51
|
Raschzok N, Reutzel-Selke A, Schmuck RB, Tannus LM, Morgul MH, Dietel C, Leder A, Struecker B, Lippert S, Sallmon H, Schmelzle M, Bartels M, Jonas S, Pratschke J, Sauer IM. microRNA signatures in peripheral blood fail to detect acute cellular rejection after liver transplantation. Biomarkers 2016; 21:699-707. [DOI: 10.3109/1354750x.2016.1172113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Nathanael Raschzok
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Anja Reutzel-Selke
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Rosa B. Schmuck
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Laura M. Tannus
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Mehmet H. Morgul
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Department of Visceral, Transplantation, Thoracic, and Vascular Surgery, University of Leipzig, Leipzig, Germany
| | - Corinna Dietel
- Department of Visceral, Transplantation, Thoracic, and Vascular Surgery, University of Leipzig, Leipzig, Germany
| | - Annekatrin Leder
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Benjamin Struecker
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Steffen Lippert
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Hannes Sallmon
- Department of Neonatology, Charité – Universitätsmedizin Berlin, Germany
| | - Moritz Schmelzle
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Department of Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| | - Michael Bartels
- Department of Visceral, Transplantation, Thoracic, and Vascular Surgery, University of Leipzig, Leipzig, Germany
| | - Sven Jonas
- Centre Hépato-Biliaire, Hôpital Paul Brousse, Université Paris Sud, Villejuif, France
| | - Johann Pratschke
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| | - Igor M. Sauer
- Department of General, Visceral, and Transplantation Surgery, Experimental Surgery and Regenerative Medicine, Charité – Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
| |
Collapse
|
52
|
Venner JM, Famulski KS, Reeve J, Chang J, Halloran PF. Relationships among injury, fibrosis, and time in human kidney transplants. JCI Insight 2016; 1:e85323. [PMID: 27699214 DOI: 10.1172/jci.insight.85323] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Kidney transplant biopsies offer an opportunity to understand the pathogenesis of organ fibrosis. We studied the relationships between the time of biopsy after transplant (TxBx), histologic fibrosis, diseases, and transcript expression. METHODS Expression microarrays from 681 kidney transplant indication biopsies taken either early (n = 282, <1 year) or late (n = 399, >1 year) after transplant were used to analyze the molecular landscape of fibrosis in relationship to histologic fibrosis and diseases. RESULTS Fibrosis was absent at transplantation but was present in some early biopsies by 4 months after transplant, apparently as a self-limited response to donation implantation injury not associated with progression to failure. The molecular phenotype of early biopsies represented the time sequence of the response to wounding: immediate expression of acute kidney injury transcripts, followed by fibrillar collagen transcripts after several weeks, then by the appearance of immunoglobulin and mast cell transcripts after several months as fibrosis appeared. Fibrosis in late biopsies correlated with injury, fibrillar collagen, immunoglobulin, and mast cell transcripts, but these were independent of time. Pathway analysis revealed epithelial response-to-wounding pathways such as Wnt/β-catenin. CONCLUSION Fibrosis in late biopsies had different associations because many kidneys had potentially progressive diseases and subsequently failed. Molecular correlations with fibrosis in late biopsies were independent of time, probably because ongoing injury obscured the response-to-wounding time sequence. The results indicate that fibrosis in kidney transplants is driven by nephron injury and that progression to failure reflects continuing injury, not autonomous fibrogenesis. TRIAL REGISTRATION INTERCOM study (www.clinicalTrials.gov; NCT01299168). FUNDING Canada Foundation for Innovation and Genome Canada.
