1
|
Reikvam H, Hatfield K, Sandnes M, Bruserud Ø. Future biomarkers for acute graft-versus-host disease: potential roles of nucleic acids, metabolites, and immune cell markers. Expert Rev Clin Immunol 2025; 21:305-321. [PMID: 39670445 DOI: 10.1080/1744666x.2024.2441246] [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: 07/09/2024] [Revised: 10/04/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION Acute graft versus host disease (aGVHD) is a potentially lethal complication after allogeneic stem cell transplantation. Biomarkers are used to estimate the risk of aGVHD and evaluate response to treatment. The most widely used biomarkers are systemic levels of various protein mediators involved in immunoregulation or reflecting tissue damage. However, systemic levels of other molecular markers such as nucleic acids or metabolites, levels of immunocompetent cells or endothelial cell markers may also be useful biomarkers in aGVHD. AREAS COVERED This review is based on selected articles from the PubMed database. We review and discuss the scientific basis for further studies to evaluate nucleic acids, metabolites, circulating immunocompetent cell subsets or endothelial markers as biomarkers in aGVHD. EXPERT OPINION A wide range of interacting and communicating cells are involved in the complex pathogenesis of aGVHD. Both nucleic acids and metabolites function as soluble mediators involved in communication between various subsets of immunocompetent cells and between immunocompetent cells and other neighboring cells. Clinical and experimental studies suggest that both neutrophils, monocytes, and endothelial cells are involved in the early stages of aGVHD pathogenesis. In our opinion, the possible clinical use of these molecular and cellular biomarkers warrants further investigation.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Kimberley Hatfield
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, Bergen, Norway
| | - Miriam Sandnes
- Division for Hematology, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
2
|
Huang Z, Wang L, Li W, Liao N, Heng J, Qin Y, Li L, Bian Z, Cao W, Xia L, Zhang R. The role of lncRNA NEAT1 in acute graft-versus-host disease: Regulation of macrophage polarization and inflammatory cytokine secretion via JNK/NLRP3 pathway. Int Immunopharmacol 2025; 146:113857. [PMID: 39721453 DOI: 10.1016/j.intimp.2024.113857] [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: 01/11/2024] [Revised: 11/23/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Acute graft-versus-host disease (aGVHD) is a complication of allogeneic hematopoietic stem cell transplantation (allo-HSCT). The role of macrophages as proficient antigen-presenting cells in aGVHD is a prominent area of investigation in contemporary research. The association between long noncoding RNA nuclear enriched abundant transcript 1 (lncRNA NEAT1) and the macrophage function is of significant interest. However, the role of lncRNA NEAT1 in aGVHD needs to be further explored. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from patients with or without aGVHD (non-aGVHD) after allo-HSCT. RAW264.7 cells and bone marrow-derived macrophages (BMDMs) were transduced with NEAT1 lentiviral vector or transfected with NEAT1 small interfering RNA to change the expression level of lncRNA NEAT1. Finally, an aGVHD mouse model was established to evaluate the role of JNK inhibitor or NLRP3 inhibitor in aGVHD. RESULTS Compared with non-aGVHD patients, lncRNA NEAT1 was significantly up-regulated in the PBMCs of aGVHD patients. ROC and AUC analysis confirmed that the expression of lncRNA NEAT1 was correlated with the occurrence of aGVHD. The overexpression of lncRNA NEAT1 in RAW264.7 could significantly promote the proliferation, migration, and differentiation into M1 macrophages. Knockdown of lncRNA NEAT1 could significantly decrease the proportion of M1 macrophages, regulate pro-inflammatory cytokines secretion, and affect the JNK/NLRP3 pathway in lipopolysaccharides (LPS)-induced BMDMs. Correspondingly, JNK and NLRP3 inhibitors reduced LPS-induced pro-inflammatory responses in macrophages. Furthermore, JNK and NLRP3 inhibitors regulated macrophage polarization and improved symptoms in aGVHD mice. CONCLUSIONS The aforementioned data suggest that lncRNA NEAT1 potentially plays a significant role in macrophage polarization and the secretion of inflammatory cytokines through its modulation of the JNK/NLRP3 pathway. Consequently, this study establishes a foundation for the development of novel therapeutic approaches targeting aGVHD.
Collapse
Affiliation(s)
- Zhenli Huang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ni Liao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingjing Heng
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Qin
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
3
|
Sevcikova A, Fridrichova I, Nikolaieva N, Kalinkova L, Omelka R, Martiniakova M, Ciernikova S. Clinical Significance of microRNAs in Hematologic Malignancies and Hematopoietic Stem Cell Transplantation. Cancers (Basel) 2023; 15:cancers15092658. [PMID: 37174123 PMCID: PMC10177548 DOI: 10.3390/cancers15092658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/14/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
Hematologic malignancies are a group of neoplastic conditions that can develop from any stage of the hematopoiesis cascade. Small non-coding microRNAs (miRNAs) play a crucial role in the post-transcriptional regulation of gene expression. Mounting evidence highlights the role of miRNAs in malignant hematopoiesis via the regulation of oncogenes and tumor suppressors involved in proliferation, differentiation, and cell death. In this review, we provide current knowledge about dysregulated miRNA expression in the pathogenesis of hematological malignancies. We summarize data about the clinical utility of aberrant miRNA expression profiles in hematologic cancer patients and their associations with diagnosis, prognosis, and the monitoring of treatment response. Moreover, we will discuss the emerging role of miRNAs in hematopoietic stem cell transplantation (HSCT), and severe post-HSCT complications, such as graft-versus-host disease (GvHD). The therapeutical potential of the miRNA-based approach in hemato-oncology will be outlined, including studies with specific antagomiRs, mimetics, and circular RNAs (circRNAs). Since hematologic malignancies represent a full spectrum of disorders with different treatment paradigms and prognoses, the potential use of miRNAs as novel diagnostic and prognostic biomarkers might lead to improvements, resulting in a more accurate diagnosis and better patient outcomes.
Collapse
Affiliation(s)
- Aneta Sevcikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Ivana Fridrichova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Nataliia Nikolaieva
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Lenka Kalinkova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 74 Nitra, Slovakia
| | - Sona Ciernikova
- Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
| |
Collapse
|
4
|
Molecular Advances in Sinusoidal Obstruction Syndrome/Veno-Occlusive Disease. Int J Mol Sci 2023; 24:ijms24065620. [PMID: 36982695 PMCID: PMC10051970 DOI: 10.3390/ijms24065620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/07/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD) detected in the liver has been considered a severe complication of hematopoietic stem cell transplantation (HSCT). SOS/VOD is characterized by hepatomegaly, right upper quadrant pain, jaundice, and ascites. The severe forms of the disease may result in multi-organ dysfunction (MOD) with a high mortality rate (>80%). The development of SOS/VOD can be rapid and unpredictable. Therefore, early identification and severity assessment is crucial in facilitating prompt diagnosis and timely treatment. Effective treatment and potential prophylaxis with defibrotide highlight the need for characterizing a sub-group of patients at high risk for SOS/VOD. Moreover, antibodies that are conjugated with calicheamicin, gemtuzumab, and inotuzumab ozogamicin, have led to renewed interest in this syndrome. Evaluation and management of serious adverse events associated with gemtuzumab and inotuzumab ozogamicin are recommended. We review hepatic-, transplant- and patient-related risk factors, criteria for diagnosis and grading classification, and SOS/VOD potential biomarkers. Furthermore, we examine pathogenesis, clinical presentation, diagnostic criteria, risk factors, prophylaxis, and treatment of SOS/VOD occurring post HSCT. Moreover, we aim to provide an up-to-date summary of molecular advances in the diagnosis and management of SOS/VOD. We performed a comprehensive review of the literature and examined the recently available data, mostly using the PubMed and Medline search engines for original articles published over the last decade. In the era of precision medicine, our review provides up-to-date knowledge of genetic or sera markers for SOS/VOD with the goal of identifying a subset of high-risk patients.
