151
|
Diaz-Miron J, Sun R, Choi P, Sommovilla J, Guo J, Erwin CR, Mei J, Scott Worthen G, Warner BW. The effect of impaired angiogenesis on intestinal function following massive small bowel resection. J Pediatr Surg 2015; 50:948-53. [PMID: 25818317 PMCID: PMC4439276 DOI: 10.1016/j.jpedsurg.2015.03.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/10/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE Intestinal adaptation involves villus lengthening, crypt deepening, and increased capillary density following small bowel resection (SBR). Mice lacking the proangiogenic chemokine CXCL5 have normal structural adaptation but impaired angiogenesis. This work evaluates the impact of incomplete adaptive angiogenesis on the functional capacity of the intestine after SBR. METHODS CXCL5 knockout (KO) and C57BL/6 wild-type (WT) mice underwent 50% SBR. Magnetic resonance imaging measured weekly body composition. Intestinal absorptive capacity was evaluated through fecal fat analysis. Gene expression profiles for select macronutrient transporters were measured via RT-PCR. Postoperative crypt and villus measurements were assessed for structural adaptation. Submucosal capillary density was measured through CD31 immunohistochemistry. RESULTS Comparable postoperative weight gain occurred initially. Diminished weight gain, impaired fat absorption, and elevated steatorrhea occurred in KO mice after instituting high-fat diet. Greater postoperative upregulation of ABCA1 fat transporter occurred in WT mice, while PEPT1 protein transporter was significantly downregulated in KO mice. KO mice had impaired angiogenesis but intact structural adaptation. CONCLUSION After SBR, KO mice display an inefficient intestinal absorption profile with perturbed macronutrient transporter expression, impaired fat absorption, and slower postoperative weight gain. In addition to longer villi and deeper crypts, an intact angiogenic response may be required to achieve functional adaptation to SBR.
Collapse
Affiliation(s)
- Jose Diaz-Miron
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Raphael Sun
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Pamela Choi
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Joshua Sommovilla
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Jun Guo
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Christopher R Erwin
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO
| | - Junjie Mei
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - G Scott Worthen
- Division of Neonatology, The Children's Hospital of Philadelphia, Philadelphia, PA
| | - Brad W Warner
- Division of Pediatric Surgery, St Louis Children's Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO.
| |
Collapse
|
152
|
Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 2015; 26:311-27. [DOI: 10.1016/j.cytogfr.2014.11.009] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
|
153
|
Wu F, Zhao Y, Jiao T, Shi D, Zhu X, Zhang M, Shi M, Zhou H. CXCR2 is essential for cerebral endothelial activation and leukocyte recruitment during neuroinflammation. J Neuroinflammation 2015; 12:98. [PMID: 25990934 PMCID: PMC4438521 DOI: 10.1186/s12974-015-0316-6] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 04/30/2015] [Indexed: 01/05/2023] Open
Abstract
Background Chemokines and chemokine receptors cooperate to promote immune cell recruitment to the central nervous system (CNS). In this study, we investigated the roles of CXCR2 and CXCL1 in leukocyte recruitment to the CNS using a murine model of neuroinflammation. Methods Wild-type (WT), CXCL1−/−, and CXCR2−/− mice each received an intracerebroventricular (i.c.v.) injection of lipopolysaccharide (LPS). Esterase staining and intravital microscopy were performed to examine neutrophil recruitment to the brain. To assess endothelial activation in these mice, the expression of adhesion molecules was measured via quantitative real-time polymerase chain reaction (PCR) and Western blotting. To identify the cellular source of functional CXCR2, chimeric mice were generated by transferring bone marrow cells between the WT and CXCR2−/− mice. Results Expression levels of the chemokines CXCL1, CXCL2, and CXCL5 were significantly increased in the brain following the i.c.v. injection of LPS. CXCR2 or CXCL1 deficiency blocked neutrophil infiltration and leukocyte recruitment in the cerebral microvessels. In the CXCR2−/− and CXCL1−/− mice, the cerebral endothelial expression of adhesion molecules such as P-selectin and VCAM-1 was dramatically reduced. Furthermore, the bone marrow transfer experiments demonstrated that CXCR2 expression on CNS-residing cells is essential for cerebral endothelial activation and leukocyte recruitment. Compared with microglia, cultured astrocytes secreted a much higher level of CXCL1 in vitro. Astrocyte culture conditioned medium significantly increased the expression of VCAM-1 and ICAM-1 in cerebral endothelial cells in a CXCR2-dependent manner. Additionally, CXCR2 messenger RNA (mRNA) expression in cerebral endothelial cells but not in microglia or astrocytes was increased following tumor necrosis factor-α (TNF-α) stimulation. The intravenous injection of the CXCR2 antagonist SB225002 significantly inhibited endothelial activation and leukocyte recruitment to cerebral microvessels. Conclusions CXCL1 secreted by astrocytes and endothelial CXCR2 play essential roles in cerebral endothelial activation and subsequent leukocyte recruitment during neuroinflammation.