Collapse
Affiliation(s)
- Jeffery M Venner
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, Edmonton, Alberta, Canada
| | - Konrad S Famulski
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Jeff Reeve
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Jessica Chang
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada
| | - Philip F Halloran
- Alberta Transplant Applied Genomics Centre, Edmonton, Alberta, Canada.,Department of Medicine, Division of Nephrology and Transplant Immunology, Edmonton, Alberta, Canada
| |
Collapse
|
53
|
Bruneau S, Wedel J, Fakhouri F, Nakayama H, Boneschansker L, Irimia D, Daly KP, Briscoe DM. Translational implications of endothelial cell dysfunction in association with chronic allograft rejection. Pediatr Nephrol 2016; 31:41-51. [PMID: 25903640 PMCID: PMC4619184 DOI: 10.1007/s00467-015-3094-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/03/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Advances in therapeutics have dramatically improved short-term graft survival, but the incidence of chronic rejection has not changed in the past 20 years. New insights into mechanism are sorely needed at this time and it is hoped that the development of predictive biomarkers will pave the way for the emergence of preventative therapeutics. In this review, we discuss a paradigm suggesting that sequential changes within graft endothelial cells (EC) lead to an intragraft microenvironment that favors the development of chronic rejection. Key initial events include EC injury, activation and uncontrolled leukocyte-induced angiogenesis. We propose that all of these early changes in the microvasculature lead to abnormal blood flow patterns, local tissue hypoxia, and an associated overexpression of HIF-1α-inducible genes, including vascular endothelial growth factor. We also discuss how cell intrinsic regulators of mTOR-mediated signaling within EC are of critical importance in microvascular stability and may thus have a role in the inhibition of chronic rejection. Finally, we discuss recent findings indicating that miRNAs may regulate EC stability, and we review their potential as novel non-invasive biomarkers of allograft rejection. Overall, this review provides insights into molecular events, genes, and signals that promote chronic rejection and their potential as biomarkers that serve to support the future development of interruption therapeutics.
Collapse
Affiliation(s)
- Sarah Bruneau
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- INSERM UMR S-1064, Institut de Transplantation Urologie-Nephrologie (ITUN), Centre Hospitalier Universitaire (CHU) de Nantes, University of Nantes, Nantes, France
| | - Johannes Wedel
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Fadi Fakhouri
- INSERM UMR S-1064, Institut de Transplantation Urologie-Nephrologie (ITUN), Centre Hospitalier Universitaire (CHU) de Nantes, University of Nantes, Nantes, France
| | - Hironao Nakayama
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Leo Boneschansker
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA
| | - Daniel Irimia
- Department of Surgery, BioMEMS Resource Center, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA, USA
| | - Kevin P Daly
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David M Briscoe
- Transplant Research Program, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
54
|
|
55
|
Identification of Circulating miRNAs in a Mouse Model of Nerve Allograft Transplantation under FK506 Immunosuppression by Illumina Small RNA Deep Sequencing. DISEASE MARKERS 2015; 2015:863192. [PMID: 26435568 PMCID: PMC4578739 DOI: 10.1155/2015/863192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/08/2015] [Indexed: 12/24/2022]
Abstract
Background. This study aimed to establish the expression profile of circulating microRNAs (miRNAs) during nerve allotransplantation in the presence and absence of FK506 immunosuppression. Methods. A 1 cm BALB/c donor sciatic nerve graft was transplanted into the sciatic nerve gaps created in recipient C57BL/6 mice with or without daily FK506 immunosuppression [1 mg/(kg·d)]. At 3, 7, and 14 d after nerve allotransplantation, serum samples were collected for miRNA expression analysis by Illumina small RNA deep sequencing. Results. Sequence analysis showed that the dominant size of circulating small RNAs after nerve allotransplantation was 22 nucleotides, followed by 23-nucleotide sequences. Nine upregulated circulating miRNAs (let-7e-5p, miR-101a-3p, miR-151-5p, miR-181a-5p, miR-204-5p, miR-340-5p, miR-381-3p, miR-411-5p, miR-9-5p, and miR-219-2-3p) were identified at 3 d, but none was identified at 7 or 14 d. Among them, miR-9-5p had the highest fold-change of >50-fold, followed by miR-340-5p with 38.8-fold. The presence of these nine miRNAs was not significant at 7 and 14 d after nerve allotransplantation with or without immunosuppression, showing that these miRNAs are not ideal biomarkers for monitoring rejection of deep-buried nerve allografts, a response usually observed later. Conclusions. We identified nine upregulated circulating miRNAs, which may have a biological function, particularly during the early stages after nerve allotransplantation under FK506 immunosuppression.
