1
|
Zheng L, Han S, Enriquez J, Martinez OM, Krams SM. Graft-derived extracellular vesicles transport miRNAs to modulate macrophage polarization after heart transplantation. Am J Transplant 2025; 25:682-694. [PMID: 39586401 PMCID: PMC11972891 DOI: 10.1016/j.ajt.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Heart transplantation, a crucial intervention for saving lives of those with end-stage cardiac failure, often faces complications from acute allograft rejection. This study focuses on the intricate dynamics of immune cell interactions and specific communication pathways between organs, which are not yet well understood. Our study investigates this interplay using a murine heterotopic transplant model, using single-cell RNA sequencing to examine CD45+ immune cells from both the heart grafts and spleens. We conduct a comprehensive analysis focused on functional enrichment, cell trajectory, and interorgan communication in heart transplants, highlighting dynamic interactions between monocyte/macrophage subtypes that is mediated by extracellular vesicles (EVs). We use unsupervised clustering and elucidate the complex cellular interactions that influence allograft outcomes. Notably, we discovered that microRNA-363 and microRNA-709, carried by EVs from CD63+ graft macrophages, can induce M1 polarization within the recipient's spleen via the Fcho2/Notch1 signaling pathway. These insights illuminate the nuanced immune responses during acute cardiac rejection and suggest that targeting EVs from graft-resident macrophages may offer a new strategy to mitigate transplant rejection.
Collapse
Affiliation(s)
- Lei Zheng
- Transplant Immunology Lab, Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA; Department of General Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China
| | - Shuling Han
- Transplant Immunology Lab, Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jeanna Enriquez
- Transplant Immunology Lab, Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Olivia M Martinez
- Transplant Immunology Lab, Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Sheri M Krams
- Transplant Immunology Lab, Division of Abdominal Transplantation, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
2
|
Pei G, Chen L, Wang Y, He C, Fu C, Wei Q. Role of miR-182 in cardiovascular and cerebrovascular diseases. Front Cell Dev Biol 2023; 11:1181515. [PMID: 37228653 PMCID: PMC10203221 DOI: 10.3389/fcell.2023.1181515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023] Open
Abstract
The treatment of cardiovascular and cerebrovascular diseases have undergone major advances in recent decades, allowing for a more effective prevention of cardiovascular and cerebrovascular events. However, cardiac and cerebral atherothrombotic complications still account for substantial morbidity and mortality worldwide. Novel therapeutic strategies are critical to improve patient outcomes following cardiovascular diseases. miRNAs are small non-coding RNAs, that regulate gene expression. Here, we discuss the role of miR-182 in regulating myocardial proliferation, migration, hypoxia, ischemia, apoptosis and hypertrophy in atherosclerosis, CAD, MI, I/R injury, organ transplant, cardiac hypertrophy, hypertension, heart failure, congenital heart disease and cardiotoxicity. Besides, we also summarize the current progress of miR-182 therapeutics in clinical development and discuss challenges that will need to be overcome to enter the clinic for patients with cardiac disease.
