1
|
Zou Y, Zhou C, Xu H, Yu J, Ye P, Zhang H, Chen S, Zhao J, Le S, Cui J, Jiang L, Wu J, Xia J. Glibenclamide ameliorates transplant-induced arteriosclerosis and inhibits macrophage migration and MCP-1 expression. Life Sci 2019; 241:117141. [PMID: 31811853 DOI: 10.1016/j.lfs.2019.117141] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/20/2019] [Accepted: 12/01/2019] [Indexed: 01/28/2023]
Abstract
AIMS Glibenclamide, a diabetes mellitus type 2 medication, has anti-inflammatory and autoimmune properties. This study investigated the effects of glibenclamide on transplant-induced arteriosclerosis as well as the underlying molecular events. METHODS Male C57Bl/6 (H-2b) and BALB/c (H-2d) mice were used for aorta transplantation. We used hematoxylin and eosin (HE) and Elastic Van Gieson (EVG) staining for histological assessment, and qRT-PCR and ELISA to measure mRNA and protein levels. Mouse peritoneal macrophages were isolated for lipopolysaccharide (LPS) stimulation and glibenclamide treatment followed by ELISA, Western blot, and Transwell assays. RESULTS Glibenclamide inhibited transplant-induced arteriosclerosis in vivo. Morphologically, glibenclamide reduced inflammatory cell accumulation and collagen deposition in the aortas. At the gene level, glibenclamide suppressed aortic cytokine mRNA levels, including interleukin-1β (IL-1β; 10.64 ± 3.19 vs. 23.77 ± 5.72; P < .05), tumor necrosis factor-α (TNF-α; 4.59 ± 0.78 vs. 13.89 ± 5.42; P < .05), and monocyte chemoattractant protein-1 (MCP-1; 202.66 ± 23.44 vs. 1172.73 ± 208.80; P < .01), while IL-1β, TNF-α, and MCP-1 levels were also reduced in the mouse sera two weeks after glibenclamide treatment (IL-1β, 39.40 ± 13.56 ng/ml vs. 78.96 ± 9.39 ng/ml; P < .01; TNF-α, 52.60 ± 13.00 ng/ml vs. 159.73 ± 6.76 ng/ml; P < .01; and MCP-1, 56.60 ± 9.07 ng/ml vs. 223.07 ± 36.28 ng/ml; P < .001). Furthermore, glibenclamide inhibited macrophage expression and secretion of inflammatory factors in vitro through suppressing activation of the nuclear factor-κB (NF-κB) pathway and MCP-1 production. CONCLUSION Glibenclamide protected against aorta transplantation-induced arteriosclerosis by reducing inflammatory factors in vivo and inhibited macrophage migration and MCP-1 production in vitro.
Collapse
Affiliation(s)
- Yanqiang Zou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Cheng Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Heng Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ping Ye
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei 430022, China
| | - Hao Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Shanshan Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jing Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Sheng Le
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jikai Cui
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Lang Jiang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China.
| |
Collapse
|
2
|
Mok D, Black M, Gupta N, Arefanian H, Tredget E, Rayat GR. Early immune mechanisms of neonatal porcine islet xenograft rejection. Xenotransplantation 2019; 26:e12546. [DOI: 10.1111/xen.12546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/29/2019] [Accepted: 07/09/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Dereck Mok
- Department of Surgery, Ray Rajotte Surgical‐Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Mazzen Black
- Department of Surgery, Ray Rajotte Surgical‐Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Nancy Gupta
- Department of Surgery, Ray Rajotte Surgical‐Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Hossein Arefanian
- Microbiology & Immunology Unit Dasman Diabetes Institute Dasman Kuwait
| | - Eric Tredget
- Department of Surgery, Ray Rajotte Surgical‐Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| | - Gina R. Rayat
- Department of Surgery, Ray Rajotte Surgical‐Medical Research Institute, Alberta Diabetes Institute, Faculty of Medicine and Dentistry University of Alberta Edmonton Alberta Canada
| |
Collapse
|
3
|
Merani S, Truong WW, Hancock W, Anderson CC, Shapiro AMJ. Chemokines and Their Receptors in Islet Allograft Rejection and as Targets for Tolerance Induction. Cell Transplant 2017; 15:295-309. [PMID: 28863747 DOI: 10.3727/000000006783981963] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Graft rejection is a major barrier to successful outcome of transplantation surgery. Islet transplantation introduces insulin secreting tissue into type 1 diabetes mellitus recipients, relieving patients from exogenous insulin injection. However, insulitis of grafted tissue and allograft rejection prevent long-term insulin independence. Leukocyte trafficking is necessary for the launch of successful immune responses to pathogen or allograft. Chemokines, small chemotactic cytokines, direct the migration of leukocytes through their interaction with chemokine receptors found on cell surfaces of immune cells. Unique receptor expression of leukocytes, and the specificity of chemokine secretion during various states of immune response, suggest that the extracellular chemokine milieu specifically homes certain leukocyte subsets. Thus, only those leukocytes required for the current immune task are attracted to the inflammatory site. Chemokine blockade, using antagonists and monoclonal antibodies directed against chemokine receptors, is an emerging and specific immunosuppressive strategy. Importantly, chemokine blockade may potentiate tolerance induction regimens to be used following transplantation surgery, and prevent the need for life-long immunosuppression of islet transplant recipients. Here, the role for chemokine blockade in islet transplant rejection and tolerance is reviewed.
Collapse
Affiliation(s)
- Shaheed Merani
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| | - Wayne W Truong
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| | - Wayne Hancock
- Department of Pathology and Laboratory Medicine, Joseph Stokes, Jr. Research Institute and Biesecker Pediatric Liver Center, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Colin C Anderson
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| | - A M James Shapiro
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton AB, Canada
| |
Collapse
|
4
|
Foroughi F, Amirzargar A, Ahmadpoor P, Noorbakhsh F, Nafar M, Yekaninejad MS, Hosseinzadeh M, Shabaz SK, Barabadi M, Ghorbanpour M, Sarrafnejad A. Increased levels of CD4(+) and CD8(+) T cells expressing CCR1 in patients developing allograft dysfunction; a cohort study. Transpl Immunol 2016; 38:67-74. [PMID: 27234431 DOI: 10.1016/j.trim.2016.05.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Leukocyte infiltration into the graft has pivotal effects on kidney transplantation outcome. The present study sought to determine whether the expression of sequential chemokine receptors on CD4(+) and CD8(+) T cells in human renal allograft can predict clinical episodes. METHODS Blood samples from 52 consecutive renal transplant patients were evaluated at the time of transplantation and at three times (2, 90 and 180days) after transplantation to analyze the expression of CCR1 and CXCR3 on CD4(+) and CD8(+) T cells by flowcytometry. A total of 30 biopsies, including protocol biopsy (n=24) and cause biopsy (n=6), were investigated according to the Banff criteria. RESULTS The mean percentage of CD4(+) and CD8(+) T cells expressing CCR1 was significantly increased in patients with allograft dysfunction (n=25) (p=0.006, p=0.004). The mean fluorescence intensity of CXCR3 on CD4(+) and CD8(+) T cells were found to be significantly higher in graft dysfunction than that in well-functioning grafts. (p<0.001, p=0.007). Receiver Operating Characteristic (ROC) Curve Analysis showed that the calculated AUC was 0.86 at the third month for CD4(+)CCR1(+) and CD8(+)CCR1(+) (p<0.001). Multiple logistic regression analysis showed that an increase in CD4(+) expressing CXCR3 leads to a lower risk of graft dysfunction (OR=0.37), while an increase in CD8(+) expressing CCR1 results in a higher risk of graft dysfunction (OR=3.66). CONCLUSION During renal transplantation, CD4(+) and CD8(+) T cells expressing CCR1 were increased in patients who developed graft dysfunction. These findings may prospectively predict allograft dysfunction, and help elucidate the underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Farshad Foroughi
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Amirzargar
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Molecular Immunology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Pedram Ahmadpoor
- Chronic Kidney Disease Research Center, Department of Nephrology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nafar
- Chronic Kidney Disease Research Center, Department of Nephrology, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Hosseinzadeh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sanaz Keshavarz Shabaz
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehri Barabadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mina Ghorbanpour
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdolfattah Sarrafnejad
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW This review summarizes the phenotype and function of macrophages in the context of solid organ transplantation and will focus on fundamental insights into their paradoxical pro-inflammatory versus suppressive function. We will also discuss the therapeutic potential of regulatory macrophages in tolerance induction. RECENT FINDINGS Macrophages are emerging as an essential element of solid organ transplantation. Macrophages are involved in the pathogenesis of ischemia reperfusion injury, as well as both acute and chronic rejection, exacerbating injury through secretion of inflammatory effectors and by amplifying adaptive immune responses. Notably, not all responses associated with macrophages are deleterious to the graft, and graft protection can in fact be conferred by macrophages. This has been attributed to the presence of macrophages with tissue-repair capabilities, as well as the effects of regulatory macrophages. SUMMARY The explosion of new information on the role of macrophages in solid organ transplantation has opened up new avenues of research and the possibility of therapeutic intervention. However, the role of myeloid cells in graft rejection, resolution of rejection and tissue repair remains poorly understood. A better understanding of plasticity and regulation of monocyte polarization is vital for the development of new therapies for the treatment of acute and chronic transplant rejection.