Collapse
Affiliation(s)
- Fengjiao Wu
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Yawei Zhao
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Tian Jiao
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Xingxing Zhu
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Mingshun Zhang
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| | - Meiqing Shi
- Division of Immunology, Virginia-Maryland College of Veterinary Medicine, University of Maryland, College Park, Maryland, MD, 20742, USA.
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, JS, 210029, China.
| |
Collapse
|
154
|
LASP-1: a nuclear hub for the UHRF1-DNMT1-G9a-Snail1 complex. Oncogene 2015; 35:1122-33. [PMID: 25982273 PMCID: PMC4651668 DOI: 10.1038/onc.2015.166] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 04/09/2015] [Accepted: 04/12/2015] [Indexed: 12/30/2022]
Abstract
Nuclear LASP-1 has a direct correlation with overall survival of breast cancer patients. In this study, immunohistochemical analysis of a human breast TMA showed that LASP-1 is absent in normal human breast epithelium but the expression increases with malignancy and is highly nuclear in aggressive breast cancer. We investigated whether the chemokines and growth factors present in the tumor microenvironment could trigger nuclear translocation of LASP-1.Treatment of human breast cancer cells with CXCL12, EGF and Heregulin and HMEC-CXCR2 cells with CXCL8 facilitated nuclear shuttling of LASP-1. Data from the biochemical analysis of the nuclear and cytosolic fractions further confirmed the nuclear translocation of LASP-1 upon chemokine and growth factor treatment. CXCL12-dependent nuclear import of LASP-1 could be blocked by CXCR4 antagonist, AMD-3100. Knock down of LASP-1 resulted in alterations in gene expression leading to an increased level of cell junction and extracellular matrix proteins and an altered cytokine secretory profile. Three dimensional cultures of human breast cancer cells on Matrigel revealed an altered colony growth, morphology and arborization pattern in LASP-1 knock down cells. Functional analysis of the LASP-1 knock down cells revealed increased adhesion to collagen IV and decreased invasion through the Matrigel. Proteomics analysis of immunoprecipitates of LASP-1 and subsequent validation approaches revealed that LASP-1associated with the epigenetic machinery especially UHRF1, DNMT1, G9a and the transcription factor Snail1. Interestingly, LASP-1 associated with UHRF1, G9a, Snail1 and di- and tri-methylated histoneH3 in a CXCL12-dependent manner based on immunoprecipitation and proximity ligation assays. LASP-1 also directly bound to Snail1 which may stabilize Snail1. Thus, nuclear LASP-1 appears to functionally serve as a hub for the epigenetic machinery.
Collapse
|
155
|
Andrejecsk JW, Chang WG, Pober JS, Saltzman WM. Controlled protein delivery in the generation of microvascular networks. Drug Deliv Transl Res 2015; 5:75-88. [PMID: 25767747 PMCID: PMC4354697 DOI: 10.1007/s13346-012-0122-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Rapid induction and stabilization of new microvascular networks is essential for the proper functioning of engineered tissues. Many efforts to achieve this goal have used proangiogenic proteins-such as vascular endothelial growth factors-to induce the formation of new microvessels. These proteins have demonstrated promise in improving vascularization, but it is also clear that the spatial and temporal presentation of these signals is important for achieving proper vascular function. Delivery systems that present proteins in a localized and sustained manner, can promote the formation and stabilization of microvascular networks by precisely presenting proangiogenic proteins at desired locations, and for specified durations. Further, these systems allow for some control over the sequence of release of multiple proteins, and it has become clear that such coordination is critical for the development of fully functional and mature vascular structures. This review focuses on the actions of proangiogenic proteins and the innovations in controlled release technologies that precisely deliver these to stimulate microvascular network formation and stabilization.
Collapse
Affiliation(s)
| | - William G Chang
- Department of Medicine and Section of Nephrology, Yale University School of Medicine, New Haven, CT 06520
| | - Jordan S Pober
- Departments of Immunobiology, Pathology, and Dermatology, Yale University School of Medicine, New Haven, CT 06520
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511
| |
Collapse
|
156
|
Zhao Y, Zhu L, Zhou T, Zhang Q, Shi S, Liu L, Lv J, Zhang H. Urinary CXCL1: a novel predictor of IgA nephropathy progression. PLoS One 2015; 10:e0119033. [PMID: 25816025 PMCID: PMC4376727 DOI: 10.1371/journal.pone.0119033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 01/09/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND IgA nephropathy (IgAN) is the most common form of primary glomerulonephritis worldwide. In recent years, consistent efforts have been made to develop new non-invasive biomarkers for IgAN progression. In our previous in vitro study we found mesangial derived CXCL1 as a contributor for kidney injury, and observed higher urinary CXCL1 levels in patients with IgAN. It implied that the urinary CXCL1 might be a potential biomarker. METHODS In the present study, we enrolled 425 IgAN patients with follow-up data and detected their urinary CXCL1 levels at the time of renal biopsy, to explore the predictive value of urinary CXCL1 in IgAN progression. Urinary CXCL1 levels were measured using enzyme-linked immunosorbent assay. RESULTS Urinary CXCL1 levels were associated with presently well established predictors of IgAN progression, including SBP (r = 0.138, p = 0.004), DBP (r = 0.114, p = 0.019), proteinuria (r = 0.155, p = 0.001), eGFR (r = -0.259, p<0.001) and tubular atrophy and interstitial fibrosis (r = 0.181, p<0.001). After adjusted for them, higher levels of urinary CXCL1 were independently associated with a greater risk of deterioration in renal function (HR, per s.d. increment of natural log-transformed CXCL1: 1.748; 95% CI: 1.222-2.499, P = 0.002). Furthermore, time-dependent receiver operating characteristic (ROC) curve showed that urinary CXCL1, when combined with proteinuria and eGFR, could enhance the prognostic value of these traditional predictors for IgAN progression. CONCLUSIONS The results in our present study suggested urinary CXCL1 as a new non-invasive predictor of IgAN progression.
