1
|
Chichelnitskiy E, Goldschmidt I, Ruhl L, Rübsamen N, Jaeger VK, Karch A, Beushausen K, Keil J, Götz JK, D'Antiga L, Debray D, Hierro L, Kelly D, McLin V, Pawlowska J, Mikolajczyk RT, Bravi M, Klaudel-Dreszler M, Demir Z, Lloyd C, Korff S, Baumann U, Falk CS. Plasma immune signatures can predict rejection-free survival in the first year after pediatric liver transplantation. J Hepatol 2024; 81:862-871. [PMID: 38821361 DOI: 10.1016/j.jhep.2024.05.032] [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: 09/18/2023] [Revised: 05/06/2024] [Accepted: 05/17/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND & AIMS After pediatric liver transplantation (pLT), children undergo life-long immunosuppression since reliable biomarkers for the assessment of rejection probability are scarce. In the multicenter (n = 7) prospective clinical cohort "ChilSFree" study, we aimed to characterize longitudinal dynamics of soluble and cellular immune mediators during the first year after pLT and identify early biomarkers associated with outcome. METHODS Using a Luminex-based multiplex technique paired with flow cytometry, we characterized longitudinal dynamics of soluble immune mediators (SIMs, n = 50) and immune cells in the blood of 244 patients at eight visits over 1 year: before, and 7/14/21/28 days and 3/6/12 months after pLT. RESULTS The unsupervised clustering of patients based on SIM profiles revealed six unique SIM signatures associated with clinical outcome. From three signatures linked to improved outcome, one was associated with 1-year-long rejection-free survival and stable graft function and was characterized by low levels of pro-inflammatory SIMs (CXCL8/9/10/12, CCL7, SCGF-β, sICAM-1), and high levels of regenerative (SCF, TNF-β) and pro-apoptotic (TRAIL) SIMs (all, p <0.001, fold change >100). Of note, this SIM signature appeared 2 weeks after pLT and remained stable over the entire year, pointing towards its potential as a novel early biomarker for minimizing or weaning immunosuppression. In the blood of these patients, a higher frequency of CD56bright natural killer cells (p <0.01), a known hallmark also associated with operationally tolerant pLT patients, was detected. The concordance of the model for prediction of rejection based on identified SIM signatures was 0.715, and 0.795, in combination with living-related transplantation as a covariate, respectively. CONCLUSIONS SIM blood signatures may enable the non-invasive and early assessment of rejection risks in the first year after pLT, paving the way for improved clinical management. IMPACT AND IMPLICATIONS ChilSFree represents the largest pediatric liver transplant (pLT) cohort with paired longitudinal data on soluble immune mediators (SIMs) and immune phenotyping in the first year after pLT. SIM signatures allow for the selection of rejection-free patients 2 weeks after pLT independently of patient diagnosis, sex, or age. The SIM signatures may enable the non-invasive and early assessment of rejection risks, paving the way for minimization or withdrawal of immunosuppression after pLT.