Collapse
|
6
|
Ferreras L, Sheerin NS, Kirby JA, Ali S. Mechanisms of Renal Graft Chronic Injury and Progression to Interstitial Fibrosis. CURRENT TRANSPLANTATION REPORTS 2015. [DOI: 10.1007/s40472-015-0069-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
7
|
Proudfoot AEI, Bonvin P, Power CA. Targeting chemokines: Pathogens can, why can't we? Cytokine 2015; 74:259-67. [PMID: 25753743 DOI: 10.1016/j.cyto.2015.02.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 12/19/2022]
Abstract
Chemoattractant cytokines, or chemokines, are the largest sub-family of cytokines. About 50 distinct chemokines have been identified in humans. Their principal role is to stimulate the directional migration of leukocytes, which they achieve through activation of their receptors, following immobilization on cell surface glycosaminoglycans (GAGs). Chemokine receptors belong to the G protein-coupled 7-transmembrane receptor family, and hence their identification brought great promise to the pharmaceutical industry, since this receptor class is the target for a large percentage of marketed drugs. Unfortunately, the development of potent and efficacious inhibitors of chemokine receptors has not lived up to the early expectations. Several approaches to targeting this system will be described here, which have been instrumental in establishing paradigms in chemokine biology. Whilst drug discovery programs have not yet elucidated how to make successful drugs targeting the chemokine system, it is now known that certain parasites have evolved anti-chemokine strategies in order to remain undetected by their hosts. What can we learn from them?
Collapse
Affiliation(s)
- Amanda E I Proudfoot
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève and NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland.
| | - Pauline Bonvin
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève and NovImmune S.A., 14 chemin des Aulx, 1228 Plan-les-Ouates, Geneva, Switzerland.
| | - Christine A Power
- Geneva Research Centre, Merck Serono S.A., 9 chemin des Mines, 1202 Genève, Switzerland.
| |
Collapse
|
8
|
Huang Y, Jiao S, Tao X, Tang Q, Jiao W, Xiao J, Xu X, Zhang Y, Liang G, Wang H. Met-CCL5 represents an immunotherapy strategy to ameliorate rabies virus infection. J Neuroinflammation 2014; 11:146. [PMID: 25182681 PMCID: PMC4243955 DOI: 10.1186/s12974-014-0146-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 08/05/2014] [Indexed: 12/25/2022] Open
Abstract
Background Infection of rabies virus (RABV) causes central nervous system (CNS) dysfunction and results in high mortality in human and animals. However, it is still unclear whether and how CNS inflammation and immune response contribute to RABV infection. Methods Suckling mice were intracerebrally infected with attenuated RABV aG and CTN strains, followed by examination of chemokine or cytokine production, inflammatory cell infiltration and neuron apoptosis in the brain. Furthermore, the suckling mice and adult mice that were intracerebrally infected with aG and the adult mice that were intramuscularly infected with street RABV HN10 were treated with CCL5 antagonist (Met-CCL5) daily beginning on day 2 postinfection. The survival rates and inflammation responses in the CNS of these mice were analyzed. Results Excessive CCL5 in the CNS was associated with CNS dysfunction, inflammation, and macrophage or lymphocyte infiltration after attenuated or street RABV infection. Administration of exogenous CCL5 induced excessive infiltration of immune cells into the CNS and enhanced inflammatory chemokine and cytokine production. Met-CCL5 treatment significantly prolonged survival time of the suckling mice inoculated with aG and adult mice infected with aG and HN10. Conclusions These results suggest that CCL5 in the CNS is a key regulator involved in inducing rabies encephalomyelitis. Furthermore, treatment with the CCL5 antagonist Met-CCL5 prolongs survival time of the mice infected with attenuated or street RABVs, which might represent a novel therapeutic strategy to ameliorate RABV infection.
Collapse
|
9
|
Gene transfer of the S24F regulated on activation normal T-cell expressed and secreted-chemokine ligand 5 variant attenuates cardiac allograft rejection. Transplantation 2014; 97:1233-9. [PMID: 24825519 DOI: 10.1097/tp.0000000000000122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Regulated on activation normal T-cell expressed and secreted (RANTES)-chemokine ligand 5 plays a key role in mediating heart transplant rejection. Suppression of RANTES-mediated signals can reduce leukocyte recruitment and mitigate transplant rejection severity. The present study describes the construction of an adenovirus overexpression vector encoding a natural S24F RANTES variant as a means of reducing leukocyte recruitment, resulting in the prevention of allograft rejection. METHODS The in vitro transendothelial chemotaxis assay was used to compare RANTES-induced transmigration of peripheral blood mononuclear cells across human umbilical vein endothelial cells cultured on the upper Transwell chamber. Intracoronary delivery of Ad-S24F, Ad-Null, or phosphate-buffered saline was performed in BALB/c donor hearts that were transplanted into the abdominal cavity of C57BL/6 recipients as a measure of allograft survival. Intragraft inflammatory cell infiltrates and associated proinflammatory cytokine expression profiles were detected by immunohistochemistry and quantitative real-time polymerase chain reaction on day 6 after transplantation, respectively. RESULTS Regulated on activation normal T-cell expressed and secreted-induced peripheral blood mononuclear cell transendothelial chemotaxis is inhibited by S24F (Ad-S24F, 9.2%±0.02%; Ad-Null, 17.7%±0.02%; medium control, 15.1%±0.01%; P<0.05). Cardiac allograft survival was prolonged after delivery of 1×10 plaque-forming units of Ad-S24F (13.00±0.33 days compared with 9.38±0.60 and 9.00±0.38 days after Ad-Null or phosphate-buffered saline treatment, respectively, P<0.05). S24F gene transfer reduced the number of intragraft CD8 T lymphocytes, monocyte-macrophages, and T-cell receptor αβ cell infiltrates (P<0.05) and decreased transcripts for RANTES and interferon-γ (P<0.05). CONCLUSION S24F is an important component of the chemokine network involved in regulating the biologic activity of RANTES, and its expression can be used in the prevention and treatment of cardiac allograft rejection.
Collapse
|
10
|
Kwan T, Wu H, Chadban SJ. Macrophages in renal transplantation: Roles and therapeutic implications. Cell Immunol 2014; 291:58-64. [PMID: 24973994 DOI: 10.1016/j.cellimm.2014.05.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/24/2014] [Accepted: 05/27/2014] [Indexed: 01/15/2023]
Abstract
The presence of macrophages within transplanted renal allografts has been appreciated for some time, whereby macrophages were viewed primarily as participants in the process of cell-mediated allograft rejection. Recent insights into macrophage biology have greatly expanded our conceptual understanding of the multiple roles of macrophages within the allograft. Distinct macrophage subsets are present within the kidney and these sub-serve discrete functions in promoting and attenuating inflammation, immune modulation and tissue repair. Unraveling the complex roles macrophages play in transplantation will allow identification of potential therapeutic targets to prevent and treat allograft rejection and maximize graft longevity.
Collapse
Affiliation(s)
- Tony Kwan
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, Australia; Collaborative Transplant Research Group, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Huiling Wu
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, Australia; Collaborative Transplant Research Group, Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Steven J Chadban
- Department of Renal Medicine, Royal Prince Alfred Hospital, Sydney, Australia; Collaborative Transplant Research Group, Sydney Medical School, The University of Sydney, Sydney, Australia.
| |
Collapse
|
11
|
Knoop K, Schwenk N, Dolp P, Willhauck MJ, Zischek C, Zach C, Hacker M, Göke B, Wagner E, Nelson PJ, Spitzweg C. Stromal targeting of sodium iodide symporter using mesenchymal stem cells allows enhanced imaging and therapy of hepatocellular carcinoma. Hum Gene Ther 2013; 24:306-16. [PMID: 23402366 DOI: 10.1089/hum.2012.104] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The tumor-homing property of mesenchymal stem cells (MSC) has lead to their use as delivery vehicles for therapeutic genes. The application of the sodium iodide symporter (NIS) as therapy gene allows noninvasive imaging of functional transgene expression by (123)I-scintigraphy or PET-imaging, as well as therapeutic application of (131)I or (188)Re. Based on the critical role of the chemokine RANTES (regulated on activation, normal T-cell expressed and presumably secreted)/CCL5 secreted by MSCs in the course of tumor stroma recruitment, use of the RANTES/CCL5 promoter should allow tumor stroma-targeted expression of NIS after MSC-mediated delivery. Using a human hepatocellular cancer (HCC) xenograft mouse model (Huh7), we investigated distribution and tumor recruitment of RANTES-NIS-engineered MSCs after systemic injection by gamma camera imaging. (123)I-scintigraphy revealed active MSC recruitment and CCL5 promoter activation in the tumor stroma of Huh7 xenografts (6.5% ID/g (123)I, biological half-life: 3.7 hr, tumor-absorbed dose: 44.3 mGy/MBq). In comparison, 7% ID/g (188)Re was accumulated in tumors with a biological half-life of 4.1 hr (tumor-absorbed dose: 128.7 mGy/MBq). Administration of a therapeutic dose of (131)I or (188)Re (55.5 MBq) in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved survival without significant differences between (131)I and (188)Re. These data demonstrate successful stromal targeting of NIS in HCC tumors by selective recruitment of NIS-expressing MSCs and by use of the RANTES/CCL5 promoter. The resulting tumor-selective radionuclide accumulation was high enough for a therapeutic effect of (131)I and (188)Re opening the exciting prospect of NIS-mediated radionuclide therapy of metastatic cancer using genetically engineered MSCs as gene delivery vehicles.