Collapse
Affiliation(s)
- Yanfeng Zhao
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Li Zhu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
- * E-mail:
| | - Tong Zhou
- Renal Division, Department of Medicine, The First People’s Hospital of Aksu District, Xinjiang, China
| | - Qingxian Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Sufang Shi
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Lijun Liu
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Jicheng Lv
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| | - Hong Zhang
- Renal Division, Department of Medicine, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment, Peking University, Ministry of Education, Beijing, China
| |
Collapse
|
157
|
Herlea-Pana O, Yao L, Heuser-Baker J, Wang Q, Wang Q, Georgescu C, Zou MH, Barlic-Dicen J. Chemokine receptors CXCR2 and CX3CR1 differentially regulate functional responses of bone-marrow endothelial progenitors during atherosclerotic plaque regression. Cardiovasc Res 2015; 106:324-37. [PMID: 25765938 DOI: 10.1093/cvr/cvv111] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/07/2015] [Indexed: 01/03/2023] Open
Abstract
AIMS Atherosclerosis manifests itself as arterial plaques, which lead to heart attacks or stroke. Treatments supporting plaque regression are therefore aggressively pursued. Studies conducted in models in which hypercholesterolaemia is reversible, such as the Reversa mouse model we have employed in the current studies, will be instrumental for the development of such interventions. Using this model, we have shown that advanced atherosclerosis regression occurs when lipid lowering is used in combination with bone-marrow endothelial progenitor cell (EPC) treatment. However, it remains unclear how EPCs home to regressing plaques and how they augment atherosclerosis reversal. Here we identify molecules that support functional responses of EPCs during plaque resolution. METHODS AND RESULTS Chemokines CXCL1 and CX3CL1 were detected in the vascular wall of atheroregressing Reversa mice, and their cognate receptors CXCR2 and CX3CR1 were observed on adoptively transferred EPCs in circulation. We tested whether CXCL1-CXCR2 and CX3CL1-CX3CR1 axes regulate functional responses of EPCs during plaque reversal. We show that pharmacological inhibition of CXCR2 or CX3CR1, or genetic inactivation of these two chemokine receptors interfered with EPC-mediated advanced atherosclerosis regression. We also demonstrate that CXCR2 directs EPCs to regressing plaques while CX3CR1 controls a paracrine function(s) of these cells. CONCLUSION CXCR2 and CX3CR1 differentially regulate EPC functional responses during atheroregression. Our study improves understanding of how chemokines and chemokine receptors regulate plaque resolution, which could determine the effectiveness of interventions reducing complications of atherosclerosis.
Collapse
Affiliation(s)
- Oana Herlea-Pana
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Longbiao Yao
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Janet Heuser-Baker
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| | - Qiongxin Wang
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Qilong Wang
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Constantin Georgescu
- Arthritis and Clinical Immunology Programs, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ming-Hui Zou
- Division of Molecular Medicine, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jana Barlic-Dicen
- Cardiovascular Biology, Oklahoma Medical Research Foundation, 825 NE 13th Street, Oklahoma City, OK 73104, USA
| |
Collapse
|
158
|
Wilson GC, Kuboki S, Freeman CM, Nojima H, Schuster RM, Edwards MJ, Lentsch AB. CXC chemokines function as a rheostat for hepatocyte proliferation and liver regeneration. PLoS One 2015; 10:e0120092. [PMID: 25756662 PMCID: PMC4355613 DOI: 10.1371/journal.pone.0120092] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/03/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Our previous in vitro studies have demonstrated dose-dependent effects of CXCR2 ligands on hepatocyte cell death and proliferation. In the current study, we sought to determine if CXCR2 ligand concentration is responsible for the divergent effects of these mediators on liver regeneration after ischemia/reperfusion injury and partial hepatectomy. METHODS Murine models of partial ischemia/reperfusion injury and hepatectomy were used to study the effect of CXCR2 ligands on liver regeneration. RESULTS We found that hepatic expression of the CXCR2 ligands, macrophage inflammatory protein-2 (MIP-2) and keratinocyte-derived chemokine (KC), was significantly increased after both I/R injury and partial hepatectomy. However, expression of these ligands after I/R injury was 30-100-fold greater than after hepatectomy. Interestingly, the same pattern of expression was found in ischemic versus non-ischemic liver lobes following I/R injury with expression significantly greater in the ischemic liver lobes. In both systems, lower ligand expression was associated with increased hepatocyte proliferation and liver regeneration in a CXCR2-dependent fashion. To confirm that these effects were related to ligand concentration, we administered exogenous MIP-2 and KC to mice undergoing partial hepatectomy. Mice received a "high" dose that replicated serum levels found after I/R injury and a "low" dose that was similar to that found after hepatectomy. Mice receiving the "high" dose had reduced levels of hepatocyte proliferation and regeneration whereas the "low" dose promoted hepatocyte proliferation and regeneration. CONCLUSIONS Together, these data demonstrate that concentrations of CXC chemokines regulate the hepatic proliferative response and subsequent liver regeneration.
