1
|
Zheng M, Yang Z, Shi L, Zhao L, Liu K, Tang N. The role of lncRNAs in AKI and CKD: Molecular mechanisms, biomarkers, and potential therapeutic targets. Genes Dis 2025; 12:101509. [PMID: 40083322 PMCID: PMC11904545 DOI: 10.1016/j.gendis.2024.101509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 02/04/2024] [Accepted: 11/02/2024] [Indexed: 03/16/2025] Open
Abstract
Exosomes, a type of extracellular vesicle, are commonly found in different body fluids and are rich in nucleic acids (circRNA, lncRNAs, miRNAs, mRNAs, tRNAs, etc.), proteins, and lipids. They are involved in intercellular communication. lncRNAs are responsible for the modulation of gene expression, thus affecting the pathological process of kidney injury. This review summarizes the latest knowledge on the roles of exosome lncRNAs and circulating lncRNAs in the pathogenesis, biomarker discovery, and treatment of chronic kidney disease, renal fibrosis, and acute kidney injury, providing an overview of novel regulatory approaches and lncRNA delivery systems.
Collapse
Affiliation(s)
- Minhui Zheng
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Zixuan Yang
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Lei Shi
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Liyuan Zhao
- Anhui University of Traditional Chinese Medicine, Hefei, Anhui 230000, China
- Yangtze Delta Drug Advanced Research Institute, Yangtze Delta Pharmaceutical College, Nantong, Jiangsu 226133, China
| | - Kelan Liu
- Intensive Care Unit, Liyang People's Hospital, Liyang, Jiangsu 213300, China
| | - Naping Tang
- Shanghai Innostar Bio-Technology Co., Ltd., China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| |
Collapse
|
2
|
Mu YF, Mao ZH, Pan SK, Liu DW, Liu ZS, Wu P, Gao ZX. Macrophage-driven inflammation in acute kidney injury: Therapeutic opportunities and challenges. Transl Res 2025; 278:1-9. [PMID: 39954848 DOI: 10.1016/j.trsl.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Acute kidney injury (AKI) is increasingly being recognized as a systemic disorder associated with significant morbidity and mortality. AKI manifests with extensive cellular damage, necrosis, and an intense inflammatory response, often leading to late-stage interstitial fibrosis. Although the mechanisms underlying renal injury and repair remain poorly understood, macrophages (pivotal inflammatory cells) play central roles in AKI. They undergo polarization into pro-inflammatory and anti-inflammatory phenotypes, contributing dynamically to both the injury and repair processes while maintaining homeostasis. Macrophages modulate microenvironmental inflammation by releasing extracellular vesicles (EVs) containing pro- or anti-inflammatory signaling molecules, thereby influencing the regulation of tissue injury. The injured tissue cells release EVs and activate local macrophages to initiate these responses. Our bibliometric analysis indicated that a shift has occurred in AKI macrophage research towards therapeutic strategies and clinical translation, focusing on macrophage-targeted therapies, including exosomes and nanoparticles. This review highlights the roles and mechanisms of macrophage activation, phenotypic polarization, and trans-differentiation in AKI and discusses macrophage-based approaches for AKI prevention and treatment. Understanding the involvement of macrophages in AKI contributes to the comprehension of related immune mechanisms and lays the groundwork for novel diagnostic and therapeutic avenues.
Collapse
Affiliation(s)
- Ya-Fan Mu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zi-Hui Mao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Shao-Kang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Dong-Wei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Zhang-Suo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China
| | - Peng Wu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| | - Zhong-Xiuzi Gao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Institute of Nephrology, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, China; Henan Province Research Center for Kidney Disease, Zhengzhou, China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, China.
