1
|
Filz von Reiterdank I, Bento R, Hyun I, Isasi R, Wolf SM, Coert JH, Mink van der Molen AB, Parekkadan B, Uygun K. Designer Organs: Ethical Genetic Modifications in the Era of Machine Perfusion. Annu Rev Biomed Eng 2025; 27:101-128. [PMID: 39874605 DOI: 10.1146/annurev-bioeng-062824-121925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Gene therapy is a rapidly developing field, finally yielding clinical benefits. Genetic engineering of organs for transplantation may soon be an option, thanks to convergence with another breakthrough technology, ex vivo machine perfusion (EVMP). EVMP allows access to the functioning organ for genetic manipulation prior to transplant. EVMP has the potential to enhance genetic engineering efficiency, improve graft survival, and reduce posttransplant complications. This will enable genetic modifications with a vast variety of applications, while raising questions on the ethics and regulation of this emerging technology. This review provides an in-depth discussion of current methodologies for delivering genetic vectors to transplantable organs, particularly focusing on the enabling role of EVMP. Organ-by-organ analysis and key characteristics of various vector and treatment options are assessed. We offer a road map for research and clinical translation, arguing that achieving scientific benchmarks while creating anticipatory governance is necessary to secure societal benefit from this technology.
Collapse
Affiliation(s)
- Irina Filz von Reiterdank
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Shriners Children's Boston, Boston, Massachusetts, USA
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Raphaela Bento
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Shriners Children's Boston, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Insoo Hyun
- Center for Life Sciences and Public Learning, Boston Museum of Science, Boston, Massachusetts, USA
| | - Rosario Isasi
- Dr. John T. Macdonald Foundation Department of Human Genetics and Institute for Human Genomics, University of Miami School of Medicine, Miami, Florida, USA
| | - Susan M Wolf
- Law School, Medical School, and Consortium on Law and Values in Health, Environment & the Life Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - J Henk Coert
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aebele B Mink van der Molen
- Department of Plastic, Reconstructive and Hand Surgery, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, USA
| | - Korkut Uygun
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Shriners Children's Boston, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Wang W, Gao T, Wang Y, Wang R, He M, Wang L, Zhou W, Ding M, Song Y, Ji X, Li X, Song Y, Zhu Y, Zhang Y, Xie Y, Chen Y, Jin Q, Xie M, Zhang L. Macrophage-Tased Dual-Phase T Cell Immunomodulation to Combat Transplant Rejection. Adv Healthc Mater 2025:e2403591. [PMID: 40264278 DOI: 10.1002/adhm.202403591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Transplant rejection remains a major challenge, driven primarily by the activation of alloreactive T cells. While enhancement of PD-L1 checkpoint molecules has exhibited potential in inhibiting T cell activity, its efficacy is often hindered by limited specificity and inadequate efficiency. Herein, a novel dual-phase immune modulation strategy is developed in which CTLA4-Ig and PD-L1 provide distinct, non-redundant inhibitory signals during the initial activation phase and the post-activation phase of T cells. PD-L1 is stably expressed on macrophages (sPD-L1 M) through lentiviral transduction, allowing them to leverage their chemotactic and antigen-presenting functions to target and deliver PD-L1 to transplant rejection sites. Notably, sPD-L1 M exhibited adaptive targeting capabilities, increasing their migration to grafts in response to heightened rejection. In an allograft skin model, the combined intravenous administration of sPD-L1 M and subcutaneous administration of CTLA4-Ig demonstrated synergistic efficacy, significantly suppressing alloreactive T cell activation, enhancing the recruitment of regulatory T cells (Tregs), downregulating pro-inflammatory cytokines, and prolonging allograft survival compared to either treatment alone. This study presents a promising strategy to effectively suppress T cell activity and prevent allogeneic immune responses without systemic immunosuppression.
Collapse
Affiliation(s)
- Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihui Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rui Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengrong He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wuqi Zhou
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengdan Ding
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yuan Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xiang Ji
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Xueke Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ye Zhu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yiwei Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yuji Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
3
|
Brown AE, Kelly YM, Zarinsefat A, Meier RPH, Worner G, Tavakol M, Sarwal MM, Laszik ZG, Stock PG, Sigdel TK. Gene expression-based molecular scoring of pancreas transplant rejection for a quantitative assessment of rejection severity and resistance to treatment. Am J Transplant 2025; 25:316-328. [PMID: 39349170 PMCID: PMC11878653 DOI: 10.1016/j.ajt.2024.09.032] [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: 11/12/2023] [Revised: 09/01/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Pancreas transplantation improves glycemic control and mortality in patients with diabetes but requires aggressive immunosuppression to control the alloimmune and autoimmune response. Recent developments in "omics" methods have provided gene transcript-based biomarkers for organ transplant rejection. The tissue Common Response Module (tCRM) score is developed to identify the severity of rejection in kidney, heart, liver, and lung transplants. Still, it has not yet been validated in pancreas transplants (PT). We evaluated the tCRM score's relevance in PT and additional markers of acute cellular rejection (ACR) for PT. An analysis of 51 pancreas biopsies with ACR identified 37 genes and 56 genes significantly upregulated in the case of grade 3 and grade 2 ACR, respectively (P < .05). Significant differences were seen with higher grades of rejection among several transcripts. Of the 22 genes differentially expressed in grade 3 ACR, 18 were also differentially expressed in grade 2 ACR. The rejection signal was attributable to activated leukocytes' infiltration. Significantly higher tCRM scores were found in grade 3 ACR (P = .007) and grade 2 ACR (P = .004), compared to normal samples. The tCRM score was able to distinguish treatment-resistant cases from those successfully treated for rejection.
Collapse
Affiliation(s)
- Audrey E Brown
- Department of Surgery, University of California, San Francisco, California, USA
| | - Yvonne M Kelly
- Department of Surgery, University of California, San Francisco, California, USA; Department of Surgery, Columbia University, New York, New York, USA
| | - Arya Zarinsefat
- Department of Surgery, University of California, San Francisco, California, USA
| | - Raphael P H Meier
- Department of Surgery, University of California, San Francisco, California, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Giulia Worner
- Department of Surgery, University of California, San Francisco, California, USA
| | - Mehdi Tavakol
- Department of Surgery, University of California, San Francisco, California, USA
| | - Minnie M Sarwal
- Department of Surgery, University of California, San Francisco, California, USA
| | - Zoltan G Laszik
- Department of Surgery, University of California, San Francisco, California, USA
| | - Peter G Stock
- Department of Surgery, University of California, San Francisco, California, USA.
| | - Tara K Sigdel
- Department of Surgery, University of California, San Francisco, California, USA.
| |
Collapse
|
4
|
Huang G. Advances in metabolomics profiling of pediatric kidney diseases: A review. BIOMOLECULES & BIOMEDICINE 2024; 24:1044-1054. [PMID: 38400839 PMCID: PMC11379015 DOI: 10.17305/bb.2024.10098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 02/26/2024]
Abstract
Pediatric renal diseases encompass a diverse array of pathological conditions, often engendering enduring ramifications. Metabolomics, an emergent branch of omics sciences, endeavors to holistically delineate alterations in metabolite compositions through the amalgamation of sophisticated analytical chemistry techniques and robust statistical methodologies. Recent advancements in metabolomics research within the realm of pediatric nephrology have been substantial, offering promising avenues for the identification of robust biomarkers, the elaboration of novel therapeutic targets, and the intricate elucidation of molecular mechanisms. The present discourse aims to critically review the progress in metabolomics profiling pertinent to pediatric renal disorders over the previous 12 years.