Collapse
|
56
|
Serum MicroRNA-99a Helps Detect Acute Rejection in Renal Transplantation. Transplant Proc 2015; 47:1683-7. [DOI: 10.1016/j.transproceed.2015.04.094] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 03/13/2015] [Accepted: 04/07/2015] [Indexed: 12/12/2022]
|
57
|
Mastoridis S, Martínez-Llordella M, Sanchez-Fueyo A. Emergent Transcriptomic Technologies and Their Role in the Discovery of Biomarkers of Liver Transplant Tolerance. Front Immunol 2015; 6:304. [PMID: 26157438 PMCID: PMC4476276 DOI: 10.3389/fimmu.2015.00304] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/27/2015] [Indexed: 01/20/2023] Open
Abstract
Liver transplantation offers a unique window into transplant immunology due, in part, to the considerable proportion of recipients who develop immunological tolerance to their allograft. Biomarkers are able to identify and predict such a state of tolerance, and thereby able to establish suitable candidates for the minimization of hazardous immunosuppressive therapies, are not only of great potential clinical benefit but might also shed light on the immunological mechanisms underlying tolerance and rejection. Here, we review the emergent transcriptomic technologies serving as drivers of biomarker discovery, we appraise efforts to identify a molecular signature of liver allograft tolerance, and we consider the implications of this work on the mechanistic understanding of immunological tolerance.
Collapse
|
58
|
Molecular monitoring of alloimmune-mediated injury in kidney transplant patients. Curr Opin Nephrol Hypertens 2015; 23:625-30. [PMID: 25202838 DOI: 10.1097/mnh.0000000000000064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Rapid progress in molecular technology has allowed development of numerous molecular tools to help the clinician to evaluate graft status in kidney transplant patients. This review highlights recent findings, describing the use of molecular approaches to monitor, diagnose, and predict alloimmune-mediated injury in kidney grafts. RECENT FINDINGS Both previously identified and newly discovered molecular markers of immune injury have been studied and validated in large multicenter studies. Recent data indicate that measuring specific gene transcripts in noninvasive samples, such as urine or peripheral blood, can identify the occurrence of acute rejection and differentiate this immune-mediated injury from other causes of graft dysfunction. Serial monitoring of urine in stable renal transplant patients may detect the onset of rejection before development of graft dysfunction. Moreover, combining gene expression analysis with conventional histopathologic assessment of grafts can enhance the accuracy of diagnosis and may also help predict graft outcomes. SUMMARY Measuring specific gene transcription in noninvasive clinical samples has the potential to become an important and standard tool to monitor alloimmune-mediated injury in kidney transplant recipients. Prospective studies are ongoing to validate these findings for use of these approaches in clinical settings.