Collapse
Affiliation(s)
- Kerstin Knoop
- Department of Internal Medicine II, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
A novel CXCL10-based GPI-anchored fusion protein as adjuvant in NK-based tumor therapy. PLoS One 2013; 8:e72749. [PMID: 24023642 PMCID: PMC3758322 DOI: 10.1371/journal.pone.0072749] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 07/12/2013] [Indexed: 01/13/2023] Open
Abstract
Background Cellular therapy is a promising therapeutic strategy for malignant diseases. The efficacy of this therapy can be limited by poor infiltration of the tumor by immune effector cells. In particular, NK cell infiltration is often reduced relative to T cells. A novel class of fusion proteins was designed to enhance the recruitment of specific leukocyte subsets based on their expression of a given chemokine receptor. The proteins are composed of an N-terminal chemokine head, the mucin domain taken from the membrane-anchored chemokine CX3CL1, and a C-terminal glycosylphosphatidylinositol (GPI) membrane anchor replacing the normal transmembrane domain allowing integration of the proteins into cell membranes when injected into a solid tumor. The mucin domain in conjunction with the chemokine head acts to specifically recruit leukocytes expressing the corresponding chemokine receptor. Methodology/Principal Findings A fusion protein comprising a CXCL10 chemokine head (CXCL10-mucin-GPI) was used for proof of concept for this approach and expressed constitutively in Chinese Hamster Ovary cells. FPLC was used to purify proteins. The recombinant proteins efficiently integrated into cell membranes in a process dependent upon the GPI anchor and were able to activate the CXCR3 receptor on lymphocytes. Endothelial cells incubated with CXCL10-mucin-GPI efficiently recruited NK cells in vitro under conditions of physiologic flow, which was shown to be dependent on the presence of the mucin domain. Experiments conducted in vivo using established tumors in mice suggested a positive effect of CXCL10-mucin-GPI on the recruitment of NK cells. Conclusions The results suggest enhanced recruitment of NK cells by CXCL10-mucin-GPI. This class of fusion proteins represents a novel adjuvant in cellular immunotherapy. The underlying concept of a chemokine head fused to the mucin domain and a GPI anchor signal sequence may be expanded into a broader family of reagents that will allow targeted recruitment of cells in various settings.
Collapse
|
13
|
Yu X, Huang C, Song B, Xiao Y, Fang M, Feng J, Wang P. CD4+CD25+ regulatory T cells-derived exosomes prolonged kidney allograft survival in a rat model. Cell Immunol 2013; 285:62-8. [PMID: 24095986 DOI: 10.1016/j.cellimm.2013.06.010] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 04/27/2013] [Accepted: 06/19/2013] [Indexed: 11/29/2022]
Abstract
CD4(+)CD25(+) regulatory T cells (Tregs) are negative regulators of the immune system that induce and maintain immune tolerance. Exosomes are natural products released from many sources and play a role in antigen presentation, immunoregulation, and signal transduction. In order to determine whether exosomes can be released from Tregs and participate in transplantation tolerance, we isolated and purified Tregs-derived exosomes and established a rat model of kidney transplantation. We then transferred the autologous exosomes into recipients to observe the effect of transplantation tolerance in vivo and in vitro. From in vivo study, serum analysis and histology showed that the function of exosomes can postpone allograft rejection and prolong the survival time of transplanted kidney. From in vitro study, exosomes possessed the capacity to suppress T cells proliferation. Taken together, these results suggest that the Tregs-derived exosomes have a suppressive role on acute rejection and inhibit T cells proliferation, especially exosomes derived from donor-type Tregs, which imply that the Tregs-derived exosomes are one of far-end regulation mechanisms of Tregs. Thus, exosomes released from Tregs could be considered as a possible immunosuppressive reagent for the treatment of transplant rejection.
Collapse
Affiliation(s)
- Xuesong Yu
- Department of Nephrology, 422 Hospital of PLA, Zhanjiang, Jiangshu 52400, China; Department of Urology, Xinqiao Hospital, Third Military Medical University, ChongQing 400037, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Macrophages: contributors to allograft dysfunction, repair, or innocent bystanders? Curr Opin Organ Transplant 2013; 17:20-5. [PMID: 22157320 DOI: 10.1097/mot.0b013e32834ee5b6] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Macrophages are members of the innate immune response. However, their role in the adaptive immune response is not known. The purpose of this review is to highlight our current understanding of macrophage structure and function and how they may participate in allograft injury. RECENT FINDINGS Studies in acute kidney injury models identify macrophages as key mediators of inflammatory injury, while more recent studies indicate that they may play a reparative role, depending on phenotype - M1 or M2 type macrophages. Mregs, generated in vitro, appear to have immune suppressive abilities and a unique phenotype. In solid-organ transplant, the emphasis of studies has been on acute or chronic injury. These data are derived from animal models using depletion of macrophages or antagonizing their activation and inflammatory responses. The relative contribution of macrophage phenotype in transplantation has not been explored. SUMMARY These studies suggest that macrophages play an injurious role in acute cellular allograft rejection, as well as in chronic injury. Infiltration of an allograft with macrophages is also associated with worse graft function and poor prognosis. Further studies are needed to understand the mechanisms of macrophage-mediated injury, explore their potential reparative role, and determine if they or their functional products are biomarkers of poor graft outcomes.
Collapse
|
15
|
Koenen R, Weber C. Chemokines and Their Receptors as Therapeutic Targets in Atherosclerosis. Atherosclerosis 2012. [DOI: 10.1201/b13723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
Proudfoot AE, Power CA, Church DJ, Soler D, Mack M. Cellular assays of chemokine receptor activation. ACTA ACUST UNITED AC 2012; Chapter 12:Unit12.4. [PMID: 21959755 DOI: 10.1002/0471141755.ph1204s14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
This unit describes procedures for measuring the activation of chemokine receptors by their ligands. Chemokines are chemoattractant proteins and two assays are described for analyzing the chemoattractant properties (chemotaxis) of these proteins. Chemokine receptors are coupled to G proteins, and activation of the receptors results in the mobilization of intracellular Ca²⁺ stores. Two assays for the measurement of this activity are described which employ calcium-sensitive dyes, one for adherent and one for non-adherent cells. In addition, an assay for quantitating chemokine receptor downmodulation and subsequent recycling events is described. The assays are applicable to both purified leukocytes and stably expressed recombinant chemokine receptors.This unit describes procedures for measuring the activation of chemokine receptors by their ligands.
Collapse
Affiliation(s)
- A E Proudfoot
- Serono Pharmaceutical Research Institute, Plan-les-Ouates, Switzerland
| | | | | | | | | |
Collapse
|
17
|
Nelson PJ, Teixeira MM. Dissection of inflammatory processes using chemokine biology: Lessons from clinical models. Immunol Lett 2012; 145:55-61. [DOI: 10.1016/j.imlet.2012.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 04/13/2012] [Indexed: 12/30/2022]
|
18
|
Pereira AB, Teixeira AL, Rezende NA, Pereira RM, Miranda DM, Oliveira EA, Teixeira MM, Simões E Silva AC. Urinary chemokines and anti-inflammatory molecules in renal transplanted patients as potential biomarkers of graft function: a prospective study. Int Urol Nephrol 2012; 44:1539-48. [PMID: 22544449 DOI: 10.1007/s11255-012-0176-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/04/2012] [Indexed: 01/05/2023]
Abstract
PURPOSE Clinical- and histopathology-based scores are the limited predictors of allograft outcome. Thus, predictors of allograft survival still remain a challenge. This study aimed to evaluate the urinary levels of chemokines and anti-inflammatory molecules at 30, 90, and 300 days after renal transplantation and to further correlate these measurements to graft function. METHODS Glomerular filtration rate (GFR) and urinary levels of MCP-1/CCL2, MIP-1α/CCL3, RANTES/CCL5, IL-8/CXCL8, IP-10/CXCL10, interleukin-1 receptor antagonist, soluble tumor necrosis factor receptor-1, and receptor-2 were determined at 30, 90, and 300 days after renal transplantation in 22 patients. Transplanted patients were also divided according to the type of donor (living donor, LD, n = 13 or deceased donor, DD, n = 9). RESULTS Urinary levels of all molecules, except MIP-1α/CCL3, remained unchanged at 30, 90, and 300 days after transplantation in our 22 patients. MIP-1α/CCL3 levels significantly reduced from 30 to 300 days and showed a negative correlation with GFR at 30 days. The comparison between LD and DD groups showed similar levels of all markers, except for MCP-1/CCL2, which presented higher values in LD than in DD at 30 days. sTNFR1 and MCP-1/CCL2 significantly reduced from 30 to 300 days in LD group, but only sTNFR2 concentrations at 30 days were negatively correlated with GFR at 300 days. On the other hand, in DD group, IL-1Ra concentrations at 30 and at 90 days were positively correlated with GFR at 300 days. CONCLUSION Urinary chemokine and anti-inflammatory molecules measurements may be a promising tool in the follow-up of renal transplanted patients.