Collapse
Affiliation(s)
- Guoping Huang
- Department of Nephrology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
5
|
Lu X, Xu Z, Shu F, Wang Y, Han Y, Yang X, Shi P, Fan C, Wang L, Yu F, Sun Q, Cheng F, Chen H. Reactive Oxygen Species Responsive Multifunctional Fusion Extracellular Nanovesicles: Prospective Treatments for Acute Heart Transplant Rejection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406758. [PMID: 38949397 DOI: 10.1002/adma.202406758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/19/2024] [Indexed: 07/02/2024]
Abstract
Heart transplantation offers life-saving treatment for patients with end-stage heart failure; however, ischemia-reperfusion injury (IRI) and subsequent immune responses remain significant challenges. Current therapies primarily target adaptive immunity, with limited options available for addressing IRI and innate immune activation. Although plant-derived vesicle-like nanoparticles show promise in managing diseases, their application in organ transplantation complications is unexplored. Here, this work develops a novel reactive oxygen species (ROS)-responsive multifunctional fusion extracellular nanovesicles carrying rapamycin (FNVs@RAPA) to address early IRI and Ly6C+Ly6G- inflammatory macrophage-mediated rejection in heart transplantation. The FNVs comprise Exocarpium Citri grandis-derived extracellular nanovesicles with anti-inflammatory and antioxidant properties, and mesenchymal stem cell membrane-derived nanovesicles expressing calreticulin with macrophage-targeting ability. A novel ROS-responsive bio-orthogonal chemistry approach facilitates the active targeting delivery of FNVs@RAPA to the heart graft site, effectively alleviating IRI and promoting the polarization of Ly6C+Ly6G- inflammatory macrophages toward an anti-inflammatory phenotype. Hence, FNVs@RAPA represents a promising therapeutic approach for mitigating early transplantation complications and immune rejection. The fusion-targeted delivery strategy offers superior heart graft site enrichment and macrophage-specific targeting, promising improved transplant outcomes.
Collapse
Affiliation(s)
- Xingyu Lu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Fan Shu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yidan Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yuhang Han
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xinrui Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Peilin Shi
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chuanqiang Fan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Fei Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qipeng Sun
- Department of Kidney Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
6
|
Jiang H, Zhao Q, Ye X. Application of nanomaterials in heart transplantation: a narrative review. J Thorac Dis 2024; 16:3389-3405. [PMID: 38883645 PMCID: PMC11170395 DOI: 10.21037/jtd-23-1506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/15/2024] [Indexed: 06/18/2024]
Abstract
Background and Objective Heart transplantation (HT) is a therapeutic option for end-stage heart disease. Still, it faces many challenges, especially the shortage of donor sources and the poor durability of grafts, which are the two critical issues. In this review, we generalize the application of existing nanomedicine technologies in donor management as well as prevention and diagnosis of post-transplantation complications, also including the current preclinical studies of nanomaterials in cardiac tissue engineering and gene-editing xeno-donor grafts. Finally, we discuss the remaining problems and future directions of nanomaterials in the field of HT. Methods A narrative review using current search of the most recent literature on the topic. The terms "nanomaterials", "nano medicine'', "Heart transplantation (HT)", "Nano-drug delivery system (NDDS)" or their combination were searched in PubMed and Google Scholar. The specified timeframe began from 1990, and we prioritized publications mainly from the last 10 years. Key Content and Findings Nano-systems integrating therapeutic and diagnostic functions have been applied to cardiovascular diseases (CVDs) with their unique advantages in multiple fields such as drug delivery, tissue engineering, gene editing, imaging, biomarker editing, and many other aspects. In terms of transplantation, the preservation, transportation, and pretreatment of donor hearts machine perfusion (MP) provide the possibility for nano-systems with unique features, and therapeutic and diagnostic functions to be directly and passively targeted in order to improve the functional status of the transplanted organs or to increase the ability to tolerate the graft of patients. The development of nano-imaging, nanosensor, and nano biomarker technologies are also being applied to monitor the status of transplant recipients for early prevention and treatment of post-transplantation-related complications. Nanomaterials combined with cardiac tissue engineering and gene editing technologies could also expand graft sources and alleviate donor shortages. Conclusions Although the overall research on nanomaterial applications in the field of HT is in its infancy, its role in improving the prognosis of transplant recipients and breaking the current dilemma of HT is clear. However, before nanotechnologies can be translated into clinical applications in the future, they must be aimed at ensuring the drug delivery system's safety and pose a challenge in the direction of the ability to intervene with multiple drugs in combination.
Collapse
Affiliation(s)
- Huaiyu Jiang
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Chen L, Yang J, Klassen H. Immune Responses to Sequential Binocular Transplantation of Allogeneic Retinal Progenitor Cells to the Vitreous Cavity in Mice. Int J Mol Sci 2023; 24:ijms24076205. [PMID: 37047179 PMCID: PMC10093920 DOI: 10.3390/ijms24076205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
Intravitreal transplantation of allogeneic human retinal progenitor cells (hRPCs) holds promise as a treatment for blinding retinal degenerations. Prior work has shown that neural progenitors are well-tolerated as allografts following single injections; however, sequential delivery of allogeneic cells raises the potential risk of host sensitization with subsequent immune rejection of grafts. The current study was designed to assess whether an immune response would be induced by repeated intravitreal transplants of allogeneic RPCs utilizing the mouse animal model. We injected murine retinal progenitor cells (gmRPCs), originally derived from donors with a C57BL/6 genetic background, into BALB/c recipient mice in order to provide safety data as to what might be expected following repeated treatment of patients with allogeneic human cell product. Immune responses to gmRPCs were mild, consisting of T cells, B cells, neutrophils, and natural killer cells, with macrophages clearly the predominating. Animals treated with repeat doses of gmRPCs did not show evidence of sensitization, nor was there immune-mediated destruction of the grafts. Despite the absence of immunosuppressive treatments, allogeneic gmRPC grafts survived following repeat dosing, thus providing support for the preliminary observation that repeated injection of allogeneic RPCs to the vitreous cavity is tolerated in patients with retinitis pigmentosa.
Collapse
|
8
|
Comparative transcriptome profile of mouse macrophages treated with the RhoA/Rock pathway inhibitors Y27632, Fingolimod (Gilenya), and Rezurock (Belumosudil, SLx-2119). Int Immunopharmacol 2023; 118:110017. [PMID: 36931169 DOI: 10.1016/j.intimp.2023.110017] [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: 12/02/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023]
Abstract
Macrophages play a crucial role in, the currently uncurable, chronic rejection of transplants. In rodent transplantation models, inhibition of the RhoA/Rock pathway disrupts actin-related functions of macrophages, preventing them from entering the graft, and reducing vessel occlusion, fibrosis, and chronic rejection. Among RhoA/Rock inhibitors that inhibit chronic rejection in mouse transplantation are Y27632, Fingolimod, and Rezurock. In a mouse model, Rezurok is more effective in preventing fibrosis and less effective in preventing vessel occlusion than Y27632 or Fingolimod. Fingolimod is FDA-approved for treating multiple sclerosis (MS) and Rezurock for chronic graft versus host disease (GVHD). Still, none had been tested for chronic rejection in humans. To explain the differences in the anti-chronic rejection properties of Y27632, Fingolimod, and Rezurock, we compared the transcriptome profile of mouse macrophages treated with these compounds separately. Treatment with Y27632 or Fingolimod downregulated GTPase and actin pathways involved in cell migration. Rezurock downregulated genes related to fibrosis, such as PTX3, CCR2, CCL2, cell cycle, DNA replication, adaptive immune response, and organelle assembly, while Fingolimod also specifically downregulated NOTCH1 at mRNA . The result of this study not only uncovers which pathways are shared or specific for these drugs but will help in the development of macrophage pathway-targeted therapies in human transplantation, MS, and GVHD. Because macrophages are the major players in immune response, tissue regeneration, renewal, and homeostasis, and development of many diseases, including cancer, the data compiled here will help in designing novel or improved therapies in many clinical applications.