Collapse
|
5
|
Khokhar M, Purohit P, Gadwal A, Tomo S, Bajpai NK, Shukla R. The Differentially Expressed Genes Responsible for the Development of T Helper 9 Cells From T Helper 2 Cells in Various Disease States: Immuno-Interactomics Study. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2023; 4:e42421. [PMID: 38935935 PMCID: PMC11135241 DOI: 10.2196/42421] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 01/18/2023] [Accepted: 01/25/2023] [Indexed: 06/29/2024]
Abstract
BACKGROUND T helper (Th) 9 cells are a novel subset of Th cells that develop independently from Th2 cells and are characterized by the secretion of interleukin (IL)-9. Studies have suggested the involvement of Th9 cells in variable diseases such as allergic and pulmonary diseases (eg, asthma, chronic obstructive airway disease, chronic rhinosinusitis, nasal polyps, and pulmonary hypoplasia), metabolic diseases (eg, acute leukemia, myelocytic leukemia, breast cancer, lung cancer, melanoma, pancreatic cancer), neuropsychiatric disorders (eg, Alzheimer disease), autoimmune diseases (eg, Graves disease, Crohn disease, colitis, psoriasis, systemic lupus erythematosus, systemic scleroderma, rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, atopic dermatitis, eczema), and infectious diseases (eg, tuberculosis, hepatitis). However, there is a dearth of information on its involvement in other metabolic, neuropsychiatric, and infectious diseases. OBJECTIVE This study aims to identify significant differentially altered genes in the conversion of Th2 to Th9 cells, and their regulating microRNAs (miRs) from publicly available Gene Expression Omnibus data sets of the mouse model using in silico analysis to unravel various pathogenic pathways involved in disease processes. METHODS Using differentially expressed genes (DEGs) identified from 2 publicly available data sets (GSE99166 and GSE123501) we performed functional enrichment and network analyses to identify pathways, protein-protein interactions, miR-messenger RNA associations, and disease-gene associations related to significant differentially altered genes implicated in the conversion of Th2 to Th9 cells. RESULTS We extracted 260 common downregulated, 236 common upregulated, and 634 common DEGs from the expression profiles of data sets GSE99166 and GSE123501. Codifferentially expressed ILs, cytokines, receptors, and transcription factors (TFs) were enriched in 7 crucial Kyoto Encyclopedia of Genes and Genomes pathways and Gene Ontology. We constructed the protein-protein interaction network and predicted the top regulatory miRs involved in the Th2 to Th9 differentiation pathways. We also identified various metabolic, allergic and pulmonary, neuropsychiatric, autoimmune, and infectious diseases as well as carcinomas where the differentiation of Th2 to Th9 may play a crucial role. CONCLUSIONS This study identified hitherto unexplored possible associations between Th9 and disease states. Some important ILs, including CCL1 (chemokine [C-C motif] ligand 1), CCL20 (chemokine [C-C motif] ligand 20), IL-13, IL-4, IL-12A, and IL-9; receptors, including IL-12RB1, IL-4RA (interleukin 9 receptor alpha), CD53 (cluster of differentiation 53), CD6 (cluster of differentiation 6), CD5 (cluster of differentiation 5), CD83 (cluster of differentiation 83), CD197 (cluster of differentiation 197), IL-1RL1 (interleukin 1 receptor-like 1), CD101 (cluster of differentiation 101), CD96 (cluster of differentiation 96), CD72 (cluster of differentiation 72), CD7 (cluster of differentiation 7), CD152 (cytotoxic T lymphocyte-associated protein 4), CD38 (cluster of differentiation 38), CX3CR1 (chemokine [C-X3-C motif] receptor 1), CTLA2A (cytotoxic T lymphocyte-associated protein 2 alpha), CTLA28, and CD196 (cluster of differentiation 196); and TFs, including FOXP3 (forkhead box P3), IRF8 (interferon regulatory factor 8), FOXP2 (forkhead box P2), RORA (RAR-related orphan receptor alpha), AHR (aryl-hydrocarbon receptor), MAF (avian musculoaponeurotic fibrosarcoma oncogene homolog), SMAD6 (SMAD family member 6), JUN (Jun proto-oncogene), JAK2 (Janus kinase 2), EP300 (E1A binding protein p300), ATF6 (activating transcription factor 6), BTAF1 (B-TFIID TATA-box binding protein associated factor 1), BAFT (basic leucine zipper transcription factor), NOTCH1 (neurogenic locus notch homolog protein 1), GATA3 (GATA binding protein 3), SATB1 (special AT-rich sequence binding protein 1), BMP7 (bone morphogenetic protein 7), and PPARG (peroxisome proliferator-activated receptor gamma, were able to identify significant differentially altered genes in the conversion of Th2 to Th9 cells. We identified some common miRs that could target the DEGs. The scarcity of studies on the role of Th9 in metabolic diseases highlights the lacunae in this field. Our study provides the rationale for exploring the role of Th9 in various metabolic disorders such as diabetes mellitus, diabetic nephropathy, hypertensive disease, ischemic stroke, steatohepatitis, liver fibrosis, obesity, adenocarcinoma, glioblastoma and glioma, malignant neoplasm of stomach, melanoma, neuroblastoma, osteosarcoma, pancreatic carcinoma, prostate carcinoma, and stomach carcinoma.
