1
|
Spector BL, Koseva BS, Sante D, Cheung WA, Alisch RS, Kats A, Bergmann P, Grundberg E, Wyckoff GJ, Willig LK. Total plasma cfDNA methylation in pediatric kidney transplant recipients provides insight into acute allograft rejection pathophysiology. Clin Immunol 2025; 275:110475. [PMID: 40107586 DOI: 10.1016/j.clim.2025.110475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Cell-free DNA (cfDNA) is a marker of organ injury and immune response. DNA methylation is an epigenetic regulator of gene expression. Here, we elucidate total plasma cfDNA methylation from kidney transplant recipients in presence versus absence of rejection. In doing so, we exploit cfDNA as a real-time biomarker to define molecular pathways of rejection. Twenty plasma cfDNA samples from pediatric kidney transplant recipients were collected at allograft biopsy. Differentially methylated cytosine residues (>20 % methylation difference, q-value <0.05) were identified in presence (N = 7) versus absence (N = 9) of acute rejection. Separate analyses were performed comparing borderline rejection (N = 4) to rejection and non-rejection. In rejection versus non-rejection, there were 1269 differentially methylated genes corresponding to 533 pathways. These numbers were 4-13× greater than comparisons against borderline samples. Enriched pathways between rejection and non-rejection samples were related to immune cell/inflammatory response, lipid metabolism, and tryptophan-kynurenine metabolism, suggesting differential methylation of these pathways contributes to rejection.
Collapse
Affiliation(s)
- Benjamin L Spector
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States; Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States.
| | - Boryana S Koseva
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Drinnan Sante
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City School of Pharmacy, Kansas City, MO, United States
| | - Warren A Cheung
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Alexander Kats
- Department of Pathology and Laboratory Medicine, Children's Mercy Hospital, Kansas City, MO, United States
| | - Phillip Bergmann
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Elin Grundberg
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| | - Gerald J Wyckoff
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City School of Pharmacy, Kansas City, MO, United States
| | - Laurel K Willig
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, MO, United States
| |
Collapse
|
2
|
Shirey KA, Joseph J, Coughlan L, Nijhuis H, Varley AW, Blanco JCG, Vogel SN. An adenoviral vector encoding an inflammation-inducible antagonist, HMGB1 Box A, as a novel therapeutic approach to inflammatory diseases. mBio 2025; 16:e0338724. [PMID: 39699172 PMCID: PMC11796352 DOI: 10.1128/mbio.03387-24] [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: 10/30/2024] [Accepted: 11/26/2024] [Indexed: 12/20/2024] Open
Abstract
Influenza, as well as other respiratory viruses, can trigger local and systemic inflammation resulting in an overall "cytokine storm" that produces serious outcomes such as acute lung injury (ALI) or acute respiratory distress syndrome (ARDS). We hypothesized that gene therapy platforms could be useful in these cases if the production of an anti-inflammatory protein reflects the intensity and duration of the inflammatory condition. The recombinant protein would be produced and released only in the presence of the inciting stimulus, avoiding immunosuppression or other unwanted side effects that may occur when treating infectious diseases with anti-inflammatory drugs. To test this hypothesis, we developed AdV.C3-Tat/HIV-Box A, an inflammation-inducible cassette that remains innocuous in the absence of inflammation but releases HMGB1 Box A, an antagonist of high mobility group box 1 (HMGB1), in response to inflammatory stimuli such as lipopolysaccharide (LPS) or influenza virus infection. We report here that this novel inflammation-inducible HMGB1 Box A construct in a non-replicative adenovirus (AdV) vector mitigates lung and systemic inflammation therapeutically in response to influenza infection. We anticipate that this strategy will apply to the treatment of multiple diseases in which HMGB1-mediated signaling is a central driver of inflammation.IMPORTANCEMany inflammatory diseases are mediated by the action of a host-derived protein, HMGB1, on Toll-like receptor 4 (TLR4) to elicit an inflammatory response. We have engineered a non-replicative AdV vector that produces HMGB1 Box A, an antagonist of HMGB1-induced inflammation, under the control of an endogenous complement component C3 (C3) promoter sequence, that is inducible by LPS and influenza in vitro and ex vivo in macrophages (Mϕ) and protects mice and cotton rats therapeutically against infection with mouse-adapted and human non-adapted influenza strains, respectively, in vivo. We anticipate that this novel strategy will apply to the treatment of multiple infectious and non-infectious diseases in which HMGB1-mediated TLR4 signaling is a central driver of inflammation.
Collapse
Affiliation(s)
- Kari Ann Shirey
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - John Joseph
- Sigmovir Biosystems Inc., Rockville, Maryland, USA
| | - Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Center for Vaccine Development and Global Health (CVD), University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Haye Nijhuis
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, Location AMC, University of Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, the Netherlands
| | | | | | - Stefanie N. Vogel
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Jacobs MME, Maas RJF, Jonkman I, Negishi Y, Tielemans Zamora W, Yanginlar C, van Heck J, Matzaraki V, Martens JHA, Baltissen M, Vermeulen M, Morla-Folch J, Ranzenigo A, Wang W, Umali M, Ochando J, van der Vlag J, Hilbrands LB, Joosten LAB, Netea MG, Mulder WJM, van Leent MMT, Mhlanga MM, Teunissen AJP, Rother N, Duivenvoorden R. Trained immunity is regulated by T cell-induced CD40-TRAF6 signaling. Cell Rep 2024; 43:114664. [PMID: 39178113 PMCID: PMC11536040 DOI: 10.1016/j.celrep.2024.114664] [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: 02/05/2024] [Revised: 07/08/2024] [Accepted: 08/06/2024] [Indexed: 08/25/2024] Open
Abstract
Trained immunity is characterized by histone modifications and metabolic changes in innate immune cells following exposure to inflammatory signals, leading to heightened responsiveness to secondary stimuli. Although our understanding of the molecular regulation of trained immunity has increased, the role of adaptive immune cells herein remains largely unknown. Here, we show that T cells modulate trained immunity via cluster of differentiation 40-tissue necrosis factor receptor-associated factor 6 (CD40-TRAF6) signaling. CD40-TRAF6 inhibition modulates functional, transcriptomic, and metabolic reprogramming and modifies histone 3 lysine 4 trimethylation associated with trained immunity. Besides in vitro studies, we reveal that single-nucleotide polymorphisms in the proximity of CD40 are linked to trained immunity responses in vivo and that combining CD40-TRAF6 inhibition with cytotoxic T lymphocyte antigen 4-immunoglobulin (CTLA4-Ig)-mediated co-stimulatory blockade induces long-term graft acceptance in a murine heart transplantation model. Combined, our results reveal that trained immunity is modulated by CD40-TRAF6 signaling between myeloid and adaptive immune cells and that this can be leveraged for therapeutic purposes.
Collapse
Affiliation(s)
- Maaike M E Jacobs
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne J F Maas
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Inge Jonkman
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yutaka Negishi
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Willem Tielemans Zamora
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cansu Yanginlar
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Julia van Heck
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Marijke Baltissen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Judit Morla-Folch
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Ranzenigo
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - William Wang
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Martin Umali
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Johan van der Vlag
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Musa M Mhlanga
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, the Netherlands; Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Chen Y, Wang J, An C, Bao S, Zhang C. The role and research progress of macrophages after heart transplantation. Heliyon 2024; 10:e33844. [PMID: 39027574 PMCID: PMC11255595 DOI: 10.1016/j.heliyon.2024.e33844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Since the 60s of the 20th century, heart transplantation has been the best treatment for patients with end-stage heart failure. Due to the increasing number of patients, how to expand the number of donor organs and enhance immune compatibility has become an urgent problem to be solved at this stage. Although current immunosuppression is effective, its side effects are also quite obvious, such as opportunistic infections and malignant tumors. In this review, we focus on the important role in macrophages after heart transplantation and their potential targets for achieving allogeneic graft tolerance, in order to improve effective graft survival and reduce infection and the occurrence of malignant tumors.
Collapse
Affiliation(s)
- Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - JianPeng Wang
- School of First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ShanQing Bao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - ChengXin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| |
Collapse
|
5
|
Vezzoli A, Mrakic-Sposta S, Brizzolari A, Balestra C, Camporesi EM, Bosco G. Oxy-Inflammation in Humans during Underwater Activities. Int J Mol Sci 2024; 25:3060. [PMID: 38474303 DOI: 10.3390/ijms25053060] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/22/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Underwater activities are characterized by an imbalance between reactive oxygen/nitrogen species (RONS) and antioxidant mechanisms, which can be associated with an inflammatory response, depending on O2 availability. This review explores the oxidative stress mechanisms and related inflammation status (Oxy-Inflammation) in underwater activities such as breath-hold (BH) diving, Self-Contained Underwater Breathing Apparatus (SCUBA) and Closed-Circuit Rebreather (CCR) diving, and saturation diving. Divers are exposed to hypoxic and hyperoxic conditions, amplified by environmental conditions, hyperbaric pressure, cold water, different types of breathing gases, and air/non-air mixtures. The "diving response", including physiological adaptation, cardiovascular stress, increased arterial blood pressure, peripheral vasoconstriction, altered blood gas values, and risk of bubble formation during decompression, are reported.
Collapse
Affiliation(s)
- Alessandra Vezzoli
- Institute of Clinical Physiology-National Research Council (CNR-IFC), 20142 Milano, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Simona Mrakic-Sposta
- Institute of Clinical Physiology-National Research Council (CNR-IFC), 20142 Milano, Italy
| | - Andrea Brizzolari
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Costantino Balestra
- Environmental, Occupational, Aging (Integrative) Physiology Laboratory, Haute Ecole Bruxelles-Brabant (HE2B), 1160 Brussels, Belgium
- Physical Activity Teaching Unit, Motor Sciences Department, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
- DAN Europe Research Division (Roseto-Brussels), 1160 Brussels, Belgium
| | | | - Gerardo Bosco
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
6
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
7
|
Tseng HT, Lin YW, Huang CY, Shih CM, Tsai YT, Liu CW, Tsai CS, Lin FY. Animal Models for Heart Transplantation Focusing on the Pathological Conditions. Biomedicines 2023; 11:biomedicines11051414. [PMID: 37239085 DOI: 10.3390/biomedicines11051414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/29/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Cardiac transplant recipients face many complications due to transplant rejection. Scientists must conduct animal experiments to study disease onset mechanisms and develop countermeasures. Therefore, many animal models have been developed for research topics including immunopathology of graft rejection, immunosuppressive therapies, anastomotic techniques, and graft preservation techniques. Small experimental animals include rodents, rabbits, and guinea pigs. They have a high metabolic rate, high reproductive rate, small size for easy handling, and low cost. Additionally, they have genetically modified strains for pathological mechanisms research; however, there is a lacuna, as these research results rarely translate directly to clinical applications. Large animals, including canines, pigs, and non-human primates, have anatomical structures and physiological states that are similar to those of humans; therefore, they are often used to validate the results obtained from small animal studies and directly speculate on the feasibility of applying these results in clinical practice. Before 2023, PubMed Central® at the United States National Institute of Health's National Library of Medicine was used for literature searches on the animal models for heart transplantation focusing on the pathological conditions. Unpublished reports and abstracts from conferences were excluded from this review article. We discussed the applications of small- and large-animal models in heart transplantation-related studies. This review article aimed to provide researchers with a complete understanding of animal models for heart transplantation by focusing on the pathological conditions created by each model.