Collapse
|
9
|
Simona MS, Alessandra V, Emanuela C, Elena T, Michela M, Fulvia G, Vincenzo S, Ilaria B, Federica M, Eloisa A, Massimo A, Maristella G. Evaluation of Oxidative Stress and Metabolic Profile in a Preclinical Kidney Transplantation Model According to Different Preservation Modalities. Int J Mol Sci 2023; 24:ijms24021029. [PMID: 36674540 PMCID: PMC9861050 DOI: 10.3390/ijms24021029] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
This study addresses a joint nuclear magnetic resonance (NMR) and electron paramagnetic resonance (EPR) spectroscopy approach to provide a platform for dynamic assessment of kidney viability and metabolism. On porcine kidney models, ROS production, oxidative damage kinetics, and metabolic changes occurring both during the period between organ retrieval and implantation and after kidney graft were examined. The 1H-NMR metabolic profile—valine, alanine, acetate, trimetylamine-N-oxide, glutathione, lactate, and the EPR oxidative stress—resulting from ischemia/reperfusion injury after preservation (8 h) by static cold storage (SCS) and ex vivo machine perfusion (HMP) methods were monitored. The functional recovery after transplantation (14 days) was evaluated by serum creatinine (SCr), oxidative stress (ROS), and damage (thiobarbituric-acid-reactive substances and protein carbonyl enzymatic) assessments. At 8 h of preservation storage, a significantly (p < 0.0001) higher ROS production was measured in the SCS vs. HMP group. Significantly higher concentration data (p < 0.05−0.0001) in HMP vs. SCS for all the monitored metabolites were found as well. The HMP group showed a better function recovery. The comparison of the areas under the SCr curves (AUC) returned a significantly smaller (−12.5 %) AUC in the HMP vs. SCS. EPR-ROS concentration (μmol·g−1) from bioptic kidney tissue samples were significantly lower in HMP vs. SCS. The same result was found for the NMR monitored metabolites: lactate: −59.76%, alanine: −43.17%; valine: −58.56%; and TMAO: −77.96%. No changes were observed in either group under light microscopy. In conclusion, a better and more rapid normalization of oxidative stress and functional recovery after transplantation were observed by HMP utilization.
Collapse
Affiliation(s)
- Mrakic-Sposta Simona
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20159 Milano, Italy
| | - Vezzoli Alessandra
- Institute of Clinical Physiology, National Research Council (IFC-CNR), 20159 Milano, Italy
- Correspondence: (V.A.); (G.M.)
| | - Cova Emanuela
- Department of Molecular Medicine, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Ticcozzelli Elena
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Montorsi Michela
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Roma, Italy
| | - Greco Fulvia
- Institute of Chemical Sciences and Technologies “G. Natta”, National Research Council (SCITEC-CNR), 20133 Milan, Italy
| | - Sepe Vincenzo
- Department of Molecular Medicine, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Benzoni Ilaria
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Meloni Federica
- Section of Pneumology, Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
| | - Arbustini Eloisa
- Centre for Inherited Cardiovascular Diseases, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Abelli Massimo
- Department of Surgery, IRCCS Foundation Policlinico San Matteo, 27100 Pavia, Italy
| | - Gussoni Maristella
- Institute of Chemical Sciences and Technologies “G. Natta”, National Research Council (SCITEC-CNR), 20133 Milan, Italy
- Correspondence: (V.A.); (G.M.)
| |
Collapse
|
10
|
Cuervo Florez M, Bruner J, Zarrinpar A. Progress and challenges in diagnosis and treatment of rejection following liver transplantation. Curr Opin Organ Transplant 2021; 26:669-674. [PMID: 34581291 DOI: 10.1097/mot.0000000000000924] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Liver biopsy remains the most widely utilized method for diagnosis of allograft rejection following liver transplantation. However, associated risks and limitations present an opportunity for emerging noninvasive diagnostic techniques to improve upon the current standard of care. This review evaluates progress toward development of new noninvasive methods for the monitoring and diagnosing of allograft rejection. RECENT FINDINGS Recent studies investigate the potential of a variety of analytes. Quantification of dd-cfDNA and of DSA show potential to indicate status of allograft rejection and aid in immunosuppression modulation. Moreover, mRNA microarray profiling of differentially expressed genes, as well as characterization of cytokine responses and immunophenotypic shifts following liver transplantation, may predict and recognize rejection events. SUMMARY Noninvasive methods are not yet ready to replace liver biopsy as the standard of care for diagnosis of allograft rejection, though several assays and biomarkers have shown promising preliminary results. As noninvasive techniques become validated in clinical settings, their integration with current diagnostic methods is likely to foster increased sensitivity, specificity, and reliability of diagnosis.
Collapse
Affiliation(s)
- Mateo Cuervo Florez
- Department of Surgery, College of Medicine, University of Florida, Florida, USA
| | | | | |
Collapse
|
11
|
New insights on the monitoring of solid-organ allografts based on immune cell signatures. Transpl Immunol 2021; 70:101509. [PMID: 34843937 DOI: 10.1016/j.trim.2021.101509] [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: 10/23/2021] [Revised: 11/17/2021] [Accepted: 11/23/2021] [Indexed: 11/23/2022]
Abstract
Attaining a fair long-term allograft survival remains a challenge for allogeneic transplantation worldwide. Although the emergence of immunosuppressants has caused noticeable progress in the management of immunologic rejection, proper application of these therapeutics and dose adjustments require delicate and real-time monitoring of recipients. Nevertheless, the majority of conventional allograft monitoring approaches are based on organ damage or functional tests that render them unable to predict the rejection events in early time points before the establishment of a functional alloimmune response. On the other hand, biopsy-based methods include invasive practices and are accompanied by serious complications. In recent years, there have been a myriad of attempts on the discovery of reliable and non-invasive approaches for the monitoring of allografts that regarding a close relationship between allografts and hosts' immune system, most of the attempts have been devoted to the studies on the immune response-associated biomarkers. The discovery of gene and protein expression patterns in immune cells along with their phenotypic characterization and secretome analysis as well as tracking the immune responses in allograft tissues and clinical specimens are among the notable attempts taken to discover the non-invasive predictive markers with a proper coincidence to the pathologic condition. Collectively, these studies suggest a list of candidate biomarkers with ideal potentials for early and non-invasive prediction of allograft rejection and shed light on the way towards developing more standardized and reproducible approaches for monitoring the allograft rejection.