Collapse
Affiliation(s)
- André Barreto Pereira
- Departamento de Clínica Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG 30130-100, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Dilek N, Poirier N, Usal C, Martinet B, Blancho G, Vanhove B. Control of transplant tolerance and intragraft regulatory T cell localization by myeloid-derived suppressor cells and CCL5. THE JOURNAL OF IMMUNOLOGY 2012; 188:4209-16. [PMID: 22450806 DOI: 10.4049/jimmunol.1101512] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature cells that are believed to inhibit immune responses in the contexts of cancer and organ transplantation, in association with regulatory T cells (Treg). However, the way in which MDSC cooperate with Treg remains elusive. In this study, we used DNA microarrays to analyze gene expression in blood-derived MDSC from rat recipients of kidney allografts. We found CCL5 (Rantes), a chemotactic C-C motif 5 chemokine, to be strongly downregulated after treatment with a tolerizing regimen. The amount of CCL5 protein was also lower in the plasma of tolerant recipients, whereas intragraft CCL5 was unchanged. Because CCL5 is chemotactic for Treg, we hypothesized that a gradient of CCL5 between the graft and peripheral blood might contribute to the intragraft localization of Treg in tolerant animals. To test this hypothesis, we treated tolerant rat recipients of kidney allografts with recombinant rat CCL5 to restore normal plasma concentrations. This led to a strong reduction in intragraft Treg monitored by immunohistofluorescence and by quantitative real-time PCR measurement of Foxp3 mRNA. Ultimately, this treatment led to an increase in serum creatinine concentrations and to kidney graft rejection after about a month. The kidney function of syngeneic grafts was not affected by a similar administration of CCL5. These data highlight the contribution of MDSC to the establishment of a graft-to-periphery CCL5 gradient in tolerant kidney allograft recipients, which controls recruitment of Treg to the graft where they likely contribute to maintaining tolerance.
Collapse
Affiliation(s)
- Nahzli Dilek
- INSERM, Unité Mixte de Recherche en Santé 1064, Nantes F-44093, France
| | | | | | | | | | | |
Collapse
|
20
|
von Toerne C, Bedke J, Safi S, Porubsky S, Gretz N, Loewe R, Nelson PJ, Gröne HJ. Modulation of Wnt and Hedgehog signaling pathways is linked to retinoic acid-induced amelioration of chronic allograft dysfunction. Am J Transplant 2012; 12:55-68. [PMID: 21992189 DOI: 10.1111/j.1600-6143.2011.03776.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Chronic renal allograft damage (CAD) is manifested by a smoldering inflammatory process that leads to transplant glomerulopathy, diffuse interstitial fibrosis and tubular atrophy with loss of tubular structures. Using a Fischer 344 (RT1lvl) to Lewis (RT1l) rat renal allograft model, transcriptomic profiling and pathway mapping, we have previously shown that dynamic dysregulation of the Wnt signaling pathways may underlie progressive CAD. Retinoic acid, an important regulator of differentiation during vertebrate embryogenesis, can moderate the damage observed in this experimental model of CAD. We show here that subsets of the Hedgehog (Hh) and canonical Wnt signaling pathways are linked to the pathophysiology of progressive fibrosis, loss of cilia in epithelia and chronic dysfunction. Oral treatment with 13cis retinoic acid (13cRA) was found to selectively ameliorate the dysregulation of the Hh and canonical Wnt pathways associated with CAD, and lead to a general preservation of cilial structures. Interplay between these pathways helps explain the therapeutic effects of retinoic acid treatment in CAD, and suggests future targets for moderating chronic fibrosing organ damage.
Collapse
Affiliation(s)
- C von Toerne
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ullevig S, Zhao Q, Lee CF, Seok Kim H, Zamora D, Asmis R. NADPH oxidase 4 mediates monocyte priming and accelerated chemotaxis induced by metabolic stress. Arterioscler Thromb Vasc Biol 2011; 32:415-26. [PMID: 22095986 DOI: 10.1161/atvbaha.111.238899] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Metabolic disorders increase monocyte chemoattractant protein-1 (MCP-1)-induced monocyte chemotaxis in mice. The goal of this study was to determine the molecular mechanisms responsible for the enhanced responsiveness of monocytes to chemoattractants induced by metabolic stress. METHODS AND RESULTS Chronic exposure of monocytes to diabetic conditions induced by human LDL plus high D-glucose concentrations (LDL+HG) promoted NADPH Oxidase 4 (Nox4) expression, increased intracellular H(2)O(2) formation, stimulated protein S-glutathionylation, and increased chemotaxis in response to MCP-1, platelet-derived growth factor B, and RANTES. Both H(2)O(2) added exogenously and overexpression of Nox4 mimicked LDL+HG-induced monocyte priming, whereas Nox4 knockdown protected monocytes against metabolic stress-induced priming and accelerated chemotaxis. Exposure of monocytes to LDL+HG promoted the S-glutathionylation of actin, decreased the F-actin/G-actin ratio, and increased actin remodeling in response to MCP-1. Preventing LDL+HG-induced protein S-glutathionylation by overexpressing glutaredoxin 1 prevented monocyte priming and normalized monocyte chemotaxis in response to MCP-1. Induction of hypercholesterolemia and hyperglycemia in C57BL/6 mice promoted Nox4 expression and protein S-glutathionylation in macrophages, and increased macrophage recruitment into MCP-1-loaded Matrigel plugs implanted subcutaneous in these mice. CONCLUSIONS By increasing actin-S-glutathionylation and remodeling, metabolic stress primes monocytes for chemoattractant-induced transmigration and recruitment to sites of vascular injury. This Nox4-dependent process provides a novel mechanism through which metabolic disorders promote atherogenesis.
Collapse
Affiliation(s)
- Sarah Ullevig
- Clinical Laboratory Sciences, School of Health Professions, University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Drive, MC 6246, San Antonio, TX 78229-3900, USA
| | | | | | | | | | | |
Collapse
|
22
|
Chevigné A, Fievez V, Schmit JC, Deroo S. Engineering and screening the N-terminus of chemokines for drug discovery. Biochem Pharmacol 2011; 82:1438-56. [DOI: 10.1016/j.bcp.2011.07.091] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 07/21/2011] [Accepted: 07/22/2011] [Indexed: 01/21/2023]
|
23
|
Raffetseder U, Liehn EA, Weber C, Mertens PR. Role of cold shock Y-box protein-1 in inflammation, atherosclerosis and organ transplant rejection. Eur J Cell Biol 2011; 91:567-75. [PMID: 21943779 DOI: 10.1016/j.ejcb.2011.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/05/2011] [Accepted: 07/08/2011] [Indexed: 12/14/2022] Open
Abstract
Chemokines (chemoattractant cytokines) are crucial regulators of immune cell extravasation from the bloodstream into inflamed tissue. Dysfunctional regulation and perpetuated chemokine gene expression are linked to progressive chronic inflammatory diseases and, in respect to transplanted organs, may trigger graft rejection. RANTES (regulated upon activation, normal T cell expressed and secreted (also known as CCL5)) is a model chemokine with relevance in numerous inflammatory diseases where the innate immune response predominates. Transcription factor Y-box binding protein-1 (YB-1) serves as a trans-regulator of CCL5 gene transcription in vascular smooth muscle cells and leucocytes. This review provides an update on YB-1 as a mediator of inflammatory processes and focuses on the role of YB-1 in CCL5 expression in diseases with monocytic cell infiltrates, albeit acute or chronic. Paradigms of such diseases encompass atherosclerosis and transplant rejection where cold shock protein YB-1 takes a dominant role in transcriptional regulation.