Collapse
Affiliation(s)
- Horng-Ta Tseng
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Wen Lin
- Institute of Oral Biology, National Yang Ming Chiao Tung University (Yangming Campus), Taipei 112304, Taiwan
| | - Chun-Yao Huang
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Ming Shih
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Ting Tsai
- Division of Cardiovascular Surgery, Tri-Service General Hospital, Defense Medical Center, Taipei 11490, Taiwan
| | - Chen-Wei Liu
- Department of Basic Medical Science, College of Medicine, University of Arizona, Phoenix, AZ 85721, USA
| | - Chien-Sung Tsai
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiovascular Surgery, Tri-Service General Hospital, Defense Medical Center, Taipei 11490, Taiwan
- Department and Graduate Institute of Pharmacology, National Defense Medical Center, Taipei 11490, Taiwan
| | - Feng-Yen Lin
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
- Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Departments of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
8
|
Lajqi T, Köstlin-Gille N, Bauer R, Zarogiannis SG, Lajqi E, Ajeti V, Dietz S, Kranig SA, Rühle J, Demaj A, Hebel J, Bartosova M, Frommhold D, Hudalla H, Gille C. Training vs. Tolerance: The Yin/Yang of the Innate Immune System. Biomedicines 2023; 11:766. [PMID: 36979747 PMCID: PMC10045728 DOI: 10.3390/biomedicines11030766] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
For almost nearly a century, memory functions have been attributed only to acquired immune cells. Lately, this paradigm has been challenged by an increasing number of studies revealing that innate immune cells are capable of exhibiting memory-like features resulting in increased responsiveness to subsequent challenges, a process known as trained immunity (known also as innate memory). In contrast, the refractory state of endotoxin tolerance has been defined as an immunosuppressive state of myeloid cells portrayed by a significant reduction in the inflammatory capacity. Both training as well tolerance as adaptive features are reported to be accompanied by epigenetic and metabolic alterations occurring in cells. While training conveys proper protection against secondary infections, the induction of endotoxin tolerance promotes repairing mechanisms in the cells. Consequently, the inappropriate induction of these adaptive cues may trigger maladaptive effects, promoting an increased susceptibility to secondary infections-tolerance, or contribute to the progression of the inflammatory disorder-trained immunity. This review aims at the discussion of these opposing manners of innate immune and non-immune cells, describing the molecular, metabolic and epigenetic mechanisms involved and interpreting the clinical implications in various inflammatory pathologies.
Collapse
Affiliation(s)
- Trim Lajqi
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Natascha Köstlin-Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Reinhard Bauer
- Institute of Molecular Cell Biology, Jena University Hospital, D-07745 Jena, Germany
| | - Sotirios G. Zarogiannis
- Department of Physiology, School of Health Sciences, Faculty of Medicine, University of Thessaly, GR-41500 Larissa, Greece
| | - Esra Lajqi
- Department of Radiation Oncology, Heidelberg University Hospital, D-69120 Heidelberg, Germany
| | - Valdrina Ajeti
- Department of Pharmacy, Alma Mater Europaea—Campus College Rezonanca, XK-10000 Pristina, Kosovo
| | - Stefanie Dietz
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Simon A. Kranig
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Jessica Rühle
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Ardian Demaj
- Faculty of Medical Sciences, University of Tetovo, MK-1200 Tetova, North Macedonia
| | - Janine Hebel
- Department of Neonatology, University of Tübingen, D-72076 Tübingen, Germany
| | - Maria Bartosova
- Center for Pediatric and Adolescent Medicine Heidelberg, University of Heidelberg, D-69120 Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, D-87700 Memmingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children’s Hospital, D-69120 Heidelberg, Germany
| |
Collapse
|
9
|
David BT, Curtin JJ, Brown JL, Scorpio K, Kandaswamy V, Coutts DJC, Vivinetto A, Bianchimano P, Karuppagounder SS, Metcalfe M, Cave JW, Hill CE. Temporary induction of hypoxic adaptations by preconditioning fails to enhance Schwann cell transplant survival after spinal cord injury. Glia 2023; 71:648-666. [PMID: 36565279 PMCID: PMC11848738 DOI: 10.1002/glia.24302] [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: 04/19/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 12/25/2022]
Abstract
Hypoxic preconditioning is protective in multiple models of injury and disease, but whether it is beneficial for cells transplanted into sites of spinal cord injury (SCI) is largely unexplored. In this study, we analyzed whether hypoxia-related preconditioning protected Schwann cells (SCs) transplanted into the contused thoracic rat spinal cord. Hypoxic preconditioning was induced in SCs prior to transplantation by exposure to either low oxygen (1% O2 ) or pharmacological agents (deferoxamine or adaptaquin). All preconditioning approaches induced hypoxic adaptations, including increased expression of HIF-1α and its target genes. These adaptations, however, were transient and resolved within 24 h of transplantation. Pharmacological preconditioning attenuated spinal cord oxidative stress and enhanced transplant vascularization, but it did not improve either transplanted cell survival or recovery of sensory or motor function. Together, these experiments show that hypoxia-related preconditioning is ineffective at augmenting either cell survival or the functional outcomes of SC-SCI transplants. They also reveal that the benefits of hypoxia-related adaptations induced by preconditioning for cell transplant therapies are not universal.
Collapse
Affiliation(s)
- Brian T. David
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Jessica J. Curtin
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Jennifer L. Brown
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Kerri Scorpio
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Veena Kandaswamy
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - David J. C. Coutts
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Ana Vivinetto
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Paola Bianchimano
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Saravanan S. Karuppagounder
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - Mariajose Metcalfe
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
| | - John W. Cave
- InVitro Cell Research, LLC, Englewood, NJ, United States
| | - Caitlin E. Hill
- Burke Neurological Institute, White Plains, NY, United States
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, NY, United States
- Neural Stem Cell Institute, Rensselaer, NY, United States
| |
Collapse
|
10
|
Abstract
Solid organ transplantation is a life-saving treatment for people with end-stage organ disease. Immune-mediated transplant rejection is a common complication that decreases allograft survival. Although immunosuppression is required to prevent rejection, it also increases the risk of infection. Some infections, such as cytomegalovirus and BK virus, can promote inflammatory gene expression that can further tip the balance toward rejection. BK virus and other infections can induce damage that resembles the clinical pathology of rejection, and this complicates accurate diagnosis. Moreover, T cells specific for viral infection can lead to rejection through heterologous immunity to donor antigen directly mediated by antiviral cells. Thus, viral infections and allograft rejection interact in multiple ways that are important to maintain immunologic homeostasis in solid organ transplant recipients. Better insight into this dynamic interplay will help promote long-term transplant survival.
Collapse
Affiliation(s)
- Lauren E Higdon
- Department of Medicine/Nephrology, Stanford University, Palo Alto, CA
| | - Jane C Tan
- Department of Medicine/Nephrology, Stanford University, Palo Alto, CA
| | - Jonathan S Maltzman
- Department of Medicine/Nephrology, Stanford University, Palo Alto, CA
- Geriatric Research Education and Clinical Center, VA Palo Alto Health Care System, Palo Alto, CA
| |
Collapse
|
11
|
Ochando J, Mulder WJM, Madsen JC, Netea MG, Duivenvoorden R. Trained immunity - basic concepts and contributions to immunopathology. Nat Rev Nephrol 2023; 19:23-37. [PMID: 36253509 PMCID: PMC9575643 DOI: 10.1038/s41581-022-00633-5] [Citation(s) in RCA: 165] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 02/08/2023]
Abstract
Trained immunity is a functional state of the innate immune response and is characterized by long-term epigenetic reprogramming of innate immune cells. This concept originated in the field of infectious diseases - training of innate immune cells, such as monocytes, macrophages and/or natural killer cells, by infection or vaccination enhances immune responses against microbial pathogens after restimulation. Although initially reported in circulating monocytes and tissue macrophages (termed peripheral trained immunity), subsequent findings indicate that immune progenitor cells in the bone marrow can also be trained (that is, central trained immunity), which explains the long-term innate immunity-mediated protective effects of vaccination against heterologous infections. Although trained immunity is beneficial against infections, its inappropriate induction by endogenous stimuli can also lead to aberrant inflammation. For example, in systemic lupus erythematosus and systemic sclerosis, trained immunity might contribute to inflammatory activity, which promotes disease progression. In organ transplantation, trained immunity has been associated with acute rejection and suppression of trained immunity prolonged allograft survival. This novel concept provides a better understanding of the involvement of the innate immune response in different pathological conditions, and provides a new framework for the development of therapies and treatment strategies that target epigenetic and metabolic pathways of the innate immune system.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain.
| | - Willem J. M. Mulder
- grid.6852.90000 0004 0398 8763Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands ,grid.59734.3c0000 0001 0670 2351Biomedical Engineering and Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Joren C. Madsen
- grid.32224.350000 0004 0386 9924Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA USA ,grid.32224.350000 0004 0386 9924Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA USA
| | - Mihai G. Netea
- grid.10417.330000 0004 0444 9382Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands ,grid.10388.320000 0001 2240 3300Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Raphaël Duivenvoorden
- Biomedical Engineering and Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
12
|
Toll-like receptors and damage-associated molecular patterns in the pathogenesis of heart transplant rejection. Mol Cell Biochem 2022; 477:2841-2850. [PMID: 35678986 DOI: 10.1007/s11010-022-04491-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
Significant strides have been made in our understanding of the immune system and its role in cardiac transplant rejection. Despite the growing knowledge of immune responses, the mortality rate following cardiac transplantation remains grim. Related to procedural and pathological complications, toll-like receptor (TLR) and damage-associated molecular pattern (DAMP) signaling is the most direct and earliest interface between tissue integration and the innate immune response. This in turn can activate an adaptive immune response that further damages myocardial tissue. Furthermore, relevant literature on the status of DAMPs in the context of heart-transplantation remains limited, warranting further attention in clinical and translational research. This review aims to critically appraise the perspectives, advances, and challenges on DAMP-mediated innate immune response in the immune-mediated rejection of cardiac transplantation. Detailed analysis of the influence of TLR and DAMP signaling in mounting the immune response against the transplanted heart holds promise for improving outcomes through early detection and prevention of varied forms of organ rejection.
Collapse
|
13
|
Ginsenoside Rh2 reduces depression in offspring of mice with maternal toxoplasma infection during pregnancy by inhibiting microglial activation via the HMGB1/TLR4/NF-κB signaling pathway. J Ginseng Res 2022; 46:62-70. [PMID: 35035240 PMCID: PMC8753429 DOI: 10.1016/j.jgr.2021.04.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 02/08/2023] Open
Abstract
Background Maternal Toxoplasma gondii (T. gondii) infection during pregnancy has been associated with various mental illnesses in the offspring. Ginsenoside Rh2 (GRh2) is a major bioactive compound obtained from ginseng that has an anti-T. gondii effect and attenuates microglial activation through toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway. GRh2 also alleviated tumor-associated or lipopolysaccharide-induced depression. However, the effects and potential mechanisms of GRh2 on depression-like behavior in mouse offspring caused by maternal T. gondii infection during pregnancy have not been investigated. Methods We examined GRh2 effects on the depression-like behavior in mouse offspring, caused by maternal T. gondii infection during pregnancy, by measuring depression-like behaviors and assaying parameters at the neuronal and molecular level. Results We showed that GRh2 significantly improved behavioral measures: sucrose consumption, forced swim time and tail suspended immobility time of their offspring. These corresponded with increased tissue concentrations of 5-hydroxytryptamine and dopamine, and attenuated indoleamine 2,3-dioxygenase or enhanced tyrosine hydroxylase expression in the prefrontal cortex. GRh2 ameliorated neuronal damage in the prefrontal cortex. Molecular docking results revealed that GRh2 binds strongly to both TLR4 and high mobility group box 1 (HMGB1). Conclusion This study demonstrated that GRh2 ameliorated the depression-like behavior in mouse offspring of maternal T. gondii infection during pregnancy by attenuating the excessive activation of microglia and neuroinflammation through the HMGB1/TLR4/NF-κB signaling pathway. It suggests that GRh2 could be considered a potential therapy in preventing and treating psychiatric disorders in the offspring mice of mothers with prenatal exposure to T. gondii infection.