Collapse
Affiliation(s)
- Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Ashita Gadwal
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Sojit Tomo
- Department of Biochemistry, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Nitin Kumar Bajpai
- Department of Nephrology, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| | - Ravindra Shukla
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Jodhpur, Jodhpur, India
| |
Collapse
|
6
|
Carneiro TX, Marrese DG, Dos Santos MG, Gonçalves MV, Novis YAS, Rizzatti EG, Rocha V, Sandes AF, de Lacerda MP, Arrais-Rodrigues C. Circulating extracellular vesicles as a predictive biomarker for acute graft-versus-host disease. Exp Hematol 2023; 117:15-23. [PMID: 36400315 DOI: 10.1016/j.exphem.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/19/2022]
Abstract
The diagnosis and management of graft-versus-host disease (GVHD) have remained important challenges in allogeneic stem cell transplantation (allo-SCT). Novel diagnostic methods and therapeutic interventions are needed to further improve on patient outcomes. Extracellular vesicles (EV) are microvesicles formed by the inversion of the phospholipid bilayer of different cellular subtypes and have been described as biomarkers of cellular damage, activation, and intercellular signaling in numerous clinical scenarios. We studied the association between the levels of EV and the incidence of acute GVHD (aGVHD). Forty patients undergoing allo-SCT for hematological malignancies had their plasma collected at neutrophil engraftment. Using flow cytometry combined with fluorescent beads, the total circulating EV count (TEV) was established with annexin V positivity; CD61 positivity was used for platelet-derived EV (PEV), and CD235 positivity, for erythrocyte-derived EV (EryEV). TEV counts greater than 516/μL were associated with a higher cumulative incidence (CI) of grade II to IV aGVHD (54% vs. 21%; p = 0.02), as were EryEV counts above 357 /μL (CI of aGVHD: 59% vs. 26%; p = 0.04). In patients who are exposed to reduced intensity conditioning (RIC), stronger associations of both high TEV and EryEV counts with aGVHD were observed (77% vs. 22%; p = 0.003 and 89% vs. 27%; p = 0.002, respectively). PEV levels were not associated with the risk of aGVHD. Our data suggest that the measurement of cell-derived EV at engraftment can be used as a preemptive biomarker for acute GVHD.
Collapse
Affiliation(s)
- Thiago Xavier Carneiro
- Centro de Oncologia, Hospital Sirio Libanes, São Paulo, São Paulo, Brazil; Disciplina de Hematologia, Universidade Federal de São Paulo/Escola Paulista de Medicina, São Paulo, Brazil.
| | - Daniella Gregolin Marrese
- Disciplina de Hematologia, Universidade Federal de São Paulo/Escola Paulista de Medicina, São Paulo, Brazil; Grupo Fleury, São Paulo, Brazil
| | - Melina Gonçalves Dos Santos
- Disciplina de Hematologia, Universidade Federal de São Paulo/Escola Paulista de Medicina, São Paulo, Brazil; Grupo Fleury, São Paulo, Brazil
| | - Matheus Vescovi Gonçalves
- Disciplina de Hematologia, Universidade Federal de São Paulo/Escola Paulista de Medicina, São Paulo, Brazil; Grupo Fleury, São Paulo, Brazil
| | | | | | - Vanderson Rocha
- Centro de Oncologia, Hospital Sirio Libanes, São Paulo, São Paulo, Brazil
| | | | | | - Celso Arrais-Rodrigues
- Centro de Oncologia, Hospital Sirio Libanes, São Paulo, São Paulo, Brazil; Disciplina de Hematologia, Universidade Federal de São Paulo/Escola Paulista de Medicina, São Paulo, Brazil
| |
Collapse
|
7
|
Vajari MK, Moradinasab S, Yousefi AM, Bashash D. Noncoding RNAs in diagnosis and prognosis of graft-versus-host disease (GVHD). J Cell Physiol 2022; 237:3480-3495. [PMID: 35842836 DOI: 10.1002/jcp.30830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/11/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a functional therapy for a plethora of hematologic malignancies and immune disorders. Graft-versus-host disease (GVHD), on the other hand, is one of the major complications ahead of a successful HSCT, contributing to transplant-associated morbidity and mortality. Notably, little is known about the underlying mechanism of this event; therefore, exploring precise biomarkers and uncovering the molecular pathogenesis of GVHD is valuable for early diagnosis and treatment optimization. Thanks to the advances in sequencing techniques, the noncoding sequences of the human genome-formerly considered "junk"-are now identified as functional molecules. Noncoding RNAs (ncRNA) control cellular responses by regulating gene expression, and previous studies have shown that these tiny molecules, especially microRNAs (miRNAs), can affect allogeneic T cell responses in both animal models and clinical experiments. The present study gives an overview of the functions of various miRNAs in regulating T cell responses in GVHD. We also provide an outlook on miRNAs and long noncoding RNAs (lncRNAs) potential role in GVHD with the hope of providing a future research direction for expanding their application as the sensitive and noninvasive diagnostic or prognostic biomarkers and also the promising therapeutic targets for improving outcomes after allogeneic HSCT.
Collapse
Affiliation(s)
- Mahdi K Vajari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Susan Moradinasab
- Iranian Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Lazana I, Vassilopoulos G. A 'waste product' to save the day in the field of transplantation: the evolving potential of extracellular vesicles. Immunology 2022; 167:154-164. [PMID: 35751500 DOI: 10.1111/imm.13532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 05/25/2022] [Indexed: 11/26/2022] Open
Abstract
Graft rejection and graft-versus-host disease constitute the leading causes of morbidity and early mortality after solid organ and hematopoietic stem cell transplantation, respectively. Despite the current advances in immunotherapy, their incidence remains significant, underlying the need for new therapies to be developed. Extracellular vesicles (EV), and particularly small EV (sEV), have emerged as significant mediators of intercellular communication and immune modulation. Depending on the parental cell, they may exert potent immunostimulatory or immunosuppressive functions, attracting a major interest in field of transplantation. An increasing number of publications, studying their role in graft dysfunction pathophysiology, early detection of graft failure and in prevention and/or therapy of graft rejection, have emerged in recent years with enthusiastic results. In this review, we discuss the role and various applications of sEV in the transplant setting and present their huge potential for clinical translation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ioanna Lazana
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Hematology Department, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - George Vassilopoulos
- Cell and Gene Therapy Laboratory, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Division of Hematology, University of Thessaly Medical School, Larissa, Greece
| |
Collapse
|
9
|
Lia G, Di Vito C, Bruno S, Tapparo M, Brunello L, Santoro A, Mariotti J, Bramanti S, Zaghi E, Calvi M, Comba L, Fascì M, Giaccone L, Camussi G, Boyle EM, Castagna L, Evangelista A, Mavilio D, Bruno B. Extracellular Vesicles as Biomarkers of Acute Graft-vs.-Host Disease After Haploidentical Stem Cell Transplantation and Post-Transplant Cyclophosphamide. Front Immunol 2022; 12:816231. [PMID: 35145514 PMCID: PMC8821147 DOI: 10.3389/fimmu.2021.816231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Even with high-dose post-transplant cyclophosphamide (PT-Cy) which was initially introduced for graft-versus-host disease (GvHD) prevention in the setting of HLA-haploidentical transplantation, both acute and chronic GvHDs remain a major clinical challenge. Despite improvements in the understanding of the pathogenesis of both acute and chronic GvHDs, reliable biomarkers that predict their onset have yet to be identified. We recently studied the potential correlation between extracellular vesicles (EVs) and the onset of acute (a)GvHD in transplant recipients from related and unrelated donors. In the present study, we further investigated the role of the expression profile of membrane proteins and their microRNA (miRNA) cargo (miRNA100, miRNA155, and miRNA194) in predicting the onset of aGvHD in haploidentical transplant recipients with PT-Cy. Thirty-two consecutive patients were included. We evaluated the expression profile of EVs, by flow cytometry, and their miRNA cargo, by real-time PCR, at baseline, prior, and at different time points following transplant. Using logistic regression and Cox proportional hazard models, a significant association between expression profiles of antigens such as CD146, CD31, CD140a, CD120a, CD26, CD144, and CD30 on EVs, and their miRNA cargo with the onset of aGvHD was observed. Moreover, we also investigated a potential correlation between EV expression profile and cargo with plasma biomarkers (e.g., ST2, sTNFR1, and REG3a) that had been associated with aGVHD previously. This analysis showed that the combination of CD146, sTNFR1, and miR100 or miR194 strongly correlated with the onset of aGvHD (AUROC >0.975). A large prospective multicenter study is currently in progress to validate our findings.