Collapse
Affiliation(s)
- Ute Raffetseder
- Department of Nephrology and Clinical Immunology, University Hospital RWTH-Aachen, Pauwelsstrasse 30, 52057 Aachen, Germany.
| | | | | | | |
Collapse
|
24
|
Gross ML, Piecha G, Bierhaus A, Hanke W, Henle T, Schirmacher P, Ritz E. Glycated and carbamylated albumin are more "nephrotoxic" than unmodified albumin in the amphibian kidney. Am J Physiol Renal Physiol 2011; 301:F476-85. [PMID: 21367923 DOI: 10.1152/ajprenal.00342.2010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
There is increasing evidence that proteins in tubular fluid are "nephrotoxic." In vivo it is difficult to study protein loading of tubular epithelial cells in isolation, i.e., without concomitant glomerular damage or changes of renal hemodynamics, etc. Recently, a unique amphibian model has been described which takes advantage of the special anatomy of the amphibian kidney in which a subset of nephrons drains the peritoneal cavity (open nephrons) so that intraperitoneal injection of protein selectively causes protein storage in and peritubular fibrosis around open but not around closed tubules. There is an ongoing debate as to what degree albumin per se is nephrotoxic and whether modification of albumin alters its nephrotoxicity. We tested the hypothesis that carbamylation and glycation render albumin more nephrotoxic compared with native albumin and alternative albumin modifications, e.g., lipid oxidation and lipid depletion. Preparations of native and modified albumin were injected into the axolotl peritoneum. The kidneys were retrieved after 10 days and studied by light microscopy as well as by immunohistochemistry [transforming growth factor (TGF)-β, PDGF, NF-κB, collagen I and IV, RAGE], nonradioactive in situ hybridization, and Western blotting. Two investigators unaware of the animal groups evaluated and scored renal histology. Compared with unmodified albumin, glycated and carbamylated albumin caused more pronounced protein storage. After no more than 10 days, selective peritubular fibrosis was seen around nephrons draining the peritoneal cavity (open nephrons), but not around closed nephrons. Additionally, more intense expression of RAGE, NF-κB, as well as PDGF, TGF-β, EGF, ET-1, and others was noted by histochemistry and confirmed by RT-PCR for fibronectin and TGF-β as well as nonradioactive in situ hybridization for TGF-β and fibronectin. The data indicate that carbamylation and glycation increase the capacity of albumin to cause tubular cell damage and peritubular fibrosis.
Collapse
Affiliation(s)
- M-L Gross
- Institute of Pathology, University of Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
25
|
Poupel L, Combadière C. [Atherosclerosis : on the trail of chemokines]. Biol Aujourdhui 2011; 204:285-293. [PMID: 21215245 DOI: 10.1051/jbio/2010026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2010] [Indexed: 05/30/2023]
Abstract
Atherosclerosis, which is more than a problem of lipid metabolism, is associated with chronic inflammation of large arteries. This is notably caused by the recruitment of circulating blood monocytes to the arterial wall. Extensive studies in humans and mice have shown that the chemokines and their receptors, responsible for leukocyte recirculation, are strongly implicated in the initial onset of atherosclerosis. Murine models have provided further proof of the role of the CCR2/CCL2, CX3CR1/CXCL16 and CCR5/CCL5 axes in the different stages of disease, as well as the preventative roles of CCR1/CCL5 and CXCR6/CXCL16. The integration at the cellular level of various signals in the chemokine network underlines the complex process of leukocyte recruitment to the lesional area. Furthermore the capacity of chemokines to modulate atherosclerosis lies not just with their chemoattractant properties but also with their influence on leukocyte homeostasis. These molecules have therefore quickly become therapeutic targets for atherosclerosis and have opened up new avenues for treating inflammatory diseases. This review principally addresses the implication of chemokines and their receptors in the initial recruitment steps of blood monocytes, and provides an overview of recent research on these molecular controllers of inflammation.
Collapse
Affiliation(s)
- Lucie Poupel
- INSERM-UPMC, Université Paris 6, UMR S 945, Laboratoire d'Immunologies Cellularie, Faculté de Médecine Pité-Salpêtrière, Paris, France
| | | |
Collapse
|
26
|
Furuichi K, Wada T, Kaneko S. Cytokines and chemokines as therapeutic targets for ischemic kidney injury. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
27
|
Vielhauer V, Kulkarni O, Reichel CA, Anders HJ. Targeting the recruitment of monocytes and macrophages in renal disease. Semin Nephrol 2010; 30:318-33. [PMID: 20620675 DOI: 10.1016/j.semnephrol.2010.03.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Macrophages convert proinflammatory or anti-inflammatory signals of tissue microenvironments into response mechanisms. These response mechanisms largely derive from evolutionary conserved defense programs of innate host defense, wound healing, and tissue homeostasis. Hence, in many settings these programs lead to renal inflammation and tissue remodeling (ie, glomerulonephritis and sclerosis or interstitial nephritis and fibrosis). There is abundant experimental evidence that blocking macrophage recruitment or macrophage activation can ameliorate renal inflammation and fibrosis. In this review we discuss experimental tools to target renal macrophage recruitment by using antagonists against selectins, chemokines, integrins, or other important cytokines that mediate renal injury via macrophage recruitment, some of these already having been used in clinical trials.
Collapse
Affiliation(s)
- Volker Vielhauer
- Klinikum der Universität, Ludwig-Maximilians-University, Munich, Germany
| | | | | | | |
Collapse
|
28
|
Berres ML, Koenen RR, Rueland A, Zaldivar MM, Heinrichs D, Sahin H, Schmitz P, Streetz KL, Berg T, Gassler N, Weiskirchen R, Proudfoot A, Weber C, Trautwein C, Wasmuth HE. Antagonism of the chemokine Ccl5 ameliorates experimental liver fibrosis in mice. J Clin Invest 2010; 120:4129-4140. [PMID: 20978355 PMCID: PMC2964968 DOI: 10.1172/jci41732] [Citation(s) in RCA: 216] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 09/01/2010] [Indexed: 12/13/2022] Open
Abstract
Activation of hepatic stellate cells in response to chronic inflammation represents a crucial step in the development of liver fibrosis. However, the molecules involved in the interaction between immune cells and stellate cells remain obscure. Herein, we identify the chemokine CCL5 (also known as RANTES), which is induced in murine and human liver after injury, as a central mediator of this interaction. First, we showed in patients with liver fibrosis that CCL5 haplotypes and intrahepatic CCL5 mRNA expression were associated with severe liver fibrosis. Consistent with this, we detected Ccl5 mRNA and CCL5 protein in 2 mouse models of liver fibrosis, induced by either injection of carbon tetrachloride (CCl4) or feeding on a methionine and choline-deficient (MCD) diet. In these models, Ccl5-/- mice exhibited decreased hepatic fibrosis, with reduced stellate cell activation and immune cell infiltration. Transplantation of Ccl5-deficient bone marrow into WT recipients attenuated liver fibrosis, identifying infiltrating hematopoietic cells as the main source of Ccl5. We then showed that treatment with the CCL5 receptor antagonist Met-CCL5 inhibited cultured stellate cell migration, proliferation, and chemokine and collagen secretion. Importantly, in vivo administration of Met-CCL5 greatly ameliorated liver fibrosis in mice and was able to accelerate fibrosis regression. Our results define a successful therapeutic approach to reduce experimental liver fibrosis by antagonizing Ccl5 receptors.
Collapse
Affiliation(s)
- Marie-Luise Berres
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Rory R. Koenen
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Anna Rueland
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Mirko Moreno Zaldivar
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Daniel Heinrichs
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Hacer Sahin
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Petra Schmitz
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Konrad L. Streetz
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Thomas Berg
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Nikolaus Gassler
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Ralf Weiskirchen
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Amanda Proudfoot
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Christian Weber
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Christian Trautwein
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| | - Hermann E. Wasmuth
- Medical Department III and
Institute of Molecular Cardiovascular Research, University Hospital Aachen, Aachen, Germany.
Department of Gastroenterology and Hepatology, Charité University Hospital Berlin, Berlin, Germany.
Institute of Pathology and
Institute of Clinical Chemistry and Pathobiochemistry, University Hospital Aachen, Aachen, Germany.
Merck Serono Geneva Research Centre, Geneva, Switzerland
| |
Collapse
|
29
|
Abstract
The tolerance state that exists between renal cell carcinoma (RCC) and the host's immune system would be an ideal situation in the setting of human kidney transplantation, in which graft tolerance is the ultimate goal of immunosuppressive therapy. On the other hand, acute rejection, as it appears in renal allografts, would be the optimal immunologic situation in patients with RCC. Analysis of the underlying mechanisms of acute allograft rejection and local pro-tumor immunosuppression could help to identify potential therapeutic targets for inducing immune tolerance in allograft recipients and immune rejection in RCC patients. Experimental kidney transplantation might be a suitable model in which to analyze these processes. Macrophages are a prominent and vital cell type in the cellular infiltrate seen in both RCC and renal allografts. Depending on their polarization, they can initiate and promote either proinflammatory or pro-tumor responses, which lead to tissue rejection or acceptance, respectively. Improved understanding of macrophage biology could lead to therapeutic modification of their function in order to promote a desirable immunologic response in either RCC or transplant tissue.
Collapse
|
30
|
Abstract
Macrophages are present within the transplanted kidney in varying numbers throughout its lifespan. Because of their prominence during acute rejection episodes, macrophages traditionally have been viewed as contributors to T-cell-directed graft injury. With growing appreciation of macrophage biology, it has become evident that different types of macrophages exist within the kidney, subserving a range of functions that include promotion or attenuation of inflammation, participation in innate and adaptive immune responses, and mediation of tissue injury and fibrosis, as well as tissue repair. A deeper understanding of how macrophages accumulate within the kidney and of what factors control their differentiation and function may identify novel therapeutic targets in transplantation.