Collapse
|
14
|
Zhao J, Huang X, Mcleod P, Jiang J, Liu W, Haig A, Jevnikar AM, Jiang Z, Zhang ZX. Toll-like receptor 3 is an endogenous sensor of cell death and a potential target for induction of long-term cardiac transplant survival. Am J Transplant 2021; 21:3268-3279. [PMID: 33784431 DOI: 10.1111/ajt.16584] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/24/2021] [Accepted: 03/22/2021] [Indexed: 01/25/2023]
Abstract
Inflammation posttransplant is directly linked to cell death programs including apoptosis and necrosis. Cell death leads to the release of cellular contents which can promote inflammation. Targeting of these pathways should be an effective strategy to prevent transplant rejection. Toll-like receptor 3 (TLR3) is emerging as a major endogenous sensor of inflammation. In this study, we assessed the role of TLR3 on cell death and transplant rejection. We showed that TLR3 is highly expressed on mouse microvascular endothelial cell (ECs) and the endothelium of cardiac grafts. We demonstrated that TLR3 interacting with dsRNA or self-RNA triggered apoptosis and necroptosis in ECs. Interestingly, TLR3-induced necroptosis led mitochondrial damage. Inhibition of the mitochondrial membrane permeability molecule Cyclophilin D prevented necroptosis in ECs. In vivo, endothelium damage and activities of caspase-3 and mixed lineage kinase domain-like protein were inhibited in TLR3-/- cardiac grafts compared with C57BL/6 grafts posttransplant (n = 5, p < .001). Importantly, TLR3-/- cardiac grafts had prolonged survival in allogeneic BALB/c mice (mean survival = 121 ± 67 vs. 31 ± 6 days of C57BL/6 grafts, n = 7, p = .002). In summary, our study suggests that TLR3 is an important cell death inducer in ECs and cardiac grafts and thus a potential therapeutic target in preventing cardiac transplant rejection.
Collapse
Affiliation(s)
- Jiangqi Zhao
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, China
- Department of Pathology, Western University, London, ON, Canada
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
| | - Patrick Mcleod
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON, Canada
| | - Winnie Liu
- Department of Pathology, Western University, London, ON, Canada
| | - Aaron Haig
- Department of Pathology, Western University, London, ON, Canada
| | - Anthony M Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON, Canada
| | - Zhenyu Jiang
- Department of Rheumatology and Immunology, The First Hospital of Jilin University, Changchun, China
| | - Zhu-Xu Zhang
- Department of Pathology, Western University, London, ON, Canada
- Matthew Mailing Centre for Translational Transplantation Studies, London, ON, Canada
- Multi-Organ Transplant Program, London Health Sciences Centre, London, ON, Canada
- Division of Nephrology, Department of Medicine, Western University, London, ON, Canada
| |
Collapse
|
15
|
Chen Y, Zhang W, Bao H, He W, Chen L. High Mobility Group Box 1 Contributes to the Acute Rejection of Liver Allografts by Activating Dendritic Cells. Front Immunol 2021; 12:679398. [PMID: 34177922 PMCID: PMC8222728 DOI: 10.3389/fimmu.2021.679398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/25/2021] [Indexed: 01/03/2023] Open
Abstract
Acute rejection induced by the recognition of donor alloantigens by recipient T cells leads to graft failure in liver transplant recipients. The role of high mobility group box 1 (HMGB1), an inflammatory mediator, in the acute allograft rejection of liver transplants is unknown. Here, rat orthotopic liver transplantation was successfully established to analyze the expression pattern of HMGB1 in liver allografts and its potential role in promoting the maturation of dendritic cells (DCs) to promote T cell proliferation and differentiation. Five and 10 days after transplantation, allografts showed a marked upregulation of HMGB1 expression accompanied by elevated levels of serum transaminase and CD3+ and CD86+ inflammatory cell infiltration. Furthermore, in vitro experiments showed HMGB1 increased the expressions of co-stimulatory molecules (CD80, CD83, and MHC class II) on bone marrow-derived DCs. HMGB1-pulsed DCs induced naive CD4+ T cells to differentiate to Th1 and Th17 subsets secreting IFN-γ and IL-17, respectively. Further in vivo experiments confirmed the administration of glycyrrhizic acid, a natural HMGB1 inhibitor, during donor liver preservation had therapeutic effects by reducing inflammation and hepatocyte damage reflected by a decline in serum transaminase and prolonged allograft survival time. These results suggest the involvement of HMBG1 in acute liver allograft rejection and that it might be a candidate therapeutic target to avoid acute rejection after liver transplantation.
Collapse
Affiliation(s)
- Yi Chen
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Wenmin Zhang
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Institute of Oncology, Fujian Medical University, Fuzhou, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Hui Bao
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Institute of Oncology, Fujian Medical University, Fuzhou, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Wubing He
- Department of Emergency, Fujian Provincial Hospital; Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lihong Chen
- Department of Pathology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Pathology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,Institute of Oncology, Fujian Medical University, Fuzhou, China.,Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| |
Collapse
|
16
|
Zou H, Ming B, Li J, Xiao Y, Lai L, Gao M, Xu Y, Tan Z, Gong F, Zheng F. Extracellular HMGB1 Contributes to the Chronic Cardiac Allograft Vasculopathy/Fibrosis by Modulating TGF-β1 Signaling. Front Immunol 2021; 12:641973. [PMID: 33777037 PMCID: PMC7988222 DOI: 10.3389/fimmu.2021.641973] [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: 12/15/2020] [Accepted: 02/16/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac allograft vasculopathy (CAV) charactered with aberrant remodeling and fibrosis usually leads to the loss of graft after heart transplantation. Our previous work has reported that extracellular high-mobility group box 1 (HMGB1) participated in the CAV progression via promoting inflammatory cells infiltration and immune damage. The aim of this study was to investigate the involvement of HMGB1 in the pathogenesis of CAV/fibrosis and potential mechanisms using a chronic cardiac rejection model in mice. We found high levels of transforming growth factor (TGF)-β1 in cardiac allografts after transplantation. Treatment with HMGB1 neutralizing antibody markedly prolonged the allograft survival accompanied by attenuated fibrosis of cardiac allograft, decreased fibroblasts-to-myofibroblasts conversion, and reduced synthesis and release of TGF-β1. In addition, recombinant HMGB1 stimulation promoted release of active TGF-β1 from cardiac fibroblasts and macrophages in vitro, and subsequent phosphorylation of Smad2 and Smad3 which were downstream of TGF-β1 signaling. These data indicate that HMGB1 contributes to the CAV/fibrosis via promoting the activation of TGF-β1/Smad signaling. Targeting HMGB1 might become a new therapeutic strategy for inhibiting cardiac allograft fibrosis and dysfunction.
Collapse
Affiliation(s)
- Huijuan Zou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Reproductive Medicine Center, Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bingxia Ming
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jun Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Xiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lai
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Gao
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Tan
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feili Gong
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences, Wuhan, China.,NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.,Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
17
|
Wu Z, Liang J, Huang W, Jiang L, Paul C, Gao X, Alam P, Kanisicak O, Xu M, Wang Y. Immunomodulatory effects of mesenchymal stem cells for the treatment of cardiac allograft rejection. Exp Biol Med (Maywood) 2020; 246:851-860. [PMID: 33327780 DOI: 10.1177/1535370220978650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Heart transplantation continues to be the gold standard clinical intervention to treat patients with end-stage heart failure. However, there are major complications associated with this surgical procedure that reduce the survival prognosis of heart transplant patients, including allograft rejection, malignancies, infections, and other complications that arise from the use of broad-spectrum immunosuppression drugs. Recent studies have demonstrated the use of mesenchymal stem cells (MSCs) against allotransplantation rejection in both in vitro and in vivo settings due to their immunomodulatory properties. Therefore, utilization of MSCs provides new and exciting strategies to improve heart transplantation and potentially reduce the use of broad-spectrum immunosuppression drugs while alleviating allograft rejection. In this review, we will discuss the current research on the mechanisms of cardiac allograft rejection, the physiological and immunological characteristics of MSCs, the effects of MSCs on the immune system, and immunomodulation of heart transplantation by MSCs.
Collapse
Affiliation(s)
- Zhichao Wu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wei Huang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Lin Jiang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Christian Paul
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Xiang Gao
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Perwez Alam
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Onur Kanisicak
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meifeng Xu
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
18
|
Frye CC, Bery AI, Kreisel D, Kulkarni HS. Sterile inflammation in thoracic transplantation. Cell Mol Life Sci 2020; 78:581-601. [PMID: 32803398 DOI: 10.1007/s00018-020-03615-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/20/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
The life-saving benefits of organ transplantation can be thwarted by allograft dysfunction due to both infectious and sterile inflammation post-surgery. Sterile inflammation can occur after necrotic cell death due to the release of endogenous ligands [such as damage-associated molecular patterns (DAMPs) and alarmins], which perpetuate inflammation and ongoing cellular injury via various signaling cascades. Ischemia-reperfusion injury (IRI) is a significant contributor to sterile inflammation after organ transplantation and is associated with detrimental short- and long-term outcomes. While the vicious cycle of sterile inflammation and cellular injury is remarkably consistent amongst different organs and even species, we have begun understanding its mechanistic basis only over the last few decades. This understanding has resulted in the developments of novel, yet non-specific therapies for mitigating IRI-induced graft damage, albeit with moderate results. Thus, further understanding of the mechanisms underlying sterile inflammation after transplantation is critical for identifying personalized therapies to prevent or interrupt this vicious cycle and mitigating allograft dysfunction. In this review, we identify common and distinct pathways of post-transplant sterile inflammation across both heart and lung transplantation that can potentially be targeted.
Collapse
Affiliation(s)
- C Corbin Frye
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Amit I Bery
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA.
| | - Daniel Kreisel
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hrishikesh S Kulkarni
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine, 4523 Clayton Avenue, Campus Box 8052, St. Louis, MO, 63110, USA
| |
Collapse
|
19
|
Kaur I, Behl T, Bungau S, Kumar A, Mehta V, Setia D, Uddin MS, Zengin G, Aleya L, Arora S. Exploring the therapeutic promise of targeting HMGB1 in rheumatoid arthritis. Life Sci 2020; 258:118164. [PMID: 32739467 DOI: 10.1016/j.lfs.2020.118164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/25/2020] [Accepted: 07/25/2020] [Indexed: 12/22/2022]
Abstract
High mobility group box-1 (HMGB1) protein is a diverse, single polypeptide moiety, present in mammalian eukaryotic cells. In response to stimuli, this nuclear protein is actively secreted in to the extracellular compartment or passively released by the necrotic cells, in order to mediate inflammatory responses, by forming complexes with IL-1α, IL-1β, LPS and other moieties, and binding to RAGE, TLR and other receptor ligands, initiating downstream, signaling processes. This molecule acts as a proinflammatory cytokine and contributes to the progression of diseases like, acute lung injury, autoimmune liver damage, graft rejection immune response and arthritis. Small concentrations of HMGB1 are released during apoptosis, which facilitates oxidative regulation on Cys106, and propagates immune inactivating tolerogenic signals in the body. The review portrays the role of HMGB1 in rheumatoid arthritis, evidently supported by pre-clinical and clinical investigations, demonstrating extensive HMGB1 expression in synovial tissue and fluid as well as serum, excessive expression of transduction receptor signaling molecules, bone remodeling and uncontrolled expression of bone destroying osteoclastogenesis, resulting in destruction of articular cartilage, bone deformation and synovial proliferation, alleviating the pathogenesis in RA disease. Moreover, the review highlights the therapeutic regime targeting HMGB1, facilitating inhibition of its actions and release into the extracellular compartment, to ameliorate the destructive events that prevail in rheumatoid arthritis.