Collapse
Affiliation(s)
- Giuseppe Lia
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Stefania Bruno
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Marta Tapparo
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Lucia Brunello
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Armando Santoro
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Jacopo Mariotti
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Stefania Bramanti
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Zaghi
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Michela Calvi
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Lorenzo Comba
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Martina Fascì
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luisa Giaccone
- Division of Hematology, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Giovanni Camussi
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Eileen M. Boyle
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
| | - Luca Castagna
- Bone Marrow Transplant Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Andrea Evangelista
- Clinical Epidemiology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University (NYU) Langone Health, New York, NY, United States
- *Correspondence: Benedetto Bruno,
| |
Collapse
|
10
|
Moreno-Castaño AB, Salas MQ, Palomo M, Martinez-Sanchez J, Rovira M, Fernández-Avilés F, Martínez C, Cid J, Castro P, Escolar G, Carreras E, Diaz-Ricart M. Early vascular endothelial complications after hematopoietic cell transplantation: Role of the endotheliopathy in biomarkers and target therapies development. Front Immunol 2022; 13:1050994. [PMID: 36479117 PMCID: PMC9720327 DOI: 10.3389/fimmu.2022.1050994] [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: 09/22/2022] [Accepted: 11/03/2022] [Indexed: 11/22/2022] Open
Abstract
This work aims to review the role of endothelial dysfunction underlying the main complications appearing early after autologous and allogeneic hematopoietic cell transplantation (HCT). The endothelial damage as the pathophysiological substrate of sinusoidal obstruction syndrome (SOS) is well established. However, there is growing evidence of the involvement of endothelial dysfunction in other complications, such as acute graft-versus-host disease (aGVHD) and transplant-associated thrombotic microangiopathy (TA-TMAs). Moreover, HCT-related endotheliopathy is not only limited to the HCT setting, as there is increasing evidence of its implication in complications derived from other cellular therapies. We also review the incidence and the risk factors of the main HCT complications and the biological evidence of the endothelial involvement and other linked pathways in their development. In addition, we cover the state of the art regarding the potential use of the biomarkers of endotheliopathy in the prediction, the early diagnosis, and the follow-up of the HCT complications and summarize current knowledge points to the endothelium and the other linked pathways described as potential targets for the prevention and treatment of HCT-complications. Lastly, the endothelium-focused therapeutic strategies that are emerging and might have a potential impact on the survival and quality of life of post-HCT-patients are additionally reviewed.
Collapse
Affiliation(s)
- Ana Belén Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic, Barcelona, Spain.,Clínic, Institut Josep Carreras, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - María Queralt Salas
- Hematology Department, Bone Marrow Transplantation Unit, Institut Clínic de Malalties Hemato-Oncològiques (ICMHO), Hospital Clínic, Barcelona, Spain
| | - Marta Palomo
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic, Barcelona, Spain.,Clínic, Institut Josep Carreras, Barcelona, Spain.,Campus Clinic, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Clínic, Institut Josep Carreras, Barcelona, Spain.,Campus Clinic, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Hematology Department, Bone Marrow Transplantation Unit, Institut Clínic de Malalties Hemato-Oncològiques (ICMHO), Hospital Clínic, Barcelona, Spain
| | - Francesc Fernández-Avilés
- Hematology Department, Bone Marrow Transplantation Unit, Institut Clínic de Malalties Hemato-Oncològiques (ICMHO), Hospital Clínic, Barcelona, Spain
| | - Carmen Martínez
- Hematology Department, Bone Marrow Transplantation Unit, Institut Clínic de Malalties Hemato-Oncològiques (ICMHO), Hospital Clínic, Barcelona, Spain
| | - Joan Cid
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Apheresis & Cellular Therapy Unit, Department of Hemotherapy and Hemostasis, Institut Clínic de Malalties Hemato-Oncològiques (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Pedro Castro
- Clínic, Institut Josep Carreras, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.,Medical Intensive Care Unit, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic, Barcelona, Spain.,Clínic, Institut Josep Carreras, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Enric Carreras
- Clínic, Institut Josep Carreras, Barcelona, Spain.,Campus Clinic, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Maribel Diaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic, Barcelona, Spain.,Clínic, Institut Josep Carreras, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| |
Collapse
|
11
|
Yang F, Hayashi I, Sato S, Saijo-Ban Y, Yamane M, Fukui M, Shimizu E, He J, Shibata S, Mukai S, Asai K, Ogawa M, Lan Y, Zeng Q, Hirakata A, Tsubota K, Ogawa Y. Eyelid blood vessel and meibomian gland changes in a sclerodermatous chronic GVHD mouse model. Ocul Surf 2021; 26:328-341. [PMID: 34715372 DOI: 10.1016/j.jtos.2021.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 10/10/2021] [Accepted: 10/21/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE To investigate pathological changes in blood vessels and meibomian glands (MGs) in the eyelids of sclerodermatous chronic graft-versus-host disease (cGVHD) model mice. METHODS We used an established major histocompatibility complex compatible, multiple minor histocompatibility antigen-mismatched sclerodermatous cGVHD mouse model. Blood vessels and MGs of eyelids from allogeneic bone marrow transplantation (allo-BMT) recipient mice and syngeneic bone marrow transplantation (syn-BMT) recipient mice were assessed by histopathology, immunohistochemistry and transmission electron microscopy. Peripheral blood samples from the recipients were examined by flow cytometry. RESULTS Allo-BMT samples showed dilating, tortuous and branching vessels and shrunk MGs in the eyelids; showed significantly higher expression of VEGFR2 (p = 0.029), CD133 (p = 0.016), GFP (p = 0.006), and α-SMA (p = 0.029) in the peripheral MG area; showed endothelial damage and activation, fibrotic change, and immune cell infiltration into MGs compared with syn-BMT samples. Fewer Ki-67+ cells were observed in allo- and syn-BMT samples than in wild-type samples (p = 0.030). Ultrastructural changes including endothelial injury and activation, fibroblast activation, granulocyte degranulation, immune cell infiltration into MGs, and necrosis, apoptosis of MG basal cells were found in allo-BMT samples compared with syn-BMT samples. CONCLUSION A series of our studies indicated that cGVHD can cause eyelid vessel and MGs changes, including endothelial injury and activation, neovascularization, early fibrotic changes, immune cell infiltration, MG basal cell necrosis and apoptosis, and resultant MG atrophy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan; Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Aier School of Ophthalmology, Central South University, Changsha, China
| | - Isami Hayashi
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan; Department of Ophthalmology, Kyorin University, School of Medicine, Tokyo, Japan
| | - Shinri Sato
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Yumiko Saijo-Ban
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Mio Yamane
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Masaki Fukui
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Jingliang He
- Eye Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University School of Medicine, Tokyo, Japan
| | - Shin Mukai
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Kazuki Asai
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Mamoru Ogawa
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan
| | - Yuqing Lan
- Department of Ophthalmology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qingyan Zeng
- Aier Eye Hosoital of Wuhan University, Wuhan, Hubei province, China
| | - Akito Hirakata
- Department of Ophthalmology, Kyorin University, School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan; Tsubota Laboratory, Inc., Tokyo, Japan.