Collapse
Affiliation(s)
- Gaiqin Pei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Li Chen
- Department of Rehabilitation, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Yang Wang
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chengqi He
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Quan Wei
- Department of Rehabilitation Medicine and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, Sichuan, China
| |
Collapse
|
3
|
Shah P, Agbor-Enoh S, Bagchi P, deFilippi CR, Mercado A, Diao G, Morales DJ, Shah KB, Najjar SS, Feller E, Hsu S, Rodrigo ME, Lewsey SC, Jang MK, Marboe C, Berry GJ, Khush KK, Valantine HA. Circulating microRNAs in cellular and antibody-mediated heart transplant rejection. J Heart Lung Transplant 2022; 41:1401-1413. [PMID: 35872109 PMCID: PMC9529890 DOI: 10.1016/j.healun.2022.06.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Noninvasive monitoring of heart allograft health is important to improve clinical outcomes. MicroRNAs (miRs) are promising biomarkers of cardiovascular disease and limited studies suggest they can be used to noninvasively diagnose acute heart transplant rejection. METHODS The Genomic Research Alliance for Transplantation (GRAfT) is a multicenter prospective cohort study that phenotyped heart transplant patients from 5 mid-Atlantic centers. Patients who had no history of rejection after transplant were compared to patients with acute cellular rejection (ACR) or antibody-mediated rejection (AMR). Small RNA sequencing was performed on plasma samples collected at the time of an endomyocardial biopsy. Differential miR expression was performed with adjustment for clinical covariates. Regression was used to develop miR panels with high diagnostic accuracy for ACR and AMR. These panels were then validated in independent samples from GRAfT and Stanford University. Receiver operating characteristic curves were generated and area under the curve (AUC) statistics calculated. Distinct ACR and AMR clinical scores were developed to translate miR expression data for clinical use. RESULTS The GRAfT cohort had a median age of 52 years, with 35% females and 45% Black patients. Between GRAfT and Stanford, we included 157 heart transplant patients: 108 controls and 49 with rejection (50 ACR and 38 AMR episodes). After differential miR expression and regression analysis, we identified 12 miRs that accurately discriminate ACR and 17 miRs in AMR. Independent validation of the miR panels within GRAfT led to an ACR AUC 0.92 (95% confidence interval [CI]: 0.86-0.98) and AMR AUC 0.82 (95% CI: 0.74-0.90). The externally validated ACR AUC was 0.72 (95% CI: 0.59-0.82). We developed distinct ACR and AMR miR clinical scores (range 0-100), a score ≥ 65, identified ACR with 86% sensitivity, 76% specificity, and 98% negative predictive value, for AMR score performance was 82%, 84% and 97%, respectively. CONCLUSIONS We identified novel miRs that had excellent performance to noninvasively diagnose acute rejection after heart transplantation. Once rigorously validated, the unique clinical ACR and AMR scores usher in an era whereby genomic biomarkers can be used to screen and diagnose the subtype of rejection. These novel biomarkers may potentially alleviate the need for an endomyocardial biopsy while facilitating the initiation of targeted therapy based on the noninvasive diagnosis of ACR or AMR.
Collapse
Affiliation(s)
- Palak Shah
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church, Virginia; Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland.
| | - Sean Agbor-Enoh
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Pramita Bagchi
- Volgenau School of Engineering, George Mason University, Fairfax, Virginia
| | | | - Angela Mercado
- Heart Failure, Mechanical Circulatory Support & Transplant, Inova Heart and Vascular Institute, Falls Church, Virginia
| | - Gouqing Diao
- Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia
| | - Dave Jp Morales
- Heart Failure & Transplantation, Stanford University, Palo Alto, California
| | - Keyur B Shah
- The Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Samer S Najjar
- Advanced Heart Failure Program, Medstar Heart and Vascular Institute, Washington Hospital Center, Washington, District of Columbia
| | - Erika Feller
- Heart Failure & Transplantation, University of Maryland, Baltimore, Maryland
| | - Steven Hsu
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Maria E Rodrigo
- The Pauley Heart Center, Virginia Commonwealth University, Richmond, Virginia
| | - Sabra C Lewsey
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Moon Kyoo Jang
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Applied Precision Genomics, National Heart, Lung and Blood Institute, Bethesda, Maryland
| | - Charles Marboe
- Department of Pathology, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York, New York
| | - Gerald J Berry
- Stanford University School of Medicine, Palo Alto, California
| | - Kiran K Khush
- Stanford University School of Medicine, Palo Alto, California
| | - Hannah A Valantine
- Genomic Research Alliance for Transplantation (GRAfT), Bethesda, Maryland; Stanford University School of Medicine, Palo Alto, California
| |
Collapse
|
4
|