Collapse
|
12
|
Nash A, Aghlara-Fotovat S, Hernandez A, Scull C, Veiseh O. Clinical translation of immunomodulatory therapeutics. Adv Drug Deliv Rev 2021; 176:113896. [PMID: 34324885 PMCID: PMC8567306 DOI: 10.1016/j.addr.2021.113896] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023]
Abstract
Immunomodulatory therapeutics represent a unique class of drug products that have tremendous potential to rebalance malfunctioning immune systems and are quickly becoming one of the fastest-growing areas in the pharmaceutical industry. For these drugs to become mainstream medicines, they must provide greater therapeutic benefit than the currently used treatments without causing severe toxicities. Immunomodulators, cell-based therapies, antibodies, and viral therapies have all achieved varying amounts of success in the treatment of cancers and/or autoimmune diseases. However, many challenges related to precision dosing, off-target effects, and manufacturing hurdles will need to be addressed before we see widespread adoption of these therapies in the clinic. This review provides a perspective on the progress of immunostimulatory and immunosuppressive therapies to date and discusses the opportunities and challenges for clinical translation of the next generation of immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Amanda Nash
- Rice University, Department of Bioengineering, Houston TX, United States
| | | | - Andrea Hernandez
- Rice University, Department of Bioengineering, Houston TX, United States
| | | | - Omid Veiseh
- Rice University, Department of Bioengineering, Houston TX, United States.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Brady CA, Williams C, Sharps MC, Shelleh A, Batra G, Heazell AEP, Crocker IP. Chronic histiocytic intervillositis: A breakdown in immune tolerance comparable to allograft rejection? Am J Reprod Immunol 2021; 85:e13373. [PMID: 33155353 PMCID: PMC7988544 DOI: 10.1111/aji.13373] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/14/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic histiocytic intervillositis (CHI) is a pregnancy disorder characterized by infiltration of maternal macrophages into the intervillous space of the human placenta, often with accompanying perivillous fibrin deposition. CHI is associated strongly with foetal growth restriction and increased risk of miscarriage and stillbirth. Although rare, affecting 6 in every 10 000 pregnancies beyond 12 weeks' gestation, the rate of recurrence is high at 25%-100%. To date, diagnosis of CHI can only be made post-delivery upon examination of the placenta due to a lack of diagnostic biomarkers, and criteria vary across publications. No treatment options have shown proven efficacy, and CHI remains a serious obstetric conundrum. Although its underlying aetiology is unclear, due to the presence of maternal macrophages and the reported increased incidence in women with autoimmune disease, CHI is hypothesized to be an inappropriate immune response to the semi-allogeneic foetus. Given this lack of understanding, treatment approaches remain experimental with limited rationale. However, there is recent evidence that immunosuppression and antithrombotic therapies may be effective in preventing recurrence of associated adverse pregnancy outcomes. With similarities noted between the pathological features of CHI and acute rejection of solid organ transplants, further investigation of this hypothesis may provide a basis for tackling CHI and other immune-related placental conditions. This review will explore parallels between CHI and allograft rejection and identify areas requiring further confirmation and exploitation of this comparison.
Collapse
Affiliation(s)
- Chloe A. Brady
- Tommy's Maternal and Fetal Health Research CentreSt. Mary’s HospitalThe University of ManchesterManchesterUK
| | - Charlotte Williams
- Tommy's Maternal and Fetal Health Research CentreSt. Mary’s HospitalThe University of ManchesterManchesterUK
- University of ExeterExeterUK
| | - Megan C. Sharps
- Tommy's Maternal and Fetal Health Research CentreSt. Mary’s HospitalThe University of ManchesterManchesterUK
| | - Amena Shelleh
- St Mary’s HospitalManchester University NHS Foundation TrustManchesterUK
| | - Gauri Batra
- Paediatric HistopathologyCentral Manchester University Hospitals NHS Foundation TrustManchesterUK
| | - Alexander E. P. Heazell
- Tommy's Maternal and Fetal Health Research CentreSt. Mary’s HospitalThe University of ManchesterManchesterUK
- St Mary’s HospitalManchester University NHS Foundation TrustManchesterUK
| | - Ian P. Crocker
- Tommy's Maternal and Fetal Health Research CentreSt. Mary’s HospitalThe University of ManchesterManchesterUK
| |
Collapse
|
16
|
Yang M, Xu Z, Yan H, Tsai HI, Su D, Yan F, Lu Q, Feng J, Zeng W, Xi L, Zha H, Ling Y, He C, Wu Y, Xu X, Zheng G, Liu G, Chen H, Cheng F. PD-L1 cellular nanovesicles carrying rapamycin inhibit alloimmune responses in transplantation. Biomater Sci 2021; 9:1246-1255. [PMID: 33367372 DOI: 10.1039/d0bm01798a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Organ transplantation has been employed upon serious injuries, but a T-cell-mediated potent inflammatory immune response often leads to graft rejection. Immunosuppressive drugs such as rapamycin (RAPA) have to be taken after organ transplantation, but long-term use of these drugs causes severe adverse effects. Immune checkpoint pathways such as the programmed death-receptor 1/programmed death-ligand 1 (PD-1/PD-L1) provides an immunosuppressive environment, preventing excessive tissue destruction due to inflammatory immune responses. In this study, we bioengineered cell membrane-derived PD-L1 nanovesicles (PD-L1 NVs) to carry low doses of RAPA. These NVs inhibited T-cell activation and proliferation in vitro, by enhancing the PD-1/PD-L1 immune co-inhibitory signaling axis and inhibiting the mTOR pathway. Importantly, PD-L1 NVs encapsulated with rapamycin exerted stronger effects on inhibiting T-cell proliferation than PD-L1 NVs or rapamycin alone. This can be recapitulated in a mouse skin transplantation model, leading to the weakened alloimmune response and allograft tolerance. We also found that PD-L1/rapamycin vesicles have additional function to induce regulatory T cells in the recipient spleens. Our study highlighted the power of combining low-dose rapamycin and PD-L1 in the nanovesicles as immunosuppressants to promote allograft acceptance.
Collapse
Affiliation(s)
- Min Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Hailan Yan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Hsiang-I Tsai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Fuxia Yan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Qiumei Lu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Jianhua Feng
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Weiwei Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Lifang Xi
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Hualian Zha
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yunzhi Ling
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Chao He
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yingyi Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Xiaowei Xu
- Clinical Neuroscience Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518000, China
| | - Gang Zheng
- XuZhou Central Hospital Affiliated to Medical School of Southeast University, XuZhou, 221000, China
| | - Gan Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
17
|
Park MY, Krishna Vasamsetti BM, Kim WS, Kang HJ, Kim DY, Lim B, Cho K, Kim JS, Chee HK, Park JH, Yang HS, Rallabandi HR, Ock SA, Park MR, Lee H, Hwang IS, Kim JM, Oh KB, Yun IJ. Comprehensive Analysis of Cardiac Xeno-Graft Unveils Rejection Mechanisms. Int J Mol Sci 2021; 22:ijms22020751. [PMID: 33451076 PMCID: PMC7828557 DOI: 10.3390/ijms22020751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
Porcine heart xenotransplantation is a potential treatment for patients with end-stage heart failure. To understand molecular mechanisms of graft rejection after heart transplantation, we transplanted a 31-day-old alpha-1,3-galactosyltransferase knockout (GTKO) porcine heart to a five-year-old cynomolgus monkey. Histological and transcriptome analyses were conducted on xenografted cardiac tissue at rejection (nine days after transplantation). The recipient monkey's blood parameters were analyzed on days -7, -3, 1, 4, and 7. Validation was conducted by quantitative real-time PCR (qPCR) with selected genes. A non-transplanted GTKO porcine heart from an age-matched litter was used as a control. The recipient monkey showed systemic inflammatory responses, and the rejected cardiac graft indicated myocardial infarction and cardiac fibrosis. The transplanted heart exhibited a total of 3748 differentially expressed genes compared to the non-transplanted heart transcriptome, with 2443 upregulated and 1305 downregulated genes. Key biological pathways involved at the terminal stage of graft rejection were cardiomyopathies, extracellular interactions, and ion channel activities. The results of qPCR evaluation were in agreement with the transcriptome data. Transcriptome analysis of porcine cardiac tissue at graft rejection reveals dysregulation of the key molecules and signaling pathways, which play relevant roles on structural and functional integrities of the heart.
Collapse
Affiliation(s)
- Min Young Park
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Bala Murali Krishna Vasamsetti
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Wan Seop Kim
- Department of Pathology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Hee Jung Kang
- Department of Laboratory Medicine, Hallym University College of Medicine, Hallym University Sacred Heart Hospital, Dongan-gu, Anyang 14068, Korea;
| | - Do-Young Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Byeonghwi Lim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
| | - Kahee Cho
- Primate Organ Transplantation Centre, Genia Inc., Sungnam 13201, Korea;
| | - Jun Seok Kim
- Department of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea; (J.S.K.); (H.K.C.)
| | - Hyun Keun Chee
- Department of Thoracic and Cardiovascular Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea; (J.S.K.); (H.K.C.)
| | - Jung Hwan Park
- Department of Nephrology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Hyun Suk Yang
- Department of Cardiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea;
| | - Harikrishna Reddy Rallabandi
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Sun A. Ock
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Mi-Ryung Park
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Heasun Lee
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - In-Sul Hwang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
| | - Jun-Mo Kim
- Department of Animal Science and Technology, College of Biotechnology and Natural Resources, Chung-Ang University, Gyeonggi-do 17546, Korea; (M.Y.P.); (D.-Y.K.); (B.L.)
