 12/13/2022]
Abstract
The burden of chronic kidney disease is dramatically rising, making it a major public health concern worldwide. Kidney transplantation is now the best treatment for patients with end-stage renal disease. Although kidney transplantation may improve survival and quality of life, its long-term results are hampered by immune- and/or non-immune-mediated complications. Thus, the identification of transplanted patients with a higher risk of posttransplant complications has become a big challenge for public health. However, current biomarkers of posttransplant complications have a poor predictive value, rising the need to explore novel approaches for the management of transplant patient. In this review we summarize the emerging literature about DNA methylation in kidney transplant complications, in order to highlight its perspectives toward biomarker identification. In the forthcoming future the monitoring of DNA methylation in kidney transplant patients could become a plausible strategy toward the prevention and/or treatment of kidney transplant complications.
Collapse
|
20
|
Schaenman J, Liao D, Phonphok K, Bunnapradist S, Karlamangla A. Predictors of Early and Late Mortality in Older Kidney Transplant Recipients. Transplant Proc 2019; 51:684-691. [PMID: 30979451 DOI: 10.1016/j.transproceed.2019.01.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 01/02/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Older kidney patients with chronic kidney disease benefit significantly from kidney transplantation. However, these older transplant recipients have greater mortality after transplantation than younger transplant recipients. Understanding the impact of comorbidities on post-transplant mortality can improve risk stratification and patient selection. METHODS A single-center analysis of 3105 kidney transplant recipients was performed over a 12-year period. Comorbidities associated with death were evaluated in older and younger transplant recipients. RESULTS The 2 most important factors associated with increased mortality in the first 100 days after transplant were recipient age ≥60 and receipt of deceased donor organs (adjusted odds ratios, 3.29 and 5.80, respectively), with no statistically significant impact of recipient comorbidities. In the later post-transplant period (after the first 100 days), recipient age ≥60 and receipt of deceased donor organs (adjusted hazard ratios [HR] of 2.14 and 2.29, respectively) remained predictors of mortality. We also found that donor age ≥60 and the recipient having cardiovascular disease and diabetes were independent predictors of increased mortality. There was a statistically significant interaction between diabetes and heart disease and recipient age ≥60, with a lesser impact on late mortality in older patients compared to younger patients. CONCLUSIONS This analysis suggests that comorbidities have a larger impact later after transplantation, with less effect on older recipients. These observations suggest that certain comorbid conditions should be evaluated differently in older patients compared to younger ones.
Collapse
Affiliation(s)
- J Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA.
| | - D Liao
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - K Phonphok
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA; Division of Nephrology, Department of Medicine, Rajavithi Hospital, Bangkok, Thailand
| | - S Bunnapradist
- Division of Nephrology, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - A Karlamangla
- Division of Geriatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
21
|
Doreille A, Dieudé M, Cardinal H. The determinants, biomarkers, and consequences of microvascular injury in kidney transplant recipients. Am J Physiol Renal Physiol 2018; 316:F9-F19. [PMID: 30379097 DOI: 10.1152/ajprenal.00163.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Independent of the initial cause of kidney disease, microvascular injury to the peritubular capillary network appears to play a central role in the development of interstitial fibrosis in both native and transplanted kidney disease. This association is explained by mechanisms such as the upregulation of profibrotic genes and epigenetic changes induced by hypoxia, capillary leakage, endothelial and pericyte transition to interstitial fibroblasts, as well as modifications in the secretome of endothelial cells. Alloimmune injury due to antibody-mediated rejection and ischemia-reperfusion injury are the two main etiologies of microvascular damage in kidney transplant recipients. The presence of circulating donor-specific anti-human leukocyte antigen (HLA) antibodies, histological findings, such as diffuse C4d staining in peritubular capillaries, and the extent and severity of peritubular capillaritis, are commonly used clinically to provide both diagnostic and prognostic information. Complement-dependent assays, circulating non-HLA antibodies, or evaluation of the microvasculature with novel imaging techniques are the subject of ongoing studies.