| |
Collapse
|
3
|
Avalos-de Leon CG, Thomson AW. Regulatory Immune Cell-derived Exosomes: Modes of Action and Therapeutic Potential in Transplantation. Transplantation 2025:00007890-990000000-00994. [PMID: 39865513 DOI: 10.1097/tp.0000000000005309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Reduced dependence on antirejection agents, improved long-term allograft survival, and induction of operational tolerance remain major unmet needs in organ transplantation due to the limitations of current immunosuppressive therapies. To address this challenge, investigators are exploring the therapeutic potential of adoptively transferred host- or donor-derived regulatory immune cells. Extracellular vesicles of endosomal origin (exosomes) secreted by these cells seem to be important contributors to their immunoregulatory properties. Twenty years ago, it was first reported that donor-derived exosomes could extend the survival of transplanted organs in rodents. Recent studies have revealed that regulatory immune cells, such as regulatory myeloid cells (dendritic cells, macrophages, or myeloid-derived suppressor cells), regulatory T cells, or mesenchymal stem/stromal cells can suppress graft rejection via exosomes that express a cargo of immunosuppressive molecules. These include cell surface molecules that interact with adaptive immune cell receptors, immunoregulatory enzymes, and micro- and long noncoding RNAs that can regulate inflammatory gene expression via posttranscriptional changes and promote tolerance through promotion of regulatory T cells. This overview analyzes the diverse molecules and mechanisms that enable regulatory immune cell-derived exosomes to modulate alloimmunity and promote experimental transplant tolerance. We also discuss the potential benefits and limitations of their application as therapeutic entities in organ transplantation.
Collapse
Affiliation(s)
- Cindy G Avalos-de Leon
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| |
Collapse
|
4
|
Lin Z, Cai W, Sun Y, Han B, Hu Y, He Z, Chen X. Mechanism and application of mesenchymal stem cells and their secreting extracellular vesicles in regulating CD4 +T cells in immune diseases. BIOPHYSICS REPORTS 2024; 10:403-415. [PMID: 39758422 PMCID: PMC11693500 DOI: 10.52601/bpr.2024.240005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/07/2024] [Indexed: 01/07/2025] Open
Abstract
Mesenchymal stem cells (MSCs) show significant promise in treating immune diseases due to their ability to differentiate into various cell types and their immunomodulatory properties. However, the mechanisms by which MSCs regulate CD4+T cells, essential for immune responses, are not yet fully understood. This study aims to provide a comprehensive overview of how MSCs and their secreted extracellular vesicles (EVs) modulate CD4+T cells in immune diseases. We begin by discussing the immunomodulatory properties of MSCs and the factors contributing to their effectiveness. Following this, we explore how MSCs interact with CD4+T cells through various pathways, including the secretion of soluble factors, direct cell-cell contact, and EV-mediated communication. A key focus is on the therapeutic potential of MSC-derived EVs, which are rich in bioactive molecules such as proteins, lipids, and nucleic acids. These molecules can regulate the phenotype and function of CD4+T cells. The challenges and future perspectives in utilizing MSCs and EVs for immune-disease therapy are also addressed. Overall, this research aims to enhance our understanding of the mechanisms behind MSC-mediated regulation of CD4+T cells and provide insights into the potential use of MSCs and EVs as therapeutic tools in immune diseases. In summary, understanding how MSCs and their EVs control CD4+T cells can offer valuable perspectives for developing innovative immunotherapeutic approaches. Leveraging the immunomodulatory capacity of MSCs and EVs holds promise for managing immune-related disorders.
Collapse
Affiliation(s)
- Zehua Lin
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Weisong Cai
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yuechen Sun
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Baoai Han
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yifan Hu
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Zuhong He
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Xiong Chen
- Department of Otolaryngology, Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
- Sleep Medicine Centre, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| |
Collapse
|
5
|
Peng XC, Ma LL, Miao JY, Xu SQ, Shuai ZW. Differential lncRNA profiles of blood plasma-derived exosomes from systemic lupus erythematosus. Gene 2024; 927:148713. [PMID: 38906394 DOI: 10.1016/j.gene.2024.148713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
INTRODUCTION Long non-coding RNAs (lncRNAs) dysregulation is key in the pathogenesis of systemic lupus erythematosus (SLE), but the role of exosomal lncRNAs in SLE has not been well studied. We elucidated the profiles of plasma exosomal lncRNAs expression in patients with SLE and predictd their potential clinical significance in SLE. METHODS In the screening stage, six newly diagnosed and untreated patients with SLE and six healthy controls were examined by high-throughput sequencing technology, and differential exosomal lncRNA profiles were constructed. In the validation phase, two differentially selected exosomal lncRNAs from 20 patients each with active and stable SLE and 20 healthy controls were verified with RT-qPCR. The correlation between the selected exosomal lncRNAs and SLE clinical indicators was examined. The diagnostic value of the selected exosomal lncRNAs in SLE was analyzed by the receiver operator characteristic (ROC) curve. RESULTS Exosomes were successfully extracted from the patients and controls. Sequencing-phase sequencing demonstrated 528 upregulated lncRNAs and 7491 downregulated lncRNAs. In the validation stage, exosomal LINC00667 and DANCR were significantly upregulated in the patients, and positively correlated with Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2 K). Exosomal DANCR expression between the active and stable SLE patients was different. The area under the curve(AUC) of exosomal LINC00667 and DANCR for SLE diagnosis was 0.815 and 0.759, respectively. CONCLUSIONS Exosomal LINC00667 and DANCR were upregulated in SLE, and might be new biomarkers thereof. Exosomal DANCR was associated with SLE activity.