Collapse
Affiliation(s)
- Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine of Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, Distt. Shimla, Himachal Pradesh, India
| | - Dhruv Setia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey
| | - Lotfi Aleya
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey; Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
20
|
Cai S, Chandraker A. Cell Therapy in Solid Organ Transplantation. Curr Gene Ther 2020; 19:71-80. [PMID: 31161989 DOI: 10.2174/1566523219666190603103840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/30/2019] [Accepted: 05/23/2019] [Indexed: 12/28/2022]
Abstract
Transplantation is the only cure for end-stage organ failure. Current immunosuppressive drugs have two major limitations: 1) non antigen specificity, which increases the risk of cancer and infection diseases, and 2) chronic toxicity. Cell therapy appears to be an innovative and promising strategy to minimize the use of immunosuppression in transplantation and to improve long-term graft survival. Preclinical studies have shown efficacy and safety of using various suppressor cells, such as regulatory T cells, regulatory B cells and tolerogenic dendritic cells. Recent clinical trials using cellbased therapies in solid organ transplantation also hold out the promise of improving efficacy. In this review, we will briefly go over the rejection process, current immunosuppressive drugs, and the potential therapeutic use of regulatory cells in transplantation.
Collapse
Affiliation(s)
- Songjie Cai
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, United States
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
21
|
Wang S, Cai S, Zhang W, Liu X, Li Y, Zhang C, Zeng Y, Xu M, Rong R, Yang T, Shi B, Chandraker A, Yang C, Zhu T. High-mobility group box 1 protein antagonizes the immunosuppressive capacity and therapeutic effect of mesenchymal stem cells in acute kidney injury. J Transl Med 2020; 18:175. [PMID: 32312307 PMCID: PMC7169035 DOI: 10.1186/s12967-020-02334-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022] Open
Abstract
Background Kidney ischemia reperfusion injury (IRI) is a common cause of acute kidney injury and an unavoidable consequence of kidney transplantation and still lacks specific therapeutics. Recently, mesenchymal stem cell (MSC) has been emerging as a promising cell-based therapy for IRI in the context of transplantation. MSC negatively regulates the secretion of pro-inflammatory as well as the activation of immune cells during IRI through its unique immunosuppressive property. Methods We employed mice kidney IRI model and MSC cell line to monitor the IRI related checkpoints. siRNAs were utilized to knock down the potential key factors for mechanistic analysis. Statistical analysis was performed by using one-way ANOVA with Tukey’s post hoc procedure by SPSS. Results The expression of high-mobility group box 1 protein (HMGB1) is increased in the acute phase as well as the recovery stage of IRI. Importantly, the HMGB1 upregulation is correlated with the injury severity. HMGB1 diminishes the MSC induced immunosuppressive capacity in the presence of pro-inflammatory cytokines in vitro. Toll like receptor 4 (TLR4)-mediated inducible nitric oxide synthase (iNOS) inhibition contributes to the negative effect of HMGB1 on MSCs. HMGB1-TLR4 signaling inhibition augments the therapeutic efficacy of MSCs in mice renal IRI model. Conclusions These findings demonstrate that HMGB1 plays a crucial role in shaping the immunoregulatory property of MSCs within the microenvironments, providing novel insights into the crosstalk between MSCs and microenvironment components, suggesting HMGB1 signals as a promising target to improve MSC-based therapy.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Songjie Cai
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, LRMC 301, Boston, MA, 02115, USA
| | - Weitao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Xigao Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yan Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Chao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Yigang Zeng
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China
| | - Ruiming Rong
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China.,Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Anil Chandraker
- Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Ave, LRMC 301, Boston, MA, 02115, USA.
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China. .,Fudan Zhangjiang Institute, Shanghai, 201203, China.
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
22
|
Yang H, Wang H, Andersson U. Targeting Inflammation Driven by HMGB1. Front Immunol 2020; 11:484. [PMID: 32265930 PMCID: PMC7099994 DOI: 10.3389/fimmu.2020.00484] [Citation(s) in RCA: 405] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/02/2020] [Indexed: 12/22/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a highly conserved, nuclear protein present in all cell types. It is a multi-facet protein exerting functions both inside and outside of cells. Extracellular HMGB1 has been extensively studied for its prototypical alarmin functions activating innate immunity, after being actively released from cells or passively released upon cell death. TLR4 and RAGE operate as the main HMGB1 receptors. Disulfide HMGB1 activates the TLR4 complex by binding to MD-2. The binding site is separate from that of LPS and it is now feasible to specifically interrupt HMGB1/TLR4 activation without compromising protective LPS/TLR4-dependent functions. Another important therapeutic strategy is established on the administration of HMGB1 antagonists precluding RAGE-mediated endocytosis of HMGB1 and HMGB1-bound molecules capable of activating intracellular cognate receptors. Here we summarize the role of HMGB1 in inflammation, with a focus on recent findings on its mission as a damage-associated molecular pattern molecule and as a therapeutic target in inflammatory diseases. Recently generated HMGB1-specific inhibitors for treatment of inflammatory conditions are discussed.
Collapse
Affiliation(s)
- Huan Yang
- Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Haichao Wang
- Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
23
|
Ochando J, Fayad ZA, Madsen JC, Netea MG, Mulder WJM. Trained immunity in organ transplantation. Am J Transplant 2020; 20:10-18. [PMID: 31561273 PMCID: PMC6940521 DOI: 10.1111/ajt.15620] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 09/11/2019] [Accepted: 09/15/2019] [Indexed: 01/25/2023]
Abstract
Consistent induction of donor-specific unresponsiveness in the absence of continuous immunosuppressive therapy and toxic effects remains a difficult task in clinical organ transplantation. Transplant immunologists have developed numerous experimental treatments that target antigen-presentation (signal 1), costimulation (signal 2), and cytokine production (signal 3) to establish transplantation tolerance. While promising results have been obtained using therapeutic approaches that predominantly target the adaptive immune response, the long-term graft survival rates remain suboptimal. This suggests the existence of unrecognized allograft rejection mechanisms that contribute to organ failure. We postulate that trained immunity stimulatory pathways are critical to the immune response that mediates graft loss. Trained immunity is a recently discovered functional program of the innate immune system, which is characterized by nonpermanent epigenetic and metabolic reprogramming of macrophages. Since trained macrophages upregulate costimulatory molecules (signal 2) and produce pro-inflammatory cytokines (signal 3), they contribute to potent graft reactive immune responses and organ transplant rejection. In this review, we summarize the detrimental effects of trained immunity in the context of organ transplantation and describe pathways that induce macrophage training associated with graft rejection.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew York,Transplant Immunology UnitNational Center of MicrobiologyInstituto de Salud Carlos IIIMadridSpain
| | - Zahi A. Fayad
- Department of RadiologyTranslational and Molecular Imaging InstituteIcahn School of Medicine at Mount SinaiNew YorkNew York
| | - Joren C. Madsen
- Center for Transplantation Sciences and Division of Cardiac SurgeryDepartment of SurgeryMassachusetts General HospitalBostonMassachusetts
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud University Medical CenterNijmegenThe Netherlands,Department for Genomics & ImmunoregulationLife and Medical Sciences Institute (LIMES)University of BonnBonnGermany
| | - Willem J. M. Mulder
- Department of Oncological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew York,Department of RadiologyTranslational and Molecular Imaging InstituteIcahn School of Medicine at Mount SinaiNew YorkNew York,Laboratory of Chemical BiologyDepartment of Biomedical EngineeringInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhovenThe Netherlands
| |
Collapse
|
24
|
Xiong A, Wang J, Mao XL, Jiang Y, Fan Y. MiR‐199a‐3p modulates the function of dendritic cells involved in transplantation tolerance by targeting CD86. HLA 2019; 94:493-503. [PMID: 31448543 DOI: 10.1111/tan.13677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 08/06/2019] [Accepted: 08/23/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Ali Xiong
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Jing Wang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Xiao Li Mao
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yi Jiang
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| | - Yue Fan
- Department of Medical Laboratory, The Central Hospital of WuhanTongJi Medical College, Huazhong University of Science and Technology WuHan China
| |
Collapse
|
25
|
Gan I, Jiang J, Lian D, Huang X, Fuhrmann B, Liu W, Haig A, Jevnikar AM, Zhang ZX. Mitochondrial permeability regulates cardiac endothelial cell necroptosis and cardiac allograft rejection. Am J Transplant 2019; 19:686-698. [PMID: 30203531 DOI: 10.1111/ajt.15112] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/17/2018] [Accepted: 09/05/2018] [Indexed: 01/25/2023]
Abstract
Transplantation is invariably associated with programmed cell death including apoptosis and necrosis, resulting in delayed graft function and organ rejection. We have demonstrated the contribution of necroptosis to mouse microvascular endothelial cell (MVEC) death and transplant rejection. Organ injury results in the opening of mitochondrial permeability transition pores (mPTPs), which can trigger apoptotic molecules release that ultimately results in cell death. The effect of mPTPs in the necroptotic pathway remains controversial; importantly, their role in transplant rejection is not clear. In this study, tumor necrosis factor-α triggered MVECs to undergo receptor-interacting protein kinase family (RIPK1/3)-dependent necroptosis. Interestingly, inhibition of mPTP opening could also inhibit necroptotic cell death. Cyclophilin-D (Cyp-D) is a key regulator of the mPTPs. Both inhibition and deficiency of Cyp-D protected MVECs from necroptosis (n = 3, P < .00001). Additionally, inhibition of Cyp-D attenuated RIPK3-downstream mixed-lineage kinase domain-like protein phosphorylation. In vivo, Cyp-D-deficient cardiac grafts showed prolonged survival in allogeneic BALB/c mice posttransplant compared with wild-type grafts (n = 7, P < .0001). Our study results suggest that the mPTPs may be important mechanistic mediators of necroptosis in cardiac grafts. There is therapeutic potential in targeting cell death via inhibition of the mPTP-regulating molecule Cyp-D to prevent cardiac graft rejection.
Collapse
Affiliation(s)
- Ingrid Gan
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
| | - Dameng Lian
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
| | - Benjamin Fuhrmann
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
- Department of Microbiology and Immunology, Western University, London, Canada
| | - Winnie Liu
- Department of Pathology and Laboratory Medicine, Western University, London, Canada
| | - Aaron Haig
- Department of Pathology and Laboratory Medicine, Western University, London, Canada
| | - Anthony M Jevnikar
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Canada
- Department of Microbiology and Immunology, Western University, London, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Canada
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, London, Canada
- Multi-Organ Transplantation Program, London Health Sciences Centre, London, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, Canada
- Division of Nephrology, Department of Medicine, Western University, London, Canada
| |
Collapse
|
26
|
Anti-Depressive Effectiveness of Baicalin In Vitro and In Vivo. Molecules 2019; 24:molecules24020326. [PMID: 30658416 PMCID: PMC6359445 DOI: 10.3390/molecules24020326] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/11/2019] [Accepted: 01/13/2019] [Indexed: 12/18/2022] Open
Abstract
Baicalin (BA), a major polyphenol compound isolated from the extracts of Scutellaria radix, has been previously reported to ameliorate depressive-like behaviors in mice with chronic unpredictable mild stress (CUMS). However, its underlying antidepressant mechanisms remain unclear. This study was designed to confirm the antidepressant-like effects of BA on CUMS induced behavioral abnormalities in mice, and sought to explore the pharmacological mechanisms in vivo and in vitro. The CUMS procedure was carried out to induce depression in mice. Afterwards, the tail suspension test (TST), forced swim test (FST), and open field test (OFT) were performed within 24 h, then sucrose preference test (SPT) was conducted. Additionally, PC12 cells were pretreated with BA for 2 h, then further stimulated with corticosterone for 24 h. The levels of Interleukin-1β (IL-1β), IL-6 and Tumor Necrosis Factor-α (TNF-α) in serum, hippocampus homogenate and cell culture medium were determined using the enzyme-linked immunosorbent assay (ELISA) method. The protein expressions of inhibition of high mobility group box 1 protein (HMGB1)/Toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) pathways in hippocampus and PC12 cells were detected. Our results showed that CUMS-treated mice presented notable depressive-like symptoms, such as decreased sucrose consumption, increased FST and TST immobility time. While BA (25, 50 mg/kg) significantly attenuated these changes. Besides, BA treatment considerably inhibited inflammatory cytokinesl (IL-1β, IL-6, TNF-α) levels in serum, hippocampus homogenate and cell culture medium. Western blot analysis indicated that BA inhibited the expressions of HMGB1, TLR4, and p-NF-κBp65 both in vivo and in vitro. In conclusion, the present study confirmed that BA possessed efficient antidepressant effects on depression, which was possibly related to the inhibition of HMGB1/TLR4/NF-κB pathways.