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University, School of Medicine, Tokyo, Japan.
| |
Collapse
|
12
|
Wang X, Zhang R, Huang Z, Zang S, Wu Q, Xia L. Inhibition of the miR-155 and protein prenylation feedback loop alleviated acute graft-versus-host disease through regulating the balance between T helper 17 and Treg cells. Transpl Immunol 2021; 69:101461. [PMID: 34487810 DOI: 10.1016/j.trim.2021.101461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 11/19/2022]
Abstract
MicroRNA-155(miR-155) and protein prenylation have been reported to participate in acute graft-versus-host disease (aGVHD) through modulating T lymphocyte differentiation, however the mechanism remains elusive. In this study, we found that the expression of miR-155 and protein prenyltransferases in peripheral blood T lymphocytes of aGVHD mice was significantly increased. Suppression of miR-155 by antagomir-155 could remarkably reduce prenyltransferases mRNA and protein expression in T lymphocytes of aGVHD mice. Conversely, prenyltransferase inhibitors significantly reduced the level of miR-155. Inhibition of this feedback loop of miR-155 and protein prenylation in aGVHD mice led to improved survival and lower aGVHD histopathology scores and significantly induced T cell deficient differentiation towards T helper 17 (Th17) cells and titled differentiation towards CD4+CD25hi regulatory T (Treg) cells. Furthermore, the immunoregulatory effects and protection from aGVHD of prenyltransferase inhibitors could be reversed by the addition of miR-155. The dual treatment of prenylation inhibitors and antagomir-155 showed synergistic effects on T polarization and protection from aGVHD. Consistent with the in vivo changes, inhibition of this feedback loop of miR-155 and protein prenylation affected Th17 and Treg cell polarization in vitro. Our data suggest that miR-155 and protein prenylation may constitute a feedback loop that amplifies immune and inflammatory responses in subjects with aGVHD, and they may serve as potential targets for aGVHD prophylaxis and treatment.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue,Wuhan 430022, China; Department of Geriatrics,Union Hospital,Tongji Medical College, Huazhong University of Science and Technology,1277 JieFang Avenue,Wuhan 430022,China; Institute of Gerontology,Union Hospital,Tongji Medical College, Huazhong University of Science and Technology,1277 JieFang Avenue,Wuhan 430022,China
| | - Ran Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhenli Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue,Wuhan 430022, China
| | - Sibin Zang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue,Wuhan 430022, China
| | - Qiuling Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue,Wuhan 430022, China.
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue,Wuhan 430022, China.
| |
Collapse
|
13
|
Lia G, Giaccone L, Leone S, Bruno B. Biomarkers for Early Complications of Endothelial Origin After Allogeneic Hematopoietic Stem Cell Transplantation: Do They Have a Potential Clinical Role? Front Immunol 2021; 12:641427. [PMID: 34093530 PMCID: PMC8170404 DOI: 10.3389/fimmu.2021.641427] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 05/04/2021] [Indexed: 12/17/2022] Open
Abstract
Endothelial cell (EC) dysfunction causes a number of early and life-threatening post hematopoietic stem cell transplant (HCT) complications that result in a rapid clinical decline. The main early complications are graft-vs.-host disease (GVHD), transplant associated thrombotic microangiopathy (TA-TMA), and sinusoidal obstruction syndrome (SOS). Post-HCT endothelial dysfunction occurs as a result of chemotherapy, infections, and allogeneic reactivity. Despite major advances in transplant immunology and improvements in supportive care medicine, these complications represent a major obstacle for successful HCT. In recent years, different biomarkers have been investigated for early detection of post-transplant endothelial cell dysfunction, but few have been validated. In this review we will define GVHD, TA-TMA and SOS, summarize the current data available in HCT biomarker research and identify promising biomarkers for detection and diagnosis of early HCT complications.
Collapse
Affiliation(s)
- Giuseppe Lia
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Luisa Giaccone
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
| | - Sarah Leone
- Department of Internal Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Benedetto Bruno
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Division of Hematology and Medical Oncology, New York University Grossman School of Medicine, Perlmutter Cancer Center, New York University Langone Health, New York, NY, United States
| |
Collapse
|
14
|
Giaccone L, Faraci DG, Butera S, Lia G, Di Vito C, Gabrielli G, Cerrano M, Mariotti J, Dellacasa C, Felicetti F, Brignardello E, Mavilio D, Bruno B. Biomarkers for acute and chronic graft versus host disease: state of the art. Expert Rev Hematol 2020; 14:79-96. [PMID: 33297779 DOI: 10.1080/17474086.2021.1860001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Despite significant advances in treatment and prevention, graft-versus-host disease (GVHD) still represents the main cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Thus, considerable research efforts have been made to find and validate reliable biomarkers for diagnosis, prognosis, and risk stratification of GVHD. AREAS COVERED In this review the most recent evidences on different types of biomarkers studied for GVHD, such as genetic, plasmatic, cellular markers, and those associated with microbiome, were summarized. A comprehensive search of peer-review literature was performed in PubMed including meta-analysis, preclinical and clinical trials, using the terms: cellular and plasma biomarkers, graft-versus-host disease, cytokines, and allogeneic hematopoietic stem cell transplantation. EXPERT OPINION In the near future, several validated biomarkers will be available to help clinicians in the diagnosis of GVHD, the identification of patients at high risk of GVHD development and in patients' stratification according to its severity. Then, immunosuppressive treatment could be tailored to each patient's real needs. However, more efforts are needed to achieve this goal. Although most of the proposed biomarkers currently lack validation with large-scale clinical data, their study led to improved knowledge of the biological basis of GVHD, and ultimately to implementation of GHVD treatment.