Collapse
Affiliation(s)
- Xin-Chen Peng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Ling-Li Ma
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Jie-Yu Miao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Sheng-Qian Xu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| | - Zong-Wen Shuai
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
6
|
Huang D, Shen H, Xie F, Hu D, Jin Q, Hu Y, Zhong T. Role of mesenchymal stem cell-derived exosomes in the regeneration of different tissues. J Biol Eng 2024; 18:36. [PMID: 38845032 PMCID: PMC11155050 DOI: 10.1186/s13036-024-00431-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/10/2024] [Indexed: 06/10/2024] Open
Abstract
Exosomes are nanovesicles with multiple components used in several applications. Mesenchymal stem cells (MSCs) are well known for their great potential in clinical applications. MSC-derived exosomes (MSC-Exos) have been shown to mediate tissue regeneration in various diseases, including neurological, autoimmune, and inflammatory diseases, cancer, ischemic heart disease, lung injury, and liver fibrosis. They can modulate the immune response by interacting with immune effector cells in the presence of anti-inflammatory compounds and are involved in intercellular communication through various types of cargo. This review summarizes the MSC-Exos-mediated tissue regeneration in various diseases, including neurological, cardiovascular, liver, kidney, articular cartilage, and oral tissue applications. In addition, we discuss the challenges and prospects of MSC-Exos in tissue regeneration.
Collapse
Affiliation(s)
- Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Die Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yuexin Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Tianyu Zhong
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
- Precision Medicine Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| |
Collapse
|
7
|
Zhong P, Bai L, Hong M, Ouyang J, Wang R, Zhang X, Chen P. A Comprehensive Review on Circulating cfRNA in Plasma: Implications for Disease Diagnosis and Beyond. Diagnostics (Basel) 2024; 14:1045. [PMID: 38786343 PMCID: PMC11119755 DOI: 10.3390/diagnostics14101045] [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: 04/14/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Circulating cfRNA in plasma has emerged as a fascinating area of research with potential applications in disease diagnosis, monitoring, and personalized medicine. Circulating RNA sequencing technology allows for the non-invasive collection of important information about the expression of target genes, eliminating the need for biopsies. This comprehensive review aims to provide a detailed overview of the current knowledge and advancements in the study of plasma cfRNA, focusing on its diverse landscape and biological functions, detection methods, its diagnostic and prognostic potential in various diseases, challenges, and future perspectives.