Collapse
Affiliation(s)
- Gregory C. Wilson
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Satoshi Kuboki
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Christopher M. Freeman
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Hiroyuki Nojima
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Rebecca M. Schuster
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Michael J. Edwards
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Alex B. Lentsch
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
159
|
Liu X, Dai LI, Zhou R. Association between preeclampsia and the CXC chemokine family (Review). Exp Ther Med 2015; 9:1572-1576. [PMID: 26136860 DOI: 10.3892/etm.2015.2337] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 02/18/2015] [Indexed: 12/30/2022] Open
Abstract
Preeclampsia is a major cause of maternal and perinatal mortality and morbidity, characterized by gestational hypertension, proteinuria, systemic endothelial cell activation and an exaggerated inflammatory response. The precise cause of preeclampsia is not currently known; however, it is widely accepted that the pathogenesis of preeclampsia involves inadequate trophoblast invasion, leading to generalized endothelial dysfunction and an exaggerated inflammatory response. Chemokines are a superfamily of structurally similar proteins that mediate cell recruitment, angiogenesis, immunity and stem cell trafficking. CXC chemokines are a family of cytokines, unique in their ability to behave in a disparate manner in the regulation of angiogenesis. The CXC chemokine family further divides into two subfamilies; CXC ELR+, which promotes angiogenesis, and CXC ELR-, which inhibits angiogenesis. Furthermore, CXC chemokines are involved in the pathogenesis of various conditions, including malignant tumors, wound repair, chronic inflammation, atherosclerosis and potentially preeclampsia.
Collapse
Affiliation(s)
- Xijing Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - L I Dai
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
160
|
Song Y, Baba T, Li YY, Furukawa K, Tanabe Y, Matsugo S, Sasaki S, Mukaida N. Gemcitabine-induced CXCL8 expression counteracts its actions by inducing tumor neovascularization. Biochem Biophys Res Commun 2015; 458:341-346. [PMID: 25646691 DOI: 10.1016/j.bbrc.2015.01.112] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 01/23/2015] [Indexed: 11/16/2022]
Abstract
Patients with pancreatic ductal adenocarcinoma (PDAC) are frequently complicated with metastatic disease or locally advanced tumors, and consequently need chemotherapy. Gemcitabine is commonly used for PDAC treatment, but with limited efficacy. The capacity of gemcitabine to generate reactive oxygen species (ROS) in human pancreatic cancer cells, prompted us to examine its effects on the expression of pro-inflammatory cytokines and chemokines. We observed that gemcitabine enhanced selectively the expression of CXCL8 in human pancreatic cancer cells through ROS generation and NF-κB activation. In vitro blocking of CXCL8 failed to modulate gemcitabine-mediated inhibition of cell proliferation in human pancreatic cancer cells. Gemcitabine also enhanced CXCL8 expression in pancreatic cancer cells in xenografted tumor tissues. Moreover, anti-CXCL8 antibody treatment in vivo attenuated tumor formation as well as intra-tumoral vascularity in nude mice, which were transplanted with Miapaca-2 cells and treated with gemcitabine. Thus, gemcitabine-induced CXCL8 may counteract the drug through inducing neovascularization.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/adverse effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Deoxycytidine/administration & dosage
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Dose-Response Relationship, Drug
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Interleukin-8/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- Neovascularization, Pathologic/chemically induced
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/metabolism
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Reactive Oxygen Species/metabolism
- Treatment Outcome
- Up-Regulation/drug effects
- Gemcitabine
Collapse
Affiliation(s)
- Yao Song
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Ying-Yi Li
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kaoru Furukawa
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan; School of Natural System Bioengineering Course, College of Science and Engineering, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yamato Tanabe
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan; School of Natural System Bioengineering Course, College of Science and Engineering, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Seiichi Matsugo
- School of Natural System Bioengineering Course, College of Science and Engineering, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Soichiro Sasaki
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan.