Collapse
Affiliation(s)
- Alice Doreille
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Université Paris-Sud , Paris , France
| | - Mélanie Dieudé
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Canadian Donation and Transplantation Research Program, Montreal, Quebec, Canada
| | - Heloise Cardinal
- Research Centre, Centre Hospitalier de l'Université de Montréal , Montreal, Quebec , Canada.,Canadian Donation and Transplantation Research Program, Montreal, Quebec, Canada
| |
Collapse
|
22
|
LCK as a Potential Therapeutic Target for Acute Rejection after Kidney Transplantation: A Bioinformatics Clue. J Immunol Res 2018; 2018:6451298. [PMID: 29977931 PMCID: PMC6011083 DOI: 10.1155/2018/6451298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/12/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022] Open
Abstract
Objectives We aim to identify the key biomarker of acute rejection (AR) after kidney transplantation via bioinformatics methods. Methods The gene expression data GSE75693 of 30 samples with stable kidney transplantation recipients and 15 AR samples were downloaded and analyzed by the limma package to identify differentially expressed genes (DEGs). Then, Gene Ontology (GO) functional enrichment analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were done to explore the biological functions and potential important pathways of DEGs. Finally, protein-protein interactions (PPIs) and literature mining were applied to construct the cocitation network and to select the hub protein. Results A total of 437 upregulated genes and 353 downregulated genes were selected according to P < 0.01 and |log2(fold change)| > 1.0. DEGs of AR are mainly located on membranes and impact the activation of receptors in immune responses. In the PPI network, Src kinase, lymphocyte kinase (LCK), CD3G, B2M, interferon-γ, CD3D, tumor necrosis factor, VAV1, and CD3E in the T cell receptor signaling pathway were selected as important factors, and LCK was identified as the hub protein. Conclusion LCK, via acting on T-cell receptor, might be a potential therapeutic target for AR after kidney transplantation.
Collapse
|
23
|
Zhao Y, Zhu Q, Sun S, Qiu Y, Li J, Liu W, Yuan G, Ma H. Renal transplantation increases angiotensin II receptor-mediated vascular contractility associated with changes of epigenetic mechanisms. Int J Mol Med 2018; 41:2375-2388. [PMID: 29393347 DOI: 10.3892/ijmm.2018.3435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 12/21/2017] [Indexed: 11/06/2022] Open
Abstract
Hypertension is one of the most common complications following renal transplantation, and it increases the risk of graft loss and other cardiovascular diseases. Previous studies have revealed that the use of angiotensin II (Ang II) blockers for preventing and treating hypertension is closely associated with higher survival following renal transplantation. However, the cellular and molecular mechanisms by which the vascular contractility of the recipient is altered in response to Ang II following renal transplantation have not been fully elucidated. In the present study, using the Fisher‑Lewis rat kidney transplantation model, the blood pressure (BP) of the conscious transplant recipient was measured following the intravenous administration of Ang II. In addition, the mechanisms underlying the Ang II-mediated vascular contractility via the type 1 and type 2 Ang II receptors (AT1R and AT2R, respectively) in large and small-resistance blood vessels were determined in the recipient after renal transplantation. The results showed that renal transplantation significantly increased the Ang II-stimulated BP of the rats. Additionally, ex vivo contractility experiments using aorta and mesenteric arteries revealed that the contractions induced by Ang II were significantly strengthened in the recipient following renal transplantation, and were associated with an increased intracellular Ca2+ concentration. Losartan almost eradicated the Ang II-induced contractions whereas PD-123319 had no apparent effects on the Ang II-induced contractions in the aorta and mesenteric arteries of the recipient. Furthermore, the expression levels of AT1R but not AT2R were significantly increased in the vasculature of the recipient following renal transplantation, which exhibited a close association with selective DNA demethylation detected in the promoter region of the vascular AT1aR gene. These results indicate that changes of recipient vascular AT1R gene expression, occurring through a mechanism involving DNA methylation, increase the vascular contractility in response to Ang II. This may lead to the increased risk of hypertension following renal transplantation.