Collapse
Affiliation(s)
- Pengqiang Zhong
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Lu Bai
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Mengzhi Hong
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Juan Ouyang
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ruizhi Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoli Zhang
- Department of Pediatrics, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Peisong Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
8
|
Kuang Y, Yang J, Sun M, Rui T, Yang Z, Shi M. Depression of LncRNA DANCR alleviates tubular injury in diabetic nephropathy by regulating KLF5 through sponge miR-214-5p. BMC Nephrol 2024; 25:130. [PMID: 38609873 PMCID: PMC11010359 DOI: 10.1186/s12882-024-03562-6] [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: 11/16/2023] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
OBJECTIVE Diabetic nephropathy (DN) manifests a critical aspect in the form of renal tubular injury. The current research aimed to determine the function and mechanism of long non-coding ribonucleic acid (LncRNA) differentiation antagonising non-protein coding RNA (DANCR), with a focus on its impact on renal tubular injury. METHODS Quantitative reverse transcription polymerase chain reaction was employed to analyze the RNA levels of DANCR in the serum of patients with DN or human proximal tubular epithelial cells (human kidney 2 [HK2]). The diagnostic significance of DANCR was assessed using a receiver operating characteristic curve. A DN model was established by inducing HK-2 cells with high glucose (HG). Cell proliferation, apoptosis, and the levels of inflammatory factors, reactive oxygen species (ROS), and malondialdehyde (MDA) were detected using the Cell Counting Kit - 8, flow cytometry, and enzyme-linked immunosorbent assay. The interaction between microRNA (miR)-214-5p and DANCR or Krüppel-like factor 5 (KLF5) was investigated using RNA immunoprecipitation and dual-luciferase reporter assays. RESULTS Elevated levels of DANCR were observed in the serum of patients with DN and HG-inducted HK-2 cells (P < 0.05). DANCR levels effectively identified patients with DN from patients with type 2 diabetes mellitus. Silencing of DANCR protected against HG-induced tubular injury by restoring cell proliferation, inhibiting apoptosis, and reducing the secretion of inflammatory factors and oxidative stress production (P < 0.05). DANCR functions as a sponge for miR-214-5p, and the mitigation of DANCR silencing on HG-induced renal tubular injury was partially attenuated with reduced miR-214-5p (P < 0.05). Additionally, KLF5 was identified as the target of miR-214-5p. CONCLUSION DANCR was identified as diagnostic potential for DN and the alleviation of renal tubular injury via the miR-214-5p/KLF5 axis, following DANCR silencing, introduces a novel perspective and approach to mitigating DN.
Collapse
Affiliation(s)
- Yongling Kuang
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China.
| | - Juan Yang
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China
| | - Meimei Sun
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China
| | - Tingting Rui
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China
| | - Zhenhua Yang
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China
| | - Meihua Shi
- Department of Nephrology, Gongli Hospital of Shanghai Pudong New Area, No. 219 Miaopu Road, Pudong New Area, 200135, Shanghai, China
| |
Collapse
|
9
|
Ramalhete L, Araújo R, Ferreira A, Calado CRC. Exosomes and microvesicles in kidney transplantation: the long road from trash to gold. Pathology 2024; 56:1-10. [PMID: 38071158 DOI: 10.1016/j.pathol.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 09/26/2023] [Accepted: 10/19/2023] [Indexed: 01/24/2024]
Abstract
Kidney transplantation significantly enhances the survival rate and quality of life of patients with end-stage kidney disease. The ability to predict post-transplantation rejection events in their early phases can reduce subsequent allograft loss. Therefore, it is critical to identify biomarkers of rejection processes that can be acquired on routine analysis of samples collected by non-invasive or minimally invasive procedures. It is also important to develop new therapeutic strategies that facilitate optimisation of the dose of immunotherapeutic drugs and the induction of allograft immunotolerance. This review explores the challenges and opportunities offered by extracellular vesicles (EVs) present in biofluids in the discovery of biomarkers of rejection processes, as drug carriers and in the induction of immunotolerance. Since EVs are highly complex structures and their composition is affected by the parent cell's metabolic status, the importance of defining standardised methods for isolating and characterising EVs is also discussed. Understanding the major bottlenecks associated with all these areas will promote the further investigation of EVs and their translation into a clinical setting.
Collapse
Affiliation(s)
- Luis Ramalhete
- Blood and Transplantation Center of Lisbon, Instituto Português do Sangue e da Transplantação, Alameda das Linhas de Torres, Lisbon, Portugal; NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal; iNOVA4Health - Advancing Precision Medicine, RG11: Reno-Vascular Diseases Group, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal.
