1
|
Park MJ, Na HS, Joo YS, Cho KH, Kim SY, Choi JW, Baek JA, Choi JY, You YK, Cho ML. Induction of liver transplant immune tolerance in an outbred rat strain model using tacrolimus. Lab Anim Res 2023; 39:5. [PMID: 36890604 PMCID: PMC9993642 DOI: 10.1186/s42826-023-00156-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Orthotopic liver transplantation is the only option for patients with end-stage liver disease and hepatocellular carcinoma. Post-transplant immunosuppressive therapy is important to prevent graft failure. We investigated the effectiveness of tacrolimus (FK506) and their mechanisms for liver transplant immune tolerance in an outbred rat LT model. RESULTS To investigate the therapeutic effect of the FK506 on outbred rat LT model, FK506 and postoperative therapy were administered subcutaneously once or twice daily to transplanted rats. Histopathological and immunohistochemical analyses were conducted for all groups. The regulation of inflammatory cytokine signaling in the spleen was analyzed by flow cytometry. FK506 attenuated allograft rejection and increased survival in rat orthotopic liver transplantation models. The FK506-treated group had reduced serum levels of alanine aminotransferase, aspartate aminotransferase and alkaline phosphatase. Furthermore, FK506 decreased the expression of inflammatory cytokines and the activation of pathogenic Th1 and Th17 cells in the liver. CONCLUSIONS Taken together, we revealed that FK506 ameliorated strong allograft rejection in outbred liver transplantation model by anti-inflammatory effect and inhibitory peroperty of pathogenic T cells.
Collapse
Affiliation(s)
- Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Sik Na
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Young-Shin Joo
- Department of Laboratory Animal Research Center, Catholic Medical Center, Institute of Biomedical Industry, The Catholic University of Korea, Banpo-daero, Seocho-gu, Seoul, Republic of Korea
| | - Keun-Hyung Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Won Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Ah Baek
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jong Young Choi
- Division of Hepatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Banpo-daero, Seocho-gu, Seoul, Republic of Korea
| | - Young Kyoung You
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Banpo-daero, Seocho-gu, Seoul, Republic of Korea.
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Impact Biotech, Seoul, 137-040, Republic of Korea.
| |
Collapse
|
2
|
Li Y, Chen Y, Zhang X, Geng L, Dai B, Lv X, Zhang P, Li H, Yang J, Huang Y, Xu F. Protective effect of electro-acupuncture on liver ischemia-reperfusion injury in rats. Exp Ther Med 2018; 16:1373-1380. [PMID: 30116387 DOI: 10.3892/etm.2018.6287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 11/23/2017] [Indexed: 01/18/2023] Open
Abstract
Liver ischemia-reperfusion injury is an important clinical complication in which excessive inflammation is a key factor; however, few studies have provided effective means of its regulation. As previous studies suggested that electro-acupuncture (EA) is able control excessive inflammation, the present study aimed to explore its effects on liver ischemia-reperfusion injury in experimental rats. The animals were randomly divided into surgery and sham groups, which were further divided into four sub-groups, including a non-treatment (NT), a non-point acupuncture (NPA), the non-selective nicotinic acetylcholine receptor (AChR) agonist 1,1-dimethyl-4-phenyl L-pioperazinium iodide (DMPPI) and an EA group. The alanine aminotransferase (ALT), serum cytokine and myeloperoxidase (MP) levels and the tissue pathology were evaluated after 90 min of ischemia followed by a 4, 8 or 24 h reperfusion. The results demonstrated that EA and DMPPI suppressed serum ALT elevation at 4 and 8 h reperfusion, whereas NPA did not. I/R induced hepatocellular necrosis, and cytoplasmic vacuolization and sinusoidal congestion was ameliorated by EA treatment after an 8 and 24 h reperfusion. In addition, EA also inhibited liver neutrophil accumulation, evidenced by a decreased MPO level at 8 h reperfusion. EA also suppressed the release of serum inflammatory factors TNF-α and IL-6 for the duration of reperfusion. However, little influence on IL-10 was observed. Mechanistically, vagus block by subphrenic vagotomy or mecamylamine hydrochloride abolished EA effect on liver damage, neutrophil accumulation and inflammatory factor release. In conclusion, it was demonstrated that EA protects the liver against I/R induced injury by inhibiting the inflammatory response, which is associated with the vagus.
Collapse
Affiliation(s)
- Yesheng Li
- Department of Hepatobiliary Surgery, Shanghai Public Health Clinical Center, Shanghai 200083, P.R. China
| | - Yi Chen
- Department of Hepatobiliary Surgery, Shanghai Public Health Clinical Center, Shanghai 200083, P.R. China
| | - Xinji Zhang
- Department of Health Statistics, Second Military Medical University, Shanghai 200433, P.R. China
| | - Li Geng
- Department of Special Treatment, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Binghua Dai
- Department of Special Treatment, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Xin Lv
- Department of Anesthesiology, Pulmonary Hospital, Tongji University, Shanghai 200438, P.R. China
| | - Ping Zhang
- Department of Experimental Research Center, Cancer Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Honghai Li
- Department of Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai 200240, P.R. China
| | - Jiamei Yang
- Department of Special Treatment, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| | - Yangqing Huang
- Department of Hepatobiliary Surgery, Shanghai Public Health Clinical Center, Shanghai 200083, P.R. China
| | - Feng Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Hospital, Second Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
3
|
Hua F, Chen Y, Yang Z, Teng X, Huang H, Shen Z. Protective action of bone marrow mesenchymal stem cells in immune tolerance of allogeneic heart transplantation by regulating CD45RB + dendritic cells. Clin Transplant 2018; 32:e13231. [PMID: 29488658 DOI: 10.1111/ctr.13231] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Bone marrow-derived mesenchymal stem cells (BMSCs) could exert a potent immunosuppressive effect and therefore may have a therapeutic potential in T-cell-dependent pathologies. We aimed to examine the effects of BMSCs on immune tolerance of allogeneic heart transplantation and the involvement of CD45RB+ dendritic cells (DCs). METHODS Bone marrow-derived DCs and BMSCs were co-cultured, with CD45RB expression on the surface of DCs measured by flow cytometry. qRT-PCR and Western blotting were used to detect mRNA and protein levels. Cytometric bead array was performed to determine the serum level of IL-10. Survival time of transplanted heart and expression of CD4+ , CD8+ , IL-2, IL-4, IL-10, IFN-γ were determined. Immunofluorescence assay was employed to determine intensity of C3d and C4d. RESULTS DCs co-cultured with BMSCs showed increased CD45RB and Foxp3 levels. CD45RB+ DCs co-cultured with T-cells CD4+ displayed increased T-cell CD4+ Foxp3 ratio and IL-10 than DCs. Both of them extended survival time of transplanted heart, decreased histopathological classification and score, intensity of C3d, C4d, proportion of CD4+ , expression levels of IL-2 and IFN-γ, and increased the CD4+ Foxp3 ratio and levels of IL-4 and IL-10. CD45RB+ DCs achieved better protective effects than DCs. CONCLUSION BMSCs increased the expression of CD45RB in the bone marrow-derived DCs, thereby strengthening immunosuppression capacity of T cells and immune tolerance of allogeneic heart transplantation.
Collapse
Affiliation(s)
- Fei Hua
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Yueqiu Chen
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Ziying Yang
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Haoyue Huang
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiac Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
4
|
Kaufmann C, Mortimer LA, Brereton HM, Irani YD, Parker DGA, Anson DS, Bachmann LM, Williams KA. Interleukin-10 Gene Transfer in Rat Limbal Transplantation. Curr Eye Res 2017; 42:1426-1434. [DOI: 10.1080/02713683.2017.1344714] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Claude Kaufmann
- Department of Ophthalmology, Lucerne Cantonal Hospital, Lucerne, Switzerland
| | - Lauren A Mortimer
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Helen M Brereton
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Yazad D Irani
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Douglas GA Parker
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Donald S Anson
- Department of Genetic Medicine, Women’s and Children’s Hospital, Adelaide, Australia
| | - Lucas M Bachmann
- Horten Centre for Patient Oriented Research, University of Zurich, Zurich, Switzerland
| | - Keryn A Williams
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| |
Collapse
|
5
|
Quaranta P, Focosi D, Freer G, Pistello M. Tweaking Mesenchymal Stem/Progenitor Cell Immunomodulatory Properties with Viral Vectors Delivering Cytokines. Stem Cells Dev 2016; 25:1321-41. [PMID: 27476883 DOI: 10.1089/scd.2016.0145] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal Stem Cells (MSCs) can be found in various body sites. Their main role is to differentiate into cartilage, bone, muscle, and fat cells to allow tissue maintenance and repair. During inflammation, MSCs exhibit important immunomodulatory properties that are not constitutive, but require activation, upon which they may exert immunosuppressive functions. MSCs are defined as "sensors of inflammation" since they modulate their ability of interfering with the immune system both in vitro and in vivo upon interaction with different factors. MSCs may influence immune responses through different mechanisms, such as direct cell-to-cell contact, release of soluble factors, and through the induction of anergy and apoptosis. Human MSCs are defined as plastic-adherent cells expressing specific surface molecules. Lack of MHC class II antigens makes them appealing as allogeneic tools for the therapy of both autoimmune diseases and cancer. MSC therapeutic potential could be highly enhanced by the expression of exogenous cytokines provided by transduction with viral vectors. In this review, we attempt to summarize the results of a great number of in vitro and in vivo studies aimed at improving the ability of MSCs as immunomodulators in the therapy of autoimmune, degenerative diseases and cancer. We will also compare results obtained with different vectors to deliver heterologous genes to these cells.