Collapse
Affiliation(s)
- Yakun Zhao
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Qingguo Zhu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Shiping Sun
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yu Qiu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Jingquan Li
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wei Liu
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Gangjun Yuan
- Department of Urinary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hua Ma
- Surgical Department, The People's Hospital of Fuyun County, Aletai, Xinjiang 836100, P.R. China
| |
Collapse
|
24
|
Jin P, Chen H, Xie J, Zhou C, Zhu X. Essential role of microRNA-650 in the regulation of B-cell CLL/lymphoma 11B gene expression following transplantation: A novel mechanism behind the acute rejection of renal allografts. Int J Mol Med 2017; 40:1840-1850. [PMID: 29039465 PMCID: PMC5716404 DOI: 10.3892/ijmm.2017.3194] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 10/06/2017] [Indexed: 12/14/2022] Open
Abstract
Kidney transplantation is an effective final therapeutic procedure for patients with end-stage kidney failure. Although advanced immunosuppressive therapy is administered following transplantation, certain patients still suffer from acute allograft rejection. MicroRNAs (miRs) have a potential diagnostic and therapeutic value for acute renal allograft rejection; however, their underlying mechanism of action is largely unknown. In the present study, an increased level of miR-650 was identified to be associated with the downregulation of B-cell CLL/lymphoma 11B (BCL11B) expression in acute renal allograft rejection. Furthermore, in vitro study using human renal glomerular endothelial cells (HRGECs) transfected with a miR-650 mimic revealed that key characteristics of acute renal allograft rejection were observed, including apoptosis, the release of cytokines and the chemotaxis of macrophages, while the effects were reduced in HRGECs transfected with a miR-650 inhibitor. The existence of a conserved miR-650 binding site on the 3'-untranslated region of BCL11B mRNA was predicted by computational algorithms and confirmed by a luciferase reporter assay. Knockdown of BCL11B with small interfering RNA (siRNA) significantly increased the apoptotic rate and significantly decreased the proliferation ability of HRGECs compared with the negative control group. HRGECs transfected with a combination of BCL11B siRNA and the miR-650 mimic demonstrated a significant increase in the rate of apoptosis compared with the control. These results suggest that the upregulation of miR-650 contributes to the development of acute renal allograft rejection by suppression of BCL11B, which leads to apoptosis and inflammatory responses. Thus, miR-650 and BCL11B may represent potential therapeutic targets for the prevention of acute renal allograft rejection.
Collapse
Affiliation(s)
- Peng Jin
- Centre of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hongxi Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jinliang Xie
- Centre of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Cheng Zhou
- Centre of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiangrong Zhu
- Centre of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
25
|
Stapleton CP, Conlon PJ, Phelan PJ. Using omics to explore complications of kidney transplantation. Transpl Int 2017; 31:251-262. [PMID: 28892567 DOI: 10.1111/tri.13067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/26/2017] [Accepted: 09/05/2017] [Indexed: 12/12/2022]
Abstract
The importance of genetic and biochemical variation in renal transplant outcomes has been clear since the discovery of the HLA in the 1950s. Since that time, there have been huge advancements in both transplantation and omics. In recent years, there has seen an increased number of genome-, proteome- and transcriptome-wide studies in the field of transplantation moving away from the earlier candidate gene/protein approaches. These areas have the potential to lead to the development of personalized treatment depending on individual molecular risk profiles. Here, we discuss recent progress and the current literature surrounding omics and renal transplant complications.
Collapse
Affiliation(s)
- Caragh P Stapleton
- Department of Molecular and Cellular Therapeutics, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Peter J Conlon
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland.,Department of Medicine, The Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Paul J Phelan
- Department of Nephrology, Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, United Kingdom
| |
Collapse
|
26
|
Renal Protection Mediated by Hypoxia Inducible Factor-1α Depends on Proangiogenesis Function of miR-21 by Targeting Thrombospondin 1. Transplantation 2017; 101:1811-1819. [PMID: 28737660 PMCID: PMC5542793 DOI: 10.1097/tp.0000000000001501] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Angiogenesis contributes to the repair process after renal ischemia/reperfusion (I/R) injury. In the present study, we tested the role of miR-21 in the angiogenesis induced by hypoxia inducible factor (HIF)-1α through inhibiting a predicted target gene thrombospondin 1 (TSP-1). Methods To stabilize HIF-1α, hypoxia (1% O2 for 24 hours) was performed in human umbilical vein endothelial cells and cobalt chloride (CoCl2) was pretreated intraperitoneally 24 hours before renal I/R in mice. Locked nucleic acid modified anti-miR-21 and scrambled control was transfected with hypoxic cells or delivered into the mice via tail vein 1 hour before CoCl2 injection. The kidneys and blood were collected at 24 hours after reperfusion. Results HIF-1α induced by hypoxia and CoCl2 upregulated vascular endothelial growth factor and miR-21, and increased angiogenesis. It was found that expression of TSP-1 was inversely related with miR-21 in vitro and in vivo. Targeting of TSP-1 by miR-21 was further confirmed in vitro. Furthermore, HIF-1α improved renal function, accompanied with increased angiogenesis after I/R injury in mice. The protective effect of HIF-1α was attenuated by inhibition of miR-21. Conclusions HIF-1α induced angiogenesis by upregulating not only vascular endothelial growth factor but also miR-21 via inhibiting a novel target gene TSP-1. Both of them may contribute to the protective effect of HIF-1α on renal I/R injury. Hypoxia induces HIF-1α which upregulates not only VEGF but also miR-21, and this last one inhibits a novel target gene, thrombospondin 1. Angiogenesis induced by hypoxia depends at least partially on production of VEGF and inhibition of thrombospondin 1 through miR-21.