Collapse
Affiliation(s)
- Evgeny Chichelnitskiy
- Institute of Transplant Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Imeke Goldschmidt
- Division of Pediatric Gastroenterology and Hepatology, MHH, Hannover, Germany; European Pediatric Liver Transplantation Network, Germany
| | - Louisa Ruhl
- Institute of Transplant Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Nicole Rübsamen
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Veronika K Jaeger
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Andre Karch
- Institute of Epidemiology and Social Medicine, University of Münster, Münster, Germany
| | - Kerstin Beushausen
- Institute of Transplant Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Jana Keil
- Institute of Transplant Immunology, Hannover Medical School (MHH), Hannover, Germany
| | - Juliane K Götz
- Division of Pediatric Gastroenterology and Hepatology, MHH, Hannover, Germany
| | - Lorenzo D'Antiga
- Pediatric Department, Hospital Papa Giovanni XXIII Bergamo, Italy; European Pediatric Liver Transplantation Network, Germany; Department of Medicine and Surgery, University of Milano - Bicocca, 20126 Milan, Italy
| | - Dominique Debray
- Pediatric liver unit, Hôpital Necker-Enfants Malades, Paris, France; European Pediatric Liver Transplantation Network, Germany
| | - Loreto Hierro
- Hospital Infantil Universitario La Paz, Madrid, Spain; European Pediatric Liver Transplantation Network, Germany
| | - Deirdre Kelly
- Birmingham Women's & Children's Hospital, and University of Birmingham, UK; European Pediatric Liver Transplantation Network, Germany
| | - Valerie McLin
- Swiss Pediatric Liver Center, Department of Pediatrics, Gynecology, and Obstetrics, University Hospitals Geneva, University of Geneva, Geneva, Switzerland; European Pediatric Liver Transplantation Network, Germany
| | - Joanna Pawlowska
- Department of Gastroenterology, Hepatology, Nutritional Disorders, and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Rafael T Mikolajczyk
- Institute of Medical Epidemiology, Biostatistics and Medical Informatics, University of Halle, Halle, Germany
| | - Michela Bravi
- Pediatric Department, Hospital Papa Giovanni XXIII Bergamo, Italy; European Pediatric Liver Transplantation Network, Germany
| | - Maja Klaudel-Dreszler
- Department of Gastroenterology, Hepatology, Nutritional Disorders, and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Zeynep Demir
- Pediatric liver unit, Hôpital Necker-Enfants Malades, Paris, France; European Pediatric Liver Transplantation Network, Germany
| | - Carla Lloyd
- Birmingham Women's & Children's Hospital, and University of Birmingham, UK; European Pediatric Liver Transplantation Network, Germany
| | - Simona Korff
- Swiss Pediatric Liver Center, Department of Pediatrics, Gynecology, and Obstetrics, University Hospitals Geneva, University of Geneva, Geneva, Switzerland; European Pediatric Liver Transplantation Network, Germany
| | - Ulrich Baumann
- Division of Pediatric Gastroenterology and Hepatology, MHH, Hannover, Germany; European Pediatric Liver Transplantation Network, Germany
| | - Christine S Falk
- Institute of Transplant Immunology, Hannover Medical School (MHH), Hannover, Germany; German Centre for Infection Research, TTU-IICH Hannover, Hannover, Germany.
| |
Collapse
|
2
|
Shi H, Yuan M, Cai J, Shi J, Li Y, Qian Q, Dong Z, Pan G, Zhu S, Wang W, Zhou J, Zhou X, Liu J. Exploring personalized treatment for cardiac graft rejection based on a four-archetype analysis model and bioinformatics analysis. Sci Rep 2024; 14:6529. [PMID: 38499711 PMCID: PMC10948767 DOI: 10.1038/s41598-024-57097-9] [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: 10/22/2023] [Accepted: 03/14/2024] [Indexed: 03/20/2024] Open
Abstract
Heart transplantation is the gold standard for treating patients with advanced heart failure. Although improvements in immunosuppressive therapies have significantly reduced the frequency of cardiac graft rejection, the incidences of T cell-mediated rejection (TCMR) and antibody-mediated rejection remain almost unchanged. A four-archetype analysis (4AA) model, developed by Philip F. Halloran, illustrated this problem well. It provided a new dimension to improve the accuracy of diagnoses and an independent system for recalibrating the histology guidelines. However, this model was based on the invasive method of endocardial biopsy, which undoubtedly increased the postoperative risk of heart transplant patients. Currently, little is known regarding the associated genes and specific functions of the different phenotypes. We performed bioinformatics analysis (using machine-learning methods and the WGCNA algorithm) to screen for hub-specific genes related to different phenotypes, based Gene Expression Omnibus accession number GSE124897. More immune cell infiltration was observed with the ABMR, TCMR, and injury phenotypes than with the stable phenotype. Hub-specific genes for each of the four archetypes were verified successfully using an external test set (accession number GSE2596). Logistic-regression models based on TCMR-specific hub genes and common hub genes were constructed with accurate diagnostic utility (area under the curve > 0.95). RELA, NFKB1, and SOX14 were identified as transcription factors important for TCMR/injury phenotypes and common genes, respectively. Additionally, 11 Food and Drug Administration-approved drugs were chosen from the DrugBank Database for each four-archetype model. Tyrosine kinase inhibitors may be a promising new option for transplant rejection treatment. KRAS signaling in cardiac transplant rejection is worth further investigation. Our results showed that heart transplant rejection subtypes can be accurately diagnosed by detecting expression of the corresponding specific genes, thereby enabling precise treatment or medication.