Collapse
Affiliation(s)
- Luisa Giaccone
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Danilo Giuseppe Faraci
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Sara Butera
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Giuseppe Lia
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (Biometra), University of Milan , Milan, Italy
| | - Giulia Gabrielli
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Marco Cerrano
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| | - Jacopo Mariotti
- Bone Marrow Transplant Unit, Humanitas Clinical and Research Center, IRCCS , Rozzano, Italy
| | - Chiara Dellacasa
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy
| | - Francesco Felicetti
- Transition Unit for Childhood Cancer Survivors, A.O.U. Città Della Salute E Della Scienza Di Torino , University of Torino , Torino, Italy
| | - Enrico Brignardello
- Transition Unit for Childhood Cancer Survivors, A.O.U. Città Della Salute E Della Scienza Di Torino , University of Torino , Torino, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center , Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (Biometra), University of Milan , Milan, Italy
| | - Benedetto Bruno
- Department of Oncology/Hematology, Stem Cell Transplant Program, A.O.U. Città Della Salute E Della Scienza Di Torino, Presidio Molinette , Torino, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Torino , Torino, Italy
| |
Collapse
|
15
|
Crossland RE, Perutelli F, Bogunia-Kubik K, Mooney N, Milutin Gašperov N, Pučić-Baković M, Greinix H, Weber D, Holler E, Pulanić D, Wolff D, Dickinson AM, Inngjerdingen M, Grce M. Potential Novel Biomarkers in Chronic Graft-Versus-Host Disease. Front Immunol 2020; 11:602547. [PMID: 33424849 PMCID: PMC7786047 DOI: 10.3389/fimmu.2020.602547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Prognostic, diagnostic or predictive biomarkers are urgently needed for assessment of chronic graft-versus-host disease (cGvHD), a major risk for patients undergoing allogeneic hematopoietic stem cell transplantation. The main goal of this review generated within the COST Action EUROGRAFT "Integrated European Network on Chronic Graft Versus Host Disease" was to identify potential novel biomarkers for cGvHD besides the widely accepted molecular and cellular biomarkers. Thus, the focus was on cellular biomarkers, alloantibodies, glycomics, endothelial derived particles, extracellular vesicles, microbiome, epigenetic and neurologic changes in cGvHD patients. Both host-reactive antibodies in general, and particularly alloantibodies have been associated with cGvHD and require further consideration. Glycans attached to IgG modulate its activity and represent a promising predictive and/or stratification biomarker for cGVHD. Furthermore, epigenetic changes such as microRNAs and DNA methylation represent potential biomarkers for monitoring cGvHD patients and novel targets for developing new treatment approaches. Finally, the microbiome likely affects the pathophysiology of cGvHD; bacterial strains as well as microbial metabolites could display potential biomarkers for dysbiosis and risk for the development of cGvHD. In summary, although there are no validated biomarkers currently available for clinical use to better inform on the diagnosis, prognosis or prediction of outcome for cGvHD, many novel sources of potential markers have shown promise and warrant further investigation using well characterized, multi-center patient cohorts.
Collapse
Affiliation(s)
- Rachel E. Crossland
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Francesca Perutelli
- Department of Molecular Biotechnology and Health Sciences, School of Medicine, University of Torino, Torino, Italy
| | - Katarzyna Bogunia-Kubik
- Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland
| | - Nuala Mooney
- INSERM U976, Human Immunology, Pathophysiology and Immunotherapies, Hôpital Saint Louis, Paris, France
| | | | | | - Hildegard Greinix
- Division of Hematology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Daniela Weber
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Ernst Holler
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Dražen Pulanić
- Division of Hematology, Department of Internal Medicine, University Hospital Centre Zagreb, Medical School, University of Zagreb, Zagreb, Croatia
| | - Daniel Wolff
- Department of Internal Medicine III, Faculty of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Anne M. Dickinson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Marit Inngjerdingen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Magdalena Grce
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
16
|
Noulsri E. Effects of Cell-Derived Microparticles on Immune Cells and Potential Implications in Clinical Medicine. Lab Med 2020; 52:122-135. [PMID: 32816040 DOI: 10.1093/labmed/lmaa043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the past few years, interest has increased in cell-derived microparticles (MPs), which are defined by their size of from 0.1 to 1 μm, and can be derived from various cell types, including endothelial cells, leukocytes, red blood cells (RBCs), and platelets. These MPs carry negatively charged phosphatidylserine (PS) on their surfaces and proteins packaged from numerous cellular components. MPs that have been shed by the body can play important roles in the pathophysiology of diseases and can affect various biological systems. Among these systems, the immune components have been shown to be modulated by MPs. Therefore, understanding the roles of MPs in the immune system is crucial to developing alternative therapeutic treatments for diseases. This review describes the effects of MPs on various immune cells and provides plausible potential applications of the immune-modulating properties of MPs in clinical medicine.
Collapse
Affiliation(s)
- Egarit Noulsri
- Research Division, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
17
|
Chao X, Yi L, Lan LL, Wei HY, Wei D. Long-term PM 2.5 exposure increases the risk of non-small cell lung cancer (NSCLC) progression by enhancing interleukin-17a (IL-17a)-regulated proliferation and metastasis. Aging (Albany NY) 2020; 12:11579-11602. [PMID: 32554855 PMCID: PMC7343463 DOI: 10.18632/aging.103319] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/28/2020] [Indexed: 05/03/2023]
Abstract
PM2.5 is a class of airborne particles and droplets with sustained high levels in many developing countries. Epidemiological studies have indicated that PM2.5 is closely associated with the increased morbidity and mortality of lung cancer in the world. Unfortunately, the effects of PM2.5 on lung cancer are largely unknown. In the present study, we attempted to explore the role of PM2.5 in the etiology of NSCLC. Here, we found that long-term PM2.5 exposure led to significant pulmonary injury. Epithelial-mesenchymal transition (EMT) and cancer stem cells (CSC) properties were highly induced by PM2.5 exposure. EMT was evidenced by the significant up-regulation of MMP2, MMP9, TGF-β1, α-SMA, Fibronectin and Vimentin. Lung cancer progression was associated with the increased expression of Kras, c-Myc, breast cancer resistance protein BCRP (ABCG2), OCT4, SOX2 and Aldh1a1, but the decreased expression of p53 and PTEN. Importantly, mice with IL-17a knockout (IL-17a-/-) showed significantly alleviated lung injury and CSC properties following PM2.5 exposure. Also, IL-17a-/--attenuated tumor growth was recovered in PM2.5-exposed mice injected with recombinant mouse IL-17a, accompanied with significantly restored lung metastasis. Taken together, these data revealed that PM2.5 could promote the progression of lung cancer by enhancing the proliferation and metastasis through IL-17a signaling.
