1
|
Immune response associated with ischemia and reperfusion injury during organ transplantation. Inflamm Res 2022; 71:1463-1476. [PMID: 36282292 PMCID: PMC9653341 DOI: 10.1007/s00011-022-01651-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background Ischemia and reperfusion injury (IRI) is an ineluctable immune-related pathophysiological process during organ transplantation, which not only causes a shortage of donor organs, but also has long-term and short-term negative consequences on patients. Severe IRI-induced cell death leads to the release of endogenous substances, which bind specifically to receptors on immune cells to initiate an immune response. Although innate and adaptive immunity have been discovered to play essential roles in IRI in the context of organ transplantation, the pathway and precise involvement of the immune response at various stages has not yet to be elucidated. Methods We combined “IRI” and “organ transplantation” with keywords, respectively such as immune cells, danger signal molecules, macrophages, neutrophils, natural killer cells, complement cascade, T cells or B cells in PubMed and the Web of Science to search for relevant literatures. Conclusion Comprehension of the immune mechanisms involved in organ transplantation is promising for the treatment of IRI, this review summarizes the similarities and differences in both innate and adaptive immunity and advancements in the immune response associated with IRI during diverse organ transplantation.
Collapse
|
2
|
Lopez KJ, Cross-Najafi AA, Farag K, Obando B, Thadasina D, Isidan A, Park Y, Zhang W, Ekser B, Li P. Strategies to induce natural killer cell tolerance in xenotransplantation. Front Immunol 2022; 13:941880. [PMID: 36072599 PMCID: PMC9441937 DOI: 10.3389/fimmu.2022.941880] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Eliminating major xenoantigens in pig cells has drastically reduced human antibody-mediated hyperacute xenograft rejection (HXR). Despite these advancements, acute xenograft rejection (AXR) remains one of the major obstacles to clinical xenotransplantation, mediated by innate immune cells, including macrophages, neutrophils, and natural killer (NK) cells. NK cells play an 'effector' role by releasing cytotoxicity granules against xenogeneic cells and an 'affecter' role on other immune cells through cytokine secretion. We highlight the key receptor-ligand interactions that determine the NK cell response to target cells, focusing on the regulation of NK cell activating receptor (NKG2D, DNAM1) and inhibitory receptor (KIR2DL1-4, NKG2A, and LIR-1) signaling pathways. Inhibition of NK cell activity may protect xenografts from cytotoxicity. Recent successful approaches to reducing NK cell-mediated HXR and AXR are reviewed, including genetic modifications of porcine xenografts aimed at improving pig-to-human compatibility. Future directions to promote xenograft acceptance are discussed, including NK cell tolerance in pregnancy and NK cell evasion in viral infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ping Li
- *Correspondence: Ping Li, ; Burcin Ekser,
| |
Collapse
|
3
|
Baldwin WM, Valujskikh A, Fairchild RL. C1q as a potential tolerogenic therapeutic in transplantation. Am J Transplant 2021; 21:3519-3523. [PMID: 34058061 PMCID: PMC8564585 DOI: 10.1111/ajt.16705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/10/2021] [Accepted: 05/26/2021] [Indexed: 01/25/2023]
Abstract
In 1963, Lepow and colleagues resolved C1, the first component of the classical pathway, into three components, which they named C1q, C1r, and C1s. All three of these components were demonstrated to be involved in causing hemolysis in vitro. For over 30 years after that seminal discovery, the primary function attributed to C1q was as part of the C1 complex that initiated the classical pathway of the complement cascade. Then, a series of papers reported that isolated C1q could bind to apoptotic cells and facilitate their clearance by macrophages. Since then, rheumatologists have recognized that C1q is an important pattern recognition receptor (PRR) that diverts autoantigen containing extracellular vesicles from immune recognition. This critical function of C1q as a regulator of immune recognition has been largely overlooked in transplantation. Now that extracellular vesicles released from transplants have been identified as a major agent of immune recognition, it is logical to consider the potential impact of C1q on modulating the delivery of allogeneic extracellular vesicles to antigen presenting cells. This concept has clinical implications in the possible use of C1q or a derivative as a biological therapeutic to down-modulate immune responses to transplants.
Collapse
Affiliation(s)
- William M Baldwin
- Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA
| | - Anna Valujskikh
- Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA
| | - Robert L Fairchild
- Inflammation and Immunity, Lerner Research Institute, Cleveland, Ohio, USA
| |
Collapse
|
4
|
Vázquez-Carballo C, Guerrero-Hue M, García-Caballero C, Rayego-Mateos S, Opazo-Ríos L, Morgado-Pascual JL, Herencia-Bellido C, Vallejo-Mudarra M, Cortegano I, Gaspar ML, de Andrés B, Egido J, Moreno JA. Toll-Like Receptors in Acute Kidney Injury. Int J Mol Sci 2021; 22:ijms22020816. [PMID: 33467524 PMCID: PMC7830297 DOI: 10.3390/ijms22020816] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/16/2022] Open
Abstract
Acute kidney injury (AKI) is an important health problem, affecting 13.3 million individuals/year. It is associated with increased mortality, mainly in low- and middle-income countries, where renal replacement therapy is limited. Moreover, survivors show adverse long-term outcomes, including increased risk of developing recurrent AKI bouts, cardiovascular events, and chronic kidney disease. However, there are no specific treatments to decrease the adverse consequences of AKI. Epidemiological and preclinical studies show the pathological role of inflammation in AKI, not only at the acute phase but also in the progression to chronic kidney disease. Toll-like receptors (TLRs) are key regulators of the inflammatory response and have been associated to many cellular processes activated during AKI. For that reason, a number of anti-inflammatory agents targeting TLRs have been analyzed in preclinical studies to decrease renal damage during AKI. In this review, we updated recent knowledge about the role of TLRs, mainly TLR4, in the initiation and development of AKI as well as novel compounds targeting these molecules to diminish kidney injury associated to this pathological condition.
Collapse
Affiliation(s)
- Cristina Vázquez-Carballo
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Melania Guerrero-Hue
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Cristina García-Caballero
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Sandra Rayego-Mateos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
| | - José Luis Morgado-Pascual
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Carmen Herencia-Bellido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
| | - Mercedes Vallejo-Mudarra
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
| | - Isabel Cortegano
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - María Luisa Gaspar
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - Belén de Andrés
- Immunobiology Department, Carlos III Health Institute, 28220 Majadahonda (Madrid), Spain; (I.C.); (M.L.G.); (B.d.A.)
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (C.V.-C.); (S.R.-M.); (L.O.-R.); (C.H.-B.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), 28040 Madrid, Spain
- Correspondence: (J.E.); (J.A.M.); Tel.: +34-915504800 (J.E.); +34-957-218039 (J.A.M.)
| | - Juan Antonio Moreno
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain; (M.G.-H.); (C.G.-C.); (J.L.M.-P.); (M.V.-M.)
- Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 28029 Madrid, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 140471 Cordoba, Spain
- Correspondence: (J.E.); (J.A.M.); Tel.: +34-915504800 (J.E.); +34-957-218039 (J.A.M.)
| |
Collapse
|
5
|
Abstract
The human liver is an organ with a diverse array of immunologic functions. Its unique anatomic position that leads to it receiving all the mesenteric venous blood, combined with its unique micro anatomy, allows it to serve as a sentinel for the body's immune system. Hepatocytes, biliary epithelial cells, Kupffer cells, stellate cells, and liver sinusoidal endothelial cells express key molecules that recruit and activate innate and adaptive immunity. Additionally, a diverse array of lymphoid and myeloid immune cells resides within and traffics to the liver in specific circumstances. Derangement of these trafficking mechanisms underlies the pathophysiology of autoimmune liver diseases, nonalcoholic steatohepatitis, and liver transplantation. Here, we review these pathways and interactions along with potential targets that have been identified to be exploited for therapeutic purposes.
Collapse
|
6
|
Zhao Y, Sun X, Yang X, Zhang B, Li S, Han P, Zhang B, Wang X, Li S, Chang Y, Wei W. Tolerogenic Dendritic Cells Generated by BAFF Silencing Ameliorate Collagen-Induced Arthritis by Modulating the Th17/Regulatory T Cell Balance. THE JOURNAL OF IMMUNOLOGY 2019; 204:518-530. [PMID: 31843958 DOI: 10.4049/jimmunol.1900552] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
Abstract
Tolerogenic dendritic cells (tolDCs) have received much attention because of their capacity to restore immune homeostasis. RNA interference techniques have been used in several studies to generate tolDCs by inactivating certain molecules that regulate DC maturation and immunologic function. BAFF is a key B cell survival factor that is not only essential for B cell function but also T cell costimulation, and DCs are the major source of BAFF. In this study, we determined whether BAFF gene silencing in mature DCs could lead to a tolerogenic phenotype as well as the potential therapeutic effect of BAFF-silenced DCs on collagen-induced arthritis (CIA) in mice. Meanwhile, CRISPR/Cas9-mediated BAFF-/- DC2.4 cells were generated to verify the role of BAFF in DC maturation and functionality. BAFF-silenced DCs and BAFF-/- DC2.4 cells exhibited an immature phenotype and functional state. Further, the transplantation of BAFF-silenced DCs significantly alleviated CIA severity in mice, which correlated with a reduction in Th17 populations and increased regulatory T cells. In vitro, BAFF-silenced DCs promoted Foxp3 mRNA and IL-10 expression but inhibited ROR-γt mRNA and IL-17A expression in CD4+ T cells. Together, BAFF-silenced DCs can alleviate CIA, partly by inducing Foxp3+ regulatory T cells and suppressing Th17 subsets. Collectively, BAFF plays an important role in interactions between DCs and T cells, which might be a promising genetic target to generate tolDCs for autoimmune arthritis treatment.