| | - Ruben Araújo
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Aníbal Ferreira
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal; Centro Hospitalar Universitário Lisboa Central, Hospital Curry Cabral, Serviço de Nefrologia, NOVA Medical School, Lisbon, Portugal
| | - Cecília R C Calado
- ISEL - Instituto Superior de Engenharia de Lisboa, Instituto Politécnico de Lisboa, R. Conselheiro Emídio Navarro 1, Lisbon, Portugal; CIMOSM - Centro de Investigação em Modelação e Otimização de Sistemas Multifuncionais, Lisbon, Portugal
| |
Collapse
|
10
|
Caserta S, Stagno F, Gangemi S, Allegra A. Highlights on the Effects of Non-Coding RNAs in the Osteonecrosis of the Jaw. Int J Mol Sci 2024; 25:1598. [PMID: 38338876 PMCID: PMC10855359 DOI: 10.3390/ijms25031598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Osteonecrosis of the jaw is the progressive loss and destruction of bone affecting the maxilla or mandible in patients treated with antiresorptive and antiangiogenic agents without receiving prior radiation therapy. The pathogenesis involves the inflammatory pathway of receptor activator of nuclear factor NF-kB ligand and the macrophage colony-stimulating factor, essential for osteoclast precursors survival and proliferation and acting through its receptor c-Fms. Evidence has shown the role of non-coding RNAs in the pathogenesis of osteonecrosis of the jaw and this finding might be useful in diagnosis since these small RNAs could be considered as biomarkers of apoptotic activity in bone. Interestingly, it has been proved that miR-29 and miR-31-5p, acting on specific targets such as CALCR and RhoA, promote programmed-cell death and consequently the necrosis of bone tissue. Specific long non-coding RNAs, instead, have been detected both at reduced levels in patients with multiple myeloma and osteonecrosis, and associated with suppression of osteoblast differentiation, with consequences in the progression of mandible lesions. Among non-coding genic material, circular RNAs have the capability to modify the expression of specific mRNAs responsible for the inhibition of bisphosphonates activity on osteoclastogenesis.
Collapse
Affiliation(s)
- Santino Caserta
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Fabio Stagno
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Alessandro Allegra
- Hematology Unit, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy; (S.C.); (A.A.)
| |
Collapse
|
11
|
Abinti M, Favi E, Alfieri CM, Zanoni F, Armelloni S, Ferraresso M, Cantaluppi V, Castellano G. Update on current and potential application of extracellular vesicles in kidney transplantation. Am J Transplant 2023; 23:1673-1693. [PMID: 37517555 DOI: 10.1016/j.ajt.2023.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/28/2023] [Accepted: 07/17/2023] [Indexed: 08/01/2023]
Abstract
Kidney transplantation (KT) is the best treatment for end-stage kidney disease. However, early diagnosis of graft injury remains challenging, mainly because of the lack of accurate and noninvasive diagnostic techniques. Improving graft outcomes is equally demanding, as is the development of innovative therapies. Many research efforts are focusing on extracellular vesicles, cellular particles free in each body fluid that have shown promising results as precise markers of damage and potential therapeutic targets in many diseases, including the renal field. In fact, through their receptors and cargo, they act in damage response and immune modulation. In transplantation, they may be used to determine organ quality and aging, the presence of delayed graft function, rejection, and many other transplant-related pathologies. Moreover, their low immunogenicity and safe profile make them ideal for drug delivery and the development of therapies to improve KT outcomes. In this review, we summarize current evidence about extracellular vesicles in KT, starting with their characteristics and major laboratory techniques for isolation and characterization. Then, we discuss their use as potential markers of damage and as therapeutic targets, discussing their promising use in clinical practice as a form of liquid biopsy.
Collapse
Affiliation(s)
- Matteo Abinti
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Evaldo Favi
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Carlo Maria Alfieri
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Francesca Zanoni
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Division of Nephrology, Department of Medicine, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA
| | - Silvia Armelloni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Mariano Ferraresso
- Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Vincenzo Cantaluppi
- Nephrology and Kidney Transplant Unit, Department of Translational Medicine (DIMET), University of Piemonte Orientale (UPO), "Maggiore della Carita" University Hospital, Novara, Italy
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
12
|
van Zonneveld AJ, Zhao Q, Rotmans JI, Bijkerk R. Circulating non-coding RNAs in chronic kidney disease and its complications. Nat Rev Nephrol 2023; 19:573-586. [PMID: 37286733 DOI: 10.1038/s41581-023-00725-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/09/2023]
Abstract
Post-transcriptional regulation by non-coding RNAs (ncRNAs) can modulate the expression of genes involved in kidney physiology and disease. A large variety of ncRNA species exist, including microRNAs, long non-coding RNAs, piwi-interacting RNAs, small nucleolar RNAs, circular RNAs and yRNAs. Despite early assumptions that some of these species may exist as by-products of cell or tissue injury, a growing body of literature suggests that these ncRNAs are functional and participate in a variety of processes. Although they function intracellularly, ncRNAs are also present in the circulation, where they are carried by extracellular vesicles, ribonucleoprotein complexes or lipoprotein complexes such as HDL. These systemic, circulating ncRNAs are derived from specific cell types and can be directly transferred to a variety of cells, including endothelial cells of the vasculature and virtually any cell type in the kidney, thereby affecting the function of the host cell and/or its response to injury. Moreover, chronic kidney disease itself, as well as injury states associated with transplantation and allograft dysfunction, is associated with a shift in the distribution of circulating ncRNAs. These findings may provide opportunities for the identification of biomarkers with which to monitor disease progression and/or the development of therapeutic interventions.