Collapse
Affiliation(s)
- Hongjie Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Ming Yuan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jie Cai
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jiajun Shi
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Yang Li
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Qiaofeng Qian
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Zhe Dong
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Gaofeng Pan
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Shaoping Zhu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Wei Wang
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China
| | - Xianwu Zhou
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| | - Jinping Liu
- Department of Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, 430071, China.
- Hubei Provincial Engineering Research Center of Minimally Invasive Cardiovascular Surgery, Wuhan, 430071, China.
- Wuhan Clinical Research Center for Minimally Invasive Treatment of Structural Heart Disease, Wuhan, 430071, China.
| |
Collapse
|
3
|
Olsen B, Bodea J, Garcia A, Beebe K, Campbell C, Schwalbach C, Salzberg D, Miller H, Adams R, Mirea L, Castillo P, Horn B, Bansal S, Mohanakumar T, Ngwube A. Vitamin D Supplementation: Association With Serum Cytokines in Pediatric Hematopoietic Stem Cell Transplantation. Front Pediatr 2022; 10:913586. [PMID: 35911824 PMCID: PMC9326107 DOI: 10.3389/fped.2022.913586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/20/2022] [Indexed: 01/27/2023] Open
Abstract
Vitamin D deficiency is prevalent in pediatric patients presenting for hematopoietic stem cell transplantation (HSCT) and has been linked to poor clinical outcomes. Using the data from a randomized control trial, in this paper we explore the effects of vitamin D supplementation on circulating cytokine levels during pediatric HSCT (www.clinicaltrials.gov as NCT03176849). A total of 41 children, 20 received Stoss therapy and 21 children received standard of care vitamin D supplementation. Levels of 25(OH)D and 20 cytokines were assessed at baseline and day +30. Significantly (P < 0.05) higher levels of mostly proinflammatory cytokines, FGF, GCSF, TNFα, IL-2, IL-6, IP10 were detected pre-transplant for patients with low compared to those with normal vitamin D levels. In sex stratified models that compare changes in cytokines between Stoss vs. standard of care, females in the Stoss group show greater changes in mostly pro -inflammatory cytokines- IP-10 (P = 0.0047), MIG (P = 0.009), and RANTES (P = 0.0047), IL-2R (P = 0.07) and IL-6(P = 0.069). Despite a small sample size, these findings suggest vitamin D deficiency affects the pre-transplant cytokine milieu and higher doses of vitamin D (Stoss therapy) appears to influence proinflammatory cytokine responses in a sex specific manner during pediatric HSCT. Larger clinical trials are warranted to validate these results.