| |
Collapse
|
161
|
Desurmont T, Skrypek N, Duhamel A, Jonckheere N, Millet G, Leteurtre E, Gosset P, Duchene B, Ramdane N, Hebbar M, Van Seuningen I, Pruvot FR, Huet G, Truant S. Overexpression of chemokine receptor CXCR2 and ligand CXCL7 in liver metastases from colon cancer is correlated to shorter disease-free and overall survival. Cancer Sci 2015; 106:262-9. [PMID: 25580640 PMCID: PMC4376434 DOI: 10.1111/cas.12603] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 12/18/2014] [Accepted: 12/24/2014] [Indexed: 12/21/2022] Open
Abstract
Our aim was to analyze the potential role of chemokine receptors CXCR2 and CXCR4 signalling pathways in liver metastatic colorectal cancer (CRC) relapse. CXCR2, CXCR4, and their chemokine ligands were evaluated in liver metastases of colorectal cancer in order to study their correlation with overall and disease-free survival of patients having received, or not received, a neoadjuvant chemotherapy regimen. Quantitative RT-PCR and CXCR2 immunohistochemical staining were carried out using CRC liver metastasis samples. Expression levels of CXCR2, CXCR4, and their ligands were statistically analyzed according to treatment with neoadjuvant chemotherapy and patients’ outcome. CXCR2 and CXCL7 overexpression are correlated to shorter overall and disease-free survival. By multivariate analysis, CXCR2 and CXCL7 expressions are independent factors of overall and disease-free survival. Neoadjuvant chemotherapy increases significantly the expression of CXCR2: treated group 1.89 (0.02–50.92) vs 0.55 (0.07–3.22), P = 0.016. CXCL7 was overexpressed close to significance, 0.40 (0.00–7.85) vs 0.15 (0.01–7.88), P = 0.12. We show the involvement of CXCL7/CXCR2 signalling pathways as a predictive factor of poor outcome in metastatic CRC. 5-Fluorouracil-based chemotherapy regimens increase the expression of these genes in liver metastasis, providing one explanation for aggressiveness of relapsed drug-resistant tumors. Selective blockage of CXCR2/CXCL7 signalling pathways could provide new potential therapeutic opportunities.
Collapse
Affiliation(s)
- Thibault Desurmont
- Inserm, U837, Team-5 (Mucins, Epithelial Differentiation, and Carcinogenesis), Jean-Pierre Aubert Research Centre, Université Lille, France; Department of Digestive and Visceral Surgery, GHICL, Saint-Vincent de Paul Hospital, Lille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
162
|
Erin N, Nizam E, Tanrıöver G, Köksoy S. Autocrine control of MIP-2 secretion from metastatic breast cancer cells is mediated by CXCR2: a mechanism for possible resistance to CXCR2 antagonists. Breast Cancer Res Treat 2015; 150:57-69. [DOI: 10.1007/s10549-015-3297-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 02/03/2015] [Indexed: 02/06/2023]
|
163
|
Sharma B, Nannuru KC, Varney ML, Singh RK. Host Cxcr2-dependent regulation of mammary tumor growth and metastasis. Clin Exp Metastasis 2015; 32:65-72. [PMID: 25511644 PMCID: PMC4821540 DOI: 10.1007/s10585-014-9691-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/05/2014] [Indexed: 12/29/2022]
Abstract
Host-derived angiogenic and inflammatory tumor supportive microenvironment regulates progression and metastasis, but the molecular mechanism(s) underlying host-tumor interactions remains unclear. Tumor expression of CXCR2 and its ligands have been shown to regulate angiogenesis, invasion, tumor growth, and metastasis. In this report, we hypothesized that host-derived Cxcr2-dependent signaling plays an important role in breast cancer growth and metastasis. Two mammary tumor cell lines Cl66 and 4T1 cells were orthotopically implanted into the mammary fat pad of wild-type and Cxcr2(-/-) female BALB/c mice. Tumor growth and spontaneous lung metastasis were monitored. Immunohistochemical analyses of the tumor tissues were performed to analyze proliferation, angiogenesis, apoptosis and immune cell infiltration. Our results demonstrated that knock-down of host Cxcr2 decreases tumor growth and metastasis by reducing angiogenesis, proliferation and enhancing apoptosis. Host Cxcr2 plays an important role in governing the pro-inflammatory response in mammary tumors as evaluated by decreased Gr1(+) tumor-associated granulocytes, F4/80(+) tumor associated macrophages, and CD11b(+)Gr1(+) myeloid derived suppressor cells in Cxcr2(-/-) mice as compared to control wild-type mice. Together, these results demonstrate that host Cxcr2-dependent signaling regulates mammary tumor growth and metastasis by promoting angiogenesis and pro-inflammatory responses.