Collapse
Affiliation(s)
- Paola Quaranta
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy
| | - Daniele Focosi
- 2 North-Western Tuscany Blood Bank, Pisa University Hospital , Pisa, Italy
| | - Giulia Freer
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| | - Mauro Pistello
- 1 Department of Translational Research and New Technologies in Medicine and Surgery, Virology Section and Retrovirus Center, University of Pisa , Pisa, Italy .,3 Virology Unit, Pisa University Hospital , Pisa, Italy
| |
Collapse
|
6
|
Franz M, Doll F, Grün K, Richter P, Köse N, Ziffels B, Schubert H, Figulla HR, Jung C, Gummert J, Renner A, Neri D, Berndt A. Targeted delivery of interleukin-10 to chronic cardiac allograft rejection using a human antibody specific to the extra domain A of fibronectin. Int J Cardiol 2015; 195:311-22. [PMID: 26056964 DOI: 10.1016/j.ijcard.2015.05.144] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 05/13/2015] [Accepted: 05/26/2015] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND AIMS Management of chronic rejection is challenging since there are not sufficient preventive or therapeutic strategies. The rejection process leads to overexpression of ED-A(+) fibronectin (ED-A(+) Fn). The human antibody F8, specific to ED-A(+) Fn, may serve as a vehicle for targeted delivery of bioactive payloads, e.g. interleukin 10 (IL-10). The aim of this study was to investigate the therapeutic effects of the fusion protein F8-interleukin-10 (F8-IL10) in the process of chronic rejection development. METHODS A heterotopic rat heart transplantation model was used to induce chronic rejection. For therapeutic interventions, the immunocytokines F8-humanIL10 (DEKAVIL), F8-ratIL10 as well as KSF-humanIL10 (irrelevant antigen-specificity) were used. Treatment was performed weekly for 10 weeks starting at day 7 after transplantation (1mg/animal). RESULTS In the cardiac allografts, treatment with F8-huIL10 or F8-ratIL10 was associated with increased heart weights, a higher grade of chronic rejection, increased CIF, higher protein expression levels of alpha-smooth muscle actin (α-SMA), an augmented infiltration with inflammatory cells (CD4+, CD8+ and CD68+ cells) and higher serum levels of brain natriuretic peptide (BNP) compared to the control groups. CONCLUSIONS All observed treatment effects are transplantation-specific since the F8 antibody is specific to ED-A(+) Fn that is not expressed in healthy hearts. A clear targeting effect of F8-huIL10 as well as F8-ratIL10 could be proven. Against that background, a further study is needed to address the question, if F8-IL10 treatment is capable to reduce CAV and CIF starting at a time point when chronic rejection has fully developed (therapeutic approach).
Collapse
Affiliation(s)
- Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany.
| | - Fabia Doll
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Katja Grün
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Petra Richter
- Institute of Pathology, Jena University Hospital, Jena, Germany
| | - Nilay Köse
- Institute of Pathology, Jena University Hospital, Jena, Germany
| | - Barbara Ziffels
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Harald Schubert
- Institute of Laboratory Animal Science and Welfare, Jena University Hospital, Jena, Germany
| | - Hans R Figulla
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Christian Jung
- Department of Internal Medicine I, Jena University Hospital, Jena, Germany
| | - Jan Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart Center North Rhine-Westphalia, Ruhr-University of Bochum, Bad Oeynhausen, Germany
| | - André Renner
- Clinic for Thoracic and Cardiovascular Surgery, Heart Center North Rhine-Westphalia, Ruhr-University of Bochum, Bad Oeynhausen, Germany
| | - Dario Neri
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | | |
Collapse
|
7
|
Niu J, Yue W, Song Y, Zhang Y, Qi X, Wang Z, Liu B, Shen H, Hu X. Prevention of acute liver allograft rejection by IL-10-engineered mesenchymal stem cells. Clin Exp Immunol 2014; 176:473-84. [PMID: 24527865 DOI: 10.1111/cei.12283] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2014] [Indexed: 01/02/2023] Open
Abstract
Hepatic allograft rejection remains a challenging problem, with acute rejection episode as the major barrier for long-term survival in liver transplant recipients. To explore a strategy to prevent allograft rejection, we hypothesized that mesenchymal stem cells (MSCs) genetically engineered with interleukin-10 (IL-10) could produce beneficial effects on orthotopic liver transplantation (OLT) in the experimental rat model. Syngeneic MSCs transduced with IL-10 were delivered via the right jugular vein 30 min post-orthotopic transplantation in the rat model. To evaluate liver morphology and measure cytokine concentration, the blood and liver samples from each animal group were collected at different time-points (3, 5 and 7 days) post-transplantation. The mean survival time of the rats treated with MSCs-IL-10 was shown to be much longer than those treated with saline. According to Banff scheme grading, the saline group scores increased significantly compared with those in the MSCs-IL-10 group. Retinoid acid receptor-related orphan receptor gamma t (RORγt) expression was more increased in the saline group compared to those in the MSCs-IL-10 group in a time-dependent manner; forkhead box protein 3 (FoxP3) expression also decreased significantly in the saline group compared with those in the MSCs-IL-10 group in a time-dependent manner. The expression of cytokines [IL-17, IL-23, IL-6, interferon (IFN)-γ and tumour necrosis factor (TNF)-α] in the saline groups increased significantly compared with the time-point-matched MSCs-IL-10 group, whereas cytokine expression of (IL-10, TGF-β1) was deceased markedly compared to that in the MSCs-IL-10 group. These results suggest a potential role for IL-10-engineered MSC therapy to overcome clinical liver transplantation rejection.
Collapse
Affiliation(s)
- J Niu
- General Surgery of the Hospital Affiliated Xuzhou Medical College, Xuzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Prope tolerance to heart allografts in mice associated with persistence of donor interleukin-10-transduced stem cells. Transplantation 2012; 93:761-8. [PMID: 22487811 DOI: 10.1097/tp.0b013e318248372b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We previously reported that transduction of the human interleukin (IL)-10 gene into the total fetal liver stem cells (hIL-10-TFLs) of mice protects against their rejection in an allogeneic host. In this study, we explored the effects of these cells in two different models of organ transplantation. METHODS Balb/c mice were sublethally irradiated before receiving skin or vascularized heterotopic heart grafts from C57Bl/6 mice. TFLs from C57Bl/6 mice transduced with hIL-10 or untransduced TFLs were injected on the day of transplantation into recipient mice once or also every 20 days thereafter. RESULTS Skin allograft survival was prolonged for up to 17.8±0.6 days, vs. 9.0±0.4 days, in mice that received hIL-10-TFLs or untransduced TFLs, respectively. Allogeneic heart transplants survived for 86.25±13.8, 46.3±4.6, 28.1±6.1, or 11.5±0.6 days in mice that received repeated injections of hIL-10-TFLs, a single injection of hIL-10-TFLs, repeated injections of untransduced TFLs, or controls, respectively. Histological analyses of the grafts showed fewer inflammatory foci and CD8+ infiltrating cells in mice injected with hIL-10-TFLs compared with untreated mice. Expressions of H-2b and hIL-10 were found in several organs, including the thymus, liver, and the transplant, in hIL-10-TFL-injected mice. Finally, in hIL-10-TFL-injected mice, FoxP3 T cells were present inside the transplanted heart as late as 140 days after transplantation. CONCLUSIONS In this study, we showed that repeated injections of hIL-10-TFLs are efficient in mitigating transplant rejection. This "prope" tolerance was associated with survival of donor hematopoietic cells in the host.