Collapse
Affiliation(s)
- Yingjie Zhao
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Xiaojing Sun
- Anhui Maternity and Child Health Care Hospital, Hefei 230001, China
| | - Xuezhi Yang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Bingjie Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Siyu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Ping Han
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Binbin Zhang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Xinwei Wang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Susu Li
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Yan Chang
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei 230032, China; and
| |
Collapse
|
7
|
Khalil MAM, Khalil MAU, Khan TFT, Tan J. Drug-Induced Hematological Cytopenia in Kidney Transplantation and the Challenges It Poses for Kidney Transplant Physicians. J Transplant 2018; 2018:9429265. [PMID: 30155279 PMCID: PMC6093016 DOI: 10.1155/2018/9429265] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 06/04/2018] [Accepted: 06/25/2018] [Indexed: 12/14/2022] Open
Abstract
Drug-induced hematological cytopenia is common in kidney transplantation. Various cytopenia including leucopenia (neutropenia), thrombocytopenia, and anemia can occur in kidney transplant recipients. Persistent severe leucopenia or neutropenia can lead to opportunistic infections of various etiologies. On the contrary, reducing or stopping immunosuppressive medications in these events can provoke a rejection. Transplant clinicians are often faced with the delicate dilemma of balancing cytopenia and rejection from adjustments of immunosuppressive regimen. Differentials of drug-induced cytopenia are wide. Identification of culprit medication and subsequent modification is also challenging. In this review, we will discuss individual drug implicated in causing cytopenia and correlate it with corresponding literature evidence.
Collapse
Affiliation(s)
| | | | - Taqi F. Taufeeq Khan
- King Salman Armed Forces Hospital, Tabuk King Abdul Aziz Rd., Tabuk 47512, Saudi Arabia
| | - Jackson Tan
- RIPAS Hospital, Bandar Seri Begawan BA1710, Brunei Darussalam
| |
Collapse
|
8
|
Obregon C, Kumar R, Pascual MA, Vassalli G, Golshayan D. Update on Dendritic Cell-Induced Immunological and Clinical Tolerance. Front Immunol 2017; 8:1514. [PMID: 29250057 PMCID: PMC5715373 DOI: 10.3389/fimmu.2017.01514] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) as highly efficient antigen-presenting cells are at the interface of innate and adaptive immunity. As such, they are key mediators of immunity and antigen-specific immune tolerance. Due to their functional specialization, research efforts have focused on the characterization of DCs subsets involved in the initiation of immunogenic responses and in the maintenance of tissue homeostasis. Tolerogenic DCs (tolDCs)-based therapies have been designed as promising strategies to prevent and control autoimmune diseases as well as allograft rejection after solid organ transplantation (SOT). Despite successful experimental studies and ongoing phase I/II clinical trials using autologous tolDCs in patients with type 1 diabetes, rheumatoid arthritis, multiple sclerosis, and in SOT recipients, additional basic research will be required to determine the optimal DC subset(s) and conditioning regimens for tolDCs-based treatments in vivo. In this review, we discuss the characteristics of human DCs and recent advances in their classification, as well as the role of DCs in immune regulation and their susceptibility to in vitro or in vivo manipulation for the development of tolerogenic therapies, with a focus on the potential of tolDCs for the treatment of autoimmune diseases and the prevention of allograft rejection after SOT.
Collapse
Affiliation(s)
- Carolina Obregon
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Rajesh Kumar
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Manuel Antonio Pascual
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.,Department of Surgery, Transplantation Centre, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Vassalli
- Département coeur-vaisseaux, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland.,Fondazione Cardiocentro Ticino, Swiss Institute of Regenerative Medicine (SIRM), Lugano, Switzerland
| | - Déla Golshayan
- Department of Medicine, Transplantation Centre and Transplantation Immunopathology Laboratory, Service of Immunology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland.,Department of Surgery, Transplantation Centre, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Lin CM, Plenter RJ, Coulombe M, Gill RG. Interferon Gamma and Contact-dependent Cytotoxicity Are Each Rate Limiting for Natural Killer Cell-Mediated Antibody-dependent Chronic Rejection. Am J Transplant 2016; 16:3121-3130. [PMID: 27163757 PMCID: PMC5083186 DOI: 10.1111/ajt.13865] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 01/25/2023]
Abstract
Natural killer (NK) cells are key components of the innate immune system. In murine cardiac transplant models, donor-specific antibodies (DSA), in concert with NK cells, are sufficient to inflict chronic allograft vasculopathy independently of T and B cells. In this study, we aimed to determine the effector mechanism(s) required by NK cells to trigger chronic allograft vasculopathy during antibody-mediated rejection. Specifically, we tested the relative contribution of the proinflammatory cytokine interferon gamma (IFN-γ) versus the contact-dependent cytotoxic mediators of perforin and the CD95/CD95L (Fas/Fas ligand [FasL]) pathway for triggering these lesions. C3H/HeJ cardiac allografts were transplanted into immune-deficient C57BL/6 rag-/- γc-/- recipients, who also received monoclonal anti-major histocompatibility complex (MHC) class I DSA. The combination of DSA and wild-type NK cell transfer triggered aggressive chronic allograft vasculopathy. However, transfer of IFN-γ-deficient NK cells or host IFN-γ neutralization led to amelioration of these lesions. Use of either perforin-deficient NK cells or CD95 (Fas)-deficient donors alone did not alter development of vasculopathy, but simultaneous disruption of NK cell-derived perforin and allograft Fas expression resulted in prevention of these abnormalities. Therefore, both NK cell IFN-γ production and contact-dependent cytotoxic activity are rate-limiting effector pathways that contribute to this form of antibody-induced chronic allograft vasculopathy.
Collapse
Affiliation(s)
- C M Lin
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO.
| | - R J Plenter
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Aurora, CO
| | - M Coulombe
- Department of Surgery, University of Colorado, Aurora, CO
| | - R G Gill
- Department of Surgery, University of Colorado, Aurora, CO
| |
Collapse
|
10
|
Shao Z, Jiao B, Liu T, Cheng Y, Liu H, Liu Y. TAK-242 treatment ameliorates liver ischemia/reperfusion injury by inhibiting TLR4 signaling pathway in a swine model of Maastricht-category-III cardiac death. Biomed Pharmacother 2016; 84:495-501. [PMID: 27685793 DOI: 10.1016/j.biopha.2016.09.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 08/16/2016] [Accepted: 09/11/2016] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES This study aims to test the effects of TAK-242 on liver transplant viability in a model of swine Maastricht-category-III cardiac death. METHODS A swine DCD Maastricht-III model of cardiac death was established, and TAK-242 was administered prior to the induction of cardiac death. The protein and mRNA level of TLR4 signaling pathway molecules and cytokines that are important in mediating immune and inflammatory responses were assessed at different time points following the induction of cardiac death. RESULTS After induction of cardiac death, both the mRNA and protein levels of key molecules (TLR4, TRAF6, NF-ϰB, ICAM-1, MCP-1 and MPO), TNF-α and IL-6 increased significantly. Infusion of TAK-242 1h before induction of cardiac death blocked the increase of immune and inflammatory response molecules. However, the increase of TLR4 level was not affected by infusion of TAK-242. Histology study showed that infusion of TAK-242 protect liver tissue from damage during cardiac death. CONCLUSIONS These results indicates that TLR4 signaling pathway may contribute to ischemia/reperfusion injury in the liver grafts, and blocking TLR4 pathway with TAk-242 may reduce TLR4-mediated tissue damage.