Collapse
|
27
|
Wang Z, Yang H, Suo C, Wei J, Tan R, Gu M. Application of Ultrasound Elastography for Chronic Allograft Dysfunction in Kidney Transplantation. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:1759-1769. [PMID: 28503746 DOI: 10.1002/jum.14221] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 11/28/2016] [Indexed: 05/21/2023]
Abstract
Interstitial fibrosis is the main characteristic of chronic allograft dysfunction, which remains the key factor affecting long-term allograft survival after kidney transplantation. Ultrasound elastography (UE), including real-time elastography, transient elastography, and acoustic radiation force impulse, has been applied widely in breast, thyroid, and liver diseases, especially in the assessment of liver fibrosis. Recently, numerous studies have reported the efficacy of UE methods in evaluating renal allograft fibrosis. This review aims to investigate the clinical applications, limitations, and future roles of UE in current clinical practice in light of changing management paradigms. In current clinical practice, UE methods, especially transient elastographic measurement, appear to be useful for ruling out fibrosis but do not have sufficient accuracy to distinguish between various stages of allograft fibrosis. Moreover, there remain considerable issues to be solved for the application of UE in kidney transplantation. Thus, UE methods cannot replace the crucial role of renal allograft biopsy in the diagnosis and evaluation of allograft fibrosis in kidney transplantation. Perhaps UE methods could be of more importance in the long-term observation and evaluation of allograft fibrosis during follow-up.
Collapse
Affiliation(s)
- Zijie Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Haiwei Yang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanjian Suo
- Department of Pharmacy, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jifu Wei
- Department of Pharmacy, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Gu
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Suarez-Alvarez B, Rodriguez RM, Ruiz-Ortega M, Lopez-Larrea C. BET Proteins: An Approach to Future Therapies in Transplantation. Am J Transplant 2017; 17:2254-2262. [PMID: 28173625 DOI: 10.1111/ajt.14221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 01/13/2017] [Accepted: 01/31/2017] [Indexed: 01/25/2023]
Abstract
In order to develop new efficient therapies for organ transplantation, it is essential to acquire a comprehensive knowledge of the molecular mechanisms and processes, such as immune activation, chronic inflammation, and fibrosis, which lead to rejection and long-term graft loss. Recent efforts have shed some light on the epigenetic regulation associated with these processes. In this context, the bromo and extraterminal (BET) family of bromodomain proteins (BRD2, BRD3, BRD4, and BRDT) have emerged as major epigenetic players, connecting chromatin structure with gene expression changes. These proteins recognize acetylated lysines in histones and master transcription factors to recruit regulatory complex and, finally, modify the transcriptional program. Recent studies indicate that BET proteins are essential in the NF-kB-mediated inflammatory response, during the activation and differentiation of Th17-immune cells, and in profibrotic processes. Here, we review this new body of data and highlight the efficiency of BET inhibitors in several models of diseases. The promising results obtained from these preclinical models indicate that it may be time to translate these outcomes to the transplantation field, where epigenetics will be of increasing value in the coming years.