Collapse
|
9
|
Oishi H, Okada Y, Kikuchi T, Hoshikawa Y, Sado T, Noda M, Endo C, Sakurada A, Matsumura Y, Kondo T. Transbronchial human interleukin-10 gene transfer reduces acute inflammation associated with allograft rejection and intragraft interleukin-2 and tumor necrosis factor-alpha gene expression in a rat model of lung transplantation. J Heart Lung Transplant 2010; 29:360-7. [PMID: 20202600 DOI: 10.1016/j.healun.2009.10.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 10/03/2009] [Accepted: 10/04/2009] [Indexed: 10/19/2022] Open
Abstract
BACKGROUND The ability to express genes with potential immunoregulatory capacity could reduce allograft rejection (AR). This study examined the effect of ex vivo lipid-mediated transbronchial human interleukin-10 (hIL-10) gene transfer on AR and the intragraft cytokine profile in a rat model of lung transplantation. METHODS Left single lung transplants were performed between a highly histoincompatible combination of inbred rats. The donor left lung was extracted and intrabronchially instilled with a plasmid encoding hIL-10 (IL-10 group) or Escherichia coli beta-galactosidase (control group), mixed with a cationic lipid. At 3 and 6 days after transplantation, the degree of AR was graded histologically (stage 1-4) and several pathologic categories of inflammation were scored on a scale of 0 to 4 according to the percentage of involvement. Intragraft cytokine profile was examined by real-time reverse transcription polymerase chain reaction. RESULTS The stage of AR (3.1 +/- 0.4 vs 3.8 +/- 0.4) and the pathologic scores for edema (2.3 +/- 0.8 vs 3.2 +/- 0.4), intraalveolar hemorrhage (0.3 +/- 0.5 vs 2.2 +/- 0.8), and necrosis (0.3 +/- 0.5 vs 1.2 +/- 0.4) in the IL-10 group were significantly decreased compared with the control group at Day 6. IL-2 and tumor necrosis factor-alpha messenger RNA expression levels on Day 3 were significantly decreased in the IL-10 group. CONCLUSIONS Ex vivo lipid-mediated transbronchial hIL-10 gene transfer attenuated acute inflammation associated with AR, which was related to decreased levels of proinflammatory cytokine gene expression in a rat model of lung transplantation.
Collapse
Affiliation(s)
- Hisashi Oishi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Kim YH, Lim DG, Wee YM, Kim JH, Yun CO, Choi MY, Park YH, Kim SC, Han DJ. Viral IL-10 gene transfer prolongs rat islet allograft survival. Cell Transplant 2008; 17:609-18. [PMID: 18819249 DOI: 10.3727/096368908786092694] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Islet transplantation is a potential cure for diabetes. However, allotransplant rejection severely limits its clinical application. In this study, we sought to transfect rat islets with an adenoviral vector containing the viral IL-10 (vIL-10) gene and examine its efficacy in preventing graft rejection. The immunosuppressive effect of vIL-10 is reported but its efficacy is somehow debatable in transplantation model. vIL-10 transfected islets were transplanted into streptozotocin-induced diabetic rats. Blood glucose, serum vIL-10 concentration, graft histology, and graft cytokine expression were used to monitor graft function up to day 21 after transplantation. Transfected islets released a large amount of vIL-10 protein without affecting their viability and functional integrity. When we transplanted the transfected islets into allogeneic hosts, the survival of grafted islets was not significantly increased. However, the combined use of vIL-10 and subtherapeutic doses of CsA (cyclosporine) significantly prolonged graft survival beyond that achieved with either agent alone (p < 0.001). vIL-10 and CsA-treated rats contain high level of vIL-10 in serum, which is evidenced by the inhibition of allogeneic mixed lymphocyte reaction (MLR). Histological analysis additionally revealed the presence of viable islets up to 21 days. IL-10 mRNA expression in grafted liver was higher and IFN-gamma mRNA was lower in vIL-10 and CsA-treated animals, compared with other groups. The synergistic effect of this combination therapy is potentially correlated with the induction of inhibitory cytokine secretion and downregulation of proinflammatory cytokine secretion from host cells.
Collapse
Affiliation(s)
- Yang-Hee Kim
- Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, 138-736, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Novel immunosuppression: R348, a JAK3- and Syk-inhibitor attenuates acute cardiac allograft rejection. Transplantation 2008; 85:885-92. [PMID: 18360272 DOI: 10.1097/tp.0b013e318166acc4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Janus kinase (JAK)3 is crucial for signal transduction downstream of various cytokine receptors in immune cells. This is the first report on the novel JAK3 inhibitor R348. METHODS (1) Detailed pharmacokinetic data were obtained in rats; (2) multiple in vitro enzyme inhibition assays were performed to characterize the drug; (3) prevention of acute rejection was investigated in animals treated with different doses of R348 or rapamycin for 5 days; and (4) cardiac allograft survival after a 10-day treatment period was studied for various regimens of R348, tacrolimus, or rapamycin; combination indices were calculated to evaluate drug interactions. RESULTS (1) Plasma levels of R348's active metabolite R333 sustained high for 8 hr or more, depending on the dose. (2) In vitro enzyme assays showed potent inhibition of JAK3- and Syk-dependent pathways. (3) R348 40 mg/kg preserved graft function, significantly reduced graft infiltration, and decreased histologic ISHLT rejection scores on postoperative day 5. Results were similar to those of rapamycin 3 mg/kg. Likewise, both drugs significantly reduced the cellular Th1 and Th2 immune responses, as determined by enzyme-linked immunosorbent assays. Intragraft inflammatory cytokine upregulation was similarly suppressed by R348 and rapamycin. R348 10 mg/kg was subtherapeutic. (4) Allograft survival was similar for R348 20 and 40 mg/kg, which was comparable with therapeutically dosed tacrolimus or rapamycin. In combination regimens, R348 demonstrated highly beneficial synergistic interactions with tacrolimus. CONCLUSIONS R348 is a promising novel JAK3/Syk-inhibitor with favorable pharmacokinetics and biological activity. It effectively diminishes acute cardiac allograft rejection and is suitable for combination regimens with tacrolimus.
Collapse
|
12
|
Zaldumbide A, Hoeben RC. How not to be seen: immune-evasion strategies in gene therapy. Gene Ther 2007; 15:239-46. [PMID: 18046427 DOI: 10.1038/sj.gt.3303082] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The development of efficient and safe vectors for gene delivery paved the way for evolution of gene therapy as a new modality for treatment of various inherited disorders and for cancer. The current vectors, viral and non-viral, have their limitations. Innate and adaptive immune responses to vector particles and components may restrict the efficiency of gene transfer and the persistence of expression of the transgene. Results from preclinical studies in animals and more recently data from clinical studies have demonstrated the potential impact of the cellular and the humoral immune response on the therapeutic efficacy. Not only the vector components, but also the transgene products may induce an immune response that negatively affects the therapeutic efficacy. The induction of a cytotoxic T-cell response to transgene-encoded peptides, as well as the production of antibodies directed against secreted proteins have been reported in preclinical and clinical studies, and these may thwart those applications that require long-term expression. Here we will review some of the options to blunt the acquired immune responses to transgene-encoded polypeptides.
Collapse
Affiliation(s)
- A Zaldumbide
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
13
|
Ke B, Shen XD, Tsuchihashi SI, Gao F, Araujo JA, Busuttil RW, Ritter T, Kupiec-Weglinski JW. Viral interleukin-10 gene transfer prevents liver ischemia-reperfusion injury: Toll-like receptor-4 and heme oxygenase-1 signaling in innate and adaptive immunity. Hum Gene Ther 2007; 18:355-66. [PMID: 17439357 DOI: 10.1089/hum.2007.181] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) contributes to early and late dysfunction of liver transplants. We have shown that sentinel Toll-like receptor-4 (TLR4) plays a key role in the activation of T cell immune responses during hepatic IRI. We have also documented that overexpression of heme oxygenase-1 (HO-1) exerts potent cytoprotective effects. This study analyzes how adenovirus (Ad)-based viral interleukin-10 (vIL-10) gene transfer affects TLR4 and HO-1 signaling in host innate and adaptive immunity during liver IRI. Using a partial lobar warm IRI model, groups of wild-type and HO-1(+/-) knockout (KO) mice were assessed for severity of hepatocellular damage after 90 min of warm ischemia followed by 6 hr of reperfusion. Both wild-type and HO-1 (+/-) KO mice treated with Ad-vIL-10 have shown improved hepatic function (serum glutamic-oxaloacetic transaminase levels), ameliorated histological signs of IRI (Suzuki's score), decreased neutrophil accumulation (myeloperoxidase activity), and depressed tumor necrosis factor-alpha/IL-1beta, IL-2/interferon-gamma, E-selectin, and macrophage inflammatory protein-2 expression. These effects were IL-10 dependent as treatment with neutralizing antibody re-created liver IRI. In contrast, untreated wild-type and HO-1 (+/-) KO mice, as well as wild-type and HO-1 (+/-) KO mice treated with Ad-beta-Gal, showed severe hepatocellular damage due to IRI. Unlike in controls, wild-type and HO-1 (+/-) KO mice treated with Ad-vIL-10 revealed markedly depressed TLR4 and NF-kappaB expression, along with increased HO-1 and Bcl-2/Bcl-x(L) expression, as compared with respective controls. Thus, vIL-10 gene transfer prevents hepatic IRI in association with depressed expression of innate TLR4, and adaptive Th1 cytokine/chemokine programs. The induction of antioxidant HO-1 and anti-apoptotic Bcl-2/Bcl-x(L) by vIL-10 exerts synergistic cytoprotective function against antigen-independent hepatic inflammatory response triggered by IRI.