Collapse
|
27
|
Koprivica I, Vujičić M, Gajić D, Saksida T, Stojanović I. Ethyl Pyruvate Stimulates Regulatory T Cells and Ameliorates Type 1 Diabetes Development in Mice. Front Immunol 2019; 9:3130. [PMID: 30687329 PMCID: PMC6335294 DOI: 10.3389/fimmu.2018.03130] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/18/2018] [Indexed: 12/31/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which a strong inflammatory response causes the death of insulin-producing pancreatic β-cells, while inefficient regulatory mechanisms allow that response to become chronic. Ethyl pyruvate (EP), a stable pyruvate derivate and certified inhibitor of an alarmin-high mobility group box 1 (HMGB1), exerts anti-oxidant and anti-inflammatory properties in animal models of rheumatoid arthritis and encephalomyelitis. To test its therapeutic potential in T1D, EP was administered intraperitoneally to C57BL/6 mice with multiple low-dose streptozotocin (MLDS)-induced T1D. EP treatment decreased T1D incidence, reduced the infiltration of cells into the pancreatic islets and preserved β-cell function. Apart from reducing HMGB1 expression, EP treatment successfully interfered with the inflammatory response within the local pancreatic lymph nodes and in the pancreas. Its effect was restricted to boosting the regulatory arm of the immune response through up-regulation of tolerogenic dendritic cells (CD11c+CD11b-CD103+) within the pancreatic infiltrates and through the enhancement of regulatory T cell (Treg) levels (CD4+CD25highFoxP3+). These EP-stimulated Treg displayed enhanced suppressive capacity reflected in increased levels of CTLA-4, secreted TGF-β, and IL-10 and in the more efficient inhibition of effector T cell proliferation compared to Treg from diabetic animals. Higher levels of Treg were a result of increased differentiation and proliferation (Ki67+ cells), but also of the heightened potency for migration due to increased expression of adhesion molecules (CD11a and CD62L) and CXCR3 chemokine receptor. Treg isolated from EP-treated mice had the activated phenotype and T-bet expression more frequently, suggesting that they readily suppressed IFN-γ-producing cells. The effect of EP on Treg was also reproduced in vitro. Overall, our results show that EP treatment reduced T1D incidence in C57BL/6 mice predominantly by enhancing Treg differentiation, proliferation, their suppressive capacity, and recruitment into the pancreas.
Collapse
Affiliation(s)
- Ivan Koprivica
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Milica Vujičić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Dragica Gajić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Tamara Saksida
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
28
|
Braza MS, van Leent MMT, Lameijer M, Sanchez-Gaytan BL, Arts RJW, Pérez-Medina C, Conde P, Garcia MR, Gonzalez-Perez M, Brahmachary M, Fay F, Kluza E, Kossatz S, Dress RJ, Salem F, Rialdi A, Reiner T, Boros P, Strijkers GJ, Calcagno CC, Ginhoux F, Marazzi I, Lutgens E, Nicolaes GAF, Weber C, Swirski FK, Nahrendorf M, Fisher EA, Duivenvoorden R, Fayad ZA, Netea MG, Mulder WJM, Ochando J. Inhibiting Inflammation with Myeloid Cell-Specific Nanobiologics Promotes Organ Transplant Acceptance. Immunity 2018; 49:819-828.e6. [PMID: 30413362 PMCID: PMC6251711 DOI: 10.1016/j.immuni.2018.09.008] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/03/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022]
Abstract
Inducing graft acceptance without chronic immunosuppression remains an elusive goal in organ transplantation. Using an experimental transplantation mouse model, we demonstrate that local macrophage activation through dectin-1 and toll-like receptor 4 (TLR4) drives trained immunity-associated cytokine production during allograft rejection. We conducted nanoimmunotherapeutic studies and found that a short-term mTOR-specific high-density lipoprotein (HDL) nanobiologic treatment (mTORi-HDL) averted macrophage aerobic glycolysis and the epigenetic modifications underlying inflammatory cytokine production. The resulting regulatory macrophages prevented alloreactive CD8+ T cell-mediated immunity and promoted tolerogenic CD4+ regulatory T (Treg) cell expansion. To enhance therapeutic efficacy, we complemented the mTORi-HDL treatment with a CD40-TRAF6-specific nanobiologic (TRAF6i-HDL) that inhibits co-stimulation. This synergistic nanoimmunotherapy resulted in indefinite allograft survival. Together, we show that HDL-based nanoimmunotherapy can be employed to control macrophage function in vivo. Our strategy, focused on preventing inflammatory innate immune responses, provides a framework for developing targeted therapies that promote immunological tolerance.
Collapse
Affiliation(s)
- Mounia S Braza
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mandy M T van Leent
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marnix Lameijer
- Department of Medical Biochemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Brenda L Sanchez-Gaytan
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rob J W Arts
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patricia Conde
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Mercedes R Garcia
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Gonzalez-Perez
- Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Manisha Brahmachary
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francois Fay
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ewelina Kluza
- Department of Medical Biochemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Susanne Kossatz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Regine J Dress
- Singapore Immunology Network (SIgN), A STAR, Singapore, Singapore
| | - Fadi Salem
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Rialdi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - Peter Boros
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustav J Strijkers
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Biomedical Engineering and Physics, Academic Medical Center, Amsterdam, the Netherlands
| | - Claudia C Calcagno
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A STAR, Singapore, Singapore
| | - Ivan Marazzi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther Lutgens
- Department of Medical Biochemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention, Ludwig-Maximilians University Munich, Munich, Germany
| | - Gerry A F Nicolaes
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Christian Weber
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Filip K Swirski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward A Fisher
- Department of Medicine (Cardiology), New York University School of Medicine, New York, NY, USA
| | - Raphaël Duivenvoorden
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Nephrology, Academic Medical Center, Amsterdam, the Netherlands; Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medical Biochemistry, Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
29
|
Chen X, Ma J, Kwan T, Stribos EGD, Messchendorp AL, Loh YW, Wang X, Paul M, Cunningham EC, Habib M, Alexander IE, Sharland AF, Chadban SJ, Wu H. Blockade of HMGB1 Attenuates Diabetic Nephropathy in Mice. Sci Rep 2018; 8:8319. [PMID: 29844451 PMCID: PMC5974129 DOI: 10.1038/s41598-018-26637-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/08/2018] [Indexed: 12/24/2022] Open
Abstract
Activation of TLR2 or TLR4 by endogenous ligands such as high mobility group box 1 (HMGB1) may mediate inflammation causing diabetic kidney injury. We determined whether blockade of HMGB1 signaling by: (1) supra-physiological production of endogenous secretory Receptor for Advanced Glycation End-products (esRAGE), a receptor for HMGB1; (2) administration of HMGB1 A Box, a specific competitive antagonist, would inhibit development of streptozotocin induced diabetic nephropathy (DN). Wild-type diabetic mice developed albuminuria, glomerular injuries, interstitial fibrosis and renal inflammation. Using an adeno-associated virus vector, systemic over-expression of esRAGE afforded significant protection from all parameters. No protection was achieved by a control vector which expressed human serum albumin. Administration of A Box was similarly protective against development of DN. To determine the mechanism(s) of protection, we found that whilst deficiency of TLR2, TLR4 or RAGE afforded partial protection from development of DN, over-expression of esRAGE provided additional protection in TLR2−/−, modest protection against podocyte damage only in TLR4−/− and no protection in RAGE−/− diabetic mice, suggesting the protection provided by esRAGE was primarily through interruption of RAGE and TLR4 pathways. We conclude that strategies to block the interaction between HMGB1 and its receptors may be effective in preventing the development of DN.
Collapse
Affiliation(s)
- Xiaochen Chen
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Jin Ma
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Tony Kwan
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Elisabeth G D Stribos
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - A Lianne Messchendorp
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Yik W Loh
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Xiaoyu Wang
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | - Moumita Paul
- Transplantation Immunobiology Group, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Eithne C Cunningham
- Transplantation Immunobiology Group, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Miriam Habib
- Transplantation Immunobiology Group, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute and The Children's Hospital at Westmead, Westmead, NSW, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Westmead, NSW, Australia
| | - Alexandra F Sharland
- Transplantation Immunobiology Group, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Steven J Chadban
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia.,Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Huiling Wu
- Kidney Node Laboratory, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia. .,Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
30
|
Lv Q, Li C, Mo Y, He L. The role of HMGB1 in heart transplantation. Immunol Lett 2018; 194:1-3. [DOI: 10.1016/j.imlet.2017.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 01/15/2023]
|
31
|
Lee S, Piao C, Kim G, Kim JY, Choi E, Lee M. Production and application of HMGB1 derived recombinant RAGE-antagonist peptide for anti-inflammatory therapy in acute lung injury. Eur J Pharm Sci 2017; 114:275-284. [PMID: 29292016 DOI: 10.1016/j.ejps.2017.12.019] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 01/10/2023]
Abstract
Acute lung injury (ALI) is an inflammatory lung disease caused by sepsis, infection, or ischemia-reperfusion. The receptor for advanced glycation end-products (RAGE) signaling pathway plays an important role in ALI. In this study, a novel RAGE-antagonist peptide (RAP) was produced as an inhibitor of the RAGE signaling pathway based on the RAGE-binding domain of high mobility group box-1 (HMGB1). Recombinant RAP was over-expressed and purified using nickel-affinity chromatography. In lipopolysaccharide- or HMGB1-activated RAW264.7 macrophage cells, RAP reduced the levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6). RAP decreased the levels of cell surface RAGE and inhibited the nuclear translocation of nuclear factor-κB (NF-κB). These results imply that RAP decreases RAGE-mediated NF-κB activation and subsequent inflammatory reaction. For in vivo evaluation, RAP was delivered to the lungs of ALI-model animals via intratracheal administration. As a result, RAGE was down-regulated in the lung tissues by pulmonary delivery of RAP. Consequently, TNF-α, IL-6, and IL-1β were also reduced in broncoalveolar lavage fluid and the lung tissues of RAP-treated animals. Hematoxylin and eosin staining indicated that inflammation was decreased in RAP-treated animals. Collectively, these results suggest that RAP may be a useful treatment for ALI.