Collapse
Affiliation(s)
- Braden Olsen
- Creighton School of Medicine, Phoenix, AZ, United States
| | - Jessica Bodea
- St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Angela Garcia
- Creighton School of Medicine, Phoenix, AZ, United States
| | - Kristen Beebe
- Phoenix Children's Hospital, Phoenix, AZ, United States.,Mayo Clinic Arizona, Phoenix, AZ, United States
| | | | | | - Dana Salzberg
- Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Holly Miller
- Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Roberta Adams
- Phoenix Children's Hospital, Phoenix, AZ, United States.,Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Lucia Mirea
- Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Paul Castillo
- UF Health Shands Children's Hospital, Gainesville, FL, United States
| | - Biljana Horn
- UF Health Shands Children's Hospital, Gainesville, FL, United States
| | - Sandhya Bansal
- St. Joseph's Hospital & Medical Center, Phoenix, AZ, United States
| | | | - Alexander Ngwube
- Creighton School of Medicine, Phoenix, AZ, United States.,Phoenix Children's Hospital, Phoenix, AZ, United States.,Mayo Clinic Arizona, Phoenix, AZ, United States
| |
Collapse
|
4
|
Wu Y, Zhang S, Yan J. IRF1 association with tumor immune microenvironment and use as a diagnostic biomarker for colorectal cancer recurrence. Oncol Lett 2020; 19:1759-1770. [PMID: 32194669 PMCID: PMC7039159 DOI: 10.3892/ol.2020.11289] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) is considered to be one of the most lethal cancer types globally, and its recurrence is a major treatment challenge. Identifying the factors involved when determining the risk of CRC recurrence is required to improve personalized therapy for patients with CRC. Based on the GSE39582 dataset, the present study demonstrated that a higher ratio of M1 macrophages and activated memory CD4+ T cells indicated a better recurrence-free survival (RFS) time for CRC, using CIBERSORT and Pearson's correlation analysis. Through weighted correlation network analysis (WGCNA), an immune-associated module was identified that was significantly positively correlated with the ratio of M1 macrophages and activated memory CD4+ T cells. In this module, using WGCNA and a protein-protein interaction network, interferon regulatory factor 1 (IRF1), chemokine ligand 5, ubiquitin/ISG15-conjugating enzyme E2 L6, guanylate binding protein 1 and interleukin 2 receptor subunit beta were identified as hub genes. Among these genes, univariate Cox and multivariate Cox analysis revealed that IRF1 may be a potential diagnostic biomarker for RFS in patients with CRC. This was further validated using The Cancer Genome Atlas data. Gene set enrichment analysis demonstrated that IRF1 influenced the genes and pathways that are associated with immune cell recruitment and activation. Additionally, the DNA methylation of cg27587780 and cg15375424 CpG sites in the IRF1 gene region was indicated to be negatively correlated with IRF1 mRNA expression and positively correlated with the recurrence of CRC. Collectively, the results of the present study demonstrated that IRF1 may be a potential diagnostic biomarker for RFS in patients with CRC.
Collapse
Affiliation(s)
- Yanfang Wu
- Department of Gastroenterology, The Fourth People's Hospital of Shaanxi, Xi'an, Shanxi 710032, P.R. China
| | - Shuju Zhang
- Hunan Children's Research Institute, Hunan Children's Hospital, University of South China, Changsha, Hunan 410007, P.R. China
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, Beijing 102218, P.R. China
| |
Collapse
|
5
|
Enhancing Mesenchymal Stromal Cell Immunomodulation for Treating Conditions Influenced by the Immune System. Stem Cells Int 2019; 2019:7219297. [PMID: 31467564 PMCID: PMC6701346 DOI: 10.1155/2019/7219297] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs), formerly known as mesenchymal stem cells, are nonhematopoietic multipotent cells and are emerging worldwide as the most clinically used and promising source for allogeneic cell therapy. MSCs, initially obtained from bone marrow, can be derived from several other tissues, such as adipose tissue, placenta, and umbilical cord. Diversity in tissue sourcing and manufacturing procedures has significant effects on MSC products. However, in 2006, a minimal set of standard criteria has been issued by the International Society of Cellular Therapy for defining derived MSCs. These include adherence to plastic in conventional culture conditions, particular phenotype, and multilineage differentiation capacity in vitro. Moreover, MSCs have trophic capabilities, a high in vitro self-renewal ability, and immunomodulatory characteristics. Thus, immunosuppressive treatment with MSCs has been proposed as a potential therapeutic alternative for conditions in which the immune system cells influence outcomes, such as inflammatory and autoimmune diseases. The precise mechanism by which MSCs affect functions of most immune effector cells is not completely understood but involves direct contact with immune cells, soluble mediators, and local microenvironmental factors. Recently, it has been shown that their homeostatic resting state requires activation, which can be achieved in vitro with various cytokines, including interferon-γ. In the present review, we focus on the suppressive effect that MSCs exert on the immune system and highlight the significance of in vitro preconditioning and its use in preclinical studies. We discuss the clinical aspects of using MSCs as an immunomodulatory treatment. Finally, we comment on the risk of interfering with the immune system in regard to cancer formation and development.