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| | - Keon Bong Oh
- Animal Biotechnology Division, National Institute of Animal Science, RDA, Jeollabukdo 55365, Korea; (B.M.K.V.); (H.R.R.); (S.A.O.); (M.-R.P.); (H.L.); (I.-S.H.)
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| | - Ik Jin Yun
- Department of Surgery, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea
- Correspondence: (J.-M.K.); (K.B.O.); (I.J.Y.); Tel.: +82-2-2030-7583 (I.J.Y.); Fax: +82-2-2030-7749 (I.J.Y.)
| |
Collapse
|
18
|
Tanimine N, Ohira M, Tahara H, Ide K, Tanaka Y, Onoe T, Ohdan H. Strategies for Deliberate Induction of Immune Tolerance in Liver Transplantation: From Preclinical Models to Clinical Application. Front Immunol 2020; 11:1615. [PMID: 32849546 PMCID: PMC7412931 DOI: 10.3389/fimmu.2020.01615] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
The liver exhibits intrinsic immune regulatory properties that maintain tolerance to endogenous and exogenous antigens, and provide protection against pathogens. Such an immune privilege contributes to susceptibility to spontaneous acceptance despite major histocompatibility complex mismatch when transplanted in animal models. Furthermore, the presence of a liver allograft can suppress the rejection of other solid tissue/organ grafts from the same donor. Despite this immune privilege of the livers, to control the undesired alloimmune responses in humans, most liver transplant recipients require long-term treatment with immune-suppressive drugs that predispose to cardiometabolic side effects and renal insufficiency. Understanding the mechanism of liver transplant tolerance and crosstalk between a variety of hepatic immune cells, such as dendritic cells, Kupffer cells, liver sinusoidas endothelial cells, hepatic stellate cells and so on, and alloreactive T cells would lead to the development of strategies for deliberate induction of more specific immune tolerance in a clinical setting. In this review article, we focus on results derived from basic studies that have attempted to elucidate the immune modulatory mechanisms of liver constituent cells and clinical trials that induced immune tolerance after liver transplantation by utilizing the immune-privilege potential of the liver.
Collapse
Affiliation(s)
- Naoki Tanimine
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masahiro Ohira
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Medical Center for Translational and Clinical Research Hiroshima University Hospital, Hiroshima, Japan
| | - Hiroyuki Tahara
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kentaro Ide
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takashi Onoe
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Kure Medical Center and Chugoku Cancer Center, National Hospital Organization, Kure, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
19
|
Leino AD, Pai MP. Maintenance Immunosuppression in Solid Organ Transplantation: Integrating Novel Pharmacodynamic Biomarkers to Inform Calcineurin Inhibitor Dose Selection. Clin Pharmacokinet 2020; 59:1317-1334. [PMID: 32720300 DOI: 10.1007/s40262-020-00923-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Calcineurin inhibitors, the primary immunosuppressive therapy used to prevent alloreactivity of transplanted organs, have a narrow therapeutic index. Currently, treatment is individualized based on clinical assessment of the risk of rejection or toxicity guided by trough concentration monitoring. Advances in immune monitoring have identified potential markers that may have value in understanding calcineurin inhibitor pharmacodynamics. Integration of these markers has the potential to complement therapeutic drug monitoring. Existing pharmacokinetic-pharmacodynamic (PK-PD) data is largely limited to correlation between the biomarker and trough concentrations at single time points. Immune related gene expression currently has the most evidence supporting PK-PD integration. Novel biomarker-based approaches to pharmacodynamic monitoring including development of enhanced PK-PD models are proposed to realize the full clinical benefit.
Collapse
Affiliation(s)
- Abbie D Leino
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, 428 Church Street, Rm 3569, Ann Arbor, MI, 48109, USA
| | - Manjunath P Pai
- Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, 428 Church Street, Rm 3569, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
20
|
Xu Z, Tsai HI, Xiao Y, Wu Y, Su D, Yang M, Zha H, Yan F, Liu X, Cheng F, Chen H. Engineering Programmed Death Ligand-1/Cytotoxic T-Lymphocyte-Associated Antigen-4 Dual-Targeting Nanovesicles for Immunosuppressive Therapy in Transplantation. ACS NANO 2020; 14:7959-7969. [PMID: 32515579 DOI: 10.1021/acsnano.9b09065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
T cell activation by immune allorecognition is a major contributing factor toward the triggering of organ rejection. Immunosuppressive drugs have to be taken after organ transplantation, but long-term use of these drugs increases the risks of infection and other serious disorders. Here, we showed dysregulation of programmed cell death-ligand 1/programmed cell death 1 (PD-L1/PD-1) and cytotoxic T-lymphocyte-associated protein 4/cluster of differentiation 80 (CTLA-4/CD80) in the spleen of two organ transplantation models. Using a bioengineering approach, cellular exosome-like nanovesicles (NVs) displaying PD-L1/CTLA-4 dual-targeting cargos were designed, and their specificity to bind their ligands PD-1 and CD80 on T cell and dendritic cell surfaces was confirmed. These NVs consequently enhanced PD-L1/PD-1 and CTLA-4/CD80 immune inhibitory pathways, two key immune checkpoints to co-inhibit T cell activation and maintain peripheral tolerance. It was also confirmed that PD-L1/CTLA-4 NVs led to the reduction of T cell activation and proliferation in vitro and in vivo. Finally, it was demonstrated that PD-L1/CTLA-4 NVs reduced density of CD8+ T cells and cytokine production, enriched regulatory T cells, and prolonged the survival of mouse skin and heart grafts. Taken together, these data supported the idea that PD-L1/CTLA-4 dual-targeting NVs exert immune inhibitory effects and may be used as a prospective immunosuppressant in organ transplantation.
Collapse
Affiliation(s)
- Zhanxue Xu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hsiang-I Tsai
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Youmei Xiao
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Yingyi Wu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Min Yang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hualian Zha
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Fuxia Yan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Xiaoyan Liu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, 518107, Shenzhen, China
| |
Collapse
|
21
|
Sigdel TK, Schroeder AW, Yang JYC, Sarwal RD, Liberto JM, Sarwal MM. Targeted Urine Metabolomics for Monitoring Renal Allograft Injury and Immunosuppression in Pediatric Patients. J Clin Med 2020; 9:jcm9082341. [PMID: 32707952 PMCID: PMC7465632 DOI: 10.3390/jcm9082341] [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: 05/04/2020] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 12/13/2022] Open
Abstract
Despite new advancements in surgical tools and therapies, exposure to immunosuppressive drugs related to non-immune and immune injuries can cause slow deterioration and premature failure of organ transplants. Diagnosis of these injuries by non-invasive urine monitoring would be a significant clinical advancement for patient management, especially in pediatric cohorts. We investigated the metabolomic profiles of biopsy matched urine samples from 310 unique kidney transplant recipients using gas chromatography-mass spectrometry (GC-MS). Focused metabolite panels were identified that could detect biopsy confirmed acute rejection with 92.9% sensitivity and 96.3% specificity (11 metabolites) and could differentiate BK viral nephritis (BKVN) from acute rejection with 88.9% sensitivity and 94.8% specificity (4 metabolites). Overall, targeted metabolomic analyses of biopsy-matched urine samples enabled the generation of refined metabolite panels that non-invasively detect graft injury phenotypes with high confidence. These urine biomarkers can be rapidly assessed for non-invasive diagnosis of specific transplant injuries, opening the window for precision transplant medicine.