Collapse
Affiliation(s)
- Bibo Ke
- Dumont-UCLA Transplant Center, Department of Surgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Oishi H, Okada Y, Kikuchi T, Sado T, Oyaizu T, Hoshikawa Y, Suzuki S, Matsumura Y, Kondo T. Lipid-Mediated Transbronchial Human Interleukin-10 Gene Transfer Decreases Acute Inflammation Associated With Allograft Rejection in a Rat Model of Lung Transplantation. Transplant Proc 2007; 39:283-5. [PMID: 17275523 DOI: 10.1016/j.transproceed.2006.10.207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Indexed: 11/21/2022]
Abstract
BACKGROUND Transferring genes with immunoregulatory capacity to transplanted organs has the potential to modify allograft rejection (AR). We examined the effect of ex vivo lipid-mediated transbronchial human interleukin-10 (hIL-10) gene transfer on acute AR in a rat model of lung transplantation. METHODS Left single lung transplantations were performed between a highly histoincompatible rat combination: Brown Norway to Lewis. The extracted donor left lung was intrabronchially instilled with a plasmid encoding hIL-10 or Escherichia coli beta-galactosidase (control), mixed with a cationic lipid. On day 6 posttransplantation, the degree of AR was graded histologically (stages 1-4) based upon pathological categories of inflammation: perivascular, peribronchial, and peribronchiolar lymphocytic infiltrates, edema, intraalveolar hemorrhage, and necrosis. RESULTS The stage of AR in the IL-10 group (3.1 +/- 0.4) was significantly lower than the control group (3.8 +/- 0.4). Pathological scores for edema, intraalveolar hemorrhage, and necrosis in the IL-10 group (2.3 +/- 0.8, 0.3 +/- 0.5, and 0.3 +/- 0.5, respectively) were also significantly decreased compared with those in the control group (3.2 +/- 0.4, 2.2 +/- 0.8, and 1.2 +/- 0.4, respectively). CONCLUSION Ex vivo lipid-mediated transbronchial hIL-10 gene transfer attenuated acute inflammation associated with AR in a rat model of lung transplantation.
Collapse
Affiliation(s)
- H Oishi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Oi K, Davies WR, Tazelaar HD, Bailey KR, Federspiel MJ, Russell SJ, McGregor CGA. Ex vivo hypothermic recirculatory adenoviral gene transfer to the transplanted pig heart. J Gene Med 2006; 8:795-803. [PMID: 16652399 DOI: 10.1002/jgm.913] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND To facilitate the application of adenoviral gene therapy in clinical heart transplantation, we developed an ex vivo hypothermic recirculatory adenoviral gene transfer method to the transplanted pig heart. METHODS Experimental animals were assigned into three groups; controls, 1x10(8) plaque-forming units (pfu)/ml group and 1x10(9) pfu/ml group. During the 30 min gene transfer perfusion, 200 ml of University of Wisconsin solution containing the adenoviral vector was recirculated through the coronary vessels. The myocardial temperature was maintained below 4 degrees C and the perfusion pressure was adjusted at 50 mmHg. RESULTS Cardiac myocyte transduction efficiencies in the 1x10(8) pfu/ml group were 0.04% and 0.07%, whereas transduction efficiencies in the 1x10(9) pfu/ml group were widely distributed from 0.45% to 22.62%. The gene transduction efficiency increased with the virus titer. Additionally, no difference in the transduction efficiency was observed between different segments of the left ventricle. The current gene transfer method at 1x10(9) pfu/ml of adenovirus titer enabled homogeneous gene transduction into the transplanted pig heart up to a maximum of 22.62%. CONCLUSIONS This model can be applied to a large isolated heart and will greatly facilitate the investigation of gene therapy in large animal models of heart transplantation.
Collapse
Affiliation(s)
- Keiji Oi
- Mayo Clinic William J. von Liebig Transplant Center, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Okada Y, Zuo XJ, Toyoda M, Marchevsky A, Matloff JM, Oishi H, Kondo T, Jordan SC. Adenovirus mediated IL-10 gene transfer to the airway of the rat lung for prevention of lung allograft rejection. Transpl Immunol 2006; 16:95-8. [PMID: 16860711 DOI: 10.1016/j.trim.2006.03.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Accepted: 03/09/2006] [Indexed: 11/22/2022]
Abstract
BACKGROUND The ability to express genes with potential immunoregulatory capacity could reduce allograft rejection (AR). We examined the feasibility of transferring the viral interleukin-10 (vIL-10) gene into rat lungs by intra-bronchial instillation and the subsequent effects of delivered vIL-10 on acute lung AR. METHODS First, the adenoviral beta-galactosidase vector (adv-beta-gal) particles were instilled into the airway of the rat lung and protein synthesis of beta-gal was examined by histochemical staining. Next, the ability of the adenoviral vIL-10 vector (adv-vIL-10) transfection to modify AR was examined in a highly histoincompatible rat lung transplant model (BN-->Lew). Donor left lungs were transfected with 3 x 10(8) pfu/0.3 mL of adv-vIL-10 (vIL-10 group) or adv-beta-gal (control group) 3 days before transplantation. On day 6 post-transplant, lung allografts were harvested and AR was graded histologically (stage 0-4). Several pathological categories of inflammation (perivascular, peribronchial, or peribronchiolar mononuclear infiltrates, edema, vasculitis, intraalveolar hemorrhage, and necrosis) were also examined and scored on a scale of 0-4 as previously described. RESULTS A successful transgene protein synthesis by adv-beta-gal in alveolar epithelial cells and alveolar macrophages was confirmed by histochemical staining with X-gal. The vIL-10 group showed a trend toward an improved stage of AR (3.75 +/- 0.5 vs. 4.0 +/- 0), and also a decreased pathological scores for edema (3.5 +/- 0.6 vs. 4.0 +/- 0), intraalveolar hemorrhage (2.3 +/- 1.0 vs. 2.5 +/- 0.6) and necrosis (1.5 +/- 0.5 vs. 1.75 +/- 1.3) compared with the control group, however, the differences in any pathological scores between the two groups did not reach a statistical significance. CONCLUSIONS 1. A successful transgene protein synthesis in alveolar epithelial cells was ensured by intra-bronchial instillation of an adenoviral vector encoding beta-galactosidase gene. 2. Transferring the vIL-10 gene into rat lungs by intra-bronchial instillation did not seem to reduce lung AR significantly, as opposed to the results of our previous experiments in a rat cardiac allograft model. This discrepancy may be explained by several potential factors including the immunogenecity of adenoviral vectors in conjunction with the nature of the lung more susceptible to immune response and inflammation.
Collapse
Affiliation(s)
- Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, 4-1 Seiryomachi, Aoba-ku, Sendai 980-8575, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Moore F, Buonocore S, Paulart F, Thielemans K, Goldman M, Flamand V. Unexpected effects of viral interleukin-10-secreting dendritic cells in vivo: preferential inhibition of TH2 responses. Transplant Proc 2005; 36:3260-6. [PMID: 15686742 DOI: 10.1016/j.transproceed.2004.10.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral interleukin (IL)-10 (vIL-10) has been widely described as an immunoregulatory cytokine that does not possess the T-cell costimulatory activities of cellular IL-10; it was therefore believed to be a more potent tolerogenic mediator. The immunosuppressive properties of this cytokine are partly attributed to its capacity to render dendritic cells (DCs) unable to undergo full maturation and to activate T cells. We reported here that myeloid DCs retrovirally transduced with vIL-10 had an impaired production of IL-12 and a decreased expression of MHC class II molecules but had minor defects in costimulatory molecule expression and no alteration on CCR5 and CCR7 expression. In mixed leukocyte reaction, vIL-10-transduced C57BL/6 bm12 (MHC class II mismatch) DCs had a reduced capacity to stimulate C57BL/6 wild-type CD4+ T-cell proliferation. We show that bm12 vIL-10-transduced DC administration in CD8-/- C57BL/6 mice promoted IFN-gamma production, down-regulated TH2-type cytokine production, and did not induce skin graft tolerance. These findings suggest that vIL-10-transduced DC may surprisingly facilitate Th1-type inflammatory responses in vivo.