Collapse
Affiliation(s)
- Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Qiao Zhao
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Joris I Rotmans
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Roel Bijkerk
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, the Netherlands.
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
13
|
Yin S, Zhou Z, Fu P, Jin C, Wu P, Ji C, Shan Y, Shi L, Xu M, Qian H. Roles of extracellular vesicles in ageing-related chronic kidney disease: demon or angel. Pharmacol Res 2023:106795. [PMID: 37211241 DOI: 10.1016/j.phrs.2023.106795] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/02/2023] [Accepted: 05/16/2023] [Indexed: 05/23/2023]
Abstract
Ageing is a universal and unavoidable phenomenon that significantly increases the risk of developing chronic kidney disease (CKD). It has been reported that ageing is associated with functional disruption and structural damage to the kidneys. Extracellular vesicles (EVs), which are nanoscale membranous vesicles containing lipids, proteins, and nucleic acids, are secreted by cells into the extracellular spaces. They have diverse functions such as repairing and regenerating different forms of ageing-related CKD and playing a crucial role in intercellular communication. This paper reviews the etiology of ageing in CKD, with particular attention paid to the roles of EVs as carriers of ageing signals and anti-ageing therapeutic strategies in CKD. In this regard, the double-edged role of EVs in ageing-related CKD is examined, along with the potential for their application in clinical settings.
Collapse
Affiliation(s)
- Siqi Yin
- Institute of Translational Medicine of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Zixuan Zhou
- Institute of Translational Medicine of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Peiwen Fu
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Chaoying Jin
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| | - Peipei Wu
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Cheng Ji
- Institute of Translational Medicine of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yunjie Shan
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Linru Shi
- Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Min Xu
- Institute of Translational Medicine of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China.
| | - Hui Qian
- Institute of Translational Medicine of Jiangsu University, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, Jiangsu, China; Key Laboratory of Laboratory Medicine of Jiangsu Province, Department of laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, China.
| |
Collapse
|
14
|
Zhang T, Lu L, Li M, Zhang D, Yu P, Zhang X, Zhang Z, Lei C. Exosome from BMMSC Attenuates Cardiopulmonary Bypass-Induced Acute Lung Injury Via YAP/β-Catenin Pathway: Downregulation of Pyroptosis. Stem Cells 2022; 40:1122-1133. [PMID: 36063391 DOI: 10.1093/stmcls/sxac063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/22/2022] [Indexed: 01/04/2023]
Abstract
Acute lung injury (ALI) accompanied with systemic inflammatory response is an important complication after cardiopulmonary bypass (CPB). Pyroptosis, which is induced by the secretion of inflammatory factors, has been implicated in ALI. However, recent studies have suggested that bone marrow mesenchymal stem cell-derived exosomes (BMMSC-Exo) can ameliorate ALI, but the mechanism is poorly understood. Therefore, we aim to examine the effects of BMMSC-Exo in CPB-induced ALI, and its underlying mechanism. CPB rat models (male Sprague-Dawley rats) were administered BMMSC-Exo intravenously before induction of ALI. Lung tissue, bronchoalveolar lavage fluid (BALF), and alveolar macrophage (AM) were collected after the treatments for further analysis, and rat AM NR8383 cells were used for in vitro study. HE staining was performed to detect macrophage infiltration. Western blot was used to detect related proteins expression. And ELISA assay was performed to investigate secretion of inflammatory factors. These results showed that BMMSC-Exo treatment ameliorated macrophage infiltration and oxidative stress, and downregulated expression of pyroptosis-related proteins, including NLRP3, cleaved caspase-1, and GSDMD-N, in the lung tissue and AM, as well as decreased the secretion of IL-18 and IL-1β in BALF. Moreover, BMMSC-Exo activated YAP/β-catenin signaling pathway. Overall, these findings of this study indicated that BMMSC-Exo suppressed CPB-induced pyroptosis in ALI by activating YAP/β-catenin axis, which could be a novel strategy for lung protection during CPB.