Collapse
Affiliation(s)
- Seonyeong Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Chunxian Piao
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Gyeungyun Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Ji Yeon Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Eunji Choi
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea.
| |
Collapse
|
32
|
Jane-Wit D, Fang C, Goldstein DR. Innate immune mechanisms in transplant allograft vasculopathy. Curr Opin Organ Transplant 2017; 21:253-7. [PMID: 27077602 DOI: 10.1097/mot.0000000000000314] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Allograft vasculopathy is the leading cause of late allograft loss following solid organ transplantation. Ischemia reperfusion injury and donor-specific antibody-induced complement activation confer heightened risk for allograft vasculopathy via numerous innate immune mechanisms, including MyD88, high-mobility group box 1 (HMGB1), and complement-induced noncanonical nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. RECENT FINDINGS The role of MyD88, a signal adaptor downstream of the Toll-like receptors (TLR), has been defined in an experimental heart transplant model, which demonstrated that recipient MyD88 enhanced allograft vasculopathy. Importantly, triggering receptor on myeloid receptor 1, a MyD88 amplifying signal, was present in rejecting human cardiac transplant biopsies and enhanced the development of allograft vasculopathy in mice. HMGB1, a nuclear protein that activates Toll-like receptors, also enhanced the development of allograft vasculopathy. Complement activation elicits assembly of membrane attack complexes on endothelial cells which activate noncanonical NF-κB signaling, a novel complement effector pathway that induces proinflammatory genes and potentiates endothelial cell-mediated alloimmune T-cell activation, processes which enhance allograft vasculopathy. SUMMARY Innate immune mediators, including HMGB1, MyD88, and noncanonical NF-κB signaling via complement activation contribute to allograft vasculopathy. These pathways represent potential therapeutic targets to reduce allograft vasculopathy after solid organ transplantation.
Collapse
Affiliation(s)
- Dan Jane-Wit
- aDepartment of Cardiovascular Medicine bDepartment of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | | | | |
Collapse
|
33
|
Kwok C, Pavlosky A, Lian D, Jiang J, Huang X, Yin Z, Liu W, Haig A, Jevnikar AM, Zhang ZX. Necroptosis Is Involved in CD4+ T Cell-Mediated Microvascular Endothelial Cell Death and Chronic Cardiac Allograft Rejection. Transplantation 2017; 101:2026-2037. [PMID: 29633982 DOI: 10.1097/tp.0000000000001578] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Despite advances in immunosuppressive therapies, the rate of chronic transplant loss remains substantial. Organ injury involves various forms of cell death including apoptosis and necrosis. We now recognize that early injury of cardiac transplants involves a newly described form of programmed necrotic cell death, termed necroptosis. Because this involves receptor-interacting protein (RIP) kinase 1/3, this study aimed to establish the role of RIP3 in chronic cardiac allograft rejection. METHODS We used major histocompatibility complex class II mismatched C57BL/6N (H-2; B6) or B6.RIP3 (H-2; RIP3) mice to B6.C-H-2 (H2-Ab1; bm12) mouse cardiac transplantation. Microvascular endothelial cells (MVEC) were developed from B6 and RIP3 cardiac grafts. RESULT CD4 T cell-mediated cardiac graft rejection is inhibited using RIP3 deficient donor grafts, with reduced cellular infiltration and vasculopathy compared with wild type cardiac grafts. Alloreactive CD4 T cell-mediated MVEC death involves TNFα, Fas ligand (FasL) and granzyme B. Although necroptosis and release of danger molecule high-mobility group box 1 are eliminated by the absence of RIP3, CD4 T cells had attenuated MVEC death through granzyme B and FasL. CONCLUSIONS CD4 T cell-mediated MVEC death involves in TNFα, FasL and granzyme B. Necroptotic cell death and release of the danger molecule may promote inflammatory responses and transplant rejection. Although loss of RIP3 does not eliminate alloimmune responses, chronic graft injury is reduced. RIP3 is an important therapeutic target but additional granzyme and caspases inhibition is required for sufficiently improving long-term graft survival.
Collapse
Affiliation(s)
- Cecilia Kwok
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| | - Alexander Pavlosky
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| | - Dameng Lian
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Jifu Jiang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Xuyan Huang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Ziqin Yin
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
| | - Weihua Liu
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| | - Aaron Haig
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| | - Anthony M Jevnikar
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| | - Zhu-Xu Zhang
- Matthew Mailing Centre for Translational Transplantation Studies, London Health Sciences Centre, London, Ontario, Canada
- Departments of Medicine, Pathology, Immunology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
34
|
Liu L, Jiang Y, Steinle JJ. Inhibition of HMGB1 protects the retina from ischemia-reperfusion, as well as reduces insulin resistance proteins. PLoS One 2017; 12:e0178236. [PMID: 28542588 PMCID: PMC5441648 DOI: 10.1371/journal.pone.0178236] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/28/2017] [Indexed: 11/20/2022] Open
Abstract
The role of inflammation in diabetic retinal amage is well accepted. While a number of cytokines and inflammatory mediators are responsible for these changes, upstream regulators are less well studied. Additionally, the role for these upstream mediators in retinal health is unclear. In this study, we hypothesized that inhibition of high mobility group box 1 (HMGB1) could restore normal insulin signaling in retinal endothelial cells (REC) grown in high glucose, as well as protect the retina against ischemia/reperfusion (I/R)-induced retinal damage. REC were grown in normal (5mM) or high glucose (25mM) and treated with Box A or glycyrrhizin, two different HMGB1 inhibitors. Western blotting was done for HMGB1, toll-like receptor 4 (TLR4), insulin receptor, insulin receptor substrate-1 (IRS-1), and Akt. ELISA analyses were done for tumor necrosis factor alpha (TNFα) and cleaved caspase 3. In addition, C57/B6 mice were treated with glycyrrhizin, both before and after ocular I/R. Two days following I/R, retinal sections were processed for neuronal changes, while vascular damage was measured at 10 days post-I/R. Results demonstrate that both Box A and glycyrrhizin reduced HMGB1, TLR4, and TNFα levels in REC grown in high glucose. This led to reduced cleavage of caspase 3 and IRS-1Ser307 phosphorylation, and increased insulin receptor and Akt phosphorylation. Glycyrrhizin treatment significantly reduced loss of retinal thickness and degenerate capillary numbers in mice exposed to I/R. Taken together, these results suggest that inhibition of HMGB1 can reduce retinal insulin resistance, as well as protect the retina against I/R-induced damage.
Collapse
Affiliation(s)
- Li Liu
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Youde Jiang
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Jena J. Steinle
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Ophthalmology Wayne State University School of Medicine, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
35
|
Bongoni AK, Klymiuk N, Wolf E, Ayares D, Rieben R, Cowan PJ. Transgenic Expression of Human Thrombomodulin Inhibits HMGB1-Induced Porcine Aortic Endothelial Cell Activation. Transplantation 2017; 100:1871-9. [PMID: 27077599 DOI: 10.1097/tp.0000000000001188] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Transgenic expression of human thrombomodulin (hTBM), which has the potential to solve the problem of coagulation dysregulation in pig-to-primate xenotransplantation, may have additional benefits by neutralizing the proinflammatory cytokine high-mobility group box 1 (HMGB1). The aim of this study was to investigate HMGB1-mediated effects on porcine aortic endothelial cells (PAEC) from wild-type (WT) and hTBM transgenic pigs. METHODS Porcine aortic endothelial cells were treated with HMGB1, human (h)TNFα or lipopolysaccharide (LPS). Procoagulant and proinflammatory responses were assessed by measuring expression of cell surface markers (adhesion molecules, fibrinogen-like protein 2, plasminogen activator inhibitor (PAI)-1), secretion of porcine cytokines and chemokines (HMGB1, TNFα, IL-8, monocyte chemotactic protein-1), and formation of PAI-1/tissue plasminogen activator complexes. Thrombin-mediated degradation of HMGB1 in the presence of PAEC was examined by Western blot and functional assay. RESULTS High-mobility group box 1 potently activated WT PAEC, increasing the expression of E-selectin, vascular cell adhesion molecule-1, intercellular adhesion molecule-1, fibrinogen-like protein 2, and PAI-1, the secretion of TNFα, IL-8, and monocyte chemotactic protein-1 and the formation of PAI-1/tissue plasminogen activator complexes. Human TNFα- or LPS-induced activation of WT PAEC was inhibited by treatment with rabbit anti-HMGB1 antibody. Transgenic expression of hTBM significantly reduced the activation of PAEC by HMGB1 or hTNFα, and significantly enhanced thrombin-induced HMGB1 cleavage. Chemically induced shedding of the lectin-like domain of TBM resulted in significantly increased HMGB1-induced PAEC activation. CONCLUSIONS High-mobility group box 1 exerts powerful proinflammatory and procoagulant effects on WT PAEC, and appears to be an important downstream mediator for the actions of hTNFα and LPS. Human thrombomodulin transgenic PAECs are less sensitive to activation by either HMGB1 or hTNFα, an effect that appears to be dependent on the lectin-like domain of TBM.
Collapse
Affiliation(s)
- Anjan K Bongoni
- 1 Immunology Research Centre, St. Vincent's Hospital Melbourne, Victoria, Australia. 2 Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilian University, Munich, Germany. 3 Revivicor, Inc., Blacksburg, VA. 4 Department of Clinical Research, University of Bern, Bern, Switzerland. 5 Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Todd JL, Palmer SM. Danger signals in regulating the immune response to solid organ transplantation. J Clin Invest 2017; 127:2464-2472. [PMID: 28530643 DOI: 10.1172/jci90594] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Endogenous danger signals, or damage-associated molecular patterns (DAMPs), are generated in response to cell stress and activate innate immunity to provide a pivotal mechanism by which an organism can respond to damaged self. Accumulating experimental and clinical data have established the importance of DAMPs, which signal through innate pattern recognition receptors (PRRs) or DAMP-specific receptors, in regulating the alloresponse to solid organ transplantation (SOT). Moreover, DAMPs may incite distinct downstream cellular responses that could specifically contribute to the development of allograft fibrosis and chronic graft dysfunction. A growing understanding of the role of DAMPs in directing the immune response to transplantation has suggested novel avenues for the treatment or prevention of allograft rejection that complement contemporary immunosuppression and could lead to improved outcomes for solid organ recipients.
Collapse
Affiliation(s)
- Jamie L Todd
- Duke University Medical Center, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Durham, North Carolina, USA.,Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Scott M Palmer
- Duke University Medical Center, Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Durham, North Carolina, USA.,Duke Clinical Research Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
37
|
Hsieh JL, Shen PC, Wu PT, Jou IM, Wu CL, Shiau AL, Wang CR, Chong HE, Chuang SH, Peng JS, Chen SY. Knockdown of toll-like receptor 4 signaling pathways ameliorate bone graft rejection in a mouse model of allograft transplantation. Sci Rep 2017; 7:46050. [PMID: 28393847 PMCID: PMC5385519 DOI: 10.1038/srep46050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 03/10/2017] [Indexed: 11/21/2022] Open
Abstract
Non-union occurring in structural bone grafting is a major problem in allograft transplantation because of impaired interaction between the host and graft tissue. Activated toll-like receptor (TLR) induces inflammatory cytokines and chemokines and triggers cell-mediated immune responses. The TLR-mediated signal pathway is important for mediating allograft rejection. We evaluated the effects of local knockdown of the TLR4 signaling pathway in a mouse segmental femoral graft model. Allografts were coated with freeze-dried lentiviral vectors that encoded TLR4 and myeloid differentiation primary response gene 88 (MyD88) short-hairpin RNA (shRNA), which were individually transplanted into the mice. They were assessed morphologically, radiographically, and histologically for tissue remodeling. Union occurred in autografted but not in allografted mice at the graft and host junctions after 4 weeks. TLR4 and MyD88 expression was up-regulated in allografted mice. TLR4 and MyD88 shRNAs inhibited TLR4 and MyD88 expression, which led to better union in the grafted sites. More regulatory T-cells in the draining lymph nodes suggested inflammation suppression. Local inhibition of TLR4 and MyD88 might reduce immune responses and ameliorate allograft rejection.