Collapse
Affiliation(s)
- F Moore
- Institute for Medical Immunology, Université Libre de Bruxelles
| | | | | | | | | | | |
Collapse
|
18
|
Zhao J, Pettigrew GJ, Bolton EM, Murfitt CR, Carmichael A, Bradley JA, Lever AML. Lentivirus-mediated gene transfer of viral interleukin-10 delays but does not prevent cardiac allograft rejection. Gene Ther 2005; 12:1509-16. [PMID: 15858608 DOI: 10.1038/sj.gt.3302547] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Human immunodeficiency virus (HIV)-based lentiviral vectors expressing viral interleukin-10 (vIL-10) were used to transduce rat cardiac allografts with the aim of extending graft survival. vIL-10 expression was first shown, by RT-PCR, to persist in transduced heart isografts for at least 28 days after transduction. Cardiac transplants were performed in a fully allogeneic rat strain combination (Lewis to DA); allografts transduced by vectors expressing vIL-10 showed significantly prolonged survival (14.5 vs 7.5 days median survival time). Mixed lymphocyte reactions (MLRs) were used to determine the influence, in vitro, of vIL-10 on alloantigen-induced T-cell proliferation. Bioactive vIL-10, produced by DA rat aortic endothelial cells transduced with HIV-PGK-vIL-10, was added to MLRs at different time points and lymphocyte proliferation was assessed by uptake of [3H]thymidine. T-cell proliferation was inhibited by >80% when vIL-10 was added to the MLR at day 1, 2 or 3 of coculture. The inhibitory effect was significantly decreased when addition of vIL-10 was delayed until day 4 or 5 (47 and 35% inhibition, respectively). The extended graft survival time is comparable to that using adenoviral vectors delivering vIL-10 in a similar rat strain combination. The limited improvement in survival may be due to lack of inhibition of the early phase of the alloimmune response as suggested by in vitro studies confirming that maximum suppression of the MLR by vIL-10 can only be achieved if the cytokine is present at the initiation of alloimmune recognition. The delay in expression of vIL-10 from the lentiviral vector means that protocols must be developed to suppress the early stages of alloimmune stimulation before vIL-10 is produced.
Collapse
Affiliation(s)
- J Zhao
- Department of Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | |
Collapse
|
19
|
|
20
|
Salgar SK, Yang D, Ruiz P, Miller J, Tzakis AG. Viral interleukin-10-engineered autologous hematopoietic stem cell therapy: a novel gene therapy approach to prevent graft rejection. Hum Gene Ther 2004; 15:131-44. [PMID: 14975186 DOI: 10.1089/104303404772679940] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Epstein-Barr virus-encoded protein BCRF1 (viral interleukin [vIL]-10) is a biologically active homologue of cellular interleukin (IL)-10. In this study, a novel gene therapy approach to prolong allograft survival was designed. Autologous (syngeneic) hematopoietic progenitor/stem cell-enriched (HSC; lineage(-ve)) population derived from CBA/J mouse bone marrow were transduced with retrovirus encoding vIL-10 gene (vIL-10-HSC), ex vivo; vIL-10-HSC were injected (4-6 x 10(6) cells intravenously) into lethally (9.5 Gy) or sublethally (4 Gy) irradiated CBA/J mice. Six weeks after vIL-10-HSC administration, vascular heterotopic heart (C57BL/6) transplantation was performed. Ex vivo, the vIL-10-HSC produced 5.4 +/- 0.5 ng of vIL-10 protein/2 x 10(5) cells per 24 hr. In vivo, serum vIL-10 production was 187 +/- 205 pg/ml during 3-10 weeks after vIL-10-HSC administration. Cardiac allograft survival was prolonged (p < 0.004) in lethally (71 +/- 40 days) and sublethally (114 +/- 15 days) irradiated mice that received vIL-10-HSC compared to controls that received unengineered (UE) HSC or vector DNA-engineered HSC (12-16 days). However, secondary skin graft (C57BL/6) survival was not prolonged in cardiac allograft-tolerant animals. In the vIL-10-HSC-administered group, graft histopathology demonstrated mild arteritis/venulitis (grade 0.7) and rejection (grade 1.0). Intragraft expression of costimulatory molecules (B7.1, B7.2), cytokines (IL-2, IL-4, mIL-10, interferon [IFN]-gamma), and inducible nitric oxide synthase (iNOS) molecules was markedly lower in vIL-10-HSC-treated tolerant grafts that survived more than 100 days compared to vector DNA-HSC- or UE-HSC-treated controls. Furthermore, T lymphocytes derived from vIL-10-HSC-treated tolerant recipients demonstrated hyporeactivity to donor antigens in mixed lymphocyte cultures. Administration of autologous vIL-10-engineered HSC prior to organ transplantation prolonged cardiac allograft survival significantly.
Collapse
|
21
|
Lucas A, McFadden G. Secreted Immunomodulatory Viral Proteins as Novel Biotherapeutics. THE JOURNAL OF IMMUNOLOGY 2004; 173:4765-74. [PMID: 15470015 DOI: 10.4049/jimmunol.173.8.4765] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Many viruses have learned to evade or subvert the host antiviral immune responses by encoding and expressing immunomodulatory proteins that protect the virus from attack by elements of the innate and acquired immune systems. Some of these viral anti-immune regulators are expressed as secreted proteins that engage specific host immune targets in the extracellular environment, where they exhibit potent anti-immune properties. We review here viral immunomodulatory proteins that have been tested as anti-inflammatory reagents in animal models of disease caused by excessive inflammation or hyperactivated immune pathways. The potential for such viral molecules for the development of novel drugs to treat immune-based or inflammatory disorders is discussed.
Collapse
Affiliation(s)
- Alexandra Lucas
- BioTherapeutics Research Group, Robarts Research Institute, London, Ontario, Canada
| | | |
Collapse
|
22
|
Abstract
Kidney-targeted gene therapy could be an ideal treatment for renal diseases since the therapeutic molecule is limited in the kidney and the systemic effect may be minimized. The technical development of the gene delivery to kidney and the identification of the responsive gene for a particular disease encourage the challenge to hereditary diseases. Collagen type IV reassembling was reported to be succeeded in Alport syndrome model by introduction of exogenous COL4A5 gene. Many gene therapies are evaluated in various glomerulonephritis models and unilateral ureteral obstruction (UUO) model, and favorable results are accumulated. Transplant kidney is an ideal target for gene therapy, by which ischemia reperfusion, acute rejection and chronic allograft nephropathy can be treated. The importation of the novel technology, for example hybrid stem cell-gene therapy could promote the gene therapy of renal diseases toward clinical application.
Collapse
Affiliation(s)
- Enyu Imai
- Division of Nephrology, Department of Internal Medicine, Osaka University Graduate School of Medicine, Japan.
| | | | | | | |
Collapse
|
23
|
Tomasoni S, Longaretti L, Azzollini N, Gagliardini E, Mister M, Buehler T, Remuzzi G, Benigni A. Favorable effect of cotransfection with TGF-beta and CTLA4Ig of the donor kidney on allograft survival. Am J Nephrol 2004; 24:275-83. [PMID: 15057025 DOI: 10.1159/000077611] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 02/12/2004] [Indexed: 12/20/2022]
Abstract
BACKGROUND/AIMS Gene transfer of viral interleukin 10 (vIL-10) or transforming growth factor beta (TGF-beta) successfully prolonged liver and heart graft survival. Here we assessed whether injection of adenovirus (Ad) coding vIL-10 (AdvIL-10) or TGF-beta3 (AdTGF-beta3) prolonged kidney allograft survival. Since we previously demonstrated that transfection of the donor kidney with CTLA4Ig significantly prolonged allograft survival, we also evaluated the effect of a combined injection of AdvIL-10 or AdTGF-beta3 with the AdCTLA4Ig. METHODS Adenoviral vectors or saline were ex vivo injected into the renal artery of Brown Norway (RT.1n) donor kidneys subsequently grafted into Lewis (RT.1(l)) rats. Graft survival, transgene expression, graft cell infiltration, and histological changes were assessed. RESULTS Allografts of saline or Ad-beta-galactosidase controls were promptly rejected (mean survival time +/- SE 7.6 +/- 0.2 and 7.8 +/- 0.3 days, respectively). AdvIL-10 significantly prolonged survival only in 2 out of 9 animals (23.2 +/- 9.9 days), with vIL-10 expression detected on day 4. Survival was prolonged in 1 out of 5 animals by AdTGF-beta3 (14.4 +/- 5.3 days) despite the fact that the transgene was still observed after 14 days. While the combined injection of AdvIL-10 with AdCTLA4Ig did not protect the kidney from rejection (17.4 +/- 4.6 days), AdTGF-beta3 added to AdCTLA4Ig consistently prolonged the allograft lifespan in all animals (70.6 +/- 39.6 days), inducing indefinite survival in 1 animal which showed long-term gene expression and T cells hyporesponsive to alloantigens. CONCLUSION Overexpression of AdTGF-beta3 concomitant with the blockade of the CD28/B7 pathway by AdCTLA4Ig induces strong immunosuppression that occasionally allows the acceptance of a fully major histocompatibility complex mismatched renal graft.