Collapse
Affiliation(s)
- Zigong Shao
- Department of Organ transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Baoping Jiao
- Department of General Surgery, The First Hospital of Liaoning Medical University, Jinzhou 121000, China
| | - Tingting Liu
- Department of Organ transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ying Cheng
- Department of Organ transplantation, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Hao Liu
- Department of Organ transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yongfeng Liu
- Department of Organ transplantation, The First Hospital of China Medical University, Shenyang 110001, China
| |
Collapse
|
11
|
Golshayan D, Wójtowicz A, Bibert S, Pyndiah N, Manuel O, Binet I, Buhler LH, Huynh-Do U, Mueller T, Steiger J, Pascual M, Meylan P, Bochud PY. Polymorphisms in the lectin pathway of complement activation influence the incidence of acute rejection and graft outcome after kidney transplantation. Kidney Int 2016; 89:927-38. [PMID: 26924055 DOI: 10.1016/j.kint.2015.11.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 09/26/2015] [Accepted: 11/12/2015] [Indexed: 01/01/2023]
Abstract
There are conflicting data on the role of the lectin pathway of complement activation and its recognition molecules in acute rejection and outcome after transplantation. To help resolve this we analyzed polymorphisms and serum levels of lectin pathway components in 710 consecutive kidney transplant recipients enrolled in the nationwide Swiss Transplant Cohort Study, together with all biopsy-proven rejection episodes and 1-year graft and patient survival. Functional mannose-binding lectin (MBL) levels were determined in serum samples, and previously described MBL2, ficolin 2, and MBL-associated serine protease 2 polymorphisms were genotyped. Low MBL serum levels and deficient MBL2 diplotypes were associated with a higher incidence of acute cellular rejection during the first year, in particular in recipients of deceased-donor kidneys. This association remained significant (hazard ratio 1.75, 95% confidence interval 1.18-2.60) in a Cox regression model after adjustment for relevant covariates. In contrast, there was no significant association with rates of antibody-mediated rejection, patient death, early graft dysfunction or loss. Thus, results in a prospective multicenter contemporary cohort suggest that MBL2 polymorphisms result in low MBL serum levels and are associated with acute cellular rejection after kidney transplantation. Since MBL deficiency is a relatively frequent trait in the normal population, our findings may lead to individual risk stratification and customized immunosuppression.
Collapse
Affiliation(s)
- Déla Golshayan
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland; Transplantation Immunopathology Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.
| | - Agnieszka Wójtowicz
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Stéphanie Bibert
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Nitisha Pyndiah
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Oriol Manuel
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland; Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Isabelle Binet
- Nephrologie und Transplantationsmedizin, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Leo H Buhler
- Centre Universitaire Romand de Transplantation, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Inselspital Bern, Bern, Switzerland
| | - Thomas Mueller
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Jürg Steiger
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Manuel Pascual
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland; Transplantation Immunopathology Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Pascal Meylan
- Transplantation Center, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland; Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Pierre-Yves Bochud
- Service of Infectious Diseases, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
12
|
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
|
13
|
Chemokines in chronic liver allograft dysfunction pathogenesis and potential therapeutic targets. Clin Dev Immunol 2013; 2013:325318. [PMID: 24382971 PMCID: PMC3870628 DOI: 10.1155/2013/325318] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 10/03/2013] [Indexed: 02/05/2023]
Abstract
Despite advances in immunosuppressive drugs, long-term success of liver transplantation is still limited by the development of chronic liver allograft dysfunction. Although the exact pathogenesis of chronic liver allograft dysfunction remains to be established, there is strong evidence that chemokines are involved in organ damage induced by inflammatory and immune responses after liver surgery. Chemokines are a group of low-molecular-weight molecules whose function includes angiogenesis, haematopoiesis, mitogenesis, organ fibrogenesis, tumour growth and metastasis, and participating in the development of the immune system and in inflammatory and immune responses. The purpose of this review is to collect all the research that has been done so far concerning chemokines and the pathogenesis of chronic liver allograft dysfunction and helpfully, to pave the way for designing therapeutic strategies and pharmaceutical agents to ameliorate chronic allograft dysfunction after liver transplantation.
Collapse
|
14
|
Absence of FcγRIII results in increased proinflammatory response in FcγRIII-KO cardiac recipients. Transplantation 2013; 96:601-8. [PMID: 23903009 DOI: 10.1097/tp.0b013e31829c2455] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Alloantibody can contribute significantly to rejection of heart transplants by activation of complement and interactions with a variety of effector cells, including macrophages and monocytes through activating FcγRI, FcγRIII, FcγRIV, the inhibitory FcγRIIB and complement receptors. These receptors link cellular and humoral immunity by bridging the antibody specificity to effector cells. Activating FcγRs are also involved in serum amyloid P component (SAP)-mediated clearance of apoptotic bodies. METHODS B10.A (H-2a) hearts were transplanted into wild-type (WT) or FcγRIII-knockout (KO) C57BL/6 (H-2b) mouse recipients. Levels of alloantibodies and SAP in the circulation were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. Intragraft cytokine mRNA expression was measured by real-time polymerase chain reaction. Intragraft deposition of C4d, von Willebrand factor, SAP, and activated caspase 3 was visualized by immunochemistry. RESULTS B10.A hearts in C57BL/6 FcγRIII-KO recipients were rejected acutely within 6 to 8 days compared with 10 to 14 days in WT. The rejection in FcγRIII-KO was accompanied by higher levels of circulating IgM/IgG alloantibodies and SAP than in WT recipients. Histology in FcγRIII-KO cardiac allograft recipients indicated perivascular margination of monocytes and neutrophils, vascular endothelial cell injury, and intense vasculocentric infiltrates with extensive apoptosis. Higher numbers of apoptotic cells, stronger C4d and SAP deposition, and extensive activated caspase 3 were found in areas of dense pockets of apoptotic blebs in FcγRIII-KO. CONCLUSIONS We propose that absence of FcγRIII is associated with the lack of efficient SAP-mediated clearance of apoptotic cells through FcγRs. Apoptotic cells become immunogenic and induce enhanced inflammation, alloantibody production, and complement activation leading to accelerated cardiac allograft rejection.
Collapse
|
15
|
|
16
|
Wei L, Wu RB, Yang CM, Zheng SY, Yu XY. Polymerised placenta haemoglobin attenuates cold ischaemia/reperfusion injury in isolated rat heart. Microvasc Res 2011; 82:430-8. [PMID: 21907723 DOI: 10.1016/j.mvr.2011.08.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 07/28/2011] [Accepted: 08/25/2011] [Indexed: 12/14/2022]
Abstract
Ischaemia/reperfusion (I/R) injury is harmful to the cardiovascular system and is responsible for the inflammatory response, which, in turn, aggravates cardiac dysfunction. This study was designed to investigate the protective effect and potential mechanism of a haemoglobin-based oxygen carrier on cold I/R-injured hearts. Isolated Sprague-Dawley rat hearts were perfused in Langendorff mode. After a 30-min basal perfusion, rat hearts were arrested and hypothermically stored at 4°C for 12h followed by a 2-h reperfusion. Compared with histidine-tryptophan-ketoglutarate solution (HTKs), polymerised placenta haemoglobin (PolyPHb) in HTKs greatly improved heart contraction and decreased infarction size, necrosis, and apoptosis, which was related to reduced expression of TLR2, TLR4, TNF-α, and IL-1β, and NF-κB activation. Our results demonstrate the cardioprotective effect of PolyPHb on cold I/R-injured hearts and revealed that this protection was mediated in large part by attenuation of TLR2 and -4/NF-κB signalling pathway and could possibly down-regulate the inflammatory response.
Collapse
Affiliation(s)
- Li Wei
- Department of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangdong Academy of Medical Sciences, 96 Dongchuan Road, Guangzhou 510080, China
| | | | | | | | | |
Collapse
|
17
|
Association of polymorphisms of interleukin-8, CXCR1, CXCR2, and selectin with allograft outcomes in kidney transplantation. Transplantation 2011; 91:57-64. [PMID: 21452410 DOI: 10.1097/tp.0b013e3181fd0195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND Both chemokines and adhesion molecules mediate allograft rejection by recruiting leukocytes into the allograft. We investigated the association of six single nucleotide polymorphisms (SNPs) located in interleukin (IL)-8, CXCR1, CXCR2, and selectin with kidney allograft outcomes. METHODS The promoter regions of CXCR1 and CXCR2 were sequenced directly to find SNPs. Reporter gene assay was performed to determine the transcriptional activity of CXCR2 promoter polymorphisms. The association of SNPs in IL-8, CXCR1, CXCR2, and selectin with both acute rejection and estimated glomerular filtration rate at 1-year posttransplant was analyzed in 216 donor-recipient pairs of kidney transplantation. RESULTS The donor GA/AA genotypes of CXCR1 -2668G/A (rs2671222) were associated with increased risk for acute rejection even after adjusting for covariates such as gender, diabetes, preemptive transplantation, immunosuppressive regimen, relationship with the donor, and human leukocyte antigen mismatch (adjusted odds ratio 3.56; 95% confidence interval 1.37-9.27; P=0.009). Although the transcriptional activity of the CXCR2 variant promoter was 2.6-fold higher than that of the wild-type promoter (P=0.039), no significant association was observed between CXCR2 polymorphisms and kidney allograft outcomes. SNPs of IL-8, L-selectin, and E-selectin were not associated with kidney allograft outcomes. CONCLUSION The donor CXCR1 -2668 GA/AA genotypes were an independent risk factor for acute rejection in kidney transplantation.