Collapse
Affiliation(s)
- Taoyuan Zhang
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China.,Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Linhe Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Man Li
- Central Medical Branch of PLA General Hospital, Beijing, People's Republic of China
| | - Ding Zhang
- Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Peng Yu
- Department of Anesthesiology, Rizhao Traditional Chinese Medicine Hospital, Rizhao, Shandong, People's Republic of China
| | - Xinhao Zhang
- Department of Anesthesiology, Rizhao International Heart Hospital, Rizhao, Shandong, People's Republic of China
| | - Zheng Zhang
- Department of Cardiology, PLA Rocket Force Characteristic Medical Center, Beijing, People's Republic of China
| | - Chong Lei
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
15
|
Lu Y, Wang L, Zhang M, Chen Z. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles: A Novel Approach for Kidney Disease Treatment. Int J Nanomedicine 2022; 17:3603-3618. [PMID: 35990308 PMCID: PMC9386173 DOI: 10.2147/ijn.s372254] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/04/2022] [Indexed: 12/24/2022] Open
Abstract
Globally, kidney disease has become a serious health challenge, with approximately 10% of adults suffering with the disease, and increasing incidence and mortality rates every year. Small extracellular vesicles (sEVs) are 30 nm-100 nm sized nanovesicles released by cells into the extracellular matrix (ECM), which serve as mediators of intercellular communication. Depending on the cell origin, sEVs have different roles which depend on internal cargoes including, nucleic acids, proteins, and lipids. Mesenchymal stem cell (MSCs) exert anti-inflammatory, anti-aging, and wound healing functions mainly via sEVs in a stable and safe manner. MSC-derived sEVs (MSC-sEVs) exert roles in several kidney diseases by transporting renoprotective cargoes to reduce oxidative stress, inhibit renal cell apoptosis, suppress inflammation, and mediate anti-fibrosis mechanisms. Additionally, because MSC-sEVs efficiently target damaged kidneys, they have the potential to become the next generation cell-free therapies for kidney disease. Herein, we review recent research data on how MSC-sEVs could be used to treat kidney disease.
Collapse
Affiliation(s)
- Yukang Lu
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Mengting Zhang
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Zhiping Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| |
Collapse
|
16
|
Fang Y, Bouari S, Hoogduijn MJ, Ijzermans JNM, de Bruin RWF, Minnee RC. Therapeutic efficacy of extracellular vesicles to suppress allograft rejection in preclinical kidney transplantation models: A systematic review and meta-analysis. Transplant Rev (Orlando) 2022; 36:100714. [PMID: 35853384 DOI: 10.1016/j.trre.2022.100714] [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: 05/21/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND Kidney transplantation is the optimal treatment of end-stage renal disease. Extracellular vesicles (EVs) have tremendous therapeutic potential, but their role in modulating immune responses in kidney transplantation remains unclear. METHODS We performed a systematic review and meta-analysis to investigate the therapeutic efficacy of EVs in preclinical kidney transplant models. Outcomes for meta-analysis were graft survival and renal function. Subgroup analysis was conducted between immune cell derived EVs (immune cell-EVs) and mesenchymal stromal cell derived EVs (MSC-EVs). RESULTS Seven studies published from 2013 to 2021 were included. The overall effects showed that EVs had a positive role in prolonging allograft survival (standardized mean difference (SMD) = 2.00; 95% confidence interval (CI), 0.79 to 3.21; P < 0.01; I2 = 94%), reducing serum creatinine (SCr) (SMD = -2.19; 95%CI, -3.35 to -1.04; P < 0.01; I2 = 93%) and blood urea nitrogen (BUN) concentrations (SMD = -1.69; 95%CI, -2.98 to -0.40; P = 0.01; I2 = 94%). Subgroup analyses indicated that only immune cell-EVs significantly prolonged graft survival and improve renal function but not MSC-EVs. CONCLUSIONS EVs are promising candidates to suppress allograft rejection and improve kidney transplant outcome. Immune cell-EVs showed their superiority over MSC-EVs in prolonging graft survival and improving renal function. For interpretation of the outcomes, additional studies are needed to validate these findings.