Collapse
|
59
|
Khorsandi SE, Quaglia A, Salehi S, Jassem W, Vilca-Melendez H, Prachalias A, Srinivasan P, Heaton N. The microRNA Expression Profile in Donation after Cardiac Death (DCD) Livers and Its Ability to Identify Primary Non Function. PLoS One 2015; 10:e0127073. [PMID: 25978529 PMCID: PMC4433116 DOI: 10.1371/journal.pone.0127073] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/10/2015] [Indexed: 01/06/2023] Open
Abstract
Donation after cardiac death (DCD) livers are marginal organs for transplant and their use is associated with a higher risk of primary non function (PNF) or early graft dysfunction (EGD). The aim was to determine if microRNA (miRNA) was able to discriminate between DCD livers of varying clinical outcome. DCD groups were categorized as PNF retransplanted within a week (n=7), good functional outcome (n=7) peak aspartate transaminase (AST) ≤ 1000 IU/L and EGD (n=9) peak AST ≥ 2500 IU/L. miRNA was extracted from archival formalin fixed post-perfusion tru-cut liver biopsies. High throughput expression analysis was performed using miRNA arrays. Bioinformatics for expression data analysis was performed and validated with real time quantitative PCR (RT-qPCR). The function of miRNA of interest was investigated using computational biology prediction algorithms. From the array analysis 16 miRNAs were identified as significantly different (p<0.05). On RT-qPCR miR-155 and miR-940 had the highest expression across all three DCD clinical groups. Only one miRNA, miR-22, was validated with marginal significance, to have differential expression between the three groups (p=0.049). From computational biology miR-22 was predicted to affect signalling pathways that impact protein turnover, metabolism and apoptosis/cell cycle. In conclusion, microRNA expression patterns have a low diagnostic potential clinically in discriminating DCD liver quality and outcome.
Collapse
Affiliation(s)
| | - Alberto Quaglia
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Siamak Salehi
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Wayel Jassem
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | | | - Andreas Prachalias
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Parthi Srinivasan
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| | - Nigel Heaton
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
- * E-mail:
| |
Collapse
|
60
|
Schlickeiser S, Boës D, Streitz M, Sawitzki B. The use of novel diagnostics to individualize immunosuppression following transplantation. Transpl Int 2015; 28:911-20. [PMID: 25611562 DOI: 10.1111/tri.12527] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 12/19/2014] [Accepted: 01/16/2015] [Indexed: 12/14/2022]
Abstract
Despite major improvements in short-term survival of organ allografts, long-term graft survival has not changed significantly. It is also known that toxic side effects of current immunosuppressive drugs (IS) especially calcineurin inhibitors (CNI) contribute to the unsatisfactory graft and patient survival following transplantation. Thus, clinicians strive to reduce or wean IS in potentially eligible patients. Research in the last 10 years has focussed on identification of biomarkers suitable for patient stratification in minimization or weaning trials. Most of the described biomarkers have been run retrospectively on samples collected within single-centre trials. Thus, often their performance has not been validated in other potentially multicentre clinical trials. Ultimately, the utility of biomarkers to identify potential weaning candidates should be investigated in large randomized prospective trials. In particular, for testing in such trials, we need more information about the accuracy, reproducibility, stability and limitations of the described biomarkers. Also, data repositories summarizing crucial information on biomarker performance in age- and gender-matched healthy individuals of different ethnicity are missing. This together with improved bioinformatics tools might help in developing better scores for patient stratification. Here, we will summarize the current results, knowledge and limitations on biomarkers for drug minimization or weaning trials.
Collapse
Affiliation(s)
- Stephan Schlickeiser
- Institute of Medical Immunology, CCM, Charité University Berlin, Berlin, Germany
| | - David Boës
- Institute of Medical Immunology, CCM, Charité University Berlin, Berlin, Germany
| | - Mathias Streitz
- Institute of Medical Immunology, CCM, Charité University Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Institute of Medical Immunology, CCM, Charité University Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), CVK, Charité University Berlin, Berlin, Germany
| |
Collapse
|
61
|
Brenu EW, Ashton KJ, Batovska J, Staines DR, Marshall-Gradisnik SM. High-throughput sequencing of plasma microRNA in chronic fatigue syndrome/myalgic encephalomyelitis. PLoS One 2014; 9:e102783. [PMID: 25238588 PMCID: PMC4169517 DOI: 10.1371/journal.pone.0102783] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 04/29/2014] [Indexed: 12/17/2022] Open
Abstract
Background MicroRNAs (miRNAs) are known to regulate many biological processes and their dysregulation has been associated with a variety of diseases including Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME). The recent discovery of stable and reproducible miRNA in plasma has raised the possibility that circulating miRNAs may serve as novel diagnostic markers. The objective of this study was to determine the role of plasma miRNA in CFS/ME. Results Using Illumina high-throughput sequencing we identified 19 miRNAs that were differentially expressed in the plasma of CFS/ME patients in comparison to non-fatigued controls. Following RT-qPCR analysis, we were able to confirm the significant up-regulation of three miRNAs (hsa-miR-127-3p, hsa-miR-142-5p and hsa-miR-143-3p) in the CFS/ME patients. Conclusion Our study is the first to identify circulating miRNAs from CFS/ME patients and also to confirm three differentially expressed circulating miRNAs in CFS/ME patients, providing a basis for further study to find useful CFS/ME biomarkers.