Collapse
|
18
|
Du Clos TW, Mold C. Pentraxins (CRP, SAP) in the process of complement activation and clearance of apoptotic bodies through Fcγ receptors. Curr Opin Organ Transplant 2011; 16:15-20. [PMID: 21150611 PMCID: PMC3153594 DOI: 10.1097/mot.0b013e32834253c7] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Ischemia/reperfusion injury and organ allograft rejection both entail excessive cell and tissue destruction. A number of innate immune proteins, including the pentraxins, participate in the removal of this potentially inflammatory and autoimmunogenic material. The classical pentraxins, C-reactive protein (CRP) and serum amyloid P component (SAP) are serum opsonins, which bind to damaged membranes and nuclear autoantigens. Understanding the role of pentraxins in inflammation has been advanced by the recent identification and structural analysis of their receptor interactions. RECENT FINDINGS The ligand-binding, complement-activating and opsonic properties of pentraxins have been known for some time. However, the establishment of Fcγ receptors as the primary receptors for pentraxins is a recent finding with important implications for CRP and SAP functions. The crystal structure of SAP in complex with FcγRIIa was recently solved, leading to new insights into function and new opportunities for pentraxin-based therapeutics. In addition, new approaches to inhibit CRP synthesis or binding are being developed based on clinical associations between CRP levels and cardiovascular disease. SUMMARY This review will summarize data on the function of pentraxins in clearance of injured tissue and cells and discuss the implications of this clearance pathway for autoimmunity and ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Terry W Du Clos
- Department of Veterans Affairs Medical Center and the University of New Mexico School of Medicine, Albuquerque, New Mexico 87108, USA.
| | | |
Collapse
|
19
|
Abstract
Antibody-mediated rejection has become critical clinically because this form of rejection is usually unresponsive to conventional anti-rejection therapy, and therefore, it has been recognized as a major cause of allograft loss. Our group developed experimental animal models of vascularized organ transplantation to study pathogenesis of antibody- and complement-mediated endothelial cell injury leading to graft rejection. In this review, we discuss mechanisms of antibody-mediated graft rejection resulting from activation of complement by C1q- and MBL (mannose-binding lectin)-dependent pathways and interactions with a variety of effector cells, including macrophages and monocytes through Fcgamma receptors and complement receptors.
Collapse
|
20
|
Arslan F, Keogh B, McGuirk P, Parker AE. TLR2 and TLR4 in ischemia reperfusion injury. Mediators Inflamm 2010; 2010:704202. [PMID: 20628516 PMCID: PMC2902053 DOI: 10.1155/2010/704202] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 04/07/2010] [Indexed: 12/15/2022] Open
Abstract
Ischemia reperfusion (I/R) injury refers to the tissue damage which occurs when blood supply returns to tissue after a period of ischemia and is associated with trauma, stroke, myocardial infarction, and solid organ transplantation. Although the cause of this injury is multifactorial, increasing experimental evidence suggests an important role for the innate immune system in initiating the inflammatory cascade leading to detrimental/deleterious changes. The Toll-like Receptors (TLRs) play a central role in innate immunity recognising both pathogen- and damage-associated molecular patterns and have been implicated in a range of inflammatory and autoimmune diseases. In this paper, we summarise the current state of knowledge linking TLR2 and TLR4 to I/R injury, including recent studies which demonstrate that therapeutic inhibition of TLR2 has beneficial effects on I/R injury in a murine model of myocardial infarction.
Collapse
Affiliation(s)
- F. Arslan
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - B. Keogh
- Opsona Therapeutics Ltd., Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
| | - P. McGuirk
- Opsona Therapeutics Ltd., Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
| | - A. E. Parker
- Opsona Therapeutics Ltd., Institute of Molecular Medicine, Trinity Centre for Health Sciences, St. James' Hospital, Dublin 8, Ireland
| |
Collapse
|
21
|
Endothelial transcripts uncover a previously unknown phenotype: C4d-negative antibody-mediated rejection. Curr Opin Organ Transplant 2010; 15:42-8. [DOI: 10.1097/mot.0b013e3283352a50] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Sis B, Jhangri GS, Bunnag S, Allanach K, Kaplan B, Halloran PF. Endothelial gene expression in kidney transplants with alloantibody indicates antibody-mediated damage despite lack of C4d staining. Am J Transplant 2009; 9:2312-23. [PMID: 19681822 DOI: 10.1111/j.1600-6143.2009.02761.x] [Citation(s) in RCA: 367] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Anti-HLA alloantibody is a risk factor for graft loss, but does not indicate which kidneys are experiencing antibody-mediated rejection (ABMR). C4d staining in biopsies is specific for ABMR but insensitive. We hypothesized that altered expression of endothelial genes due to alloantibody acting on the microcirculation would be sensitive indicator of ABMR. We identified 119 endothelial-associated transcripts (ENDATs) from literature, and studied their expression by microarrays in 173 renal allograft biopsies for cause. Mean ENDAT expression was increased in all rejection but was higher in ABMR than in T-cell-mediated rejection and correlated with histopathologic lesions of ABMR, and alloantibody. Many individual ENDATs were increased in ABMR and predicted graft loss. Kidneys with high ENDATs and antibody showed increased lesions of ABMR and worse prognosis in comparison to controls. Only 40% of kidneys with high ENDAT expression and chronic ABMR or graft loss were diagnosed by C4d positivity. High ENDAT expression with antibody predicts graft loss with higher sensitivity (77% vs. 31%) and slightly lower specificity (71% vs. 94%) than C4d. The results were validated in independent set of 82 kidneys. High renal endothelial transcript expression in patients with alloantibody is indicator of active antibody-mediated allograft damage and poor graft outcome.
Collapse
Affiliation(s)
- Banu Sis
- Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada.
| | | | | | | | | | | |
Collapse
|
23
|
Rother RP, Arp J, Jiang J, Ge W, Faas SJ, Liu W, Gies DR, Jevnikar AM, Garcia B, Wang H. C5 blockade with conventional immunosuppression induces long-term graft survival in presensitized recipients. Am J Transplant 2008; 8:1129-42. [PMID: 18444931 DOI: 10.1111/j.1600-6143.2008.02222.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We explored whether a functionally blocking anti-C5 monoclonal antibody (mAb) combined with T- and B-cell immunosuppression can successfully prevent antibody-mediated (AMR) and cell-mediated rejection (CMR) in presensitized murine recipients of life-supporting kidney allografts. To mimic the urgent clinical features of AMR experienced by presensitized patients, we designed a murine model in which BALB/c recipients were presensitized with fully MHC-mismatched C3H donor skin grafts one week prior to C3H kidney transplantation. Presensitized recipients demonstrated high levels of circulating and intragraft antidonor antibodies and terminal complement activity, rejecting grafts within 8.5 +/- 1.3 days. Graft rejection was predominantly by AMR, characterized by interstitial hemorrhage, edema and glomerular/tubular necrosis, but also demonstrated moderate cellular infiltration, suggesting CMR involvement. Subtherapeutic treatment with cyclosporine (CsA) and LF15-0195 (LF) did not significantly delay rejection. Significantly, however, the addition of anti-C5 mAb to this CsA/LF regimen prevented terminal complement activity and inhibited both AMR and CMR, enabling indefinite (>100 days) kidney graft survival despite the persistence of antidonor antibodies. Long-term surviving kidney grafts expressed the protective proteins Bcl-x(S/L) and A-20 and demonstrated normal histology, suggestive of graft accommodation or tolerance. Thus, C5 blockade combined with routine immunosuppression offers a promising approach to prevent graft loss in presensitized patients.
Collapse
Affiliation(s)
- R P Rother
- Alexion Pharmaceuticals, Inc., Cheshire, CT, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Infiltration of natural killer (NK) cells into solid organ allografts is observed in clinical and experimental transplantation. Studies suggest a role for NK cells in acute and chronic rejection of solid organ allografts; however, the effects of immunosuppressive agents on NK cells are not clearly established. Rat NK cell lines were analyzed for proliferation and cytotoxicity in the presence of cyclosporine, FK506, or rapamycin. Lewis recipients of DA liver allografts received immunosuppressive agents after transplantation. NK cells demonstrated robust function both in the absence and presence of cyclosporine and FK506. In contrast, rapamycin significantly inhibited proliferation and cytotoxicity of NK cells. NK cell numbers remained stable in graft recipients treated with cyclosporine and FK506, whereas there was a significant decrease in NK cells in rapamycin-treated recipients. These data indicate that immunosuppressive drugs have differential effects on NK cell function that may impact the immune response of transplant recipients.
Collapse
|
25
|
Morrell CN, Murata K, Swaim AM, Mason E, Martin TV, Thompson LE, Ballard M, Fox-Talbot K, Wasowska B, Baldwin WM. In vivo platelet-endothelial cell interactions in response to major histocompatibility complex alloantibody. Circ Res 2008; 102:777-85. [PMID: 18296616 DOI: 10.1161/circresaha.107.170332] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Platelets recruit leukocytes and mediate interactions between leukocytes and endothelial cells. Most studies examining this important platelet immune function have focused on the development of atherosclerosis, but similar mechanisms may contribute to acute and chronic vascular lesions in transplants. Platelets have been described as markers of transplant rejection, but little investigation has critically examined a role for platelets in transplant vasculopathy and, in particular, alloantibody-mediated transplant rejection. We now demonstrate using a skin transplant model that alloantibody indirectly induces platelet activation and rolling in vivo. Repeated IgG2a alloantibody injections result in sustained platelet-endothelial interactions and vascular pathology, including von Willebrand factor release, small platelet thrombi, and complement deposition. Maintenance of continued platelet-endothelial interactions are dependent on complement activation. Furthermore, we demonstrate that platelets recruit leukocytes to sites of alloantibody deposition and sustain leukocyte-endothelial cell interactions in vivo. Taken together, our model demonstrates an important role for platelets in alloantibody induced transplant rejection.