Collapse
Affiliation(s)
- Susanna Tomasoni
- Mario Negri Institute for Pharmacological Research, Institute for Pharmacological Research, Bergamo, Italy
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Salgar SK, Yang D, Ruiz P, Miller J, Tzakis AG. Viral interleukin-10 gene therapy to induce tolerance to solid organ transplants in mice. Transplant Proc 2004; 36:397-8. [PMID: 15050171 DOI: 10.1016/j.transproceed.2003.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
In this study, a novel gene therapy approach to prolong allograft survival was designed. Autologous (syngeneic) hematopoietic stem cell-enriched bone marrow cells (HSC; lin(-)) engineered with the vIL-10 gene (vIL-10-HSC) were injected (4 to 6 x 10(6) cells, i.v.) into lethally (9.5 Gy) or sublethally (4 Gy) irradiated CBA/J mice 6 weeks prior to allogeneic heart (C57BL/6) transplantation (Tx). Cardiac allograft survival was significantly (P <.004) prolonged in lethally (71 +/- 40 days) and sublethally (114 +/- 15 days) irradiated mice that received vIL-10-HSC compared to controls that received no HSC (11 +/- 1 days), unengineered HSC, or vector-DNA-engineered HSC (12 to 16 days). Tolerant graft histopathology demonstrated mild arteritis/venulitis (grade 0.7) and rejection (grade 1.0). Intragraft expression of costimulatory molecules (B7.1, B7.2), cytokines (IL-2, IL-4, mIL-10, IFN-gamma), and iNOS molecules were markedly lower in tolerant grafts that survived for >100 days; recipient T lymphocytes demonstrated hyporeactivity to donor and third-party antigens in mixed lymphocyte cultures. These findings have important implications and potential therapeutic applications in transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- S K Salgar
- Department of Surgery, University of Miami, Miami, FL 33136, USA.
| | | | | | | | | |
Collapse
|
25
|
Abstract
Somatic cell gene therapy has made considerable progress last five years and has shown clear success in some clinical trials. In the field of nephrology, both the elucidation of pathophysiology of renal diseases and the development of gene transfer technique have become driving force for new therapy of incurable renal diseases, such as Alport syndrome and polycystic kidney disease. Gene therapy of renal cancer, although its application is limited to advanced cancer, is the front-runner of clinical application. Erythropoietin gene therapy has provided encouraging results for the treatment of anemia in uremic rats and recently progressed to the inducible one in response to hypoxia. Gene therapy for glomerulonephritis and renal fibrosis showed prominent impact on experimental models, although the safety must be confirmed for prolonged treatment. Transplant kidney is an ideal material for gene modification and induction of tolerance in the transplant kidney is an attractive challenge. Emerging techniques are becoming available such as stem cell technology and messenger RNA silencing strategies. We believe that the future of gene therapy research is exciting and promising and it holds an enormous potential for clinical application.
Collapse
Affiliation(s)
- Enyu Imai
- Division of Nephrology, Department of Internal Medicine, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871
| | | |
Collapse
|
26
|
Fujino M, Adachi K, Kawasaki M, Kitazawa Y, Funeshima N, Okuyama T, Kimura H, Li XK. Prolonged survival of rat liver allograft with adenoviral gene transfection of human immunodeficiency virus type 1 nef. Liver Transpl 2003; 9:805-13. [PMID: 12884192 DOI: 10.1053/jlts.2003.50149] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
HIV-1 nef is believed to allow immune evasion by modifying cell surface molecules because of certain mechanisms such as downregulation of the major histocompatibility complex (MHC) class I molecule complex as well as upregulation of FasL. In the present study, we successfully generated a recombinant adenovirus vector containing HIV-1 nef. We detected the expression of nef in liver infected with AxCANef by immune staining and Western blotting, and confirmed its expression as persistent for more than 4 weeks. Furthermore, the surface expression of MHC class I was downregulated in AxCANef-infected hepatic cells. In addition, we also observed nef-induced FasL upregulation of gene-transfected hepatic cells. Using a DA-to-Lewis orthotopic liver transplantation model, we transfected AxCANef to a liver graft to determine whether nef expression could have an effect on recipient survival. AxCANef significantly prolonged recipient survival time (14.5 days) compared with the uninfected group (11 days) (P <.001) and the AxCALacZ-infected group (11 days) (P <.001). Histologic analysis showed reduction in the number of accumulated inflammatory cells and an increase in apoptotic cells in grafts expressing nef. In conclusion, we showed that the nef gene could prolong survival of rat liver allografts, and this result suggested the potential clinical use of its transfection.
Collapse
Affiliation(s)
- Masayuki Fujino
- Department of Innovative Surgery, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Interleukin (IL)-10 is an important immunoregulatory cytokine produced by many cell populations. Its main biological function seems to be the limitation and termination of inflammatory responses and the regulation of differentiation and proliferation of several immune cells such as T cells, B cells, natural killer cells, antigen-presenting cells, mast cells, and granulocytes. However, very recent data suggest IL-10 also mediates immunostimulatory properties that help to eliminate infectious and noninfectious particles with limited inflammation. Numerous investigations, including expression analyses in patients, in vitro and animal experiments suggest a major impact of IL-10 in inflammatory, malignant, and autoimmune diseases. So IL-10 overexpression was found in certain tumors as melanoma and several lymphomas and is considered to promote further tumor development. Systemic IL-10 release is a powerful tool of the central nervous system to prevent hyperinflammatory processes by activation of the neuro-endocrine axis following acute stress reactions. In contrast, a relative IL-10 deficiency has been observed and is regarded to be of pathophysiological relevance in certain inflammatory disorders characterized by a type 1 cytokine pattern such as psoriasis. Recombinant human IL-10 has been produced and is currently being tested in clinical trials. This includes rheumatoid arthritis, inflammatory bowel disease, psoriasis, organ transplantation, and chronic hepatitis C. The results are heterogeneous. They give new insight into the immunobiology of IL-10 and suggest that the IL-10/IL-10 receptor system may become a new therapeutic target.
Collapse
Affiliation(s)
- K Asadullah
- Head of Corporate Research Business Area Dermatology, Schering AG, D-13342 Berlin, Germany.
| | | | | |
Collapse
|
28
|
Zhang AB, Zheng SS, Jia CK, Wang Y. Effect of 1,25-dihydroxyvitamin D3 on preventing allograft from acute rejection following rat orthotoic liver transplantation. World J Gastroenterol 2003; 9:1067-71. [PMID: 12717858 PMCID: PMC4611374 DOI: 10.3748/wjg.v9.i5.1067] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the mechanism and the preventive role of 1, 25-dihydroxyvitamin D3 in acute rejection following orthotopic liver transplantation.
METHODS: Rats were randomly divided as donors or recipients for orthotopic liver allotransplantation model. Four groups were designed in the study, Group I: syngenic control (Wistar to Wistar); Group II: acute rejection (SD to Wistar); Group III: acute rejection treated with cyclosporine A, and Group IV: acute rejection treated with 1,25-(OH)2 D3. Liver function, rejection activity index and mRNA of IFN-γ, IL-10 intragraft in recipients were measured on day 1, 5, 7, 15, 30 posttransplant for assessing graft function, severity of acute rejection and immune state of recipients.
RESULTS: Survival time of recipients in Group IV was significantly prolonged (4/6 recipients survived for over 100 days. vs Group II, P < 0.001; vs Group III, P > 0.05). After treatment with 1,25-(OH)2 D3, mean value of all the assay tested on each experimental time was compared, liver function in group IV was significantly improved (AST 127 ± 41 U/L-360 ± 104 U/L, BIL 13 ± 5 mmol/l-38 ± 11 mmol/l; vs Group II, P < 0.05; vs Group III, P > 0.05. Rejection activity index was significantly decreased (0-3.3 ± 1.6; vs Group II, P < 0.05; vs Group III, P > 0.05). Level of hepatic IFN-γ mRNA in group IV was decreased, while level of hepatic IL-10 mRNA was increased (vs Group II, P < 0.05; vs Group III, P > 0.05).