Collapse
Affiliation(s)
- Ekua W. Brenu
- School of Medical Science, Griffith Health Centre, Griffith University, Gold Coast, Queensland, Australia
- The National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, Queensland, Australia
- * E-mail:
| | - Kevin J. Ashton
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Jana Batovska
- Faculty of Health Sciences and Medicine, Bond University, Robina, Queensland, Australia
| | - Donald R. Staines
- The National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, Queensland, Australia
- Queensland Health, Gold Coast Public Health Unit, Robina, Gold Coast, Queensland, Australia
| | - Sonya M. Marshall-Gradisnik
- School of Medical Science, Griffith Health Centre, Griffith University, Gold Coast, Queensland, Australia
- The National Centre for Neuroimmunology and Emerging Diseases, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
62
|
Seki A, Fishbein MC. Predicting the development of cardiac allograft vasculopathy. Cardiovasc Pathol 2014; 23:253-60. [DOI: 10.1016/j.carpath.2014.05.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 12/11/2022] Open
|
63
|
Mengel M. Renalomics: Molecular Pathology in Kidney Biopsies. Surg Pathol Clin 2014; 7:443-55. [PMID: 26837449 DOI: 10.1016/j.path.2014.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this article, various omics technologies and their applications in renal pathology (native and transplant biopsies) are reviewed and discussed. Despite significant progress and novel insights derived from these applications, extensive adoption of molecular diagnostics in renal pathology has not been accomplished. Further validation of specific applications leading to increased diagnostic precision in a clinically relevant way is ongoing.
Collapse
Affiliation(s)
- Michael Mengel
- Department of Laboratory Medicine and Pathology, University of Alberta Hospital, 4B1.18 Walter Mackenzie Center, 8440-112 Street, Edmonton T6G2S2, Canada.
| |
Collapse
|
64
|
Wang Y, Tian Y, Ding Y, Wang J, Yan S, Zhou L, Xie H, Chen H, Li H, Zhang J, Zhao J, Zheng S. MiR-152 may silence translation of CaMK II and induce spontaneous immune tolerance in mouse liver transplantation. PLoS One 2014; 9:e105096. [PMID: 25133393 PMCID: PMC4136864 DOI: 10.1371/journal.pone.0105096] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 07/19/2014] [Indexed: 01/08/2023] Open
Abstract
Spontaneous immune tolerance in mouse liver transplantation has always been a hotspot in transplantation-immune research. Recent studies revealed that regulatory T cells (Tregs), hepatic satellite cells and Kupffer cells play a potential role in spontaneous immune tolerance, however the precise mechanism of spontaneous immune tolerance is still undefined. By using Microarray Chips, we investigated different immune regulatory factors to decipher critical mechanisms of spontaneous tolerance after mouse liver transplantation. Allogeneic (C57BL/6-C3H) and syngeneic (C3H-C3H) liver transplantation were performed by 6-8 weeks old male C57BL/6 and C3H mice. Graft samples (N = 4 each group) were collected from 8 weeks post-operation mice. 11 differentially expressed miRNAs in allogeneic grafts (Allografts) vs. syngeneic grafts (Syngrafts) were identified using Agilent Mouse miRNA Chips. It was revealed that 185 genes were modified by the 11 miRNAs, furthermore, within the 185 target genes, 11 of them were tightly correlated with immune regulation after Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and Genbank data cross-comparison. Verified by real-time PCR and western blot, our results indicated that mRNA expression levels of IL-6 and TAB2 were respectively down regulated following miR-142-3p and miR-155 augment. In addition, increased miR-152 just silenced mRNA of CaMK II and down-regulated translation of CaMK II in tolerated liver grafts, which may play a critical role in immune regulation and spontaneous tolerance induction of mouse liver transplantation.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Tian
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Ding
- Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingcheng Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Yan
- Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Zhou