Collapse
Affiliation(s)
- Craig N Morrell
- Department of Molecular and Comparative Pathobiology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Agrawal S, Tripathi P, Naik S. Roles and mechanism of natural killer cells in clinical and experimental transplantation. Expert Rev Clin Immunol 2008; 4:79-91. [DOI: 10.1586/1744666x.4.1.79] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
27
|
Wang H, Arp J, Liu W, Faas SJ, Jiang J, Gies DR, Ramcharran S, Garcia B, Zhong R, Rother RP. Inhibition of Terminal Complement Components in Presensitized Transplant Recipients Prevents Antibody-Mediated Rejection Leading to Long-Term Graft Survival and Accommodation. THE JOURNAL OF IMMUNOLOGY 2007; 179:4451-63. [PMID: 17878341 DOI: 10.4049/jimmunol.179.7.4451] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ab-mediated rejection (AMR) remains the primary obstacle in presensitized patients following organ transplantation, as it is refractory to anti-T cell therapy and can lead to early graft loss. Complement plays an important role in the process of AMR. In the present study, a murine model was designed to mimic AMR in presensitized patients. This model was used to evaluate the effect of blocking the fifth complement component (C5) with an anti-C5 mAb on prevention of graft rejection. BALB/c recipients were presensitized with C3H donor skin grafts 7 days before heart transplantation from the same donor strain. Heart grafts, transplanted when circulating anti-donor IgG Abs were at peak levels, were rejected in 3 days. Graft rejection was characterized by microvascular thrombosis and extensive deposition of Ab and complement in the grafts, consistent with AMR. Anti-C5 administration completely blocked terminal complement activity and local C5 deposition, and in combination with cyclosporine and short-term cyclophosphamide treatment, it effectively prevented heart graft rejection. These recipients achieved permanent graft survival for >100 days with normal histology despite the presence of systemic and intragraft anti-donor Abs and complement, suggesting ongoing accommodation. Furthermore, double-transplant experiments demonstrated that immunological alterations in both the graft and the recipient were required for successful graft accommodation to occur. These data suggest that terminal complement blockade with a functionally blocking Ab represents a promising therapeutic approach to prevent AMR in presensitized recipients.
Collapse
Affiliation(s)
- Hao Wang
- Multi-Organ Transplant Program, University Hospital, London Health Sciences Centre, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sun E. Cell death recognition model for the immune system. Med Hypotheses 2007; 70:585-96. [PMID: 17681705 DOI: 10.1016/j.mehy.2007.05.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Accepted: 05/31/2007] [Indexed: 12/11/2022]
Abstract
It is essential for the immune system to recognize markers or understand rules required for discriminating antigens that should be actively responded to from those be tolerated. Although the classic self-nonself theory over the past five decades has been challenged by "danger" model and "infectious nonself" model, etc., no theories could fit for all. Cell death is important not only for its role in homeostasis, but also for its decisive effects on the immune responses. Different ways of cell death, apoptosis or necrosis, transmit fundamentally opposite driving forces for the immune system, inducing tolerance or initiating adaptive immune responses. The progress in understanding phagocytosis and process of apoptotic and necrotic cells leads the author to propose "cell death" recognition model for the immune system. Four principles are important in this model. First, only antigens shedding from apoptotic or necrotic cells rather than those from healthy cells, can be presented to naïve T cells. Second, either apoptotic cells or necrotic cells, but not healthy cells, can attract phagocytes, namely dendritic cells (DC) or macrophages that are also antigen presenting cells (APC), to scavenge dead cells. Third, macrophages or DC residing in non-lymphoid tissues phagocytose dying/dead cells, migrate to lymphoid tissues and present antigens to naïve T cells there. Fourth, tolerance or adaptive responses are not dependent on whether the antigens are self or nonself, but on the ways of cell death during antigen presentation. Importantly, tolerance and adaptive immunity are all dominant responses and the impact of cell death on immune responses is a dynamic balance between them. "Cell death" recognition model could more easily explain various immune phenomena, including infection, self tolerance and autoimmunity, tumor immunity as well as transplant rejection. Investigation into the roles and mechanisms of cell death mediated immune responses and finding out key modulators will prompt better understanding the ways of immune recognition and provide novel strategies for the management of autoimmunity, tumors, infections as well as transplantation.
Collapse
Affiliation(s)
- Erwei Sun
- Institute of Transplant Immunology, Organ Transplantation Department, Zhujiang Hospital, Southern Medical University, 253 Gongye Road, Guangzhou 510282, China.
| |
Collapse
|
29
|
Hirayama S, Shiraishi T, Shirakusa T, Higuchi T, Miller EJ. Prevention of neutrophil migration ameliorates rat lung allograft rejection. Mol Med 2007. [PMID: 17225868 DOI: 10.2119/2006-00036.hirayama] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chemokines activate and recruit specific leukocyte subpopulations. We sought to determine whether neutrophil migration, which can contribute to the development of ischemia-reperfusion injury, correlates with lung allograft rejection. Orthotopic left lung allotransplantation was performed from Brown Norway (donor) to Fisher 344 (recipient) rats. Because the role of activated neutrophils in the development of allograft rejection is believed to be biphasic, we used specific CXC receptor inhibition with antileukinate in 2 dosing regimens. Recipients were allocated into 4 groups; A (early administration) received 2 doses of antileukinate (10.0 mg/kg) intramuscularly 24 h before and immediately after transplantation; B (continuous administration) continuously received antileukinate intraperitoneally (10.0 mg/kg/day) for 7 days after surgery. Groups A or B were compared with individual controls that received PBS alone. The progression of rejection was assessed radiographically. Histologic evaluation of allograft rejection based on pathologic rejection grade, performed on day 7, demonstrated significantly lower histologic rejection in group B compared with the control group (2.1+/-1.0 vs. 3.3+/-0.5; P=0.018), whereas there was no significant difference in group A compared with the control group. There were no significant differences between the aeration scores of groups A or B compared with their control groups. Our data suggest that neutrophils may play a promoting role in the development of allograft rejection, and blockage of neutrophil migration may suppress acute lung allograft rejection.
Collapse
Affiliation(s)
- Shin Hirayama
- Department of Surgery II, Fukuoka University School of Medicine, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
30
|
Lu CY, Hartono J, Senitko M, Chen J. The inflammatory response to ischemic acute kidney injury: a result of the 'right stuff' in the 'wrong place'? Curr Opin Nephrol Hypertens 2007; 16:83-9. [PMID: 17293682 DOI: 10.1097/mnh.0b013e3280403c4e] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Ischemic acute kidney injury may be exacerbated by an inflammatory response. How injury elicits inflammation remains a major question in understanding acute kidney injury. The present review examines the hypothesis that molecules released by injured cells elicit inflammation. RECENT FINDINGS After necrotic death, intracellular molecules find their way into the extracellular space. These molecules include heat shock proteins and HMGB1. Receptors for these proteins include TLR4, TLR2, CD91 and RAGE. These proinflammatory mechanisms may be so useful that nature has evolved mechanisms for programming necrotic death via poly(ADP-ribose) polymerase and cyclophilin D. In addition, apoptosis may also elicit inflammation. SUMMARY The concepts discussed in this review are important for clinical medicine. Drugs and genetic manipulation may ameliorate ischemic kidney injury by regulating the inflammatory response to cell injury.
Collapse
Affiliation(s)
- Christopher Y Lu
- Department of Internal Medicine, Nephrology Division, UT Southwestern Medical Center, Dallas, Texas 63110, USA.
| | | | | | | |
Collapse
|
31
|
Coupel S, Moreau A, Hamidou M, Horejsi V, Soulillou JP, Charreau B. Expression and release of soluble HLA-E is an immunoregulatory feature of endothelial cell activation. Blood 2007; 109:2806-14. [PMID: 17179229 DOI: 10.1182/blood-2006-06-030213] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Human leukocyte antigen (HLA)-E belongs, with HLA-G and HLA-F, to the non-classic major histocompatibility complex (MHC) class I (Ib) molecules, broadly defined by a limited polymorphism and a restricted pattern of cellular expression. In contrast to HLA-G, the expression and function of HLA-E and HLA-F in physiologic and pathologic processes remain poorly established. In the present study, we show that HLA-E protein expression in normal human nonlymphoid organs is mainly restricted to endothelial cells (ECs). HLA-E is also basally expressed by B and T lymphocytes, natural killer (NK) cells and by macrophages. We demonstrate that tumor necrosis factor alpha (TNFalpha), interleukin-1beta (IL-1beta), and interferon gamma (IFNgamma) up-regulate the cell-surface expression of HLA-E on ECs in vitro and induce the release of soluble HLA-E (sHLA-E). HLA-E up-regulation protects IFN-gamma-activated ECs from NK-mediated cell lysis, while sHLA-E protects bystander cells. Finally, sHLA-E is not detected in normal sera, and increased serum levels correlate with disease activity in patients with antineutrophil cytoplasmic antibody-associated systemic vasculitis. Thus, HLA-E expression and release of sHLA-E are features of EC activation and emphasize immunoregulatory functions of the endothelium. The present identification of soluble HLA-E molecules may have important implications in understanding the pathogenesis of immune-mediated vascular diseases and for the diagnosis and monitoring of patients.