Collapse
|
6
|
Zou XF, Song B, Duan JH, Hu ZD, Cui ZL, Yang T. PRINS Long Noncoding RNA Involved in IP-10–Mediated Allograft Rejection in Rat Kidney Transplant. Transplant Proc 2018; 50:1558-1565. [DOI: 10.1016/j.transproceed.2018.03.105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/01/2018] [Accepted: 03/12/2018] [Indexed: 01/10/2023]
|
7
|
Kadle RL, Abdou SA, Villarreal-Ponce AP, Soares MA, Sultan DL, David JA, Massie J, Rifkin WJ, Rabbani P, Ceradini DJ. Microenvironmental cues enhance mesenchymal stem cell-mediated immunomodulation and regulatory T-cell expansion. PLoS One 2018. [PMID: 29513756 PMCID: PMC5841747 DOI: 10.1371/journal.pone.0193178] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are known to both have powerful immunosuppressive properties and promote allograft tolerance. Determining the environmental oxygen tension and inflammatory conditions under which MSCs are optimally primed for this immunosuppressive function is essential to their utilization in promoting graft tolerance. Of particular interest is the mechanisms governing the interaction between MSCs and regulatory T cells (Tregs), which is relatively unknown. We performed our experiments utilizing rat bone marrow derived MSCs. We observed that priming MSCs in hypoxia promotes maintenance of stem-like characteristics, with greater expression of typical MSC cell-surface markers, increased proliferation, and maintenance of differentiation potential. Addition of autologous MSCs to CD4+/allogeneic endothelial cell (EC) co-culture increases regulatory T cell (Treg) proliferation, which is further enhanced when MSCs are primed in hypoxia. Furthermore, MSC-mediated Treg expansion does not require direct cell-cell contact. The expression of indolamine 2,3-dioxygenase, a mediator of MSC immunomodulation, increases when MSCs are primed in hypoxia, and inhibition of IDO significantly decreases the expansion of Tregs. Priming with inflammatory cytokines IFNγ and TNFα increases also expression of markers associated with MSC immunomodulatory function, but decreases MSC proliferation. The expression of IDO also increases when MSCs are primed with inflammatory cytokines. However, there is no increase in Treg expansion when MSCs are primed with IFNγ, suggesting an alternate mechanism for inflammatory-stimulated MSC immunomodulation. Overall, these results suggest that MSCs primed in hypoxia or inflammatory conditions are optimally primed for immunosuppressive function. These results provide a clearer picture of how to enhance MSC immunomodulation for clinical use.