Collapse
|
22
|
Singh R, Peters-Sengers H, Remmerswaal EBM, Yapici U, van der Pant KAMI, van der Weerd NC, Roelofs JJTH, van Lier RAW, Bemelman FJ, Florquin S, Ten Berge IJM. Clinical consequences of primary CMV infection after renal transplantation: a case-control study. Transpl Int 2020; 33:1116-1127. [PMID: 32480425 PMCID: PMC7540315 DOI: 10.1111/tri.13667] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/11/2020] [Accepted: 05/25/2020] [Indexed: 12/15/2022]
Abstract
The impact of primary cytomegalovirus infection (pCMV) on renal allograft function and histology is controversial. We evaluated the influence on incidence of acute rejection, allograft loss, allograft function and interstitial fibrosis/tubular atrophy (IF/TA). Retrospective case-control study, recipients transplanted between 2000 and 2014. Risk of acute rejection and allograft loss for those who experienced pCMV infection compared with those who did not, within an exposure period of two months after transplantation. Besides, its influence on allograft function and histology at one to three years after transplantation. Of 113 recipients experienced pCMV infection, 306 remained CMV seronegative. pCMV infection in the exposure period could not be proven as increasing the risk for acute rejection [HR = 2.18 (95% CI 0.80-5.97) P = 0.13] or allograft loss [HR = 1.11 (95%CI 0.33-3.72) P = 0.87]. Combination of pCMV infection and acute rejection posed higher hazard for allograft loss than acute rejection alone [HR = 3.69 (95% CI 1.21-11.29) P = 0.02]. eGFR(MDRD) values did not significantly differ at years one [46 vs. 50], two [46 vs. 51] and three [46 vs. 52]. No association between pCMV infection and IF/TA could be demonstrated [OR = 2.15 (95%CI 0.73-6.29) P = 0.16]. pCMV infection was not proven to increase the risk for acute rejection or allograft loss. However, it increased the risk for rejection-associated allograft loss. In remaining functioning allografts, it was not significantly associated with decline in function nor with presence of IF/TA.
Collapse
Affiliation(s)
- Ramandeep Singh
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Hessel Peters-Sengers
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - Ester B M Remmerswaal
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
| | - Unsal Yapici
- Department of Pathology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - Karlijn A M I van der Pant
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Neelke C van der Weerd
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - René A W van Lier
- Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Fréderike J Bemelman
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Sandrine Florquin
- Department of Pathology, Amsterdam University Medical Centers, location Academic Medical Center, Amsterdam, The Netherlands
| | - Ineke J M Ten Berge
- Renal Transplant Unit, Department of Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Thongprayoon C, Vaitla P, Craici IM, Leeaphorn N, Hansrivijit P, Salim SA, Bathini T, Cabeza Rivera FH, Cheungpasitporn W. The Use of Donor-Derived Cell-Free DNA for Assessment of Allograft Rejection and Injury Status. J Clin Med 2020; 9:E1480. [PMID: 32423115 PMCID: PMC7290747 DOI: 10.3390/jcm9051480] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/06/2023] Open
Abstract
Patient monitoring after kidney transplantation (KT) for early detection of allograft rejection remains key in preventing allograft loss. Serum creatinine has poor predictive value to detect ongoing active rejection as its increase is not sensitive, nor specific for acute renal allograft rejection. Diagnosis of acute rejection requires allograft biopsy and histological assessment, which can be logistically challenging in some cases and carries inherent risk for complications related to procedure. Donor-derived cell-free DNA (dd-cfDNA), DNA of donor origin in the blood of KT recipient arising from cells undergoing injury and death, has been examined as a potential surrogate marker for allograft rejection. A rise in dd-cfDNA levels precedes changes in serum creatinine allows early detections and use as a screening tool for allograft rejection. In addition, when used in conjunction with donor-specific antibodies (DSA), it increases the pre-biopsy probability of antibody-mediated rejection (ABMR) aiding the decision-making process. Advancements in noninvasive biomarker assays such as dd-cfDNA may offer the opportunity to improve and expand the spectrum of available diagnostic tools to monitor and detect risk for rejection and positively impact outcomes for KT recipients. In this this article, we discussed the evolution of dd-cfDNA assays and recent evidence of assessment of allograft rejection and injury status of KT by the use of dd-cfDNA.
Collapse
Affiliation(s)
- Charat Thongprayoon
- Division of Nephrology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (C.T.); (I.M.C.)
| | - Pradeep Vaitla
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Iasmina M. Craici
- Division of Nephrology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA; (C.T.); (I.M.C.)
| | - Napat Leeaphorn
- Renal Transplant Program, University of Missouri-Kansas City School of Medicine/Saint Luke’s Health System, Kansas City, MO 64111, USA;
| | - Panupong Hansrivijit
- Department of Internal Medicine, University of Pittsburgh Medical Center Pinnacle, Harrisburg, PA 17105, USA;
| | - Sohail Abdul Salim
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Tarun Bathini
- Department of Internal Medicine, University of Arizona, Tucson, AZ 85724, USA;
| | - Franco H. Cabeza Rivera
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA; (P.V.); (S.A.S.); (F.H.C.R.)
| |
Collapse
|
24
|
Abstract
Early detection of graft injury after kidney transplantation is key to maintaining long-term good graft function. Graft injury could be due to a multitude of factors including ischaemia reperfusion injury, cell or antibody-mediated rejection, progressive interstitial fibrosis and tubular atrophy, infections and toxicity from the immunosuppressive drugs themselves. The current gold standard for assessing renal graft dysfunction is renal biopsy. However, biopsy is usually late when triggered by a change in serum creatinine and of limited utility in diagnosis of early injury when histological changes are equivocal. Therefore, there is a need for timely, objective and non-invasive diagnostic techniques with good early predictive value to determine graft injury and provide precision in titrating immunosuppression. We review potential novel plasma and urine biomarkers that offer sensitive new strategies for early detection and provide major insights into mechanisms of graft injury. This is a rapidly expanding field, but it is likely that a combination of biomarkers will be required to provide adequate sensitivity and specificity for detecting graft injury.
Collapse
|
25
|
Andrikovics H, Őrfi Z, Meggyesi N, Bors A, Varga L, Kövy P, Vilimszky Z, Kolics F, Gopcsa L, Reményi P, Tordai A. Current Trends in Applications of Circulatory Microchimerism Detection in Transplantation. Int J Mol Sci 2019; 20:E4450. [PMID: 31509957 PMCID: PMC6769866 DOI: 10.3390/ijms20184450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 02/07/2023] Open
Abstract
Primarily due to recent advances of detection techniques, microchimerism (the proportion of minor variant population is below 1%) has recently gained increasing attention in the field of transplantation. Availability of polymorphic markers, such as deletion insertion or single nucleotide polymorphisms along with a vast array of high sensitivity detection techniques, allow the accurate detection of small quantities of donor- or recipient-related materials. This diagnostic information can improve monitoring of allograft injuries in solid organ transplantations (SOT) as well as facilitate early detection of relapse in allogeneic hematopoietic stem cell transplantation (allo-HSCT). In the present review, genetic marker and detection platform options applicable for microchimerism detection are discussed. Furthermore, current results of relevant clinical studies in the context of microchimerism and SOT or allo-HSCT respectively are also summarized.