Collapse
Affiliation(s)
- Jeng-Long Hsieh
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Po-Chuan Shen
- Department of Orthopedics, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Po-Ting Wu
- Department of Orthopedics, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - I-Ming Jou
- Department of Orthopedics, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Liang Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ai-Li Shiau
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chrong-Reen Wang
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Earn Chong
- Department of Medical Laboratory Science and Biotechnology, Chung Hwa University of Medical Technology, Tainan, Taiwan
| | - Shu-Han Chuang
- Department of Pharmacology, Institute for Drug Evaluation Platform, Development Center for Biotechnology, Taipei, Taiwan
| | - Jia-Shiou Peng
- Department of Nursing, Chung-Jen Junior College of Nursing, Health Sciences and Management, Chiayi, Taiwan
| | - Shih-Yao Chen
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
38
|
Land WG, Agostinis P, Gasser S, Garg AD, Linkermann A. DAMP-Induced Allograft and Tumor Rejection: The Circle Is Closing. Am J Transplant 2016; 16:3322-3337. [PMID: 27529775 DOI: 10.1111/ajt.14012] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/28/2016] [Accepted: 07/31/2016] [Indexed: 01/25/2023]
Abstract
The pathophysiological importance of the immunogenicity of damage-associated molecular patterns (DAMPs) has been pinpointed by their identification as triggers of allograft rejection following release from dying cells, such as after ischemia-reperfusion injury. In cancers, however, this strong trigger of a specific immune response gives rise to the success of cancer immunotherapy. Here, we review the recently literature on the pathophysiological importance of DAMP release and discuss the implications of these processes for allograft rejection and cancer immunotherapy, revealing a striking mechanistic overlap. We conclude that these two fields share a common mechanistic basis of regulated necrosis and inflammation, the molecular characterization of which may be helpful for both oncologists and the transplant community.
Collapse
Affiliation(s)
- W G Land
- German Academy of Transplantation Medicine, Munich, Germany.,Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, Plateforme GENOMAX, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,LabexTRANSPLANTEX, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - P Agostinis
- Cell Death Research and Therapy (CDRT) Lab, Department of Cellular and Molecular Medicine, KU Leuven, University of Leuven, Leuven, Belgium
| | - S Gasser
- Immunology Programme and Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - A D Garg
- Cell Death Research and Therapy (CDRT) Lab, Department of Cellular and Molecular Medicine, KU Leuven, University of Leuven, Leuven, Belgium
| | - A Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.,Cluster of Excellence EXC306, Inflammation at Interfaces, Schleswig-Holstein, Germany.,Clinic for Nephrology and Hypertension, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
39
|
Ischémie–reperfusion. Liquides de conservation et machines de perfusion en transplantation rénale. Prog Urol 2016; 26:964-976. [DOI: 10.1016/j.purol.2016.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/20/2016] [Accepted: 08/22/2016] [Indexed: 12/12/2022]
|
40
|
Ming B, Gao M, Zou H, Chen H, Sun Y, Xiao Y, Lai L, Xiong P, Xu Y, Tan Z, Wang J, Chen G, Gong F, Xia J, Zheng F. HMGB1 blockade differentially impacts pulmonary inflammation and defense responses in poly(I:C)/LPS-exposed heart transplant mice. Mol Immunol 2016; 76:80-9. [PMID: 27387278 DOI: 10.1016/j.molimm.2016.06.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/09/2016] [Accepted: 06/17/2016] [Indexed: 01/05/2023]
Abstract
A large number of recipients are in a compromised immune defense condition because of the routine application of immunosuppressive regimens after heart transplantation. Our previous work demonstrated that blockade of high-mobility group box 1 (HMGB1) prolongs the graft survival. Whether and how HMGB1 blockade impacts respiratory responses against pathogen-like challenge in organ transplant recipients when it improves cardiac graft are not elucidated. At the present study, after abdominal heterotopic heart transplantation, the recipient mice were treated with HMGB1 mAb, and then challenged with poly(I:C) or LPS intratracheally on day 7 post transplantation. We found that the level of bronchoalveolar lavage (BAL) HMGB1 was elevated after heart transplantation, and aggravated responses to respiratory tract poly(I:C)/LPS challenge were observed. HMGB1 neutralizing mAb treatment in poly(I:C)-challenged recipient mice alleviated pulmonary histopathological changes, neutrophil infiltration and inflammatory cytokine release, but unaffected the level of IFN-β, the distribution of CD11b(+)CD27(+)/CD11b(+)CD27(-) NK cell subsets, and CD8(+) T cell responses. In LPS-exposed recipient mice, HMGB1 mAb treatment ameliorated pulmonary inflammatory damage and enhanced the phagocytosis of phagocytic cells. Thus, this study may establish a basis for the application of HMGB1 blockade to improve the outcomes of heart transplant recipients because HMGB1 inhibition ameliorates pulmonary inflammation, but maintains defense-associated responses.
Collapse
Affiliation(s)
- Bingxia Ming
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Gao
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijuan Zou
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huoying Chen
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Sun
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Xiao
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lai
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Xiong
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Xu
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Tan
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - Feili Gong
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Cardiovascular Surgery, Central Hospital of Wuhan, Wuhan, China
| | - Fang Zheng
- Department of immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; Key Laboratory of Organ Transplantation, Ministry of Public Health, China.
| |
Collapse
|
41
|
Braza F, Brouard S, Chadban S, Goldstein DR. Role of TLRs and DAMPs in allograft inflammation and transplant outcomes. Nat Rev Nephrol 2016; 12:281-90. [PMID: 27026348 DOI: 10.1038/nrneph.2016.41] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Graft inflammation impairs the induction of solid organ transplant tolerance and enhances acute and chronic rejection. Elucidating the mechanisms by which inflammation is induced after organ transplantation could lead to novel therapeutics to improve transplant outcomes. In this Review we describe endogenous substances--damage-associated molecular patterns (DAMPs)--that are released after allograft reperfusion and induce inflammation. We also describe innate immune signalling pathways that are activated after solid organ transplantation, with a focus on Toll-like receptors (TLRs) and their signal adaptor, MYD88. Experimental and clinical studies have yielded a large body of evidence that TLRs and MYD88 are instrumental in initiating allograft inflammation and promoting the development of acute and chronic rejection. Ongoing clinical studies are testing TLR inhibition strategies in solid organ transplantation, although avoiding compromising host defence to pathogens is a key challenge. Further elucidation of the mechanisms by which sterile inflammation is induced, maintained and amplified within the allograft has the potential to lead to novel anti-inflammatory treatments that could improve outcomes for solid organ transplant recipients.
Collapse
Affiliation(s)
- Faouzi Braza
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande, 2780-156 Oeiras, Portugal
| | - Sophie Brouard
- INSERM, UMR 1064, CHU de Nantes, ITUN, 30 Bd Jean Monnet Nantes F-44093, France
| | - Steve Chadban
- Renal Medicine and Transplantation, Royal Prince Alfred Hospital, Missenden Road Camperdown, NSW 2050, Sydney, Australia.,Kidney Node, Charles Perkins Centre, University of Sydney, Missenden Road, Camperdown, NSW 2093, Australia
| | - Daniel R Goldstein
- Department of Internal Medicine, 333 Cedar St, Yale School of Medicine, New Haven, Connecticut 06525, USA.,Department of Immunobiology, 300 Cedar St, Yale School of Medicine, New Haven, Connecticut 06525, USA
| |
Collapse
|
42
|
Park HS, Kim EN, Kim MY, Lim JH, Kim HW, Park CW, Yang CW, Jin DC, Choi BS. The protective effect of neutralizing high-mobility group box1 against chronic cyclosporine nephrotoxicity in mice. Transpl Immunol 2015; 34:42-9. [PMID: 26603313 DOI: 10.1016/j.trim.2015.11.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 11/11/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND High-mobility group box1 (HMGB1) is known to be involved in innate immune response through interaction with receptor for advanced glycation end products (RAGE) and toll-like receptors (TLRs), besides its proper role within the nucleus. Immunological pathways, including TLR signaling, are also involved in chronic cyclosporine (CsA) nephrotoxicity. This study was designed to determine whether neutralizing HMGB1 prevents chronic CsA nephrotoxicity. METHODS Chronic CsA nephrotoxicity was induced by CsA subcutaneous injection daily for 4weeks under salt-depletion in mice. Anti-HMGB1 neutralizing antibody for HMGB1 blockade (600mcg/mouse) was administered weekly to mice in the anti-HMGB1 treatment group. The effects of HMGB1 neutralization were evaluated in terms of renal function as well as histological and immunopathological examination. RESULTS Anti-HMGB1 administration prevented the increases in serum creatinine and 24h albuminuria and the decrease in creatinine clearance associated with CsA treatment. Increased tubulointerstitial fibrosis and transforming growth factor (TGF)-β immunohistochemical staining associated with CsA treatment were also prevented by anti-HMGB1 administration. Anti-HMGB1 administration prevented the activation of the TLR4 signaling pathway, which resulted in the reduction of nuclear factor kappa B (NF-κB) expression. In cultured tubular cells, anti-HMGB1 pretreatment also prevented the increases in fibronectin and collagen IV levels associated with CsA treatment. CONCLUSIONS Neutralizing HMGB1 with an anti-HMGB1 antibody ameliorated chronic CsA nephrotoxicity via inhibition of the TLR4 signaling pathway. Our study suggests that HMGB1 blockade can be beneficial for increasing allograft survival in renal transplant recipients by protecting against calcineurin inhibitor-induced nephrotoxicity.
Collapse
Affiliation(s)
- Hoon Suk Park
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Nim Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Young Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hee Lim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung Wook Kim
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chul Woo Yang
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Chan Jin
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Bum Soon Choi
- Division of Nephrology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
43
|
GE WENSONG, FAN JIANGAO, CHEN YINGWEI, XU LEIMING. Expression and purification of functional HMGB1 A box by fusion with SUMO. Mol Med Rep 2015; 12:6527-6532. [PMID: 26352592 PMCID: PMC4626187 DOI: 10.3892/mmr.2015.4308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/17/2015] [Indexed: 01/14/2023] Open
Abstract
High‑mobility‑group‑box chromosomal protein 1 (HMGB1) is a ubiquitous and abundant nuclear protein in eukaryotic cells. Nuclear HMGB1 serves an important role in maintaining nuclear stability under stress. However, extracellular HMGB1 exerts actions, which are distinctly different compared with these intracellular functions. HMGB1, when released extracellularly, is a potent innate signal, which initiates host defense mechanisms or tissue regeneration. HMGB1 has two DNA‑binding domains: HMG A box and B box. The HMGB1 A box exhibits an antagonistic, anti‑inflammatory effect, and is a potential therapeutic target, however, the large‑scale expression and purification of the HMGB1 A box with high efficiency remains to be reported. In the present study, a SUMO‑fusion expression system was used to express and purify high levels of functional HMGB1 A box to meet the requirements of therapeutic protein production.