Collapse
Affiliation(s)
- Yitian Fang
- Erasmus MC Transplant Institute, Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Sarah Bouari
- Erasmus MC Transplant Institute, Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Martin J Hoogduijn
- Erasmus MC Transplant Institute, Nephrology and Transplantation, Department of Internal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Jan N M Ijzermans
- Erasmus MC Transplant Institute, Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Ron W F de Bruin
- Erasmus MC Transplant Institute, Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Robert C Minnee
- Erasmus MC Transplant Institute, Division of HPB and Transplant Surgery, Department of Surgery, Erasmus MC University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
17
|
Proteomics for Biomarker Discovery for Diagnosis and Prognosis of Kidney Transplantation Rejection. Proteomes 2022; 10:proteomes10030024. [PMID: 35893765 PMCID: PMC9326686 DOI: 10.3390/proteomes10030024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023] Open
Abstract
Renal transplantation is currently the treatment of choice for end-stage kidney disease, enabling a quality of life superior to dialysis. Despite this, all transplanted patients are at risk of allograft rejection processes. The gold-standard diagnosis of graft rejection, based on histological analysis of kidney biopsy, is prone to sampling errors and carries high costs and risks associated with such invasive procedures. Furthermore, the routine clinical monitoring, based on urine volume, proteinuria, and serum creatinine, usually only detects alterations after graft histologic damage and does not differentiate between the diverse etiologies. Therefore, there is an urgent need for new biomarkers enabling to predict, with high sensitivity and specificity, the rejection processes and the underlying mechanisms obtained from minimally invasive procedures to be implemented in routine clinical surveillance. These new biomarkers should also detect the rejection processes as early as possible, ideally before the 78 clinical outputs, while enabling balanced immunotherapy in order to minimize rejections and reducing the high toxicities associated with these drugs. Proteomics of biofluids, collected through non-invasive or minimally invasive analysis, e.g., blood or urine, present inherent characteristics that may provide biomarker candidates. The current manuscript reviews biofluids proteomics toward biomarkers discovery that specifically identify subclinical, acute, and chronic immune rejection processes while allowing for the discrimination between cell-mediated or antibody-mediated processes. In time, these biomarkers will lead to patient risk stratification, monitoring, and personalized and more efficient immunotherapies toward higher graft survival and patient quality of life.
Collapse
|
18
|
Xiang X, Zhu J, Dong G, Dong Z. Epigenetic Regulation in Kidney Transplantation. Front Immunol 2022; 13:861498. [PMID: 35464484 PMCID: PMC9024296 DOI: 10.3389/fimmu.2022.861498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/17/2022] [Indexed: 12/29/2022] Open
Abstract
Kidney transplantation is a standard care for end stage renal disease, but it is also associated with a complex pathogenesis including ischemia-reperfusion injury, inflammation, and development of fibrosis. Over the past decade, accumulating evidence has suggested a role of epigenetic regulation in kidney transplantation, involving DNA methylation, histone modification, and various kinds of non-coding RNAs. Here, we analyze these recent studies supporting the role of epigenetic regulation in different pathological processes of kidney transplantation, i.e., ischemia-reperfusion injury, acute rejection, and chronic graft pathologies including renal interstitial fibrosis. Further investigation of epigenetic alterations, their pathological roles and underlying mechanisms in kidney transplantation may lead to new strategies for the discovery of novel diagnostic biomarkers and therapeutic interventions.
Collapse
Affiliation(s)
- Xiaohong Xiang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States.,Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiefu Zhu
- Center of Nephrology and Dialysis, Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guie Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| | - Zheng Dong
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital of Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veteran Affairs (VA) Medical Center, Augusta, GA, United States
| |
Collapse
|