Collapse
Affiliation(s)
- Hajnalka Andrikovics
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
- Department of Pathophysiology, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltán Őrfi
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Nóra Meggyesi
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - András Bors
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Lívia Varga
- School of PhD Studies, Semmelweis University, 1085 Budapest, Hungary
- Hungarian National Blood Transfusion Service, 1113 Budapest, Hungary
| | - Petra Kövy
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
- School of PhD Studies, Semmelweis University, 1085 Budapest, Hungary
| | - Zsófia Vilimszky
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Fanni Kolics
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - László Gopcsa
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Péter Reményi
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest National Institute of Hematology and Infectious Diseases, 1097 Budapest, Hungary
| | - Attila Tordai
- Department of Pathophysiology, Semmelweis University, 1089 Budapest, Hungary.
- Department of Transfusion Medicine, Semmelweis University, 1089 Budapest, Hungary.
| |
Collapse
|
26
|
Sharbafi MH, Assadiasl S, Pour‐reza‐gholi F, Barzegari S, Mohammadi Torbati P, Samavat S, Nicknam MH, Amirzargar A. TLR‐2, TLR‐4 and MyD88 genes expression in renal transplant acute and chronic rejections. Int J Immunogenet 2019; 46:427-436. [DOI: 10.1111/iji.12446] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/17/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022]
Affiliation(s)
| | - Sara Assadiasl
- Molecular Immunology Research Center Tehran University of Medical Sciences Tehran Iran
| | - Fatemeh Pour‐reza‐gholi
- Chronic Kidney Disease Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Saeed Barzegari
- Department of health information technology, Amol Faculty of Paramedical Sciences Mazandaran University of Medical Sciences Sari Iran
| | - Peyman Mohammadi Torbati
- Department of Pathology Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Shiva Samavat
- Chronic Kidney Disease Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mohammad Hossein Nicknam
- Department of Immunology, School of Medicine Tehran University of Medical Sciences Tehran Iran
- Molecular Immunology Research Center Tehran University of Medical Sciences Tehran Iran
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine Tehran University of Medical Sciences Tehran Iran
- Molecular Immunology Research Center Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
27
|
Wosik J, Chen W, Qin K, Ghobrial RM, Kubiak JZ, Kloc M. Magnetic Field Changes Macrophage Phenotype. Biophys J 2019; 114:2001-2013. [PMID: 29694876 DOI: 10.1016/j.bpj.2018.03.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/26/2018] [Accepted: 03/06/2018] [Indexed: 01/26/2023] Open
Abstract
Macrophages play a crucial role in homeostasis, regeneration, and innate and adaptive immune responses. Functionally different macrophages have different shapes and molecular phenotypes that depend on the actin cytoskeleton, which is regulated by the small GTPase RhoA. The naive M0 macrophages are slightly elongated, proinflammatory M1 are round, and M2 antiinflammatory macrophages are elongated. We have recently shown in the rodent model system that genetic or pharmacologic interference with the RhoA pathway deregulates the macrophage actin cytoskeleton, causes extreme macrophage elongation, and prevents macrophage migration. Here, we report that an exposure of macrophages to a nonuniform magnetic field causes extreme elongation of macrophages and has a profound effect on their molecular components and organelles. Using immunostaining and Western blotting, we observed that magnetic force rearranges the macrophage actin cytoskeleton, the Golgi complex, and the cation channel receptor TRPM2, and modifies the expression of macrophage molecular markers. We have found that the magnetic-field-induced alterations are very similar to changes caused by RhoA interference. We also analyzed magnetic-field-induced forces acting on macrophages and found that the location and alignment of magnetic-field-elongated macrophages correlate very well with the simulated distribution and orientation of such magnetic force lines.
Collapse
Affiliation(s)
- Jarek Wosik
- Electrical and Computer Engineering Department, University of Houston, Houston, Texas; Texas Center for Superconductivity, University of Houston, Houston, Texas.
| | - Wei Chen
- The Houston Methodist Research Institute, Houston, Texas; Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Kuang Qin
- Electrical and Computer Engineering Department, University of Houston, Houston, Texas; Texas Center for Superconductivity, University of Houston, Houston, Texas
| | - Rafik M Ghobrial
- The Houston Methodist Research Institute, Houston, Texas; Department of Surgery, The Houston Methodist Hospital, Houston, Texas
| | - Jacek Z Kubiak
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes), UMR 6290, Cell Cycle Group, Faculty of Medicine, Rennes, France; Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, Texas; Department of Surgery, The Houston Methodist Hospital, Houston, Texas; Department of Genetics, The University of Texas, M.D. Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
28
|
Sigdel TK, Archila FA, Constantin T, Prins SA, Liberto J, Damm I, Towfighi P, Navarro S, Kirkizlar E, Demko ZP, Ryan A, Sigurjonsson S, Sarwal RD, Hseish SC, Chan-On C, Zimmermann B, Billings PR, Moshkevich S, Sarwal MM. Optimizing Detection of Kidney Transplant Injury by Assessment of Donor-Derived Cell-Free DNA via Massively Multiplex PCR. J Clin Med 2018; 8:jcm8010019. [PMID: 30583588 PMCID: PMC6352163 DOI: 10.3390/jcm8010019] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/15/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Standard noninvasive methods for detecting renal allograft rejection and injury have poor sensitivity and specificity. Plasma donor-derived cell-free DNA (dd-cfDNA) has been reported to accurately detect allograft rejection and injury in transplant recipients and shown to discriminate rejection from stable organ function in kidney transplant recipients. This study used a novel single nucleotide polymorphism (SNP)-based massively multiplexed PCR (mmPCR) methodology to measure dd-cfDNA in various types of renal transplant recipients for the detection of allograft rejection/injury without prior knowledge of donor genotypes. A total of 300 plasma samples (217 biopsy-matched: 38 with active rejection (AR), 72 borderline rejection (BL), 82 with stable allografts (STA), and 25 with other injury (OI)) were collected from 193 unique renal transplant patients; dd- cfDNA was processed by mmPCR targeting 13,392 SNPs. Median dd-cfDNA was significantly higher in samples with biopsy-proven AR (2.3%) versus BL (0.6%), OI (0.7%), and STA (0.4%) (p < 0.0001 all comparisons). The SNP-based dd-cfDNA assay discriminated active from non-rejection status with an area under the curve (AUC) of 0.87, 88.7% sensitivity (95% CI, 77.7⁻99.8%) and 72.6% specificity (95% CI, 65.4⁻79.8%) at a prespecified cutoff (>1% dd-cfDNA). Of 13 patients with AR findings at a routine protocol biopsy six-months post transplantation, 12 (92%) were detected positive by dd-cfDNA. This SNP-based dd-cfDNA assay detected allograft rejection with superior performance compared with the current standard of care. These data support the feasibility of using this assay to detect disease prior to renal failure and optimize patient management in the case of allograft injury.
Collapse
Affiliation(s)
- Tara K Sigdel
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | | | | | | | - Juliane Liberto
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | - Izabella Damm
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | - Parhom Towfighi
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | | | | | | | | | | | - Reuben D Sarwal
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | - Szu-Chuan Hseish
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | - Chitranon Chan-On
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| | | | | | | | - Minnie M Sarwal
- University of California San Francisco, Department of Surgery, San Francisco, CA 94143, USA.
| |
Collapse
|
29
|
Chen W, Chen W, Li XC, Ghobrial RM, Kloc M. Coinhibition of mTORC1/mTORC2 and RhoA /ROCK pathways prevents chronic rejection of rat cardiac allografts. TRANSPLANTATION REPORTS 2018. [DOI: 10.1016/j.tpr.2018.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
30
|
Abstract
Current donor pool utilization is unable to meet the high demand for kidney transplants. Donor pool expansion using expanded-criteria donors and dual kidney transplantation are viable options. Advances in diagnosing antibody-mediated rejection and targeting immunosuppression increase long-term transplantation success. Further exploration of minimally invasive surgical techniques, kidney bioengineering, and artificial-implantable renal devices hold promise for the future.