Collapse
Affiliation(s)
- Rohini L. Kadle
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Salma A. Abdou
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | | | - Marc A. Soares
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Darren L. Sultan
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Joshua A. David
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Jonathan Massie
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - William J. Rifkin
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Piul Rabbani
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
| | - Daniel J. Ceradini
- NYU Langone Medical Center, Department of Plastic Surgery, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
8
|
Mildenberger J, Johansson I, Sergin I, Kjøbli E, Damås JK, Razani B, Flo TH, Bjørkøy G. N-3 PUFAs induce inflammatory tolerance by formation of KEAP1-containing SQSTM1/p62-bodies and activation of NFE2L2. Autophagy 2017; 13:1664-1678. [PMID: 28820283 PMCID: PMC5640206 DOI: 10.1080/15548627.2017.1345411] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Inflammation is crucial in the defense against infections but must be tightly controlled to limit detrimental hyperactivation. Our diet influences inflammatory processes and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have known anti-inflammatory effects. The balance of pro- and anti-inflammatory processes is coordinated by macrophages and macroautophagy/autophagy has recently emerged as a cellular process that dampens inflammation. Here we report that the n-3 PUFA docosahexaenoic acid (DHA) transiently induces cytosolic speckles of the autophagic receptor SQSTM1/p62 (sequestosome 1) (described as SQSTM1/p62-bodies) in macrophages. We suggest that the formation of SQSTM1/p62-bodies represents a fast mechanism of NFE2L2/Nrf2 (nuclear factor, erythroid 2 like 2) activation by recruitment of KEAP1 (kelch like ECH associated protein 1). Further, the autophagy receptor TAX1BP1 (Tax1 binding protein 1) and ubiquitin-editing enzyme TNFAIP3/A20 (TNF α induced protein 3) could be identified in DHA-induced SQSTM1/p62-bodies. Simultaneously, DHA strongly dampened the induction of pro-inflammatory genes including CXCL10 (C-X-C motif chemokine ligand 10) and we suggest that formation of SQSTM1/p62-bodies and activation of NFE2L2 leads to tolerance towards selective inflammatory stimuli. Finally, reduced CXCL10 levels were related to the improved clinical outcome in n-3 PUFA-supplemented heart-transplant patients and we propose CXCL10 as a robust marker for the clinical benefits mobilized by n-3 PUFA supplementation.
Collapse
Affiliation(s)
- Jennifer Mildenberger
- a Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences , Norwegian University of Science and Technology , Trondheim , Norway.,b Department of Biomedical Laboratory Science, Faculty of Natural Sciences , Norwegian University of Science and Technology , Trondheim , Norway
| | - Ida Johansson
- a Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences , Norwegian University of Science and Technology , Trondheim , Norway
| | - Ismail Sergin
- d Department of Medicine, Cardiovascular Division , Washington University School of Medicine , St. Louis , MO , USA
| | - Eli Kjøbli
- b Department of Biomedical Laboratory Science, Faculty of Natural Sciences , Norwegian University of Science and Technology , Trondheim , Norway
| | - Jan Kristian Damås
- a Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences , Norwegian University of Science and Technology , Trondheim , Norway.,c Department of Infectious Diseases , St Olav University Hospital , Trondheim , Norway
| | - Babak Razani
- d Department of Medicine, Cardiovascular Division , Washington University School of Medicine , St. Louis , MO , USA.,e Department of Pathology & Immunology , Washington University School of Medicine , St. Louis , MO , USA
| | - Trude Helen Flo
- a Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences , Norwegian University of Science and Technology , Trondheim , Norway
| | - Geir Bjørkøy
- a Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine, Faculty of Medicine and Health Sciences , Norwegian University of Science and Technology , Trondheim , Norway.,b Department of Biomedical Laboratory Science, Faculty of Natural Sciences , Norwegian University of Science and Technology , Trondheim , Norway
| |
Collapse
|
9
|
Shino MY, Weigt SS, Li N, Derhovanessian A, Sayah DM, Huynh RH, Saggar R, Gregson AL, Ardehali A, Ross DJ, Lynch JP, Elashoff RM, Belperio JA. Impact of Allograft Injury Time of Onset on the Development of Chronic Lung Allograft Dysfunction After Lung Transplantation. Am J Transplant 2017; 17:1294-1303. [PMID: 27676455 PMCID: PMC5368037 DOI: 10.1111/ajt.14066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/16/2016] [Accepted: 09/18/2016] [Indexed: 01/25/2023]
Abstract
The impact of allograft injury time of onset on the risk of chronic lung allograft dysfunction (CLAD) remains unknown. We hypothesized that episodes of late-onset (≥6 months) allograft injury would produce an augmented CXCR3/ligand immune response, leading to increased CLAD. In a retrospective single-center study, 1894 transbronchial biopsy samples from 441 lung transplant recipients were reviewed for the presence of acute rejection (AR), lymphocytic bronchiolitis (LB), diffuse alveolar damage (DAD), and organizing pneumonia (OP). The association between the time of onset of each injury pattern and CLAD was assessed by using multivariable Cox models with time-dependent covariates. Bronchoalveolar lavage (BAL) CXCR3 ligand concentrations were compared between early- and late-onset injury patterns using linear mixed-effects models. Late-onset DAD and OP were strongly associated with CLAD: adjusted hazard ratio 2.8 (95% confidence interval 1.5-5.3) and 2.0 (1.1-3.4), respectively. The early-onset form of these injury patterns did not increase CLAD risk. Late-onset LB and acute rejection (AR) predicted CLAD in univariable models but lost significance after multivariable adjustment for late DAD and OP. AR was the only early-onset injury pattern associated with CLAD development. Elevated BAL CXCR3 ligand concentrations during late-onset allograft injury parallel the increase in CLAD risk and support our hypothesis that late allograft injuries result in a more profound CXCR3/ligand immune response.