Mahtal N, Lenoir O, Tinel C, Anglicheau D, Tharaux PL. MicroRNAs in kidney injury and disease. Nat Rev Nephrol 2022; 18:643-662. [PMID: 35974169 DOI: 10.1038/s41581-022-00608-6] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2022] [Indexed: 11/09/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression by degrading or repressing the translation of their target messenger RNAs. As miRNAs are critical regulators of cellular homeostasis, their dysregulation is a crucial component of cell and organ injury. A substantial body of evidence indicates that miRNAs are involved in the pathophysiology of acute kidney injury (AKI), chronic kidney disease and allograft damage. Different subsets of miRNAs are dysregulated during AKI, chronic kidney disease and allograft rejection, which could reflect differences in the physiopathology of these conditions. miRNAs that have been investigated in AKI include miR-21, which has an anti-apoptotic role, and miR-214 and miR-668, which regulate mitochondrial dynamics. Various miRNAs are downregulated in diabetic kidney disease, including the miR-30 family and miR-146a, which protect against inflammation and fibrosis. Other miRNAs such as miR-193 and miR-92a induce podocyte dedifferentiation in glomerulonephritis. In transplantation, miRNAs have been implicated in allograft rejection and injury. Further work is needed to identify and validate miRNAs as biomarkers of graft function and of kidney disease development and progression. Use of combinations of miRNAs together with other molecular markers could potentially improve diagnostic or predictive power and facilitate clinical translation. In addition, targeting specific miRNAs at different stages of disease could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Nassim Mahtal
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France
| | - Olivia Lenoir
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| | - Claire Tinel
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Dany Anglicheau
- Service de Néphrologie et Transplantation Adulte, Hôpital Necker-Enfants Malades, Université Paris Cité, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut Necker-Enfants Malades, Inserm, Université Paris Cité, Paris, France
| | - Pierre-Louis Tharaux
- Paris Cardiovascular Research Center - PARCC, Inserm, Université Paris Cité, Paris, France.
| |
Collapse
|
5
|
Ma C, He D, Tian P, Wang Y, He Y, Wu Q, Jia Z, Zhang X, Zhang P, Ying H, Jin ZB, Hu G. miR-182 targeting reprograms tumor-associated macrophages and limits breast cancer progression. Proc Natl Acad Sci U S A 2022; 119:e2114006119. [PMID: 35105806 PMCID: PMC8833194 DOI: 10.1073/pnas.2114006119] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
The protumor roles of alternatively activated (M2) tumor-associated macrophages (TAMs) have been well established, and macrophage reprogramming is an important therapeutic goal. However, the mechanisms of TAM polarization remain incompletely understood, and effective strategies for macrophage targeting are lacking. Here, we show that miR-182 in macrophages mediates tumor-induced M2 polarization and can be targeted for therapeutic macrophage reprogramming. Constitutive miR-182 knockout in host mice and conditional knockout in macrophages impair M2-like TAMs and breast tumor development. Targeted depletion of macrophages in mice blocks the effect of miR-182 deficiency in tumor progression while reconstitution of miR-182-expressing macrophages promotes tumor growth. Mechanistically, cancer cells induce miR-182 expression in macrophages by TGFβ signaling, and miR-182 directly suppresses TLR4, leading to NFκb inactivation and M2 polarization of TAMs. Importantly, therapeutic delivery of antagomiR-182 with cationized mannan-modified extracellular vesicles effectively targets macrophages, leading to miR-182 inhibition, macrophage reprogramming, and tumor suppression in multiple breast cancer models of mice. Overall, our findings reveal a crucial TGFβ/miR-182/TLR4 axis for TAM polarization and provide rationale for RNA-based therapeutics of TAM targeting in cancer.
Collapse
Affiliation(s)
- Chengxin Ma
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dasa He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Pu Tian
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Wang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yunfei He
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qiuyao Wu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhenchang Jia
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xue Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peiyuan Zhang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hao Ying
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing Key Lab of Ophthalmology and Visual Sciences, Beijing 100005, China
| | - Guohong Hu
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China;
| |
Collapse
|
6
|
Abstract
Despite the overall success of heart transplantation as a definitive treatment for endstage heart failure, cardiac allograft rejection remains an important cause of morbidity and mortality. Endomyocardial biopsy has been the standard of care for rejection monitoring, but is associated with several diagnostic limitations and serious procedural complications. The use of molecular diagnostics has emerged over the past decade as a tool to potentially circumvent some of these limitations. We present an update on novel molecular approaches to detecting transplant rejection, focusing on 4 categories: microarray technology, gene expression profiling, cell-free DNA and microRNA.