Collapse
Affiliation(s)
- Bhawna Sharma
- Department of Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | | | - Michelle L. Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, USA
| |
Collapse
|
164
|
Hahn CS, Scott DW, Xu X, Roda MA, Payne GA, Wells JM, Viera L, Winstead CJ, Bratcher P, Sparidans RW, Redegeld FA, Jackson PL, Folkerts G, Blalock JE, Patel RP, Gaggar A. The matrikine N-α-PGP couples extracellular matrix fragmentation to endothelial permeability. SCIENCE ADVANCES 2015; 1:e1500175. [PMID: 26229981 PMCID: PMC4517288 DOI: 10.1126/sciadv.1500175] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 02/25/2015] [Indexed: 06/04/2023]
Abstract
The compartmentalization and transport of proteins and solutes across the endothelium is a critical biologic function altered during inflammation and disease, leading to pathology in multiple disorders. The impact of tissue damage and subsequent extracellular matrix (ECM) fragmentation in regulating this process is unknown. We demonstrate that the collagen-derived matrikine acetylated proline-glycine-proline (N-α-PGP) serves as a critical regulator of endothelial permeability. N-α-PGP activates human endothelial cells via CXC-chemokine receptor 2 (CXCR2), triggering monolayer permeability through a discrete intracellular signaling pathway. In vivo, N-α-PGP induces local vascular leak after subcutaneous administration and pulmonary vascular permeability after systemic administration. Furthermore, neutralization of N-α-PGP attenuates lipopolysaccharide-induced lung leak. Finally, we demonstrate that plasma from patients with acute respiratory distress syndrome (ARDS) induces VE-cadherin phosphorylation in human endothelial cells, and this activation is attenuated by N-α-PGP blockade with a concomitant improvement in endothelial monolayer impedance. These results identify N-α-PGP as a novel ECM-derived matrikine regulating paracellular permeability during inflammatory disease and demonstrate the potential to target this ligand in various disorders characterized by excessive matrix turnover and vascular leak such as ARDS.
Collapse
Affiliation(s)
- Cornelia S. Hahn
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David W. Scott
- Department of Cell Biology and Physiology, and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Xin Xu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mojtaba Abdul Roda
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, Netherlands
| | - Gregory A. Payne
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - J. Michael Wells
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL 35233, USA
| | - Liliana Viera
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Colleen J. Winstead
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Preston Bratcher
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rolf W. Sparidans
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, Netherlands
| | - Frank A. Redegeld
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, Netherlands
| | - Patricia L. Jackson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gert Folkerts
- Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3508 TB Utrecht, Netherlands
| | - J. Edwin Blalock
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rakesh P. Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Amit Gaggar
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Program in Protease and Matrix Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Medical Service at Birmingham VA Medical Center, Birmingham, AL 35233, USA
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
165
|
Vries MHM, Wagenaar A, Verbruggen SEL, Molin DGM, Post MJ. CXCL1 promotes arteriogenesis through enhanced monocyte recruitment into the peri-collateral space. Angiogenesis 2014; 18:163-71. [DOI: 10.1007/s10456-014-9454-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 12/03/2014] [Indexed: 12/24/2022]
|
166
|
Inconformity of CXCL3 plasma level and placenta expression in preeclampsia and its effect on trophoblast viability and invasion. PLoS One 2014; 9:e114408. [PMID: 25485631 PMCID: PMC4259324 DOI: 10.1371/journal.pone.0114408] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 11/07/2014] [Indexed: 12/31/2022] Open
Abstract
As a member of the chemokine family, CXCL3 was previously known to participate in many pathophysiological events. However, whether CXCL3 stimulates trophoblast invasion as a key process of preeclampsia pathogenesis remains largely unknown. Therefore, the aim of this study was to investigate this hypothesis and determine the effect of CXCL3 on the first trimester trophoblast. Seventy gravidas were included in this study. ELISA was used to detect CXCL3 plasma levels on preeclampsia and normal pregnant groups. CXCL3 protein and mRNA levels were detected via Western blot and real-time quantitative PCR analysis after immunolocalized in human placenta. Moreover, the CXCL3 function in HTR-8/Svneo was analyzed via WST-1 assay, flow cytometry and invasion test. The plasma CXCL3 level in preeclampsia was significantly higher than that in normal pregnancy. CXCL3 expression was observed in the cytoplasm of placental trophoblasts and vascular endothelium in all groups without significant difference between maternal and fetal sides. In addition, placenta CXCL3 expression in severe preeclampsia was significantly lower than those in normal and mild PE groups. Moreover, exogenous CXCL3 can promote the proliferation and invasion of HTR-8/Svneo; however, its effect on apoptosis remains unclear. In summary, a significant abnormality of plasma CXCL3 level and placental CXCL3 expression was discovered in severe preeclampsia; CXCL3 had a function in trophoblast invasion, which indicated its participation in shallow implantation. Therefore CXCL3 might be involved in severe preeclampsia pathogenesis.