Collapse
Affiliation(s)
- Xie Chao
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, P.R. China
| | - Liu Yi
- Centers of Disease Control and Prevention of Shandong Province, Jinan 250014, Shandong Province, P.R. China
| | - Li Lan Lan
- Affiliated Hospital of Binzhou Medical College, Binzhou 256603, Shandong Province, P.R. China
| | - Han Yun Wei
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Shihuan Province, P.R. China
| | - Dong Wei
- Department of Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, Shandong Province, P.R. China
| |
Collapse
|
18
|
Gavriilaki E, Sakellari I, Gavriilaki M, Anagnostopoulos A. A New Era in Endothelial Injury Syndromes: Toxicity of CAR-T Cells and the Role of Immunity. Int J Mol Sci 2020; 21:E3886. [PMID: 32485958 PMCID: PMC7312228 DOI: 10.3390/ijms21113886] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy with chimeric antigen receptor T (CAR-T cells) has been recently approved for patients with relapsed/refractory B-lymphoproliferative neoplasms. Along with great efficacy in patients with poor prognosis, CAR-T cells have been also linked with novel toxicities in a significant portion of patients. Cytokine release syndrome (CRS) and neurotoxicity present with unique clinical phenotypes that have not been previously observed. Nevertheless, they share similar characteristics with endothelial injury syndromes developing post hematopoietic cell transplantation (HCT). Evolution in complement therapeutics has attracted renewed interest in these life-threatening syndromes, primarily concerning transplant-associated thrombotic microangiopathy (TA-TMA). The immune system emerges as a key player not only mediating cytokine responses but potentially contributing to endothelial injury in CAR-T cell toxicity. The interplay between complement, endothelial dysfunction, hypercoagulability, and inflammation seems to be a common denominator in these syndromes. As the indications for CAR-T cells and patient populations expand, there in an unmet clinical need of better understanding of the pathophysiology of CAR-T cell toxicity. Therefore, this review aims to provide state-of-the-art knowledge on cellular therapies in clinical practice (indications and toxicities), endothelial injury syndromes and immunity, as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Ioanna Sakellari
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Maria Gavriilaki
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Achilles Anagnostopoulos
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| |
Collapse
|
19
|
Lia G, Di Vito C, Cerrano M, Brunello L, Calcaterra F, Tapparo M, Giaccone L, Mavilio D, Bruno B. Extracellular Vesicles After Allogeneic Hematopoietic Cell Transplantation: Emerging Role in Post-Transplant Complications. Front Immunol 2020; 11:422. [PMID: 32265915 PMCID: PMC7100658 DOI: 10.3389/fimmu.2020.00422] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/24/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in the cellular crosstalk by transferring bioactive molecules through biological barriers from a cell to another, thus influencing recipient cell functions and phenotype. Therefore, EVs are increasingly being explored as biomarkers of disease progression or response to therapy and as potential therapeutic agents in different contexts including in hematological malignancies. Recently, an EV role has emerged in allogeneic hematopoietic cell transplantation (allo-HCT) as well. Allogeneic hematopoietic cell transplantation often represents the only curative option in several hematological disorders, but it is associated with potentially life-threatening complications that can have a significant impact on clinical outcomes. The most common complications have been well-established and include graft-versus-host disease and infections. Furthermore, relapse remains an important cause of treatment failure. The aim of this review is to summarize the current knowledge, the potential applications, and clinical relevance of EVs in allo-HCT. Herein, we will mainly focus on the immune-modulating properties of EVs, in particular those derived from mesenchymal stromal cells, as potential therapeutic strategy to improve allo-HCT outcome. Moreover, we will briefly describe the main findings on EVs as biomarkers to monitor graft-versus-host disease onset and tumor relapse.
Collapse
Affiliation(s)
- Giuseppe Lia
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Marco Cerrano
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Lucia Brunello
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesca Calcaterra
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Marta Tapparo
- Department of Medical Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Luisa Giaccone
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Benedetto Bruno
- Stem Cell Transplant Program, Department of Oncology, A.O.U. Città della Salute e della Scienza di Torino, Turin, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Gomez I, Ward B, Souilhol C, Recarti C, Ariaans M, Johnston J, Burnett A, Mahmoud M, Luong LA, West L, Long M, Parry S, Woods R, Hulston C, Benedikter B, Niespolo C, Bazaz R, Francis S, Kiss-Toth E, van Zandvoort M, Schober A, Hellewell P, Evans PC, Ridger V. Neutrophil microvesicles drive atherosclerosis by delivering miR-155 to atheroprone endothelium. Nat Commun 2020; 11:214. [PMID: 31924781 PMCID: PMC6954269 DOI: 10.1038/s41467-019-14043-y] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 12/11/2019] [Indexed: 12/18/2022] Open
Abstract
Neutrophils are implicated in the pathogenesis of atherosclerosis but are seldom detected in atherosclerotic plaques. We investigated whether neutrophil-derived microvesicles may influence arterial pathophysiology. Here we report that levels of circulating neutrophil microvesicles are enhanced by exposure to a high fat diet, a known risk factor for atherosclerosis. Neutrophil microvesicles accumulate at disease-prone regions of arteries exposed to disturbed flow patterns, and promote vascular inflammation and atherosclerosis in a murine model. Using cultured endothelial cells exposed to disturbed flow, we demonstrate that neutrophil microvesicles promote inflammatory gene expression by delivering miR-155, enhancing NF-κB activation. Similarly, neutrophil microvesicles increase miR-155 and enhance NF-κB at disease-prone sites of disturbed flow in vivo. Enhancement of atherosclerotic plaque formation and increase in macrophage content by neutrophil microvesicles is dependent on miR-155. We conclude that neutrophils contribute to vascular inflammation and atherogenesis through delivery of microvesicles carrying miR-155 to disease-prone regions.
Collapse
Affiliation(s)
- Ingrid Gomez
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Ben Ward
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Celine Souilhol
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| | - Chiara Recarti
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Department of Molecular Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Mark Ariaans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jessica Johnston
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Amanda Burnett
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Marwa Mahmoud
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Cardiovascular Mechanobiology and Nanomedicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Le Anh Luong
- William Harvey Research Institute, Queen Mary University, London, UK
| | - Laura West
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Merete Long
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Sion Parry
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Rachel Woods
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Carl Hulston
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Birke Benedikter
- Department of Medical Microbiology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, The Netherlands
| | - Chiara Niespolo
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Rohit Bazaz
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Sheila Francis
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Endre Kiss-Toth
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Marc van Zandvoort
- Department of Molecular Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Andreas Schober
- Experimental Vascular Medicine, Ludwig-Maximilian University of Munich, Munich, Germany
| | - Paul Hellewell
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- College of Health and Life Sciences, Brunel University, London, UK
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Bateson Institute, University of Sheffield, Sheffield, UK
| | - Victoria Ridger
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK.