CONCLUSION: Our results indicated that 1,25-(OH)2 D3 induced the secretion of cytokine toward to Th2 type, which would alleviate acute rejection, protect liver function and prolong survival of recipient after orthotopic liver transplantation.
Collapse
Affiliation(s)
- Ai-Bin Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Medicine School, Zhejiang University, Hangzhou 310003, Zhejiang Province, China.
| | | | | | | |
Collapse
|
29
|
Fu S, Chen D, Mao X, Zhang N, Ding Y, Bromberg JS. Feline immunodeficiency virus-mediated viral interleukin-10 gene transfer prolongs non-vascularized cardiac allograft survival. Am J Transplant 2003; 3:552-61. [PMID: 12752311 DOI: 10.1034/j.1600-6143.2003.00076.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Previous experiments demonstrated plasmid-, retroviral-, or adenoviral-mediated vIL-10 gene transfer could prolong allograft survival, but transgene expression was rapidly extinguished. Feline immunodeficiency virus (FIV) can integrate into genomic DNA of nondividing cells, resulting in indefinite transgene expression. We hypothesized FIV-mediated gene transfer could provide long-term gene expression, and improved allograft survival. FIV-vIL-10 and FIV-beta-gal were produced using the FELIX vector system. With vector transfer to syngeneic cardiac grafts, beta-galactosidase reporter gene expression was noted as early as day 5, was strongly expressed at days 10 and 20, and persisted for 50 days after transplantation. For allografts, FIV-vIL-10 gene transfer more than doubled mean survival from 10 +/- 1.6 to 22.3 +/- 3 days. When combined with other immunosuppressants, such as anti-CD40L mAb, FTY720, or anti-CD3 mAb, the mean survival times were prolonged to 27 +/- 4.6 days, 27.8 +/- 4.6 days, and 45.5 +/- 4.9 days, respectively. Multiple chemokine and chemokine receptor genes were induced by ischemia-reperfusion injury in syngeneic grafts, and in allogeneic grafts more genes were induced and to a greater degree. In allogeneic grafts transduced with FIV-IL-10, a number of the chemokine genes were suppressed. Therefore, FIV virus-mediated vIL-10 gene transfer prolongs allograft survival and, in combination with other agents, produces an additive effect.
Collapse
Affiliation(s)
- Shuang Fu
- Carl C. Icahn Institute for Gene Therapy and Molecular Medicine, and Recanati/Miller Transplantation Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | |
Collapse
|
30
|
Suárez A, Castro P, Alonso R, Mozo L, Gutiérrez C. Interindividual variations in constitutive interleukin-10 messenger RNA and protein levels and their association with genetic polymorphisms. Transplantation 2003; 75:711-7. [PMID: 12640314 DOI: 10.1097/01.tp.0000055216.19866.9a] [Citation(s) in RCA: 170] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND Genetic variations in the interleukin (IL)-10 gene promoter have been associated with levels of induced production of IL-10, disease susceptibility, and allograft rejection. Small amounts of this cytokine are constitutively produced and are important in maintaining the physiologic function of the cytokine network. In this study, we evaluated the distribution of IL-10 basal levels and its genetic regulation in a healthy Spanish population. METHODS Polymorphisms at the -1,082, -819, and -512 positions of the IL-10 promoter were analyzed by polymerase chain reaction amplification and hybridization with fluorescent-labeled allele-specific probes in 183 Spanish people. Levels of IL-10 messenger (m)RNA were tested by real-time reverse transcription-polymerase chain reaction in 123 healthy donors. Serum concentrations of IL-10 were measured by a highly sensitive ELISA, whereas protein amounts in lipopolysaccharide culture supernatants were quantified by an in-house ELISA. RESULTS The frequency of IL-10 promoter alleles and haplotypes in our population showed remarkable differences from other Caucasian populations. Large interindividual variations were found in mRNA and protein constitutive levels of IL-10, which allowed its classification in low and intermediate/high producers. We found statistical differences in mRNA concentration between the polymorphic variant GCC/GCC and the low producer genotypes. The G allele at position -1082 was the most important genetic factor in the regulation of constitutive IL-10 mRNA levels. Similarly, we also found an association of this polymorphic position with serum concentration greater than 2 pg/mL. CONCLUSIONS Constitutive levels of IL-10 (mRNA and serum protein) displayed remarkable interindividual variations, which are genetically controlled by polymorphic variants at the cytokine gene promoter.
Collapse
Affiliation(s)
- Ana Suárez
- Department of Functional Biology, Area of Immunology, University of Oviedo, Oviedo, Spain
| | | | | | | | | |
Collapse
|
31
|
Adachi O, Yamato E, Kawamoto S, Yamamoto M, Tahara H, Tabayashi K, Miyazaki JI. High-level expression of viral interleukin-10 in cardiac allografts fails to prolong graft survival. Transplantation 2002; 74:1603-8. [PMID: 12490795 DOI: 10.1097/00007890-200212150-00019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Viral interleukin (vIL)-10, encoded in the Epstein-Barr virus genome, shares many of the anti-inflammatory properties of cellular IL-10 but is supposed to lack IL-10's immunostimulatory properties. Thus, vIL-10 is expected to offer superior immunosuppression. METHODS We established transgenic mice (vIL-10 Tg) that express vIL-10 systemically and transplanted their hearts as vascularized allografts into unmodified major histocompatibility complex (MHC) full-mismatch or MHC class II-disparate mice. RESULTS The vIL-10 Tg mice revealed high-level expression of vIL-10 in major organs including the heart. However, the heart grafts from the vIL-10 Tg mice failed to exhibit prolonged survival in combination with either the MHC full-mismatch or the class II-disparate mice. In the MHC class II-disparate mice, the vIL-10 Tg heart grafts showed severe CD8 T-cell infiltration and increased interferon (IFN)-gamma mRNA expression compared with non-Tg grafts. CONCLUSION High level expression of vIL-10 in grafts can exacerbate immunological rejection in an allogenic transplantation model.
Collapse
Affiliation(s)
- Osamu Adachi
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Bunnapradist S, Vo A, Toyoda M, Alsabeh R, Lockhart C, Puliyanda D, Jordan SC. Posttransplantation lymphoproliferative disorder presenting as a unilateral leg mass 10 years after kidney transplantation. Transplantation 2002; 74:1648-51. [PMID: 12490805 DOI: 10.1097/00007890-200212150-00029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Epstein-Barr virus (EBV)-associated lymphoproliferative disorder is a well-known complication of solid organ transplantation. The risk factors include type and degree of immunosuppression and EBV status of the donor and recipient before transplantation. The manifestations of posttransplantation lymphoproliferative disorders are myriad, from subtle findings such as tonsillar hypertrophy to fulminant manifestations such as hepatocellular failure and death. Here we report an unusual presentation of posttransplantation lymphoproliferative disorder as unilateral leg swelling in a 68-year-old man 10 years after kidney transplantation. The patient had detectable EBV-DNA by polymerase chain reaction in the peripheral blood and EBV messenger RNA by in situ hybridization in biopsy tissue. The patient's tumor did not respond to reductions in immunosuppressive medication, intravenous immunoglobulin, ganciclovir, and rituximab. Eventually the patient responded to CHOP (cyclophosphamide, Oncovin [vincristine, Eli Lilly, Indianapolis, IN], prednisone, and doxorubicin) therapy. He is currently 2 years posttreatment, with a serum creatinine of 0.8 mg/dL, on no immunosuppression.