Collapse
Affiliation(s)
- WEN-SONG GE
- Department of Gastroenterology, Shanghai Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - JIAN-GAO FAN
- Department of Gastroenterology, Shanghai Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - YING-WEI CHEN
- Department of Gastroenterology, Shanghai Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - LEI-MING XU
- Department of Gastroenterology, Shanghai Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
44
|
Li JH, Zhao B, Zhu XH, Wang L, Zou HJ, Chen S, Guo H, Ruan YL, Zheng F, Xiang Y, Ming CS, Gong FL, Chen G. Blockade of Extracellular HMGB1 Suppresses Xenoreactive B Cell Responses and Delays Acute Vascular Xenogeneic Rejection. Am J Transplant 2015; 15:2062-74. [PMID: 25943147 DOI: 10.1111/ajt.13275] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 02/16/2015] [Accepted: 02/17/2015] [Indexed: 01/25/2023]
Abstract
Blockade of extracellular high mobility group box 1 (HMGB1) can significantly prolong murine cardiac allograft survival. Here, we determined the role of HMGB1 in xenotransplantation. Sprague-Dawley rat hearts were transplanted heterotopically into BALB/c mice. Xenografts without any treatment developed predominant acute vascular rejection within 6 days. Both passively released HMGB1 from xenografts and actively secreted HMGB1 from infiltrated immune cells were significantly increased after xenotransplantation. HMGB1-neutralizing antibody treatment significantly prolonged xenograft survival and attenuated pathologic damage, immune cell infiltration, and HMGB1 expression and release in the xenografts. Compared to control IgG treatment evaluated at study endpoint, treatment with HMGB1-neutralizing antibody markedly suppressed xenoreactive B cell responses, as evidenced by the significant inhibition of anti-rat antibody production and deposition in xenografts at Day 6 posttransplant. Furthermore, treatment with anti-HMGB1 antibody suppressed B cell activation and reduced IFN-γ and IL-17A production after xenotransplantation. These results demonstrate for the first time that HMGB1 plays an important role in mediating acute xenograft rejection. Thus, we have shown that neutralization of extracellular HMGB1 can significantly inhibit xenoreactive B cell responses and delay xenograft rejection in a rat-to-mouse model of xenotransplantation, uncovering new insights in the role of HMGB1 in transplantation.
Collapse
Affiliation(s)
- J-H Li
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - B Zhao
- Department of Thoracic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - X-H Zhu
- Department of Cardiovascular Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - L Wang
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - H-J Zou
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - S Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - H Guo
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - Y-L Ruan
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - F Zheng
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - Y Xiang
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - C-S Ming
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - F-L Gong
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| | - G Chen
- Institute of Organ Transplantation, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Organ Transplantation, Ministry of Education, China.,Key Laboratory of Organ Transplantation, Ministry of Public Health, China
| |
Collapse
|
45
|
DC-SIGN(+) Macrophages Control the Induction of Transplantation Tolerance. Immunity 2015; 42:1143-58. [PMID: 26070485 DOI: 10.1016/j.immuni.2015.05.009] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 02/06/2015] [Accepted: 04/06/2015] [Indexed: 11/23/2022]
Abstract
Tissue effector cells of the monocyte lineage can differentiate into different cell types with specific cell function depending on their environment. The phenotype, developmental requirements, and functional mechanisms of immune protective macrophages that mediate the induction of transplantation tolerance remain elusive. Here, we demonstrate that costimulatory blockade favored accumulation of DC-SIGN-expressing macrophages that inhibited CD8(+) T cell immunity and promoted CD4(+)Foxp3(+) Treg cell expansion in numbers. Mechanistically, that simultaneous DC-SIGN engagement by fucosylated ligands and TLR4 signaling was required for production of immunoregulatory IL-10 associated with prolonged allograft survival. Deletion of DC-SIGN-expressing macrophages in vivo, interfering with their CSF1-dependent development, or preventing the DC-SIGN signaling pathway abrogated tolerance. Together, the results provide new insights into the tolerogenic effects of costimulatory blockade and identify DC-SIGN(+) suppressive macrophages as crucial mediators of immunological tolerance with the concomitant therapeutic implications in the clinic.
Collapse
|
46
|
Im KI, Kim N, Lim JY, Nam YS, Lee ES, Kim EJ, Kim HJ, Kim SH, Cho SG. The Free Radical Scavenger NecroX-7 Attenuates Acute Graft-versus-Host Disease via Reciprocal Regulation of Th1/Regulatory T Cells and Inhibition of HMGB1 Release. THE JOURNAL OF IMMUNOLOGY 2015; 194:5223-32. [PMID: 25911749 DOI: 10.4049/jimmunol.1402609] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 03/27/2015] [Indexed: 01/27/2023]
Abstract
Graft-versus-host disease (GVHD) is a major complication associated with allogeneic hematopoietic stem cell transplantation. Despite the prominent role of the adaptive immune system, the importance of controlling the innate immune system in the pathogenesis of GVHD has recently been rediscovered. High-mobility group box 1 (HMGB1) is a crucial damage-associated molecular pattern signal that functions as a potent innate immune mediator in GVHD. In the present study, we investigated treatment of experimental GVHD through HMGB1 blockade using the compound cyclopentylamino carboxymethylthiazolylindole (NecroX)-7. Treated animals significantly attenuated GVHD-related mortality and inhibited severe tissue damage. These protective effects correlated with the decrease in HMGB1 expression and lower levels of reactive oxidative stress. Additionally, NecroX-7 inhibited the HMGB1-induced release of TNF and IL-6, as well as the expression of TLR-4 and receptor for advanced glycation end products. We also observed increased regulatory T cell numbers, which may be associated with regulation of differentiation signals independent of HMGB1. Taken together, these data indicate that NecroX-7 protects mice against lethal GVHD by reciprocal regulation of regulatory T/Th1 cells, attenuating systemic HMGB1 accumulation and inhibiting HMGB1-mediated inflammatory response. Our results indicate the possibility of a new use for a clinical drug that is effective for the treatment of GVHD.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Jung-Yeon Lim
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Young-Sun Nam
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Sol Lee
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Eun-Jung Kim
- Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea
| | - Hyoung Jin Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Soon Ha Kim
- Strategy and Development, LG Life Sciences Ltd., Seoul 2305-738, Korea; and
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, Catholic Research Institute of Medical Science, Catholic University of Korea, Seoul 137-701, Korea; Laboratory of Immune Regulation, Convergent Research Consortium for Immunologic Disease, Catholic University of Korea College of Medicine, St. Mary's Hospital, Seoul 137-701, Korea; Department of Hematology, Catholic Blood and Marrow Transplantation Center, Seoul St. Mary's Hospital, Catholic University of Korea College of Medicine, Seoul 137-701, Korea
| |
Collapse
|
47
|
Shen H, Heuzey E, Mori DN, Wong CK, Colangelo CM, Chung LM, Bruce C, Slizovskiy IB, Booth CJ, Kreisel D, Goldstein DR. Haptoglobin enhances cardiac transplant rejection. Circ Res 2015; 116:1670-9. [PMID: 25801896 DOI: 10.1161/circresaha.116.305406] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 03/23/2015] [Indexed: 11/16/2022]
Abstract
RATIONALE Early graft inflammation enhances both acute and chronic rejection of heart transplants, but it is unclear how this inflammation is initiated. OBJECTIVE To identify specific inflammatory modulators and determine their underlying molecular mechanisms after cardiac transplantation. METHODS AND RESULTS We used a murine heterotopic cardiac transplant model to identify inflammatory modulators of early graft inflammation. Unbiased mass spectrometric analysis of cardiac tissue before and ≤72 hours after transplantation revealed that 22 proteins including haptoglobin, a known antioxidant, are significantly upregulated in our grafts. Through the use of haptoglobin-deficient mice, we show that 80% of haptoglobin-deficient recipients treated with perioperative administration of the costimulatory blocking agent CTLA4 immunoglobulin exhibited >100-day survival of full major histocompatibility complex mismatched allografts, whereas all similarly treated wild-type recipients rejected their transplants by 21 days after transplantation. We found that haptoglobin modifies the intra-allograft inflammatory milieu by enhancing levels of the inflammatory cytokine interleukin-6 and the chemokine MIP-2 (macrophage inflammatory protein 2) but impair levels of the immunosuppressive cytokine interleukin-10. Haptoglobin also enhances dendritic cell graft recruitment and augments antidonor T-cell responses. Moreover, we confirmed that the protein is present in human cardiac allograft specimens undergoing acute graft rejection. CONCLUSIONS Our findings provide new insights into the mechanisms of inflammation after cardiac transplantation and suggest that, in contrast to its prior reported antioxidant function in vascular inflammation, haptoglobin is an enhancer of inflammation after cardiac transplantation. Haptoglobin may also be a key component in other sterile inflammatory conditions.
Collapse
Affiliation(s)
- Hua Shen
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Elizabeth Heuzey
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel N Mori
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Christine K Wong
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Christopher M Colangelo
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Lisa M Chung
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Can Bruce
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Ilya B Slizovskiy
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Carmen J Booth
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel Kreisel
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO
| | - Daniel R Goldstein
- From the Department of Internal Medicine (H.S., E.H., D.N.M., C.K.W., D.R.G.), Department of Immunobiology (H.S., D.N.M., C.K.W., D.R.G.), W.M. Keck Biotechnology Resource Laboratory (C.M.C., L.M.C.), Center for Medical Informatics (C.B.), and Section of Comparative Medicine (I.B.S., C.J.B.), Yale School of Medicine, New Haven, CT; Sciomix, Woodbridge, CT (C.B.); Department of Surgery (D.K.) and Department of Immunology (D.K.), Washington University School of Medicine, St Louis, MO.
| |
Collapse
|
48
|
Mori DN, Kreisel D, Fullerton JN, Gilroy DW, Goldstein DR. Inflammatory triggers of acute rejection of organ allografts. Immunol Rev 2015; 258:132-44. [PMID: 24517430 DOI: 10.1111/imr.12146] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Solid organ transplantation is a vital therapy for end stage diseases. Decades of research have established that components of the adaptive immune system are critical for transplant rejection, but the role of the innate immune system in organ transplantation is just emerging. Accumulating evidence indicates that the innate immune system is activated at the time of organ implantation by the release of endogenous inflammatory triggers. This review discusses the nature of these triggers in organ transplantation and also potential mediators that may enhance inflammation resolution after organ implantation.
Collapse
Affiliation(s)
- Daniel N Mori
- Departments of Internal Medicine and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
Vertebrates mount strong adaptive immune responses to transplanted organs (allografts), but the mechanisms by which the innate immune system initiates this response are not completely understood. In anti-microbial immunity, non-self molecules associated with pathogens but not present in the host induce the maturation of innate antigen-presenting cells (APCs) by binding to germ-line-encoded receptors. Mature APCs then initiate the adaptive immune response by presenting microbial antigen and providing costimulatory signals to T cells. How allografts activate APCs, however, is less clear, because allografts are presumably sterile. A widely accepted view is that inflammatory or 'danger' molecules released by dying graft cells at the time of transplantation trigger APC maturation and the T-cell response that follows. Alternatively, it has been proposed that the introduction of microbial products during the surgical procedure could also alert the innate immune system to the presence of the transplanted organ. Here, we review why these hypotheses fail to fully explain how the alloimmune response is initiated after transplantation and summarize evidence that recognition of allogeneic non-self by monocytes is a key event in triggering alloimmunity and graft rejection.
Collapse
Affiliation(s)
- Martin H Oberbarnscheidt
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine & University of Pittsburgh Medical Center, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh School of Medicine & University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | |
Collapse
|
50
|
Béland S, Désy O, Vallin P, Basoni C, De Serres SA. Innate immunity in solid organ transplantation: an update and therapeutic opportunities. Expert Rev Clin Immunol 2015; 11:377-89. [PMID: 25644774 DOI: 10.1586/1744666x.2015.1008453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innate immunity is increasingly recognized as a major player in transplantation. In addition to its role in inflammation in the early post-transplant period, innate immunity shapes the differentiation of cells of adaptive immunity, with a capacity to promote either rejection or tolerance. Emerging data indicate that innate allorecognition, a characteristic previously limited to lymphocytes, is involved in allograft rejection. This review briefly summarizes the physiology of each component of the innate immune system in the context of transplantation and presents the current or promising therapeutic applications, such as cellular, anticomplement and anticytokine therapies.
Collapse
Affiliation(s)
- Stéphanie Béland
- Transplantation Unit, Renal Division, Department of Medicine, CHU de Québec Research Center, Faculty of Medicine, Laval University, 11 Côte du Palais, Québec, QC, Canada
| | | | | | | | | |
Collapse
|