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haiyang Xie
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Chen
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Li
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhua Zhang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiacong Zhao
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- * E-mail:
| |
Collapse
|
65
|
Knockdown of microRNA-155 in Kupffer cells results in immunosuppressive effects and prolongs survival of mouse liver allografts. Transplantation 2014; 97:626-35. [PMID: 24531847 DOI: 10.1097/tp.0000000000000061] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Our previous studies have shown that Kupffer cells (KCs) play a crucial role in postoperative pathologic changes. Recent reports have demonstrated that microRNA-155 (miR-155) is associated with inflammation and upregulation of proinflammatory mediators in the peripheral blood and allografts of transplant patients. However, the precise mechanism for this remains unknown. METHODS KCs isolated from BALB/c mice were transfected with miR-155 mimic or inhibitor. Levels of suppressor of cytokine signaling 1/Janus kinase/signal transducer and activator of transcription (SOCS1/JAK/STAT) proteins and surface molecules (MHC-II, CD40, and CD86) were then measured. T-cell proliferation and apoptosis were evaluated in mixed lymphocyte reactions. Orthotopic liver transplantation was performed in mice after miR-155 short hairpin RNA lentivirus treatment, and postoperative survival, liver function and histology, and mRNA and protein expression were analyzed. RESULTS miR-155 knockdown in KCs decreased MHC-II, CD40, and CD86 expression, suppressed antigen-presenting function, and affected SOCS1/JAK/STAT inflammatory pathways. In addition, KCs transfected with miR-155 inhibitor and cocultured with T lymphocytes showed reduced T-cell responses but a greater number of apoptotic T cells. Finally, miR-155 suppression in graft liver prolonged liver allograft survival and improved liver function. The changes were closely associated with the levels of T helper 1 and 2 (Th1/Th2) cytokines and T-cell apoptosis, but a direct mechanistic link in vivo was not established. CONCLUSION These data suggest miR-155 regulates the balance of Th1/Th2 cytokines and the maturation and function of KCs in mice. miR-155 repression in KCs positively regulates KC function toward immunosuppression and prolongs liver allograft survival.
Collapse
|
66
|
Abstract
BACKGROUND Acute pyelonephritis (APN) versus acute rejection (AR) is a frequently encountered diagnostic and therapeutic dilemma in kidney transplants. Variable culture results, overlapping histologic features, and persistent graft dysfunction despite antibiotics are frequently encountered. Therefore, we explored the utility of intragraft microRNA profiles to distinguish between allograft APN and AR. MATERIALS AND METHODS Between 2003 and 2011, we identified 49 patients with biopsy features of APN, within the first 2 years posttransplant. MicroRNA profiling was performed on 20 biopsies (normal kidney, n=4; unequivocal AR, n=5; features of APN, n=11). RESULTS Only 32% (16/49) of the patients had concomitant positive urine cultures at biopsy, and in 8 of 16 patients, colony count was less than 10 CFU/mL. In 14 of 49 patients, positive urine culture did not coincide with the biopsy, and in 19 of 49 patients, urine cultures were negative. On microRNA profiling, good clustering was seen among the normal kidneys and among AR biopsies. Among the 11 biopsies with features of APN, 4 biopsies showed good clustering with a pattern distinct from AR; (these patients recovered graft function with antibiotics); 7 of 11 biopsies showed heterogeneity in microRNA profiles and variable outcomes with antibiotic treatment. We identified a panel of 25 microRNAs showing statistical difference in expression between AR and APN. MiR-99b, miR-23b let-7b-5p, miR-30a, and miR-145 were validated using qPCR. CONCLUSION Allograft pyelonephritis can be a diagnostic and therapeutic challenge. A gestalt approach is required. In addition to histology and cultures, differential intragraft microRNA expression may prove helpful to distinguish APN from AR in renal allograft biopsies.