Collapse
Affiliation(s)
- Garrett Salmon
- Nursing, Middle Tennessee State University, 1301 East Main Street, Murfreesboro, TN 37132, USA.
| | - Eric Salmon
- Natural Sciences, Motlow State Community College, 6015 Ledford Mill Road, Lynchburg, TN 37352, USA
| |
Collapse
|
31
|
Chen W, Chen S, Chen W, Li XC, Ghobrial RM, Kloc M. Screening RhoA/ROCK inhibitors for the ability to prevent chronic rejection of mouse cardiac allografts. Transpl Immunol 2018; 50:15-25. [DOI: 10.1016/j.trim.2018.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/18/2022]
|
32
|
McGill TJ, Stoddard J, Renner LM, Messaoudi I, Bharti K, Mitalipov S, Lauer A, Wilson DJ, Neuringer M. Allogeneic iPSC-Derived RPE Cell Graft Failure Following Transplantation Into the Subretinal Space in Nonhuman Primates. Invest Ophthalmol Vis Sci 2018; 59:1374-1383. [PMID: 29625461 PMCID: PMC5846443 DOI: 10.1167/iovs.17-22467] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose To characterize the intraocular immune response following transplantation of iPS-derived allogeneic RPE cells into the subretinal space of non–immune-suppressed rhesus macaques. Methods GFP-labeled allogeneic iPS-derived RPE cells were transplanted into the subretinal space of one eye (n = 6), and into the contralateral eye 1 day to 4 weeks later, using a two-stage transretinal and transscleral approach. Retinas were examined pre- and post-surgery by color fundus photography, fundus autofluorescence, and optical coherence tomography (OCT) imaging. Animals were euthanized between 2 hours and 7 weeks following transplantation. T-cell (CD3), B-cell (CD20), and microglial (Iba1) responses were assessed immunohistochemically. Results Cells were delivered into the subretinal space in all eyes without leakage into the vitreous. Transplanted RPE cells were clearly visible at 4 days after surgery but were no longer detectable by 3 weeks. In localized areas within the bleb containing transplanted cells, T- and B-cell infiltrates and microglia were observed in the subretinal space and underlying choroid. A T-cell response predominated at 4 days, but converted to a B-cell response at 3 weeks. By 7 weeks, few infiltrates or microglia remained. Host RPE and choroid were disrupted in the immediate vicinity of the graft, with fibrosis in the subretinal space. Conclusions Engraftment of allogeneic RPE cells failed following transplantation into the subretinal space of rhesus macaques, likely due to rejection by the immune system. These data underscore the need for autologous cell sources and/or confirmation of adequate immune suppression to ensure survival of transplanted RPE cells.
Collapse
Affiliation(s)
- Trevor J McGill
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States.,Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Jonathan Stoddard
- Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Lauren M Renner
- Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, United States
| | - Kapil Bharti
- Unit on Ocular and Stem Cell Translational Research, National Eye Institute/National Institutes of Health, Bethesda, Maryland, United States
| | - Shoukhrat Mitalipov
- Department of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| | - Andreas Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - David J Wilson
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Martha Neuringer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States.,Department of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States
| |
Collapse
|
33
|
Sigdel TK, Sarwal MM. Assessment of Circulating Protein Signatures for Kidney Transplantation in Pediatric Recipients. Front Med (Lausanne) 2017; 4:80. [PMID: 28670579 PMCID: PMC5472654 DOI: 10.3389/fmed.2017.00080] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/31/2017] [Indexed: 11/17/2022] Open
Abstract
Identification and use of non-invasive biomarkers for kidney transplantation monitoring is an unmet need. A total of 121 biobanked sera collected from 111 unique kidney transplant (KT) patients (children and adolescent) and 10 age-matched healthy normal controls were used to profile serum proteins using semi-quantitative proteomics. The proteomics data were analyzed to identify panels of serum proteins that were specific to various transplant injuries, which included acute rejection (AR), BK virus nephropathy (BKVN), and chronic allograft nephropathy (CAN). Gene expression data from matching peripheral blood mononuclear cells were interrogated to investigate the association between soluble serum proteins and altered gene expression of corresponding genes in different injury phenotypes. Analysis of the proteomics data identified from different patient phenotypes, with criteria of false discovery rate <0.05 and at least twofold changes in either direction, resulted in a list of 10 proteins that distinguished KT injury from no injury. Similar analyses to identify proteins specific to chronic injury, acute injury, and AR after kidney transplantation identified 22, 6, and 10 proteins, respectively. Elastic-Net logistic regression method was applied on the 137 serum proteins to classify different transplant injuries. This algorithm has identified panels of 10 serum proteins specific for AR, BKVN, and CAN with classification rates 93, 93, and 95%, respectively. The identified proteins could prove to be potential surrogate biomarkers for routine monitoring of the injury status of pediatric KT patients.
Collapse
Affiliation(s)
- Tara K Sigdel
- University of California, San Francisco, San Francisco, CA, United States
| | - Minnie M Sarwal
- University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
34
|
Liu Y, Kubiak JZ, Li XC, Ghobrial RM, Kloc M. Macrophages and RhoA Pathway in Transplanted Organs. Results Probl Cell Differ 2017; 62:365-376. [PMID: 28455717 DOI: 10.1007/978-3-319-54090-0_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
RhoA is a small GTPase that, via its downstream effectors, regulates a variety of cell functions such as cytokinesis, cell migration, vesicular trafficking, and phagocytosis. As such the RhoA pathway is also pivotal for proper functioning of immune cells including macrophages. By controlling actin cytoskeleton organization, RhoA pathway modulates macrophage's polarity and basic functions: phagocytosis, migration, and extracellular matrix degradation. Numerous studies indicate that macrophages are very important effectors contributing to acute and chronic rejection of transplanted organs. In this review we discuss the role of RhoA pathway in governance of macrophage's functions in terms of transplanted organs.
Collapse
Affiliation(s)
- Yianzhu Liu
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St, Houston, TX, 77030, USA
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jacek Z Kubiak
- CNRS UMR 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, IFR 140 GFAS, Rennes, France
- Faculty of Medicine, University of Rennes 1, 35043, Rennes, France
- Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| | - Xian C Li
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St, Houston, TX, 77030, USA
| | - Rafik M Ghobrial
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St, Houston, TX, 77030, USA
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston, TX, USA
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St, Houston, TX, 77030, USA.
| |
Collapse
|
35
|
Cellular and molecular profiling of graft injury post renal transplantation. Curr Opin Organ Transplant 2016; 22:36-45. [PMID: 27941467 DOI: 10.1097/mot.0000000000000377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Continues advancements in assessing methods for biomolecules that have assisted to identify surrogate candidate biomarkers that can be used to monitor the transplanted organ. These high-throughput methods can help researchers to significantly speed up the identification and the validation steps, which are crucial factors for biomarker discovery efforts. However, this task in transplantation confronts multiple limitations. The review summarizes main findings using 'omics approaches in the evaluation of different types of allograft injury with the overarching aim of evaluating the next steps for transferring the available data to the clinical setting. RECENT FINDINGS Significant discoveries have been made about the molecular and cellular mechanisms that associate with graft injury that may lead to early biomarkers of graft injury (prediction and diagnosis) with the goal of improving long-term outcomes by extending the lifespan of the graft and/or identifying new therapeutic targets. SUMMARY Common efforts among researchers are needed for transferring biomarkers to the clinical setting and, moreover, elucidate pathways that may allow for early interventions to avoid fibrosis progression and graft loss. Large and prospective studies for validation of current available data under strict analytical evaluation are needed to move biomarkers from the discovery phase to validation and clinical implementation.
Collapse
|