Collapse
Affiliation(s)
- Steven J Chadban
- Collaborative Transplantation Laboratory, Sydney Medical School, University of Sydney, New South Wales, Australia
| | | | | |
Collapse
|
31
|
Bedke J, Stojanovic T, Kiss E, Behnes CL, Proudfoot AE, Gröne HJ. Viral macrophage inflammatory protein-II improves acute rejection in allogeneic rat kidney transplants. World J Urol 2010; 28:537-42. [PMID: 20401660 PMCID: PMC2908759 DOI: 10.1007/s00345-010-0556-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Accepted: 04/07/2010] [Indexed: 11/28/2022] Open
Abstract
Purpose During rejection, leukocytes are recruited from the peripheral circulation into the graft leading to the damage of endothelial cells, capillary perfusion failure and graft loss. Chemokines play a pivotal role in the recruitment of leukocytes to the endothelium. Viral macrophage inflammatory protein-II (vMIP-II), a human herpes virus-8 DNA-encoded protein, is a broad-spectrum chemokine antagonist. The aim of the study was to prove the beneficial activity of vMIP-II treatment on acute rat kidney allograft damage. Methods Heterotopic rat kidney transplantation was performed in the Fischer 344 to Lewis transplantation model and animals were treated with vMIP-II (2 × 15 µg or 100 µg/day) for 7 days. Rejection-induced damage was analyzed by histology, and microcirculatory changes within the graft were analyzed by in vivo microscopy. Results Viral macrophage inflammatory protein-II significantly improved acute glomerular damage and tubulointerstitial inflammation and lowered the extent of vascular and tubulointerstitial damage of the treated allografts. Functional microcirculation of peritubular capillaries was significantly improved in vivo, and the firm adherence of leukocytes was significantly reduced by vMIP-II treatment. Conclusions The administration of the broad-spectrum antagonist vMIP-II improved acute renal allograft damage, mainly by a reduction in leukocyte recruitment with a subsequently improved renal cortical microcirculation in vivo.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Urology, Eberhard Karls University Tuebingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
32
|
Koenen RR, Weber C. Therapeutic targeting of chemokine interactions in atherosclerosis. Nat Rev Drug Discov 2010; 9:141-53. [PMID: 20118962 DOI: 10.1038/nrd3048] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall that is characterized by a disturbed equilibrium of immune responses and lipid accumulation, leading to the development of plaques. The atherogenic influx of mononuclear cells is orchestrated by chemokines and their receptors. Studies using gene-deficient mice and antagonists based on peptides and small molecules have generated insight into targeting chemokine-receptor axes for treating atherosclerosis, which might complement lipid-lowering strategies and risk factor modulation. Combined inhibition of multiple chemokine axes could interfere with the contributions of chemokines to disease progression at specific cells, stages or sites. In addition, the recently characterized heterophilic interactions of chemokines might present a novel target for the treatment and prevention of inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Rory R Koenen
- The Institute for Molecular Cardiovascular Research, Uni ver sitäts klinikum Aachen, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Pauwelsstrasse 30, 52074 Aachen, Germany
| | | |
Collapse
|
33
|
Sharma M. Chemokines and their receptors: orchestrating a fine balance between health and disease. Crit Rev Biotechnol 2010. [DOI: 10.3109/07388550903187418] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
34
|
Jin H, Kagiampakis I, Li P, LiWang PJ. Structural and functional studies of the potent anti-HIV chemokine variant P2-RANTES. Proteins 2010; 78:295-308. [PMID: 19722264 PMCID: PMC4306592 DOI: 10.1002/prot.22542] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The N-terminal region of the chemokine RANTES is critical for its function. A synthesized N-terminally modified analog of RANTES, P2-RANTES, was discovered using a phage display selection against living CCR5-expressing cells, and has been reported to inhibit HIV-1 env-mediated cell-cell fusion at subnanomolar levels (Hartley et al. J Virol 2003;77:6637-6644). In the present study we produced this protein using E. coli overexpression and extensively studied its structure and function. The x-ray crystal structure of P2-RANTES was solved and refined at 1.7 A resolution. This protein was found to be predominantly a monomer in solution by analytical ultracentrifugation, but a tetramer in the crystal. In studies of glycosaminoglycan binding, P2-RANTES was found to be significantly less able to bind heparin than wild type RANTES. We also tested this protein for receptor internalization where it was shown to be functional, in cell-cell fusion assays where recombinant P2-RANTES was a potent fusion inhibitor (IC(50) = 2.4 +/- 0.8 nM), and in single round infection assays where P2-RANTES inhibited at subnanomolar levels. Further, in a modified fusion assay designed to test specificity of inhibition, P2-RANTES was also highly effective, with a 65-fold improvement over the fusion inhibitor C37, which is closely related to the clinically approved inhibitor T-20. These studies provide detailed structural and functional information for this novel N-terminally modified chemokine mutant. This information will be very useful in the development of more potent anti-HIV agents. PDB Accession Number: 2vxw.
Collapse
Affiliation(s)
- Hongjun Jin
- Texas A & M University, Department of Biochemistry & Biophysics, College Station, TX 77843-2128 U.S.A
| | - Ioannis Kagiampakis
- Texas A & M University, Department of Biochemistry & Biophysics, College Station, TX 77843-2128 U.S.A
| | - Pingwei Li
- Texas A & M University, Department of Biochemistry & Biophysics, College Station, TX 77843-2128 U.S.A
| | - Patricia J. LiWang
- University of California Merced, School of Natural Sciences Merced, CA 95343
| |
Collapse
|
35
|
Bedke J, Nelson PJ, Kiss E, Muenchmeier N, Rek A, Behnes CL, Gretz N, Kungl AJ, Gröne HJ. A novel CXCL8 protein-based antagonist in acute experimental renal allograft damage. Mol Immunol 2009; 47:1047-57. [PMID: 20004976 DOI: 10.1016/j.molimm.2009.11.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 11/06/2009] [Accepted: 11/13/2009] [Indexed: 01/11/2023]
Abstract
Acute renal allograft damage is caused by early leukocyte infiltration which is mediated in part by chemokines presented by glycosaminoglycan (GAG) structures on endothelial surfaces. CXCL8 can recruit neutrophils and induce the firm arrest of monocytes on activated endothelial cells. A human CXCL8-based antagonist (dnCXCL8) designed to generate a dominant-negative mutant protein with enhanced binding to GAG structures and reduced CXCR1/2 receptor binding ability was tested in models of early allograft injury. The agent displayed enhanced binding to GAG structures in vitro and could antagonize CXCL8-induced firm adhesion of monocytes as well as neutrophils to activated microvascular endothelium in physiologic flow assays. In a rat model of acute renal damage, dnCXCL8 treatment limited proximal tubular damage and reduced granulocyte infiltration. In a Fischer 344 (RT1(lvl)) to Lewis (RT1(l)) rat acute renal allograft model, dnCXCL8 was found to reduce monocyte and CD8+ T-cell infiltration into glomeruli and to limit tubular interstitial inflammation and tubulitis in vivo. Early treatment of allografts with agents like dnCXCL8 may help reduce acute allograft damage and preserve renal morphology and thereby help limit chronic dysfunction.
Collapse
Affiliation(s)
- Jens Bedke
- Department of Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Silverstein DM. Inflammation after renal transplantation: Role in the development of graft dysfunction and potential therapies. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/17471060802302339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
37
|
Stojanovic T, Wagner AH, Wang S, Kiss E, Rockstroh N, Bedke J, Gröne HJ, Hecker M. STAT-1 decoy oligodeoxynucleotide inhibition of acute rejection in mouse heart transplants. Basic Res Cardiol 2009; 104:719-29. [PMID: 19352584 PMCID: PMC3085763 DOI: 10.1007/s00395-009-0028-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2008] [Revised: 03/16/2009] [Accepted: 03/25/2009] [Indexed: 12/02/2022]
Abstract
During acute rejection of cardiac transplants endothelial cell-leukocyte interaction fueled by co-stimulatory molecules like CD40/CD154 may ultimately lead to graft loss. One key player in up-regulating the expression of such pro-inflammatory gene products is the interferon-gamma-dependent transcription factor STAT-1. Hence down-regulating interferon-gamma-stimulated pro-inflammatory gene expression in the graft endothelial cells by employing a decoy oligodeoxynucleotide (dODN) neutralising STAT-1 may protect the graft. To verify this hypothesis, heterotopic mouse heart transplantation was performed in the allogeneic B10.A(2R) to C57BL/6 and syngeneic C57BL/6 to C57BL/6 strain combination without immunosuppression. Graft vessels were pre-treated with STAT-1 dODN, mutant control ODN (10 muM each) or vehicle (Ringer solution). Cellular rejection (vascular and interstitial component) was graded histologically and CD40, ICAM-1, VCAM-1, MCP-1, E-selectin and RANTES expression in the graft monitored by real time PCR 24 h and 9 days post-transplantation. Nine days after transplantation both rejection scores were significantly diminished by 85 and 70%, respectively, in STAT-1 dODN-treated allografts as compared to mutant control ODN-treated allografts. According to immunohistochemistry analysis, this was accompanied by a reduced infiltration of monocyte/macrophages and T cells into the graft myocardium. In addition, pro-inflammatory gene expression was strongly impaired by more than 80% in STAT-1 dODN-treated allografts 24 h post-transplantation but not in mutant control ODN or vehicle-treated allografts. This inhibitory effect on pro-inflammatory gene expression was no longer detectable 9 days post-transplantation. Single periprocedural treatment with a STAT-1 dODN thus effectively reduces cellular rejection in mouse heart allografts. This effect is associated both with an early decline in pro-inflammatory gene expression and a later drop in mononuclear cell infiltration.