Collapse
|
67
|
|
68
|
Hashmi SK, Baranov E, Gonzalez A, Olthoff K, Shaked A. Genomics of liver transplant injury and regeneration. Transplant Rev (Orlando) 2014; 29:23-32. [PMID: 24746681 DOI: 10.1016/j.trre.2014.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/19/2014] [Indexed: 12/21/2022]
Abstract
While improved surgical techniques, post-operative care, and immunosuppression regimens have reduced morbidity and mortality associated with orthotopic liver transplantation (OLT), further improvement of outcomes requires personalized treatment and a better understanding of genomic mechanisms involved. Gene expression profiles of ischemia/reperfusion (I/R) injury, regeneration, and rejection, may suggest mechanisms for development of better predictive tools and treatments. The liver is unique in its regenerative potential, recovering lost mass and function after injury from ischemia, resection, and rejection. I/R injury, an inevitable consequence of perfusion cessation, cold storage, and reperfusion, is regulated by the interaction of the immune system, inflammatory cytokines, and reduced microcirculatory blood flow in the liver. Rejection, a common post-operative complication, is mediated by the recipient's immune system through T-cell-dependent responses activating proinflammatory and apoptotic pathways. Characterizing distinctive gene expression signatures for these events can identify therapies to reduce injury, promote regeneration, and improve outcomes. While certain markers of liver injury and regeneration have been observed in animals, many of these are unverified in human studies. Further investigation of these genomic signatures and mechanisms through new technology offers promise, but continues to pose a significant challenge. An overview of the current fund of knowledge in this area is reviewed.
Collapse
Affiliation(s)
- Sohaib Khalid Hashmi
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Esther Baranov
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Ana Gonzalez
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Kim Olthoff
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Abraham Shaked
- Penn Transplant Institute, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
69
|
Abstract
The immune management of organ transplant recipients is imperfect. Beyond general dosing guidelines for immunosuppressive agents and clinical diagnostic tests for rejection or infection, there are few objective tools to determine the aggregate status of a patient's alloimmune response or protective immune capacity. The lack of prognostic precision significantly contributes to patient morbidity and reduces long-term allograft survival after kidney transplantation. Noninvasive biomarkers that could serve as predictive tools or surrogate end points for rejection might help clinicians individualize immunosuppression and allow for early intervention, ideally prior to clinically evident organ dysfunction. Although the growing understanding of organ rejection has provided numerous candidate biomarkers, none has been confirmed in robust validation studies as sufficiently useful to guide clinical practice independent of traditional clinical methods. In this Review, the general characteristics of biomarkers and surrogate end points; current biomarkers under active clinical investigation; and the prominent barriers to the translation of biomarkers into clinical practice are discussed.
Collapse
Affiliation(s)
- Denise J Lo
- Emory Transplant Center, Emory University, 101 Woodruff Circle, #5105-WMB, Atlanta, GA 30322, USA
| | - Bruce Kaplan
- University of Kansas Medical Center, Center for Transplantation, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - Allan D Kirk
- Emory Transplant Center, Emory University, 101 Woodruff Circle, #5105-WMB, Atlanta, GA 30322, USA
| |
Collapse
|
70
|
Abstract
Acute graft-versus-host disease (aGVHD) is the leading cause of morbidity and mortality after allogeneic hematopoietic cell transplantation (HCT). Approximately 35% to 50% of HCT recipients develop aGVHD; however, there are no validated diagnostic and predictive blood biomarkers for aGVHD in clinical use. Here, we show that plasma samples from aGVHD patients have a distinct microRNA (miRNA) expression profile. We found that 6 miRNAs (miR-423, miR-199a-3p, miR-93*, miR-377, miR-155, and miR-30a) were significantly upregulated in the plasma of aGVHD patients (n = 116) when compared with non-GVHD patients (n = 52) in training and validation phases. We have developed a model including 4 miRNAs (miR-423, miR-199a-3p, miR-93*, and miR-377) that can predict the probability of aGVHD with an area under the curve of 0.80. Moreover, these elevated miRNAs were detected before the onset of aGVHD (median = 16 days before diagnosis). In addition, the levels of these miRNAs were positively associated with aGVHD severity, and high expression of the miRNA panel was associated with poor overall survival. Furthermore, the miRNA signature for aGVHD was not detected in the plasma of lung transplant or nontransplant sepsis patients. Our results have identified a specific plasma miRNA signature that may serve as an independent biomarker for the prediction, diagnosis, and prognosis of aGVHD.