Collapse
Affiliation(s)
- Suphamai Bunnapradist
- Center for Kidney Transplantation, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Khanna A, Plummer M, Bromberek K, Woodliff J, Hariharan S. Immunomodulation in stable renal transplant recipients with concomitant tacrolimus and sirolimus therapy. MEDICAL IMMUNOLOGY (LONDON, ENGLAND) 2002; 1:3. [PMID: 12495444 PMCID: PMC149406 DOI: 10.1186/1476-9433-1-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2002] [Accepted: 11/19/2002] [Indexed: 11/18/2022]
Abstract
BACKGROUND: Long term treatment with immunosuppressive agents results in nephrotoxicity in renal transplant recipients. We explored the effect of combination of Tacrolimus (TAC) and Sirolimus (SRL) on the immune system in renal transplant recipients. METHODS: 10 stable renal transplant recipients were selected to participate in a pharmacokinetic study with a combination of TAC and SRL. Blood was drawn on day zero and 14 days post treatment. Lymphocyte proliferation was quantified by 3H-thymidine uptake assay (results expressed as counts per minute). The mRNA expression was studied by RT-PCR and serum levels of cytokines were quantified by ELISA and a cytokine bead array system. RESULTS: Lymphocyte proliferative response to PHA (p < 0.05), Con A (p < 0.006) and Anti-CD3 (p <0.005) were significantly decreased in patients who received both TAC and SRL compared to TAC alone. The mRNA expression of proinflammatory cytokines TNF-alpha (p < 0.05), cyclins G (p < 0.01) and E (p < 05) were decreased, and of TGF-beta (p < 0.03) and p21 (p < 0.05) were increased in patients treated with this combination. Circulating levels of IFN-gamma (p < 0.04), IL-4 (p < 0.02), and Il-2 (p < 0.03) were significantly inhibited and elevation of TGF-beta (p < 0.04) was observed in patients treated with TAC and SRL combination. CONCLUSION: These novel findings demonstrate that addition of SRL to TAC therapy enhances immuno modulation and causes increased immunosuppression providing a rationale for this concomitant therapy.
Collapse
Affiliation(s)
- Ashwani Khanna
- Department of Medicine (Nephrology), Medical College of Wisconsin, Milwaukee WI-53226, USA
| | - Matthew Plummer
- Department of Medicine (Nephrology), Medical College of Wisconsin, Milwaukee WI-53226, USA
| | - Katherine Bromberek
- Department of Medicine (Nephrology), Medical College of Wisconsin, Milwaukee WI-53226, USA
| | - Jeffrey Woodliff
- Flow Cytometry Core Lab, Medical College of Wisconsin, Milwaukee WI-53226, USA
| | - Sundaram Hariharan
- Department of Medicine (Nephrology), Medical College of Wisconsin, Milwaukee WI-53226, USA
| |
Collapse
|
34
|
Chiffoleau E, Bériou G, Dutartre P, Usal C, Soulillou JP, Cuturi MC. Induction of donor-specific allograft tolerance by short-term treatment with LF15-0195 after transplantation. Evidence for a direct effect on T-cell differentiation. Am J Transplant 2002; 2:745-57. [PMID: 12243495 DOI: 10.1034/j.1600-6143.2002.20808.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A 20-day treatment with LF15-0195, a deoxyspergualine analog, induced long-term heart allograft survival in the rat without signs of chronic rejection. LF15-0195-treated recipients did not develop an anti-donor alloantibody response. Analysis of graft-infiltrating cells, IL10, TNFalpha, IFNgamma mRNA and iNOS protein expression in allografts, 5 days after transplantation, showed that they were markedly decreased in allografts from LF15-0195-treated recipients compared with allografts from untreated recipients. Surprisingly, spleen T cells from LF15-0195 recipients, 5days after grafting, were able to proliferate strongly in vitro, when stimulated with donor cells, but had reduced mRNA expression for IFNy compared with spleen T cells from untreated graft recipients. Furthermore, when T cells from naive animals were stimulated in vitro, using anti-CD3 and anti-CD28, LF15-0195 also increased T-cell proliferation in a dose-dependent fashion: however, these cells expressed less of the Th1 -related cytokines, IFNgamma and IL2, compared with untreated cells, suggesting that LF15-0195 could act on T-cell differentiation. In conclusion, we show here that a short-term treatment with LF15-0195 induced long-term allograft tolerance, decreasing the in situ anti-donor response, and we illustrate evidence for the development of regulatory mechanisms.
Collapse
Affiliation(s)
- Elise Chiffoleau
- INSERM Unité 437 and Institut de Transplantation et de Recherche en Transplantation, CHU Hotel Dieu, Nantes, France
| | | | | | | | | | | |
Collapse
|
35
|
Dinh TP, Carpenter D, Leslie FM, Freund TF, Katona I, Sensi SL, Kathuria S, Piomelli D. Brain monoglyceride lipase participating in endocannabinoid inactivation. Proc Natl Acad Sci U S A 2002; 99:10819-24. [PMID: 12136125 PMCID: PMC125056 DOI: 10.1073/pnas.152334899] [Citation(s) in RCA: 1014] [Impact Index Per Article: 44.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The endogenous cannabinoids (endocannabinoids) are lipid molecules that may mediate retrograde signaling at central synapses and other forms of short-range neuronal communication. The monoglyceride 2-arachidonoylglycerol (2-AG) meets several criteria of an endocannabinoid substance: (i) it activates cannabinoid receptors; (ii) it is produced by neurons in an activity-dependent manner; and (iii) it is rapidly eliminated. 2-AG inactivation is only partially understood, but it may occur by transport into cells and enzymatic hydrolysis. Here we tested the hypothesis that monoglyceride lipase (MGL), a serine hydrolase that converts monoglycerides to fatty acid and glycerol, participates in 2-AG inactivation. We cloned MGL by homology from a rat brain cDNA library. Its cDNA sequence encoded for a 303-aa protein with a calculated molecular weight of 33,367 daltons. Northern blot and in situ hybridization analyses revealed that MGL mRNA is heterogeneously expressed in the rat brain, with highest levels in regions where CB(1) cannabinoid receptors are also present (hippocampus, cortex, anterior thalamus, and cerebellum). Immunohistochemical studies in the hippocampus showed that MGL distribution has striking laminar specificity, suggesting a presynaptic localization of the enzyme. Adenovirus-mediated transfer of MGL cDNA into rat cortical neurons increased MGL expression and attenuated N-methyl-D-aspartate/carbachol-induced 2-AG accumulation in these cells. No such effect was observed on the accumulation of anandamide, another endocannabinoid lipid. The results suggest that hydrolysis by means of MGL is a primary mechanism for 2-AG inactivation in intact neurons.
Collapse
Affiliation(s)
- T P Dinh
- Department of Pharmacology, University of California, Irvine, CA 92697-4625, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Redaelli CA, Wagner M, Günter-Duwe D, Tian YH, Stahel PF, Mazzucchelli L, Schmid RA, Schilling MK. 1alpha,25-dihydroxyvitamin D3 shows strong and additive immunomodulatory effects with cyclosporine A in rat renal allotransplants. Kidney Int 2002; 61:288-96. [PMID: 11786111 DOI: 10.1046/j.1523-1755.2002.00101.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Vitamin D3 and its metabolites have long been found to exert immunosuppressive effects both in vivo and in vitro. The present study investigated the effect of 1alpha,25-dihydroxycholecalciferol (1,25DHC) on vascularized renal allografts in rats. METHODS Three days prior to transplantation, two groups of animals were subjected to 1,25DHC (1 microg/kg/day IP) and a low calcium diet, which was continued until the end of the experiments. Recipient organs were removed and single allografts were transplanted in a high responder strain combination (ACI --> Lewis). Following transplantation, low-dose cyclosporine A (3.2 mg/kg/day CsA) administration was started in two experimental groups of recipients (one group receiving 1,25 DHC additionally) whereas the control allograft recipients received no immunosuppression (control III). Graft survival and renal function was monitored until death or the end of experiments and allograft rejection was assessed histologically using the Banff classification. RESULTS 1,25DHC significantly prolonged allograft survival in comparison to control III (9.6 +/- 1 vs. 5.7 +/- 0.2 days; P=0.009). In addition, a combination of 1,25DHC and low-dose CsA increased allograft survival compared to CsA administration alone (24 +/- 0.9 vs. 13 +/- 0.3 days; P=0.008). 1,25DHC preserved renal creatinine clearance and decreased proteinuria in comparison to control III, and the combination of 1,25DHC and low-dose CsA again showed an additive effect on preservation of renal function. 1,25DHC and low-dose CsA both decreased interleukin (IL)-2 and IL-12 expression levels in serum and allografts, and a combination treatment produced the strongest attenuation of IL-2 and IL-12 expression. In addition, 1,25DHC increased IL-4 and IL-10 expression levels in allografts, whereas CsA alone did not alter IL-4 and IL-10 expression. In contrast, combination of 1,25DHC and low-dose CsA showed a significant increase in IL-10 expression levels whereas IL-4 expression was not elevated. CONCLUSION Monotherapy with 1,25DHC significantly prolongs survival of renal allografts and preserves graft function in rats. A combination of 1,25DHC and CsA caused an additive effect on graft survival with differential regulation of pro- and anti-inflammatory cytokines, as compared to 1,25DHC administration alone.
Collapse
Affiliation(s)
- Claudio A Redaelli
- Department of Visceral and Transplantation Surgery, University of Bern, Bern, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|