Collapse
Affiliation(s)
- Tomislav Stojanovic
- Department of Heart, Thoracic and Vascular Surgery, University of Göttingen, Robert-Koch-Str 40, 37073 Göttingen, Germany
| | - Andreas H. Wagner
- Institute of Physiology und Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Shijun Wang
- Department of Cellular and Molecular Pathology, German Cancer Research Centre, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Eva Kiss
- Department of Cellular and Molecular Pathology, German Cancer Research Centre, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nicolas Rockstroh
- Institute of Physiology und Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Jens Bedke
- Department of Urology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076 Tübingen, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Centre, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Markus Hecker
- Institute of Physiology und Pathophysiology, Division of Cardiovascular Physiology, University of Heidelberg, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
38
|
von Toerne C, Schmidt C, Adams J, Kiss E, Bedke J, Porubsky S, Gretz N, Lindenmeyer MT, Cohen CD, Gröne HJ, Nelson PJ. Wnt pathway regulation in chronic renal allograft damage. Am J Transplant 2009; 9:2223-39. [PMID: 19681821 DOI: 10.1111/j.1600-6143.2009.02762.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The Wnt signaling pathway, linked to development, has been proposed to be recapitulated during the progressive damage associated with chronic organ failure. Chronic allograft damage following kidney transplantation is characterized by progressive fibrosis and a smoldering inflammatory infiltrate. A modified, Fischer 344 (RT1(lvl)) to Lewis (RT1(l)) rat renal allograft model that reiterates many of the major pathophysiologic processes seen in patients with chronic allograft failure was used to study the progressive disease phenotype and specific gene product expression by immunohistochemistry and transcriptomic profiling. Central components of the Tgfb, canonical Wnt and Wnt-Ca2+ signaling pathways were significantly altered with the development of chronic damage. In the canonical Wnt pathway, Wnt3, Lef1 and Tcf1 showed differential regulation. Target genes Fn1, Cd44, Mmp7 and Nos2 were upregulated and associated with the progression of renal damage. Changes in the Wnt-Ca2+ pathway were evidenced by increased expression of Wnt6, Wnt7a, protein kinase C, Cam Kinase II and Nfat transcription factors and the target gene vimentin. No evidence for alterations in the Wnt planar cell polarity (PCP) pathway was detected. Overall results suggest cross talk between the Wnt and Tgfb signaling pathways during allograft inflammatory damage and present potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- C von Toerne
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Esposito C, Grosjean F, Torreggiani M, Maggi N, Esposito V, Migotto C, Mangione F, Tinelli C, Dal Canton A. Increased asymmetric dimethylarginine serum levels are associated with acute rejection in kidney transplant recipients. Transplant Proc 2009; 41:1570-3. [PMID: 19545681 DOI: 10.1016/j.transproceed.2009.03.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2008] [Revised: 03/01/2009] [Accepted: 03/11/2009] [Indexed: 11/19/2022]
Abstract
Asymmetric dimethylarginine (ADMA) has been identified as a marker of endothelial dysfunction and an independent risk factor for cardiovascular events in uremic subjects. This study evaluated ADMA plasma levels in kidney transplant recipients. ADMA levels were serially measured during the first year posttransplantation in 41 recipients treated with cyclosporine regimen (CY), sirolimus (SIR), or low-dose cyclosporine plus everolimus (E). Homocysteine, C reactive protein (CRP), nitric oxide (NO), and standard routine laboratory analyses were determined serially. ADMA significantly increased at 6 months posttransplantation, but was significantly lower among patients on SIR or E. NO was only slightly reduced in patients with increased ADMA levels. Interestingly, ADMA was significantly increased during the first 4 days posttransplantation in patients who experienced acute rejection during the first 6 months after transplantation. The same group of patients demonstrated higher levels of CRP and systolic blood pressure before transplantation. Our results demonstrated that ADMA was increased in patients on CY at 6 months. When increased soon after transplantation ADMA may be associated with episodes of acute rejection in kidney transplant recipients. The presence of elevated systolic blood pressure, as well as CRP and ADMA levels, suggested a role for endothelial dysfunction in the development of acute rejection episodes among deceased donor kidney transplant recipients.
Collapse
Affiliation(s)
- C Esposito
- Unit of Nephrology, Dialysis and Transplantation, Policlinico San Matteo, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Renal tubular epithelial cells as immunoregulatory cells in renal allograft rejection. Transplant Rev (Orlando) 2009; 23:129-38. [DOI: 10.1016/j.trre.2009.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
42
|
Cha RH, Yang SH, Kim HS, Kim SM, Park MH, Ha J, Kim YS. Genetic interactions between the donor and the recipient for susceptibility to acute rejection in kidney transplantation: polymorphisms of CCR5. Nephrol Dial Transplant 2009; 24:2919-25. [PMID: 19561149 DOI: 10.1093/ndt/gfp317] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acute rejection (AR) contributes to the development of chronic allograft nephropathy that is the major cause of graft failure. We analyzed the 59029G>A polymorphism and an internal 32 bp deletion (CCR5 32) of CCR chemokine receptor 5 (CCR5) Delta and tried to prove the hypothesis that genetic interactions between the donor and the recipient influence the development of AR. METHODS We detected genetic polymorphisms by the TaqMan(R) method and by sizing PCR amplicons (n = 486). The primary outcomes were early acute rejection (EAR) and repeated early acute rejection (RR). We defined EAR as the occurrence of a biopsy-proven AR within 3 months after transplantation. RESULTS The development of EAR was dependent on the number of A alleles in recipients and showed a dose-response relationship (P = 0.002). When we combined the number of A alleles in both donor and recipient, episodes of EAR and RR were more prevalent as the allelic number increased (A allelic number 0 & 1, 2 versus 3 & 4, P = 0.048; 0 & 1 versus 3 & 4, P = 0.006). Statistical significance was preserved after multivariate analysis of sex, HLA mismatch and type of donor with the recipient's age as the continuous term. Also, graft survival was different according to the presence of the A allele, i.e. recipients carrying A allele (+) grafts showed poor graft survival (P = 0.008 by a log-rank test). Again, the number of A alleles affected graft survival as the recipients who carried more A alleles had poor graft survival (A allele number 0 & 1 versus 2 versus 3 & 4, P = 0.011; 0 & 1 versus 3 & 4, P = 0.08; 0 & 1 versus 2, P = 0.002; by a log-rank test). All of the participants were wild-type homozygotes for CCR5Delta32. CONCLUSIONS The A allele of CCR5 59029G>A was a risk factor for EAR and RR. As the number of A alleles increased, episodes of EAR were more frequently observed.
Collapse
Affiliation(s)
- Ran-hui Cha
- Department of Internal Medicine, Seoul National University College of Medicine, Korea
| | | | | | | | | | | | | |
Collapse
|
43
|
Schaier M, Vorwalder S, Sommerer C, Dikow R, Hug F, Gross ML, Waldherr R, Zeier M. Role of FTY720 on M1 and M2 macrophages, lymphocytes, and chemokines in 5/6 nephrectomized rats. Am J Physiol Renal Physiol 2009; 297:F769-80. [PMID: 19535570 DOI: 10.1152/ajprenal.90530.2008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal injury is accompanied by the presence of infiltrating inflammatory cells in the glomerulus and tubulointerstitium. FTY720 modifies lymphocyte migration into injured tissues by lymphocyte sequestration to secondary lymphoid organs. The purpose of this study was to examine the potential of FTY720 to influence the inflammatory response in a nonimmunological model of renal failure. Sham-operated and 5/6 nephrectomized (SNX) Sprague-Dawley rats received two different doses of FTY720 or vehicle orally for 14 wk. Treatment with FTY720 reduced glomerular and tubulointerstitial damage in SNX rats but failed to stabilize creatinine clearance. The increase in gene expression of chemokine receptors CCR1, CCR2, and CCR5 in kidneys of vehicle-treated SNX rats was significantly attenuated by high-dose FTY720. Treatment with high-dose FTY720 tended to normalize RANTES and MCP-1 renal gene expression. FTY720 affected not only glomerular and tubulointerstitial lymphocytes, but M1 and M2 phenotype macrophages were also reduced. FTY720 significantly reduced key mediators of renal inflammation and fibrosis. FTY720 also decreased immunoregulation of M2 macrophages, which are beneficial for tissue remodeling and repair.