Collapse
|
71
|
Rau CS, Yang JCS, Wu SC, Chen YC, Lu TH, Lin MW, Wu YC, Tzeng SL, Wu CJ, Hsieh CH. Profiling circulating microRNA expression in a mouse model of nerve allotransplantation. J Biomed Sci 2013; 20:64. [PMID: 24011263 PMCID: PMC3844622 DOI: 10.1186/1423-0127-20-64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 08/13/2013] [Indexed: 01/09/2023] Open
Abstract
Background The lack of noninvasive biomarkers of rejection remains a challenge in the accurate monitoring of deeply buried nerve allografts and precludes optimization of therapeutic intervention. This study aimed to establish the expression profile of circulating microRNAs (miRNAs) during nerve allotransplantation with or without immunosuppression. Results Balb/c mice were randomized into 3 experimental groups, that is, (1) untreated isograft (Balb/c → Balb/c), (2) untreated allograft (C57BL/6 → Balb/c), and (3) allograft (C57BL/6 → Balb/c) with FK506 immunosuppression. A 1-cm Balb/c or C57BL/6 donor sciatic nerve graft was transplanted into sciatic nerve gaps created in recipient mice. At 1, 3, 7, 10, and 14 d after nerve transplantation, nerve grafts, whole blood, and sera were obtained for miRNA expression analysis with an miRNA array and subsequent validation with quantitative real-time PCR (qRT-PCR). Three circulating miRNAs (miR-320, miR-762, and miR-423-5p) were identified in the whole blood and serum of the mice receiving an allograft with FK506 immunosuppression, within 2 weeks after nerve allotransplantation. However, these 3 circulating miRNAs were not expressed in the nerve grafts. The expression of all these 3 upregulated circulating miRNAs significantly decreased at 2, 4, and 6 d after discontinuation of FK506 immunosuppression. In the nerve graft, miR-125-3b and miR-672 were significantly upregulated in the mice that received an allograft with FK506 only at 7 d after nerve allotransplantation. Conclusions We identified the circulating miR-320, miR-762, and miR-423-5p as potential biomarkers for monitoring the immunosuppression status of the nerve allograft. However, further research is required to investigate the mechanism behind the dysregulation of these markers and to evaluate their prognostic value in nerve allotransplantation.
Collapse
Affiliation(s)
- Cheng-Shyuan Rau
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Center for Vascularized Composite Allotransplantation, No, 123, Ta-Pei Road, Kaohsiung City, Niao-Sung District, 833, Taiwan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Zununi S, Ardalan M. MicroRNA and Renal Allograft Monitoring. Nephrourol Mon 2013; 5:783-786. [PMID: 24282786 PMCID: PMC3830902 DOI: 10.5812/numonthly.12722] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Accepted: 06/10/2013] [Indexed: 12/24/2022] Open
Affiliation(s)
- Sepide Zununi
- Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, IR Iran
| | - Mohammadreza Ardalan
- Chronic Kidney Disease Research Center, Tabriz University of Medical Sciences, Tabriz, IR Iran
| |
Collapse
|