Collapse
Affiliation(s)
- Stéphanie Coupel
- Institut National de la Santé et de la Recherche Médicale, Unit 643, Institut de Transplantation et de Recherche en Transplantation, Nantes, France
| | | | | | | | | | | |
Collapse
|
32
|
Dimitrov JD, Roumenina LT, Doltchinkova VR, Vassilev TL. Iron Ions and Haeme Modulate the Binding Properties of Complement Subcomponent C1q and of Immunoglobulins. Scand J Immunol 2007; 65:230-9. [PMID: 17309777 DOI: 10.1111/j.1365-3083.2006.01893.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The complement system and circulating antibodies play a major role in the defence against infection. They act at the sites of inflammation, where the harsh microenvironment and the oxidative stress lead to the release of free iron ions and haeme. The aim of this study was to analyse the consequences of the exposure of C1q and immunoglobulins to iron ions or haeme. The changes in target recognition by C1q and in the rheumatoid factor activity of the immunoglobulins were investigated. The exposure of C1q to ferrous ions increased its binding to IgG and to IgM. In contrast, haeme inhibited C1q binding to all studied targets, especially to IgG1 and C-reactive protein. Thus, the haeme released as a result of tissue damage and oxidative stress may act as a negative feedback regulator of an inappropriate complement triggering as seen in ischaemia-reperfusion tissue injury. The results also show that iron ions and haeme were able to reveal rheumatoid factor activity of IgG. The modulation of the C1q-target binding as well as the revealing of rheumatoid factor activity of IgG by exposure to redox-active agents released at the sites of inflammation may have important consequences for the understanding of the immunopathological mechanisms of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- J D Dimitrov
- Department of Immunology, Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | | | | | | |
Collapse
|
33
|
Lin T, Zhou W, Farrar CA, Hargreaves REG, Sheerin NS, Sacks SH. Deficiency of C4 from donor or recipient mouse fails to prevent renal allograft rejection. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1241-8. [PMID: 16565498 PMCID: PMC1606553 DOI: 10.2353/ajpath.2006.050360] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Complement effector products generated in the transplanted kidney are known to mediate transplant rejection, but which of the three main activation pathways of complement trigger this response is unclear. Here we assessed the role of the classical and lectin pathways by studying the common component C4 in mouse kidney transplant rejection. We transplanted wild-type or C4-null H-2(b) donor kidneys into H-2(k) or H-2(d) recipients, or vice-versa, to assess the roles of donor kidney and recipient expression of complement. Intragraft C4 gene expression rose substantially during rejection. However, we found no significant association between graft acceptance and the presence of C4 in either the donor kidney or recipient mouse. At the time of rejection, we found no significant differences in alloantibody response in the different groups. Tubular deposition of C3 to C9 occurred regardless of the absence or presence of C4 in either the donor or recipient mouse, indicating that C4 was dispensable for complement activation at this site. These data suggest that complement activation and renal allograft rejection are independent of the classical and lectin pathways in these models, implying that in the absence of these pathways the alternative pathway is the main trigger for complement-mediated rejection.
Collapse
Affiliation(s)
- Tao Lin
- Department of Nephrology and Transplantation, 5th Floor, Thomas Guy House, Guy's Hospital, St. Thomas St., London SE1 9RT, UK
| | | | | | | | | | | |
Collapse
|
34
|
Maurus CF, Schneider MKJ, Schmidt D, Zünd G, Seebach JD. Activation of human microvascular endothelial cells with TNF-alpha and hypoxia/reoxygenation enhances NK-cell adhesion, but not NK-Cytotoxicity. Transplantation 2006; 81:1204-11. [PMID: 16641609 DOI: 10.1097/01.tp.0000205175.53938.bd] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Ischemia/reperfusion injury (I/R) and cellular rejection in solid organ transplantation are characterized by adhesion molecule up-regulation on the graft endothelium, a prerequisite for leukocyte recruitment. The contribution of NK cells to I/R and allograft rejection is not well understood. The aim of the present study was to investigate allogeneic interactions between human NK cells and microvascular endothelial cells (MVEC) with special regard to the differential impact of TNF-alpha and hypoxia/reoxygenation in an in vitro model of I/R. METHODS MVEC were stimulated in vitro for 8 h with TNF-alpha, exposed to hypoxia (1% O2), hypoxia/reoxygenation, and combinations thereof in a hypoxia chamber. Cell surface expression of adhesion molecules on MVEC was analyzed by flow cytometry, and adhesion molecule shedding by ELISA. NK cell adhesion on MVEC was determined under shear stress, and NK cytotoxicity using Cr-release assays. RESULTS Surface expression of ICAM-1, VCAM-1, and E-/P-selectin on MVEC was up-regulated by TNF-alpha but unaffected by hypoxia/reoxygenation in the absence of TNF-alpha. ICAM-1 expression was further increased by a combination of TNF-alpha and hypoxia/reoxygenation, whereas TNF-alpha-induced E-/P-selectin expression was strongly reversed by hypoxia/reoxygenation. NK cell adhesion increased after exposing MVEC to TNF-alpha and hypoxia/reoxygenation. Susceptibility of MVEC to NK cytotoxicity was enhanced by TNF-alpha and slighty reduced by hypoxia/reoxygenation. CONCLUSIONS Endothelial activation with TNF-alpha, but not hypoxia/reoxygenation, induced NK cytotoxicity whereas the combination thereof induced the strongest NK cell adhesion. Our findings suggesting a role for NK cells in allograft responses support the development of anti-inflammatory treatment strategies to prevent I/R.
Collapse
Affiliation(s)
- Christine F Maurus
- Laboratory for Transplantation Immunology, Department for Internal Medicine, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | |
Collapse
|
35
|
Kitchens WH, Uehara S, Chase CM, Colvin RB, Russell PS, Madsen JC. The changing role of natural killer cells in solid organ rejection and tolerance. Transplantation 2006; 81:811-7. [PMID: 16570001 DOI: 10.1097/01.tp.0000202844.33794.0e] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Natural killer (NK) cells have emerged as a particular focus of interest in transplantation due to their ability to distinguish allogeneic major histocompatibility complex (MHC) antigens and their potent cytolytic effector mechanisms. Once relegated to the field of bone marrow transplantation, NK cells have recently been shown to participate in the immune response against solid organ allo- and xenografts. These new findings suggest that the role of NK cells in solid organ rejection and tolerance needs to be reexamined.
Collapse
Affiliation(s)
- William H Kitchens
- Division of Transplantation, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | | | | | | | | | | |
Collapse
|
36
|
Neuringer IP, Chalermskulrat W, Aris R. Obliterative bronchiolitis or chronic lung allograft rejection: a basic science review. J Heart Lung Transplant 2005; 24:3-19. [PMID: 15653373 DOI: 10.1016/j.healun.2004.01.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2003] [Revised: 12/12/2003] [Accepted: 01/06/2004] [Indexed: 01/06/2023] Open
Affiliation(s)
- Isabel P Neuringer
- Division of Pulmonary and Critical Care Medicine and Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA.
| | | | | |
Collapse
|
37
|
Smith RN, Brousaides N, Grazette L, Saidman S, Semigran M, Disalvo T, Madsen J, Dec GW, Perez-Atayde AR, Collins AB. C4d Deposition in Cardiac Allografts Correlates With Alloantibody. J Heart Lung Transplant 2005; 24:1202-10. [PMID: 16143234 DOI: 10.1016/j.healun.2004.07.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2004] [Revised: 06/16/2004] [Accepted: 07/04/2004] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The presence of C4d along the peritubular capillaries in kidney allografts correlates with the presence of anti-donor serum alloantibodies. We applied C4d staining to cardiac allograft and non-allograft biopsies to determine if C4d staining in heart allografts correlates with anti-donor serum alloantibodies. METHODS We stained for C4d all available frozen tissue biopsies from cardiac transplant recipients between 1997 and 2002, including autopsies. Two hundred twenty-one tissue samples from 124 patients were analyzed. Included in both groups were a variety of International Society for Heart and Lung Transplantation (ISHLT) grades of rejection plus post-implant cardiac ischemic injury (PIMI), and biopsies from patients who had received OKT3. Patients were matched by age, gender and interval after transplantation. Forty-four additional controls were included from patients biopsied for non-transplant-related cardiac disease. RESULTS C4d staining of the myocardial capillaries correlated well with the presence of anti-donor alloantibodies. Twenty-one of 25 biopsies from patients with anti-donor alloantibodies showed C4d staining (84%), whereas only 7 of 60 without anti-donor alloantibodies stained for C4d. C4d staining did not correlate with ischemia or OKT3 therapy. Only 4 of 44 non-transplant biopsies stained for C4d (9%). An example of the clinical utility of C4d staining in patient care is presented. CONCLUSIONS C4d staining of the capillaries in cardiac allografts correlates well with anti-donor serum alloantibodies, is a useful assay to verify alloantibody deposition, and can be used to establish one of the criteria for antibody-mediated cardiac rejections.