Collapse
Affiliation(s)
- Matthias Schaier
- Dept. of Nephrology, Univ. of Heidelberg, Im Neuenheimer Feld 162, 69120 Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Kiss DL, Longden J, Fechner GA, Avery VM. The functional antagonist Met-RANTES: a modified agonist that induces differential CCR5 trafficking. Cell Mol Biol Lett 2009; 14:537-47. [PMID: 19448977 PMCID: PMC6275935 DOI: 10.2478/s11658-009-0017-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 05/06/2009] [Indexed: 11/20/2022] Open
Abstract
CC chemokine receptor 5 (CCR5) is a pro-inflammatory chemokine receptor that is expressed on cells of the immune system, and specializes in cell migration in response to inflammation and tissue damage. Due to its key role in cell communication and migration, this receptor is involved in various inflammatory and autoimmune diseases, in addition to HIV infection. Met-RANTES is a modified CCR5 ligand that has previously been shown to antagonize CCR5 activation and function in response to its natural ligands in vitro. In vivo, Met-RANTES is able to reduce inflammation in models of induced inflammatory and autoimmune diseases. However, due to the fact that Met-RANTES is also capable of partial agonist activity regarding receptor signaling and internalization, it is clear that Met-RANTES does not function as a conventional receptor antagonist. To further elucidate the effect of Met-RANTES on CCR5, receptor trafficking was investigated in a CHO-CCR5-GFP cell line using the Opera confocal plate reader. The internalization response of CCR5 was quantified, and showed that Met-RANTES internalized CCR5 in a slower, less potent manner than the agonists CCL3 and CCL5. Fluorescent organelle labeling and live cell imaging showed CCL3 and CCL5 caused CCR5 to traffic through sorting endosomes, recycling endosomes and the Golgi apparatus. In contrast, Met-RANTES caused CCR5 to traffic through sorting endosomes and the Golgi apparatus in a manner that was independent of recycling endosomes. As receptor trafficking impacts on cell surface expression and the ability of the receptor to respond to more ligand, this information may indicate an alternative regulation of CCR5 by Met-RANTES that allows the modified ligand to reduce inflammation through stimulation of a pro-inflammatory receptor.
Collapse
Affiliation(s)
- Debra L. Kiss
- Discovery Biology, Eskitis Institute for Cell and Molecular Therapies, Brisbane Innovation Park, Griffith University, Don Young Road, Nathan, QLD 4111 Australia
| | - James Longden
- Discovery Biology, Eskitis Institute for Cell and Molecular Therapies, Brisbane Innovation Park, Griffith University, Don Young Road, Nathan, QLD 4111 Australia
| | - Gregory A. Fechner
- Discovery Biology, Eskitis Institute for Cell and Molecular Therapies, Brisbane Innovation Park, Griffith University, Don Young Road, Nathan, QLD 4111 Australia
| | - Vicky M. Avery
- Discovery Biology, Eskitis Institute for Cell and Molecular Therapies, Brisbane Innovation Park, Griffith University, Don Young Road, Nathan, QLD 4111 Australia
| |
Collapse
|
45
|
Chemokine/chemokine receptor-mediated inflammation regulates pathologic changes from acute kidney injury to chronic kidney disease. Clin Exp Nephrol 2009; 13:9-14. [DOI: 10.1007/s10157-008-0119-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 09/09/2008] [Indexed: 11/26/2022]
|
46
|
Kumar D, Hosse J, von Toerne C, Noessner E, Nelson PJ. JNK MAPK Pathway Regulates Constitutive Transcription of CCL5 by Human NK Cells through SP1. THE JOURNAL OF IMMUNOLOGY 2009; 182:1011-20. [DOI: 10.4049/jimmunol.182.2.1011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
47
|
Fischer-Maas L, Schneppenheim R, Oyen F, Grabhorn E, Richter A, Fischer L, Ganschow R. Analysis of the CC chemokine receptor 5Delta32 polymorphism in pediatric liver transplant recipients. Pediatr Transplant 2008; 12:769-72. [PMID: 18194353 DOI: 10.1111/j.1399-3046.2007.00900.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
In adult liver graft recipients, it has been shown that certain chemokine polymorphisms (CCR5Delta32) may correspond to ischemic type biliary lesions leading to chronic graft dysfunction. The aim of our present study was to assess the importance of CCR5Delta32 polymorphism in a cohort of pediatric liver graft recipients with regard to acute or chronic graft dysfunction. A total of 137 children post-liver transplantation have been included for genetic analysis (CCR5Delta32 polymorphism), and the incidence of acute and chronic graft dysfunction was analyzed. The most common diagnosis leading to LTx was biliary atresia (56.2%), the median age was 14 months, and 33.5% of the patients received a living-related graft. In all, 110 of the subjects were found to have the CCR5 wild type, 25 children were heterozygous for CCR5Delta32, and two patients were homozygous. Of 137, 44 (32.1%) developed acute graft rejection, nine out of 137 (6.6%) chronic graft dysfunction (vanishing bile duct syndrome), and 84 (61.3%) children had neither acute nor chronic graft rejection. There was no significant correlation between acute graft rejection or chronic graft dysfunction and the CCR5Delta32 allele in the study population. We conclude that CCR5Delta32 polymorphism may not play a role in acute or chronic liver graft dysfunction in children.
Collapse
Affiliation(s)
- Louise Fischer-Maas
- Department of Pediatrics, Pediatrics Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
48
|
Raffetseder U, Rauen T, Djudjaj S, Kretzler M, En-Nia A, Tacke F, Zimmermann HW, Nelson PJ, Frye BC, Floege J, Stefanidis I, Weber C, Mertens PR. Differential regulation of chemokine CCL5 expression in monocytes/macrophages and renal cells by Y-box protein-1. Kidney Int 2008; 75:185-96. [PMID: 18800033 DOI: 10.1038/ki.2008.457] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Y-box protein-1 (YB-1) belongs to the family of cold shock proteins that have pleiotropic functions such as gene transcription, RNA splicing, and mRNA translation. YB-1 has a critical role in atherogenesis due to its regulatory effects on chemokine CCL5 (RANTES) gene transcription in vascular smooth muscle cells. Since CCL5 is a key mediator of kidney transplant rejection, we determined whether YB-1 is involved in allograft rejection by manipulating its expression. In human kidney biopsies, YB-1 transcripts were amplified 17-fold in acute and 21-fold in chronic allograft rejection with a close correlation between CCL5 and YB-1 mRNA expression in both conditions. Among three possible YB-1 binding sites in the CCL5 promoter, a critical element was mapped at -28/-10 bps. This site allowed up-regulation of CCL5 transcription in monocytic THP-1 and HUT78 T-cells and in human primary monocytes; however, it repressed transcription in differentiated macrophages. Conversely, YB-1 knockdown led to decreased CCL5 transcription and secretion in monocytic cells. We show that YB-1 is a cell-type specific regulator of CCL5 expression in infiltrating T-cells and monocytes/macrophages and acts as an adaptive controller of inflammation during kidney allograft rejection.
Collapse
Affiliation(s)
- Ute Raffetseder
- Department of Nephrology and Clinical Immunology, University Hospital RWTH-Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
A key event during T cell-mediated rejection of allografts is the trafficking of donor antigen-primed effector T cells from the lymphoid tissue to the graft. This trafficking is mediated in part by chemokine produced in the graftengaging receptors on the T cells and other graftinfiltrating leukocytes. The presence of specific sets of chemokines and chemokine receptors is detectable in rejecting allografts. In animal models, allograft rejection is delayed when chemokine-chemokine receptor function is absent or antagonized but cellular infiltration and graft survival eventually occur, suggesting that T cells and other leukocytes use several trafficking mechanisms during rejection. The use of chemokines as footprints of rejection may be of considerable value as noninvasive biomarkers in transplantation.
Collapse
Affiliation(s)
- Austin D. Schenk
- NB3-59, Department of Immunology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Department of Pathology, Case Western Reserve University School of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Joshua M. Rosenblum
- NB3-59, Department of Immunology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Department of Pathology, Case Western Reserve University School of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Robert L. Fairchild
- NB3-59, Department of Immunology, Glickman Urological and Kidney Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- NB3-59, Department of Immunology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Department of Pathology, Case Western Reserve University School of Medicine, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
50
|
Abstract
Transplant vasculopathy (TV) remains the leading cause of late death among heart transplant recipients. Transplant vasculopathy is characterized by progressive neointimal proliferation, leading to ischemic failure of the allograft. Multiple experimental and clinical studies have shown that injury to the graft at various stages of transplantation can be a risk factor for development of transplant vasculopathy. The hallmark of cardiac allograft injury is the infiltration of leukocytes. Recruitment of leukocytes requires intercellular communication between infiltrating cells, endothelium, parenchymal cells, and components of extracellular matrix. These events are mediated via the generation of adhesion molecules, cytokines, and chemokines. The chemokines, by virtue of their specific cell receptor expression, can selectively mediate the local recruitment/activation of distinct leukocytes/cells, allowing for migration across the endothelium and beyond the vascular compartment. This report provides a comprehensive review of the chemokines that participate in the development of transplant vasculopathy.
Collapse
Affiliation(s)
- John A Belperio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Los Angeles, CA 90024-1922
| | - Abbas Ardehali
- Division of Cardiothoracic Surgery, Department of Surgery at the David Geffen School of Medicine at UCLA, Los Angeles, CA 90024-1922
| |
Collapse
|