Collapse
|
167
|
Ryan BM, Robles AI, McClary AC, Haznadar M, Bowman ED, Pine SR, Brown D, Khan M, Shiraishi K, Kohno T, Okayama H, Modali R, Yokota J, Harris CC. Identification of a functional SNP in the 3'UTR of CXCR2 that is associated with reduced risk of lung cancer. Cancer Res 2014; 75:566-75. [PMID: 25480945 DOI: 10.1158/0008-5472.can-14-2101] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Global changes in gene expression accompany the development of cancer. Thus, inherited variants in miRNA-binding sites are likely candidates for conferring inherited susceptibility. Using an in silico approach, we compiled a comprehensive list of SNPs predicted to modulate miRNA binding in genes from several key lung cancer pathways. We then investigated whether these SNPs were associated with lung cancer risk in two independent populations. In general, SNPs in miRNA-binding sites are rare. However, some allelic variation was observed. We found that rs1126579 in CXCR2 was associated with a reduced risk of lung cancer in both European American [ORTT vs. CC 0.56 (0.37-0.88); P = 0.008] and Japanese [ORTT vs. CC 0.62 (0.38-1.00); P = 0.049] populations. Furthermore, we found that the SNP disrupted a novel binding site for miR-516a-3p, led to a moderate increase in CXCR2 mRNA and protein expression, and increased MAPK signaling. Moreover, analysis of rs1126579 with serum levels of IL8, its endogenous ligand, supported an interaction whereby rs1126579-T and high serum IL8 conferred synergistic protection from lung cancer. Our findings demonstrate a function for a 3'UTR SNP in modulating CXCR2 expression, signaling, and susceptibility to lung cancer.
Collapse
Affiliation(s)
- Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Ana I Robles
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Andrew C McClary
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland. Department of Pathology, Stanford University Hospital and Clinics, Stanford, California
| | - Majda Haznadar
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Elise D Bowman
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sharon R Pine
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey
| | - Derek Brown
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Mohammed Khan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Kouya Shiraishi
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Hirokazu Okayama
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland. Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | | | - Jun Yokota
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Curtis C Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
168
|
Raman D, Sai J, Hawkins O, Richmond A. Adaptor protein2 (AP2) orchestrates CXCR2-mediated cell migration. Traffic 2014; 15:451-69. [PMID: 24450359 DOI: 10.1111/tra.12154] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/16/2014] [Accepted: 01/22/2014] [Indexed: 12/14/2022]
Abstract
The chemokine receptor CXCR2 is vital for inflammation, wound healing, angiogenesis, cancer progression and metastasis. Adaptor protein 2 (AP2), a clathrin binding heterotetrameric protein comprised of α, β2, μ2 and σ2 subunits, facilitates clathrin-mediated endocytosis. Mutation of the LLKIL motif in the CXCR2 carboxyl-terminal domain (CTD) results in loss of AP2 binding to the receptor and loss of ligand-mediated receptor internalization and chemotaxis. AP2 knockdown also results in diminished ligand-mediated CXCR2 internalization, polarization and chemotaxis. Using knockdown/rescue approaches with AP2-μ2 mutants, the binding domains were characterized in reference to CXCR2 internalization and chemotaxis. When in an open conformation, μ2 Patch 1 and Patch 2 domains bind tightly to membrane PIP2 phospholipids. When AP2-μ2, is replaced with μ2 mutated in Patch 1 and/or Patch 2 domains, ligand-mediated receptor binding and internalization are not lost. However, chemotaxis requires AP2-μ2 Patch 1, but not Patch 2. AP2-σ2 has been demonstrated to bind dileucine motifs to facilitate internalization. Expression of AP2-σ2 V88D and V98S dominant negative mutants resulted in loss of CXCR2 mediated chemotaxis. Thus, AP2 binding to both membrane phosphatidylinositol phospholipids and dileucine motifs is crucial for directional migration or chemotaxis. Moreover, AP2-mediated receptor internalization can be dissociated from AP2-mediated chemotaxis.
Collapse
Affiliation(s)
- Dayanidhi Raman
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37212, USA; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | | | | | | |
Collapse
|
169
|
Sisto M, Lisi S. New Insights Into ADAMs Regulation of the GRO-α/CXCR2 System: Focus on Sjögren's Syndrome. Int Rev Immunol 2014; 34:486-99. [DOI: 10.3109/08830185.2014.975892] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
170
|
Leclair HM, Dubois SM, Azzi S, Dwyer J, Bidère N, Gavard J. Control of CXCR2 activity through its ubiquitination on K327 residue. BMC Cell Biol 2014; 15:38. [PMID: 25339290 PMCID: PMC4209453 DOI: 10.1186/s12860-014-0038-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 10/09/2014] [Indexed: 11/23/2022] Open
Abstract
Background The interleukin-8 chemokine (IL-8) G-protein coupled receptor CXCR2 governs pro-inflammatory and pro-angiogenic responses in leukocytes and endothelial cells. At a molecular standpoint, CXCR2 is widely reported to operate through calcium flux, phosphoinoisitide 3 kinase (PI3K) and mitogen-activated protein kinase (MAPK). While CXCR2 trafficking is suspected to be intertwined with its signaling, the exact mechanism is not fully elucidated. Results Here, we identified the lysine 327 within the CXCR2 C-terminal tail as a key residue for ubiquitination, internalization, and signaling. First, the substitution to an arginine of K327 mutation was associated with a reduction in CXCR2 poly-ubiquitination. While WT CXCR2 was rapidly internalized following IL-8 administration, K327R mutant remained at the plasma membrane. Finally, K327R mutant failed to promote the recruitment of β-arrestin2, as estimated by imagery and bioluminescence resonance transfer. As a consequence, the activation of intracellular signaling, including both early events such as ERK phosphorylation and the increase in calcium flux, and the latter activation of the AP1 and NF-κB transcription factors, was blunted. Conclusions Overall, our results demonstrate that CXCR2 ubiquitination on K327 residue modulates agonist-activated CXCR2 cell sorting and intracellular signaling. Thus, the inhibition of K327 ubiquitination might emerge as an effective mean to curb exacerbated CXCR2 signaling in several pathological conditions, such as inflammatory diseases and cancer.