Collapse
Affiliation(s)
- Lillian Benck
- Department of Cardiology, Cedars-Sinai Smidt Heart Institute
| | - Takuma Sato
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine
| | - Jon Kobashigawa
- Department of Cardiology, Cedars-Sinai Smidt Heart Institute
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW In kidney transplantation, microRNAs (miRNAs) have been extensively studied over the past decade, and panels of differentially expressed miRNAs have been identified from various body fluids/tissues, including blood, plasma, urine, or allograft biopsies, and in various conditions, such as acute T-cell-mediated and antibody-mediated rejections, chronic allograft rejection, interstitial fibrosis and tubular atrophy, acute tubular necrosis or BKV nephropathy. RECENT FINDINGS This review outlines our current knowledge regarding the complexity of miRNA regulation in fine-tuning expression of two-thirds of the human genome and the potential of miRNAs as biomarkers, based on an increasing number of case--control studies with, however, no evidence of short-term clinical development. Instead, a progressive change in study objectives is reported, with the most recent literature using miRNA-targeted genes as entry points for studying disease pathways. SUMMARY Our nascent understanding of their presumed roles in alloimmunity suggests that miRNAs are key regulators in many allograft injuries. Future directions should investigate how the integration of miRNAs with other layers of molecular data, such as genomic, transcriptomic, or proteomic data, could help to characterize the cellular interactions involved in allograft rejection and whether miRNA-based therapy could be of relevance for transplant medicine.
Collapse
|
8
|
Novák J, Macháčková T, Krejčí J, Bienertová-Vašků J, Slabý O. MicroRNAs as theranostic markers in cardiac allograft transplantation: from murine models to clinical practice. Theranostics 2021; 11:6058-6073. [PMID: 33897899 PMCID: PMC8058726 DOI: 10.7150/thno.56327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/21/2021] [Indexed: 12/11/2022] Open
Abstract
Congestive heart failure affects about 23 million people worldwide, and cardiac allograft transplantation remains one of the last options for patients with terminal refractory heart failure. Besides the infectious or oncological complications, the prognosis of patients after heart transplantation is affected by acute cellular or antibody-mediated rejection and allograft vasculopathy development. Current monitoring of both conditions requires the performance of invasive procedures (endomyocardial biopsy sampling and coronary angiography or optical coherence tomography, respectively) that are costly, time-demanding, and non-comfortable for the patient. Within this narrative review, we focus on the potential pathophysiological and clinical roles of microRNAs (miRNAs, miRs) in the field of cardiac allograft transplantation. Firstly, we provide a general introduction about the status of cardiac allograft function monitoring and the discovery of miRNAs as post-transcriptional regulators of gene expression and clinically relevant biomarkers found in the extracellular fluid. After this general introduction, information from animal and human studies are summarized to underline the importance of miRNAs both in the pathophysiology of the rejection process, the possibility of its modulation by altering miRNAs levels, and last but not least, about the use of miRNAs in the clinical practice to diagnose or predict the rejection occurrence.