Collapse
Affiliation(s)
- B Suarez-Alvarez
- Department of Immunology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - R M Rodriguez
- Department of Immunology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - M Ruiz-Ortega
- Cellular Biology of Renal Disease Laboratory, Nephrology Department, Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - C Lopez-Larrea
- Department of Immunology, Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
29
|
Peters FS, Peeters AMA, Hofland LJ, Betjes MGH, Boer K, Baan CC. Interferon-Gamma DNA Methylation Is Affected by Mycophenolic Acid but Not by Tacrolimus after T-Cell Activation. Front Immunol 2017; 8:822. [PMID: 28747916 PMCID: PMC5506181 DOI: 10.3389/fimmu.2017.00822] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022] Open
Abstract
Immunosuppressive drug therapy is required to treat patients with autoimmune disease and patients who have undergone organ transplantation. The main targets of the immunosuppressive drugs tacrolimus and mycophenolic acid (MPA; the active metabolite of mycophenolate mofetil) are T cells. It is currently unknown whether these immunosuppressive drugs have an effect on DNA methylation—an epigenetic regulator of cellular function. Here, we determined the effect of tacrolimus and MPA on DNA methylation of the gene promoter region of interferon gamma (IFNγ), a pro-inflammatory cytokine. Total T cells, naive T cells (CCR7+CD45RO−), and memory T cells (CD45RO+ and CCR7−CD45RO−) were isolated from CMV seropositive healthy controls and stimulated with α-CD3/CD28 in the presence or absence of tacrolimus or MPA. DNA methylation of the IFNγ promoter region was quantified by pyrosequencing at 4 h, days 1, 3, and 4 after stimulation. In parallel, T-cell differentiation, and IFNγ protein production were analyzed by flow cytometry at days 1 and 3 after stimulation. Our results show that MPA induced changes in IFNγ DNA methylation of naive T cells; MPA counteracted the decrease in methylation after stimulation. Tacrolimus did not affect IFNγ DNA methylation of naive T cells. In the memory T cells, both immunosuppressive drugs did not affect IFNγ DNA methylation. Differentiation of naive T cells into a central-memory-like phenotype (CD45RO+) was inhibited by both immunosuppressive drugs, while differentiation of memory T cells remained unaffected by both MPA and tacrolimus. IFNγ protein production was suppressed by tacrolimus. Our results demonstrate that MPA influenced IFNγ DNA methylation of naive T cells after stimulation of T cells, while tacrolimus had no effect. Both tacrolimus and MPA did not affect IFNγ DNA methylation of memory T cells.
Collapse
Affiliation(s)
- Fleur S Peters
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Annemiek M A Peeters
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Leo J Hofland
- Endocrinology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Michiel G H Betjes
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Karin Boer
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| | - Carla C Baan
- Nephrology and Transplantation, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
30
|
Presence of Cytotoxic Extracellular Histones in Machine Perfusate of Donation After Circulatory Death Kidneys. Transplantation 2017; 101:e93-e101. [PMID: 27906828 DOI: 10.1097/tp.0000000000001590] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Extracellular histones are cytotoxic molecules that are related to cell stress and death. They have been shown to play a crucial role in multiple pathophysiologic processes like sepsis, inflammation, vascular dysfunction, and thrombosis. Their role in organ donation and graft function and survival is still unknown. The aim of this study was to assess whether an association exists between the presence of extracellular histones in machine perfusates and deceased donor kidney viability. METHODS Machine perfusates of 390 donations after circulatory death kidneys were analyzed for histone concentration, and corresponding graft function and survival were assessed. RESULTS Extracellular histone concentrations were significantly higher in perfusates of kidneys with posttransplant graft dysfunction (primary nonfunction and delayed graft function) and were an independent risk factor for delayed graft function (odds ratio, 2.152; 95% confidence interval [95% CI], 1.199-3.863) and 1 year graft failure (hazard ratio, 1.386; 95% CI, 1.037-1.853), but not for primary nonfunction (odds ratio, 1.342; 95% CI, 0.900-2.002). One year graft survival was 12% higher in the group with low histone concentrations (P = 0.008) as compared with the group that contained higher histone concentrations. CONCLUSIONS This study warrants future studies to probe for a possible role of cytotoxic extracellular histones in organ viability and suggests that quantitation of extracellular histones might contribute to assessment of posttransplant graft function and survival.
Collapse
|
31
|
Bontha SV, Maluf DG, Mueller TF, Mas VR. Systems Biology in Kidney Transplantation: The Application of Multi-Omics to a Complex Model. Am J Transplant 2017; 17:11-21. [PMID: 27214826 DOI: 10.1111/ajt.13881] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/15/2016] [Accepted: 05/12/2016] [Indexed: 01/25/2023]
Abstract
In spite of reduction of rejection rates and improvement in short-term survival post-kidney transplantation, modest progress has occurred in long-term graft attrition over the years. Timely identification of molecular events that precede clinical and histopathological changes might help in early intervention and thereby increase the graft half-life. Evolution of "omics" tools has enabled systemic investigation of the influence of the whole genome, epigenome, transcriptome, proteome and microbiome on transplant function and survival. In this omics era, systemic approaches, in-depth clinical phenotyping and use of strict validation methods are the key for further understanding the complex mechanisms associated with graft function. Systems biology is an interdisciplinary holistic approach that focuses on complex and dynamic interactions within biological systems. The complexity of the human kidney transplant is unlikely to be captured by a reductionist approach. It appears essential to integrate multi-omics data that can elucidate the multidimensional and multilayered regulation of the underlying heterogeneous and complex kidney transplant model. Herein, we discuss studies that focus on genetic biomarkers, emerging technologies and systems biology approaches, which should increase the ability to discover biomarkers, understand mechanisms and stratify patients and responses post-kidney transplantation.