| |
Collapse
|
21
|
MicroRNAs in graft-versus-host disease: a review of the latest data. Bone Marrow Transplant 2019; 55:1014-1020. [DOI: 10.1038/s41409-019-0764-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 11/14/2019] [Accepted: 11/20/2019] [Indexed: 12/12/2022]
|
22
|
Motaei J, Yaghmaie M, Ahmadvand M, Pashaiefar H, Kerachian MA. MicroRNAs as Potential Diagnostic, Prognostic, and Predictive Biomarkers for Acute Graft-versus-Host Disease. Biol Blood Marrow Transplant 2019; 25:e375-e386. [PMID: 31419566 DOI: 10.1016/j.bbmt.2019.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
Successful treatment of various hematologic diseases with allogeneic hematopoietic stem cell transplantation is often limited due to the occurrence of acute graft-versus-host disease (aGVHD). So far, there are no approved molecular biomarkers for the diagnosis and prediction of aGVHD at the clinical level due to our incomplete understanding of the molecular biology of the disease. Various studies have been conducted on animal models and humans to investigate the role of microRNAs in aGVHD pathogenesis to implicate them as biomarkers and therapeutic targets. Because of their high stability, tissue specificity, ease of measurement, low cost, and simplicity, they are excellent targets for biomarkers. In this review, we focused on microRNA expression profiling studies that were performed recently in both animal models and human cases of aGVHD to identify diagnostic and predictive biomarkers for this disease. The expression pattern of microRNAs can be specific to cells and tissues. Because aGVHD affects several organs, microRNA signatures in target tissues may help to understand the molecular pathology of the disease. Identification of organ-specific microRNAs in aGVHD can be promising to categorize patients for organ-specific therapies. Thus, microRNAs can be used as noninvasive diagnostic tests in clinic to improve prophylaxis, predict incidence and severity, and reduce morbidity.
Collapse
Affiliation(s)
- Jamshid Motaei
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marjan Yaghmaie
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ahmadvand
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Pashaiefar
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Cancer Genetics Research Unit, Reza Radiotherapy and Oncology Center, Mashhad, Iran.
| |
Collapse
|
23
|
Gavriilaki E, Gkaliagkousi E, Sakellari I, Anyfanti P, Douma S, Anagnostopoulos A. Early Prediction of Cardiovascular Risk after Hematopoietic Cell Transplantation: Are We There Yet? Biol Blood Marrow Transplant 2019; 25:e310-e316. [PMID: 31310812 DOI: 10.1016/j.bbmt.2019.07.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/03/2019] [Accepted: 07/10/2019] [Indexed: 12/14/2022]
Abstract
Cardiovascular (CV) events have emerged as a major cause of morbidity and mortality among hematopoietic cell transplantation (HCT) survivors. Accumulating evidence supports the presence of increased CV risk in HCT recipients. Most studies have focused mainly on traditional CV risk factors, such as the metabolic syndrome and hypertension. However, detection of these factors suggests the development of irreversible overt clinical atherosclerosis. Therefore, earlier prediction of CV risk is needed to prevent CV morbidity and mortality in these patients. In the field of CV research, endothelial dysfunction is considered an early event in the pathophysiology of CV risk factors, and a number of markers have been proposed for its assessment. In addition, markers of subclinical target organ damage have been introduced to implement CV risk prediction and early preventive or intensive therapeutic interventions. Furthermore, a number of CV models have been suggested aiming for optimal stratification of patients. Preliminary studies have indicated excess CV risk using these early markers in HCT recipients. However, their role in the pathophysiology and clinical practice in HCT survivors remains largely understudied. Taking into account the need for increased awareness from treating physicians in this evolving setting, we conducted a state-of-the-art review aiming to summarize current knowledge on endothelial dysfunction, subclinical target organ damage, and CV risk prediction in HCT survivors.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece.
| | - Eugenia Gkaliagkousi
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Sakellari
- Hematology Department-BMT Unit, G. Papanicolaou Hospital, Thessaloniki, Greece
| | - Panagiota Anyfanti
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Stella Douma
- 3rd Department of Internal Medicine, Papageorgiou Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | |
Collapse
|
24
|
Zheng Y, Dong C, Yang J, Jin Y, Zheng W, Zhou Q, Liang Y, Bao L, Feng G, Ji J, Feng X, Gu Z. Exosomal microRNA‐155‐5p from PDLSCs regulated Th17/Treg balance by targeting sirtuin‐1 in chronic periodontitis. J Cell Physiol 2019; 234:20662-20674. [PMID: 31016751 DOI: 10.1002/jcp.28671] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 03/25/2019] [Accepted: 04/02/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Ya Zheng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Chen Dong
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Junling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yi Jin
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenjie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Qiao Zhou
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Yi Liang
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Liuliu Bao
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Guijuan Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Juan Ji
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xingmei Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Zhifeng Gu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
25
|
Słomka A, Urban SK, Lukacs-Kornek V, Żekanowska E, Kornek M. Large Extracellular Vesicles: Have We Found the Holy Grail of Inflammation? Front Immunol 2018; 9:2723. [PMID: 30619239 PMCID: PMC6300519 DOI: 10.3389/fimmu.2018.02723] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 11/05/2018] [Indexed: 12/17/2022] Open
Abstract
The terms microparticles (MPs) and microvesicles (MVs) refer to large extracellular vesicles (EVs) generated from a broad spectrum of cells upon its activation or death by apoptosis. The unique surface antigens of MPs/MVs allow for the identification of their cellular origin as well as its functional characterization. Two basic aspects of MP/MV functions in physiology and pathological conditions are widely considered. Firstly, it has become evident that large EVs have strong procoagulant properties. Secondly, experimental and clinical studies have shown that MPs/MVs play a crucial role in the pathophysiology of inflammation-associated disorders. A cardinal feature of these disorders is an enhanced generation of platelets-, endothelial-, and leukocyte-derived EVs. Nevertheless, anti-inflammatory effects of miscellaneous EV types have also been described, which provided important new insights into the large EV-inflammation axis. Advances in understanding the biology of MPs/MVs have led to the preparation of this review article aimed at discussing the association between large EVs and inflammation, depending on their cellular origin.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Sabine Katharina Urban
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Hospital of the Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Ewa Żekanowska
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Miroslaw Kornek
- Department of Oncology, Hematology and Rheumatology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
26
|
Peltier D, Reddy P. Non-Coding RNA Mediated Regulation of Allogeneic T Cell Responses After Hematopoietic Transplantation. Front Immunol 2018; 9:1110. [PMID: 29963039 PMCID: PMC6013767 DOI: 10.3389/fimmu.2018.01110] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 05/03/2018] [Indexed: 12/21/2022] Open
Abstract
Allogeneic bone marrow transplantation (BMT) is an effective therapy for several malignant and non-malignant disorders. The precise control of allogeneic T cells is critical for successful outcomes after BMT. The mechanisms governing desirable (graft-versus-leukemia) versus undesirable (graft-versus-host disease) allogeneic responses remain incompletely understood. Non-coding RNAs (ncRNA) are controllers of gene expression that fine-tune cellular responses. Multiple microRNAs (miRNAs), a type of ncRNA, have recently been shown to influence allogeneic T cell responses in both murine models and clinically. Here, we review the role of various miRNAs that regulate T cell responses, either positively or negatively, to allo-stimulation and highlight their potential relevance as biomarkers and as therapeutic targets for improving outcomes after allogeneic BMT.
Collapse
Affiliation(s)
- Daniel Peltier
- Division of Hematology and Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, United States
| | - Pavan Reddy
- Division of Hematology and Oncology, Department of Internal Medicine, Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|