Collapse
Affiliation(s)
- Jan Novák
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5-A18, 625 00, Brno, Czech Republic
- Second Department of Internal Medicine, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691, Brno, Czech Republic
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
| | - Táňa Macháčková
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
| | - Jan Krejčí
- Department of Cardiovascular Diseases, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691, Brno, Czech Republic
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5-A18, 625 00, Brno, Czech Republic
- RECETOX, Faculty of Sciences, Masaryk University, Kamenice 5-A29, 625 00, Brno, Czech Republic
| | - Ondřej Slabý
- Central European Institute of Technology, Masaryk University, Kamenice 5-A35, 625 00, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| |
Collapse
|
9
|
Di Stefano AB, Pappalardo M, Moschella F, Cordova A, Toia F. MicroRNAs in solid organ and vascularized composite allotransplantation: Potential biomarkers for diagnosis and therapeutic use. Transplant Rev (Orlando) 2020; 34:100566. [PMID: 32682704 DOI: 10.1016/j.trre.2020.100566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/23/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022]
Abstract
Nowadays, solid organ transplantation (SOT) is an established treatment for patients with end-organ dysfunction, which dramatically improves the quality-of-life. Vascularized composite allotransplants (VCAs) including hand and face have been reported worldwide over the last 20 years. However, VCAs, differently to SOT, are life-enhancing instead of life-saving and are not routinely performed due to the risk of immune rejection and the adverse effects of immunosuppression. Over the past decade, although considerable improvements in short-term outcomes after allotransplantation have been registered, these results have not been translated into major progress in long-term allograft acceptance and patient survival. Recently active researches in the field of biomarker discovery have been conducted to develop individualized therapies for allograft recipients. MicroRNAs (miRNAs) are a small noncoding RNAs functioning as critical regulators of gene and protein expression by RNA interference. They have been connected in numerous biological processes and diseases. Due to their immunomodulatory functions, miRNAs have been amended as potential diagnostic and prognostic biomarker for the detection of rejection in allotransplantation. Due to their specific circulating expression profile, they could act as noninvasive predictive tools for rejection that may help clinicians in an early adjustment of the immunosuppression protocol during acute rejections episodes. Indeed, specific anti-sense oligonucleotides suppressing miRNAs expressed in rejection could reduce the rejection rate in allografts and decrease the use of immunosuppressants. We present a literature review of the immunomodulatory properties and characteristics of miRNAs. We will summarize the current knowledge on miRNAs as potential biomarkers for allograft rejection and possible application in allotransplantation monitoring. Finally, we will discuss the advances in preclinical miRNA-based therapies for immunosuppression.
Collapse
Affiliation(s)
- Anna Barbara Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Marco Pappalardo
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Francesco Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy.
| | - Adriana Cordova
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy.
| | - Francesca Toia
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Surgery Section, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy; Plastic and Reconstructive Unit, Department of Oncology, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", 90127 Palermo, Italy.
| |
Collapse
|
10
|
Nováková T, Macháčková T, Novák J, Hude P, Godava J, Žampachová V, Oppelt J, Zlámal F, Němec P, Bedáňová H, Slabý O, Bienertová-Vašků J, Špinarová L, Krejčí J. Identification of a Diagnostic Set of Endomyocardial Biopsy microRNAs for Acute Cellular Rejection Diagnostics in Patients after Heart Transplantation Using Next-Generation Sequencing. Cells 2019; 8:cells8111400. [PMID: 31698874 PMCID: PMC6912472 DOI: 10.3390/cells8111400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 11/03/2019] [Accepted: 11/04/2019] [Indexed: 12/14/2022] Open
Abstract
Introduction: Acute cellular rejection (ACR) of heart allografts represents the most common reason for graft failure. Endomyocardial biopsies (EMB) are still subject to substantial interobserver variability. Novel biomarkers enabling precise ACR diagnostics may decrease interobserver variability. We aimed to identify a specific subset of microRNAs reflecting the presence of ACR. Patients and Methods: Monocentric retrospective study. A total of 38 patients with the anamnesis of ACR were identified and for each patient three consecutive samples of EMB (with, prior and after ACR) were collected. Sixteen trios were used for next-generation sequencing (exploratory cohort); the resting 22 trios were used for validation with qRT-PCR (validation cohort). Statistical analysis was performed using R software. Results: The analysis of the exploration cohort provided the total of 11 miRNAs that were altered during ACR, the three of which (miR-144, miR-589 and miR-182) were further validated in the validation cohort. Using the levels of all 11 miRNAs and principal component analysis, an ACR score was created with the specificity of 91% and sensitivity of 68% for detecting the presence of ACR in the EMB sample. Conclusion: We identified a set of microRNAs altered in endomyocardial biopsies during ACR and using their relative levels we created a diagnostic score that can be used for ACR diagnosis.