Collapse
Affiliation(s)
- S V Bontha
- Translational Genomics Transplant Laboratory, Division of Transplant, Department of Surgery, University of Virginia, Charlottesville, VA
| | - D G Maluf
- Translational Genomics Transplant Laboratory, Division of Transplant, Department of Surgery, University of Virginia, Charlottesville, VA
| | - T F Mueller
- Division of Nephrology, University Hospital, Zürich, Switzerland
| | - V R Mas
- Translational Genomics Transplant Laboratory, Division of Transplant, Department of Surgery, University of Virginia, Charlottesville, VA
| |
Collapse
|
32
|
Boer K, de Wit LEA, Peters FS, Hesselink DA, Hofland LJ, Betjes MGH, Looman CWN, Baan CC. Variations in DNA methylation of interferon gamma and programmed death 1 in allograft rejection after kidney transplantation. Clin Epigenetics 2016; 8:116. [PMID: 27891189 PMCID: PMC5112717 DOI: 10.1186/s13148-016-0288-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/08/2016] [Indexed: 02/07/2023] Open
Abstract
Background The role of DNA methylation in the regulation of the anti-donor-directed immune response after organ transplantation is unknown. Here, we studied the methylation of two mediators of the immune response: the pro-inflammatory cytokine interferon γ (IFNγ) and the inhibitory receptor programmed death 1 (PD1) in naïve and memory CD8+ T cell subsets in kidney transplant recipients receiving immunosuppressive medication. Both recipients experiencing an episode of acute allograft rejection (rejectors) as well as recipients without rejection (non-rejectors) were included. Results CpGs in the promoter regions of both IFNγ and PD1 were significantly (p < 0.001) higher methylated in the naïve CD8+ T cells compared to the memory T cell subsets. The methylation status of both IFNγ and PD1 inversely correlated with the percentage of IFNγ or PD1-producing cells. Before transplantation, the methylation status of both IFNγ and PD1 was not significantly different from healthy donors. At 3 months after transplantation, irrespective of rejection and subsequent anti-rejection therapy, the IFNy methylation was significantly higher in the differentiated effector memory CD45RA+ (EMRA) CD8+ T cells (p = 0.01) whereas the PD1 methylation was significantly higher in all memory CD8+ T cell subsets (CD27+ memory; p = 0.02: CD27− memory; p = 0.02: EMRA; p = 0.002). Comparing the increase in methylation in the first 3 months after transplantation between rejectors and non-rejectors demonstrated a significantly more prominent increase in the PD1 methylation in the CD27− memory CD8+ T cells in rejectors (increase in rejectors 14%, increase in non-rejectors 1.9%, p = 0.04). The increase in DNA methylation in the other memory CD8+ T cells was not significantly different between rejectors and non-rejectors. At 12 months after transplantation, the methylation of both IFNγ and PD1 returned to baseline levels. Conclusions The DNA methylation of both IFNγ and PD1 increases the first 3 months after transplantation in memory CD8+ T cells in kidney transplant recipients. This increase was irrespective of a rejection episode indicating that general factors of the kidney transplantation procedure, including the use of immunosuppressive medication, contribute to these variations in DNA methylation.
Collapse
Affiliation(s)
- Karin Boer
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - L Elly A de Wit
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Fleur S Peters
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Dennis A Hesselink
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Internal Medicine, Section Endocrinology, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Michiel G H Betjes
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Caspar W N Looman
- Department of Public Health, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Section Nephrology and Transplantation, Erasmus MC, University Medical Center Rotterdam, Room Na520, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
33
|
Picascia A, Grimaldi V, Napoli C. From HLA typing to anti-HLA antibody detection and beyond: The road ahead. Transplant Rev (Orlando) 2016; 30:187-94. [DOI: 10.1016/j.trre.2016.07.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/07/2016] [Accepted: 07/22/2016] [Indexed: 01/27/2023]
|
34
|
Wang H, Huang N, Liu Y, Cang J, Xue Z. Genomic distribution of 5-Hydroxymethylcytosine in mouse kidney and its relationship with gene expression. Ren Fail 2016; 38:982-8. [DOI: 10.3109/0886022x.2016.1172973] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|