Collapse
Affiliation(s)
- Rex Neal Smith
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts 02114-2696, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Obara H, Nagasaki K, Hsieh CL, Ogura Y, Esquivel CO, Martinez OM, Krams SM. IFN-gamma, produced by NK cells that infiltrate liver allografts early after transplantation, links the innate and adaptive immune responses. Am J Transplant 2005; 5:2094-103. [PMID: 16095488 PMCID: PMC1473982 DOI: 10.1111/j.1600-6143.2005.00995.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The role of NK cells following solid organ transplantation remains unclear. We examined NK cells in acute allograft rejection using a high responder model (DA-->Lewis) of rat orthotopic liver transplantation. Recipient-derived NK cells infiltrated liver allografts early after transplantation. Since chemokines are important in the trafficking of cells to areas of inflammation, we determined the intragraft expression of chemokines known to attract NK cells. CCL3 was significantly increased in allografts at 6 h post-transplant as compared to syngeneic grafts whereas CCL2 and CXCL10 were elevated in both syngeneic and allogeneic grafts. CXCL10 and CX3CL1 were significantly upregulated in allografts by day 3 post-transplant as compared to syngeneic grafts suggesting a role for these chemokines in the recruitment of effector cells to allografts. Graft-infiltrating NK cells were shown to be a major source of IFN-gamma, and IFN-gamma levels in the serum were markedly increased, specifically in allograft recipients, by day 3 post-transplant. Accordingly, in the absence of NK cells the levels of IFN-gamma were significantly decreased. Furthermore, graft survival was significantly prolonged. These data suggest that IFN-gamma-producing NK cells are an important link between the innate and adaptive immune responses early after transplantation.
Collapse
MESH Headings
- Animals
- Chemokine CCL2/metabolism
- Chemokine CCL3
- Chemokine CCL4
- Chemokine CX3CL1
- Chemokine CXCL10
- Chemokines/metabolism
- Chemokines, CC/biosynthesis
- Chemokines, CX3C/metabolism
- Chemokines, CXC/metabolism
- Cytokines/metabolism
- Densitometry
- Enzyme-Linked Immunosorbent Assay
- Flow Cytometry
- Graft Rejection
- Immune System
- Immunity, Innate
- Inflammation
- Interferon-gamma/biosynthesis
- Interferon-gamma/metabolism
- Killer Cells, Natural/cytology
- Killer Cells, Natural/metabolism
- Liver Transplantation/methods
- Macrophage Inflammatory Proteins/biosynthesis
- Membrane Proteins/metabolism
- Models, Biological
- Rats
- Rats, Inbred Lew
- Ribonucleases/metabolism
- Time Factors
- Transplantation Immunology
- Transplantation, Homologous
- Transplantation, Isogeneic
Collapse
Affiliation(s)
- Hideaki Obara
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Kazuhito Nagasaki
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Christine L. Hsieh
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Yasuhiro Ogura
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Carlos O. Esquivel
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Olivia M. Martinez
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
| | - Sheri M. Krams
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492
- Address correspondence and reprint requests to: Dr. Sheri M. Krams, Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, MSLS P313, MC: 5492, Stanford, CA 94305-5492., Phone: (650) 498-6246, Fax: (650) 498-6250, email address:
| |
Collapse
|
39
|
Kirk AD, Mannon RB, Swanson SJ, Hale DA. Strategies for minimizing immunosuppression in kidney transplantation. Transpl Int 2005; 18:2-14. [PMID: 15612977 DOI: 10.1111/j.1432-2277.2004.00019.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunosuppression remains the cause of most morbidity following organ transplantation. However, its use is also responsible for the outstanding graft and patient survival rates commonplace in modern transplantation. Thus, the predominant challenge for transplant clinicians is to provide a level of immunosuppression that prevents graft rejection while preserving immunocompetence against environmental pathogens. This review will outline several strategies for minimizing or tailoring the use of immunosuppressive drugs. The arguments for various strategies will be based on clinical trial data rather than animal studies. A distinction will be made between conventional immunosuppressive drug reduction based on over-immunosuppression, and newer induction methods specifically designed to lessen the need for chronic immunosuppression. Based on the available data we suggest that most patients can be transplanted with less immunosuppression than is currently standard.
Collapse
Affiliation(s)
- Allan D Kirk
- Transplantation Branch, Department of Health and Human Services, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Center Drive, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
40
|
Wang H, Jiang J, Liu W, Kubelik D, Chen G, Gies D, Garcia B, Zhong R, Rother RP. Prevention of acute vascular rejection by a functionally blocking anti-C5 monoclonal antibody combined with cyclosporine. Transplantation 2005; 79:1121-7. [PMID: 15880054 DOI: 10.1097/01.tp.0000161218.58276.9a] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
BACKGROUND Inhibition of the complement cascade at C5 prevents formation of pro-inflammatory molecules C5a and C5b-9, which play a key role in allograft rejection. The present study was undertaken to determine whether blocking terminal complement with anti-C5 monoclonal antibody (mAb) alone or combined with cyclosporine (CsA) would prevent acute vascular rejection (AVR) in a mouse cardiac allograft model. METHODS C3H mouse hearts were transplanted into BALB/c mice and randomized into five groups with the following treatments: (1) no treatment; (2) CsA alone; (3) control mAb; (4) anti-C5 mAb alone; and (5) anti-C5 mAb and CsA. RESULTS Allografts in untreated or control mAb-treated recipients were rapidly rejected at 8.0+/-0.6 and 8.2+/-0.8 days, respectively. These grafts exhibited typical AVR, characterized by vasculitis, hemorrhage, and thrombosis. A high level of complement activity was also demonstrated in these animals. High-dose CsA was not able to inhibit complement activation or AVR, and grafts were rejected in 15.5+/-1.1 days. Anti-C5 monotherapy completely inhibited complement activation and attenuated AVR, but grafts were rejected in 8.3+/-0.5 days by acute cellular rejection. In contrast, a combination of anti-C5 mAb and CsA successfully achieved indefinite graft survival (>100 days). This combined therapy completely inhibited terminal complement activation and prevented both humoral- and cellular-mediated rejection. CONCLUSIONS Combination therapy of anti-C5 mAb and CsA achieves indefinite graft survival in a mouse cardiac allograft model. These data suggest that inhibition of terminal complement using anti-C5 mAb may be an effective therapeutic adjunct to prevent AVR in clinical transplantation.
Collapse
Affiliation(s)
- Hao Wang
- Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Madsen JC. Is thoracic organ transplantation ready for tolerance? Transplant Rev (Orlando) 2005. [DOI: 10.1016/j.trre.2005.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
42
|
Haun JB, Baldwin WM, Alevriadou BR. Clearance of complement by human vascular endothelial cells: effects of hypoxia/reoxygenation and IL-1beta activation. Transpl Int 2005; 18:475-82. [PMID: 15773971 DOI: 10.1111/j.1432-2277.2004.00075.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibody-mediated rejection is characterized by deposits of complement (C) C4 and C3 split products on endothelial cells (ECs). C3 split products are critical mechanistically and diagnostically because they are deposited in amplified quantities, bind covalently to ECs and act as ligands for leukocytes. This study was designed to determine whether cultured vascular human ECs could clear covalently bound C3 split products from their surface. An immunoglobulin M (IgM) antibody against beta(2)-microglobulin of major histocompatibility complex class I antigens was used to activate C in human serum. Some cells were exposed to hypoxia/reoxygenation and/or interleukin 1beta (IL-1beta) prior to incubation with antibody. C3b/iC3b and C3d deposition on the cell surface was measured by flow cytometry. Incubation with antibody followed by human serum caused a dose-dependent deposition of C3b/iC3b and C3d. Over half of deposited C3b/iC3b and one-third of C3d were cleared from the cell surface during a 3-7-h incubation period with human serum. Neither hypoxia/reoxygenation nor IL-1beta further increased the deposition of C3b/iC3b and C3d, and only slightly modulated their rates of clearance. In summary, human ECs rapidly clear iC3b and C3d from their surface. This finding may have important diagnostic and mechanistic implications to transplantation because C3d is used as a marker of antibody-mediated rejection.