Collapse
Affiliation(s)
- Tereza Nováková
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
| | - Táňa Macháčková
- Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Jan Novák
- Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
- Department of Pathological Physiology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
- Correspondence: (J.N.); (J.K.); Tel.: +420-54318-2253 (J.N.); +420-54318-3475 (J.K.)
| | - Petr Hude
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
| | - Július Godava
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
| | - Víta Žampachová
- Department of Pathology, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
| | - Jan Oppelt
- Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Filip Zlámal
- Department of Pathological Physiology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Petr Němec
- Centre of Cardiovascular Surgery and Organ Transplantation, Pekařská 53, 65691 Brno, Czech Republic
| | - Helena Bedáňová
- Centre of Cardiovascular Surgery and Organ Transplantation, Pekařská 53, 65691 Brno, Czech Republic
| | - Ondřej Slabý
- Central European Institute of Technology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Masaryk University, Kamenice 5, 62500 Brno, Czech Republic
| | - Lenka Špinarová
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
| | - Jan Krejčí
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, Pekařská 53, 65691 Brno, Czech Republic
- Correspondence: (J.N.); (J.K.); Tel.: +420-54318-2253 (J.N.); +420-54318-3475 (J.K.)
| |
Collapse
|
11
|
Sang AX, McPherson MC, Ivison GT, Qu X, Rigdon J, Esquivel CO, Krams SM, Martinez OM. Dual blockade of the PI3K/Akt/mTOR pathway inhibits posttransplant Epstein-Barr virus B cell lymphomas and promotes allograft survival. Am J Transplant 2019; 19:1305-1314. [PMID: 30549430 PMCID: PMC6482059 DOI: 10.1111/ajt.15216] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 11/23/2018] [Accepted: 11/26/2018] [Indexed: 01/25/2023]
Abstract
Posttransplant lymphoproliferative disorder (PTLD) is a serious complication of organ transplantation that often manifests as Epstein-Barr virus (EBV)-associated B cell lymphomas. Current treatments for PTLD have limited efficacy and can be associated with graft rejection or systemic toxicities. The mTOR inhibitor, rapamycin, suppresses tumor growth of EBV+ B cell lymphoma cells in vitro and in vivo; however, the efficacy is limited and clinical benefits of mTOR inhibitors for PTLD are variable. Here, we show constitutive activation of multiple nodes within the PI3K/Akt/mTOR pathway in EBV+ PTLD-derived cell lines. Inhibition of either PI3K or Akt, with specific inhibitors CAL-101 and MK-2206, respectively, diminished growth of EBV+ B cell lines from PTLD patients in a dose-dependent manner. Importantly, rapamycin combined with CAL-101 or MK-2206 had a synergistic effect in suppressing cell growth as determined by IC50 isobolographic analysis and Loewe indices. Moreover, these combinations were significantly more effective than rapamycin alone in inhibiting tumor xenograft growth in NOD-SCID mice. Finally, both CAL-101 and MK-2206 also prolonged survival of heterotopic cardiac allografts in C57BL/6 mice. Thus, combination therapy with rapamycin and a PI3K inhibitor, or an Akt inhibitor, can be an efficacious treatment for EBV-associated PTLD, while simultaneously promoting allograft survival.