Collapse
Affiliation(s)
- MY Shino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - SS Weigt
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - N Li
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA 90095-1652
| | - A Derhovanessian
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - DM Sayah
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - RH Huynh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - R Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - AL Gregson
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1688
| | - A Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1741
| | - DJ Ross
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - JP Lynch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| | - RM Elashoff
- Department of Biomathematics, University of California at Los Angeles, Los Angeles, CA 90095-1652
| | - JA Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095-1690
| |
Collapse
|
10
|
A Computational Gene Expression Score for Predicting Immune Injury in Renal Allografts. PLoS One 2015; 10:e0138133. [PMID: 26367000 PMCID: PMC4569485 DOI: 10.1371/journal.pone.0138133] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 08/25/2015] [Indexed: 12/13/2022] Open
Abstract
Background Whole genome microarray meta-analyses of 1030 kidney, heart, lung and liver allograft biopsies identified a common immune response module (CRM) of 11 genes that define acute rejection (AR) across different engrafted tissues. We evaluated if the CRM genes can provide a molecular microscope to quantify graft injury in acute rejection (AR) and predict risk of progressive interstitial fibrosis and tubular atrophy (IFTA) in histologically normal kidney biopsies. Methods Computational modeling was done on tissue qPCR based gene expression measurements for the 11 CRM genes in 146 independent renal allografts from 122 unique patients with AR (n = 54) and no-AR (n = 92). 24 demographically matched patients with no-AR had 6 and 24 month paired protocol biopsies; all had histologically normal 6 month biopsies, and 12 had evidence of progressive IFTA (pIFTA) on their 24 month biopsies. Results were correlated with demographic, clinical and pathology variables. Results The 11 gene qPCR based tissue CRM score (tCRM) was significantly increased in AR (5.68 ± 0.91) when compared to STA (1.29 ± 0.28; p < 0.001) and pIFTA (7.94 ± 2.278 versus 2.28 ± 0.66; p = 0.04), with greatest significance for CXCL9 and CXCL10 in AR (p <0.001) and CD6 (p<0.01), CXCL9 (p<0.05), and LCK (p<0.01) in pIFTA. tCRM was a significant independent correlate of biopsy confirmed AR (p < 0.001; AUC of 0.900; 95% CI = 0.705–903). Gene expression modeling of 6 month biopsies across 7/11 genes (CD6, INPP5D, ISG20, NKG7, PSMB9, RUNX3, and TAP1) significantly (p = 0.037) predicted the development of pIFTA at 24 months. Conclusions Genome-wide tissue gene expression data mining has supported the development of a tCRM-qPCR based assay for evaluating graft immune inflammation. The tCRM score quantifies injury in AR and stratifies patients at increased risk of future pIFTA prior to any perturbation of graft function or histology.
Collapse
|
11
|
Bai X, Qi Z, Song G, Zhao X, Zhao H, Meng X, Liu C, Bing W, Bi Y. Effects of Monocyte Chemotactic Protein-1 and Nuclear Factor of Kappa B Pathway in Rejection of Cardiac Allograft in Rat. Transplant Proc 2015; 47:2010-6. [DOI: 10.1016/j.transproceed.2015.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/26/2015] [Accepted: 05/14/2015] [Indexed: 01/04/2023]
|