Collapse
Affiliation(s)
- Jered B Haun
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
43
|
Xu J, von Aulock S, Lucas R, Wendel A. Potential of colony-stimulating factors to improve host defense in organ transplant recipients. Curr Opin Organ Transplant 2004. [DOI: 10.1097/01.mot.0000146562.43151.e4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
44
|
Baldwin WM, Armstrong LP, Samaniego-Picota M, Rahimi S, Zachary AA, Kasper EK, Conte JV, Hruban RH, Rodriguez ER. Antithymocyte globulin is associated with complement deposition in cardiac transplant biopsies. Hum Immunol 2004; 65:1273-80. [PMID: 15556677 DOI: 10.1016/j.humimm.2004.05.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Accepted: 05/04/2004] [Indexed: 11/16/2022]
Abstract
Polyclonal antithymocyte globulin preparations contain antibodies with reactivity to endothelial cells. Therefore, we investigated whether treatment with this reagent caused complement deposition in human cardiac transplants. Frozen tissue was available from endomyocardial biopsies of 75 patients, who were transplanted between April 1995 and April 2000. Nine of these patients were converted from cyclosporin A (CsA) to horse antithymocyte globulin (ATGAM) in the first month after transplantation. All of the biopsies were stained by immunofluorescence for C4d as evidence of activation of the classical pathway of complement. In addition, biopsies from patients treated with ATGAM and control patients were stained for deposition of horse immunoglobulin (Ig)G. All nine patients who received ATGAM had deposition of horse IgG and C4d. Two color stains demonstrated that the horse IgG colocalized with the C4d staining. No staining for horse IgG or C4d was evident in biopsies obtained before ATGAM treatment. Likewise, no staining for horse IgG was detected in seven control patients who had C4d staining. Most patients treated with ATGAM had no histologic evidence of rejection, but did have myocyte damage and macrophage infiltration. Thus prophylactic treatment with ATGAM is associated with the deposition of horse IgG and activation of complement in the transplant.
Collapse
Affiliation(s)
- William M Baldwin
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21205-2196, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rose ML. De novo production of antibodies after heart or lung transplantation should be regarded as an early warning system. J Heart Lung Transplant 2004; 23:385-95. [PMID: 15063397 DOI: 10.1016/j.healun.2003.08.028] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2003] [Revised: 07/28/2003] [Accepted: 08/02/2003] [Indexed: 11/17/2022] Open
Affiliation(s)
- Marlene L Rose
- National Heart and Lung Institute, Imperial College, Heart Science Centre, Royal Brompton and Harefield NHS Trust Hospitals, Harefield, Middlesex, UK.
| |
Collapse
|
46
|
Baldwin WM, Kasper EK, Zachary AA, Wasowska BA, Rodriguez ER. Beyond C4d: other complement-related diagnostic approaches to antibody-mediated rejection. Am J Transplant 2004; 4:311-8. [PMID: 14961982 DOI: 10.1111/j.1600-6143.2004.00348.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Complement is a multifunctional system of receptors and regulators as well as effector molecules. Both the pathogenic and diagnostic power of complement is based on the capacity of the complement system to amplify innate and adaptive immunity. This amplification is accomplished through two strategies: (1) enzymatic reactions in the complement cascade, and (2) stimulation of leukocytes, platelets and parenchymal cells through specific receptors or receptor-independent pore formation. The mechanisms by which complement mediates and modifies nonspecific inflammation, antibody-mediated injury and T-cell responses are of particular significance to the pathogenesis of transplant rejection. Understanding the mechanisms by which complement integrates the interactions of leukocytes, platelets and parenchymal cells offers opportunities to further refine the diagnosis of rejection.
Collapse
Affiliation(s)
- William M Baldwin
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | | | | | | | | |
Collapse
|
47
|
Hsieh CL, Ogura Y, Obara H, Ali UA, Rodriguez GM, Nepomuceno RR, Martinez OM, Krams SM. Identification, cloning, and characterization of a novel rat natural killer receptor, RNKP30: a molecule expressed in liver allografts. Transplantation 2004; 77:121-8. [PMID: 14724446 DOI: 10.1097/01.tp.0000110423.27977.6f] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND As a component of the innate immune system, natural killer (NK) cells may play a significant role in the early events after solid-organ transplantation. Activated NK cells have been shown to infiltrate allografts in transplant models. To better understand NK cells and the role of NK cell receptors in transplantation, we have cloned and begun characterizing a novel rat molecule, rNKp30. METHODS RNKp30 cDNA was cloned by 5' rapid amplification of cDNA ends polymerase chain reaction (PCR) and reverse transcriptase (RT)-PCR from mononuclear cells infiltrating a rejecting liver allograft. Southern blot analysis was used to determine the rNKp30 gene copy number. RT-PCR and Northern blotting were used to examine rNKp30 RNA expression in NK cells, multiple tissues, and liver grafts. Immunocytochemistry, immunoprecipitation, and Western blot analysis with two anti-rNKp30 polyclonal antibodies, CA680 and CA1071, were performed. Tunicamycin and endoglycosidase treatments determined the extent of rNKp30 glycosylation. RESULTS RNKp30 is homologous to human and macaque NKp30. It is a single copy gene with five identified single-nucleotide polymorphisms. RNKp30 is expressed by NK cells and is detectable as a single transcript by Northern blot in normal spleen, lymph node, and lung tissues. RNKp30 is a variably N-glycosylated cell surface molecule with a protein backbone of approximately 21 kDa. Elevated transcript expression of rNKp30 is detected in both rejected and spontaneously accepted liver allografts, but not in syngeneic or cyclosporine A-treated allografts. CONCLUSIONS RNKp30 is a glycosylated surface NK cell receptor with limited polymorphism. This putative activation receptor is expressed in liver allografts and may participate in the innate immune response after transplantation.
Collapse
MESH Headings
- Amino Acid Sequence/genetics
- Animals
- Antigens, Surface/metabolism
- Base Sequence/genetics
- Cloning, Molecular
- DNA, Complementary/genetics
- Female
- Lectins, C-Type/metabolism
- Liver/metabolism
- Liver Transplantation
- Male
- Molecular Sequence Data
- NK Cell Lectin-Like Receptor Subfamily B
- Natural Cytotoxicity Triggering Receptor 3
- Rats
- Rats, Inbred Strains
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Transcription, Genetic/physiology
- Transplantation, Homologous
Collapse
Affiliation(s)
- Christine L Hsieh
- Department of Surgery and Program in Immunology, Stanford University School of Medicine, Stanford, California 94305-5492, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Genetic modification strategies have the potential to improve outcome following cell/organ transplantation. A unique opportunity in transplantation is that gene therapies need not be restricted to in vivo approaches and that ex vivo genetic modification of cell and/or organs can be of value. Improvements in vector design, production, and delivery should enhance transfection efficiency and optimize gene expression. Herein, we discuss potential modes of gene therapy, focusing on viral, liposome, or naked DNA-based systems for gene delivery. We suggest gene therapy targets taking into consideration the essential constituents of anti-allograft repertory. In addition to strategies that may have salutary effects in mitigating the threat of acute rejection, we suggest genetic strategies for minimizing ischemia/reperfusion injury as well as for the perennial problem of progressive functional loss of the transplanted organ. Data from pre-clinical transplant models support the idea that gene therapy may improve allograft function and survival. We are optimistic that gene therapy will be of clinical value in the near future in the management of recipients of allografts; we believe that genetic strategies would be essential for successful breaching of the formidable challenge of xenotransplantation.
Collapse
Affiliation(s)
- Dolca Thomas
- Division of Nephrology, Department of Medicine, Weill Medical College of Cornell University, New York Weill Cornell Center, 525 East 68th Street, New York, NY 10021, USA
| | | |
Collapse
|
49
|
Abstract
The complement system plays a complex role in transplantation, beginning with effects on reperfusion injury and continuing with stimulation of the adaptive immune response. Recent evidence has emphasised the importance of the late components of the complement cascade in the mediation of post-ischaemic damage, which are apparently triggered by the classical, alternative or lectin pathways of complement activation, depending on the organ affected. In studies of renal allograft rejection, the local synthesis of complement component C3 seems to influence the T-cell response more strongly than circulating complement protein, raising the possibility that there is co-operation between locally derived C3 and antigen presentation in the graft. Class switching of alloantibody to a high-affinity IgG response is also highly dependent on C3. In addition, the finding that capillary-bound C4d is a robust marker for humoral rejection has started a new investigation into the significance of alloantibodies in acute and chronic allograft rejection. There are several selective and nonselective inhibitors suitable for clinical development; clearly it is time for more concerted effort to evaluate their role in clinical transplantation.
Collapse
Affiliation(s)
- Steven H Sacks
- Department of Nephrology and Transplantation, 5th Floor, Thomas Guy House, Guy's Hospital, King's College London, University of London, SE1 9RT, London, UK.
| | | | | |
Collapse
|
50
|
Abstract
Recent studies of endothelial cell biology have provided numerous original findings relevant to transplantation. The molecular mechanisms utilized by endothelial cells to regulate cell entry into the parenchyma are becoming more clearly defined. Emerging results have additionally elucidated how endothelial cells interact with and respond to T cells and antibodies specific for transplant antigens. Progress made in deciphering the cellular and molecular basis of endothelial cell-mediated inflammation has the potential to help with the identification of novel therapeutic targets for prolonging graft survival.
Collapse
Affiliation(s)
- Anna Valujskikh
- The Department of Immunology and The Glickman Urologic Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | |
Collapse
|