Collapse
Affiliation(s)
- Adam X Sang
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Marla C McPherson
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Geoffrey T Ivison
- Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiumei Qu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph Rigdon
- Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, CA, USA
| | - Carlos O Esquivel
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sheri M Krams
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA,Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Olivia M Martinez
- Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA,Stanford Immunology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Micro-RNAs (miRNAs) are highly conserved small RNA molecules that have selective gene-regulatory functions. This posttranscriptional regulation by miRNAs is critical for many immunological processes. Many developments in establishing the biological role of miRNAs in solid organ transplantation have been generated in the last decade. Discoveries of immune regulation by miRNAs, resulting in graft prolongation and transplant tolerance, are rapidly advancing and are the subject of this review. RECENT FINDINGS Many elegant experimental studies have revealed intriguing associations between transplant tolerance and specific miRNA profiles. These findings have provided insight into the miRNAs critical for sustaining immune suppression, and have revealed common miRNA pathways that should be further investigated and/or targeted therapeutically. Further reports have strategized and corroborated different methods of manipulating miRNA expression for prolonging allograft survival, yielding promising preclinical evidence of the efficacy of miRNA-based therapies. SUMMARY The review covers these recent developments in miRNA research that can revolutionize how we implement diagnostics and prognostics and how we can strategize transplantation therapies.
Collapse
|
13
|
Haybar H, Shahrabi S, Shahjahani M, Rezaeeyan H. Diagnostic Value of HLA Typing in Pathogenesis of Cardiomyopathy. Cardiovasc Hematol Disord Drug Targets 2018; 19:132-138. [PMID: 30520385 DOI: 10.2174/1871529x19666181205151340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/14/2018] [Accepted: 11/07/2018] [Indexed: 11/22/2022]
Abstract
Development of cardiomyopathy (CM) is dependent upon several factors. However, the reaction of the immune response against myocardial tissue due to microbial and viral infections plays an important role in this disease. Therefore, the purpose of this study is to investigate the relationship between HLAs and their pathogenic mechanisms in the incidence of CM. Relevant literature was identified by a PubMed search (1989-2017) of English-language papers using the terms "Cardiomyopathy", "Human leukocyte antigen or HLA", "immune response", and "polymorphism". If CM patients are afflicted with viral and microbial infections, HLA class II molecules, which are not expressed on myocardial tissue in normal conditions, are mainly expressed on it. As a result, these HLAs present self- antigens and provoke autoimmune responses against myocardial tissue. On the other hand, the occurrence of polymorphism as well as disrupted expression of miRNAs can affect HLA expression, leading to hypertrophy and fibrosis of cardiac muscle. Finally, it is inferred that the expression evaluation of HLAs as well as identification of polymorphisms in their coding genes can be effective diagnostic factors in the detection of people susceptible to CM.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis research center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of biochemistry and hematology, faculty of medicine, Semnan University of medical sciences, Semnan, Iran
| | - Mohammad Shahjahani
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
14
|
The role of miR-183 cluster in immunity. Cancer Lett 2018; 443:108-114. [PMID: 30529154 DOI: 10.1016/j.canlet.2018.11.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/25/2018] [Accepted: 11/21/2018] [Indexed: 12/22/2022]
Abstract
MicroRNAs (miRNAs) are essential factors of an extensively conserved post-transcriptional process to regulate gene expression. MiRNAs play a pivotal role in immunity, including controlling the differentiation of various immune cells as well as their immunological functions. The miR-183 cluster, which is comprised of miR-183, -96 and -182, is a miRNA family with sequence homology. These miRNAs are usually transcribed together as a polycistronic miRNA cluster during development and are required for maturation of sensory organs. In comparison to defined sensory-specific role of these miRNAs in normal development, they are frequently over-expressed in several non-sensory diseases, including autoimmune diseases and cancers. Because individual miRNAs of miR-183 cluster have both common and unique targets within functionally interrelated pathways, they can show cooperative or opposing effects on biological processes, implying the complexity of this miR cluster-mediated gene regulation. Therefore, a better understanding of the molecular regulation of miR-183 cluster expression and its downstream networks is important for the therapeutic applications. In this review, we will discuss the characteristics of miR-183 cluster and a wide variety of evidence on its function in immune system. Newer knowledge summarized here will help readers understand the versatile role of miR-183 cluster in this field.
Collapse
|
15
|
Shah P, Bristow MR, Port JD. MicroRNAs in Heart Failure, Cardiac Transplantation, and Myocardial Recovery: Biomarkers with Therapeutic Potential. Curr Heart Fail Rep 2017; 14:454-464. [DOI: 10.1007/s11897-017-0362-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|