Collapse
Affiliation(s)
| | | | | | | | | | - Julie Gavard
- CNRS, UMR8104, 22 rue Mechain, Paris, 75014, France.
| |
Collapse
|
171
|
Shepardson KM, Jhingran A, Caffrey A, Obar JJ, Suratt BT, Berwin BL, Hohl TM, Cramer RA. Myeloid derived hypoxia inducible factor 1-alpha is required for protection against pulmonary Aspergillus fumigatus infection. PLoS Pathog 2014; 10:e1004378. [PMID: 25255025 PMCID: PMC4177996 DOI: 10.1371/journal.ppat.1004378] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/01/2014] [Indexed: 12/14/2022] Open
Abstract
Hypoxia inducible factor 1α (HIF1α) is the mammalian transcriptional factor that controls metabolism, survival, and innate immunity in response to inflammation and low oxygen. Previous work established that generation of hypoxic microenvironments occurs within the lung during infection with the human fungal pathogen Aspergillus fumigatus. Here we demonstrate that A. fumigatus stabilizes HIF1α protein early after pulmonary challenge that is inhibited by treatment of mice with the steroid triamcinolone. Utilizing myeloid deficient HIF1α mice, we observed that HIF1α is required for survival and fungal clearance early following pulmonary challenge with A. fumigatus. Unlike previously reported research with bacterial pathogens, HIF1α deficient neutrophils and macrophages were surprisingly not defective in fungal conidial killing. The increase in susceptibility of the myeloid deficient HIF1α mice to A. fumigatus was in part due to decreased early production of the chemokine CXCL1 (KC) and increased neutrophil apoptosis at the site of infection, resulting in decreased neutrophil numbers in the lung. Addition of recombinant CXCL1 restored neutrophil survival and numbers, murine survival, and fungal clearance. These results suggest that there are unique HIF1α mediated mechanisms employed by the host for protection and defense against fungal pathogen growth and invasion in the lung. Additionally, this work supports the strategy of exploring HIF1α as a therapeutic target in specific immunosuppressed populations with fungal infections.
Collapse
Affiliation(s)
- Kelly M. Shepardson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Anupam Jhingran
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Alayna Caffrey
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, United States of America
| | - Joshua J. Obar
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana, United States of America
| | - Benjamin T. Suratt
- Department of Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Brent L. Berwin
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Tobias M. Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
172
|
Sun Q, Sun F, Wang B, Liu S, Niu W, Liu E, Peng C, Wang J, Gao H, Liang B, Niu Z, Zou X, Niu J. Interleukin-8 promotes cell migration through integrin αvβ6 upregulation in colorectal cancer. Cancer Lett 2014; 354:245-53. [PMID: 25150782 DOI: 10.1016/j.canlet.2014.08.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 08/01/2014] [Accepted: 08/14/2014] [Indexed: 01/05/2023]
Abstract
Colorectal cancer (CRC), which is notorious for high morbidity and mortality around the world, shows a predilection for metastasis to liver. Interleukin-8 (IL-8), a chemokine with a defining CXC amino acid motif, has been reported to promote CRC cell migration and is associated with poor prognosis of CRC. However, the underlying molecular mechanism of IL-8-mediated migration remains obscure. In this study, we first demonstrated the cross talk between IL-8 and integrin αvβ6. We analyzed 139 human CRC samples, and found that the immunohistochemical expression of αvβ6 was significantly correlated with expression of IL-8. Furthermore, IL-8 increased the migration through integrin αvβ6 in human CRC cells, and both CXCR1 and CXCR2 were primarily involved during the process. IL-8 upregulated αvβ6 expression in a dose-dependent manner through activation of ERK and Ets-1 signaling pathway. Taken together, our results indicated that IL-8 enhances the migration of CRC cells by increasing αvβ6 integrin expression through the ERK/Ets-1 pathway. Targeting integrin αvβ6 in IL-8 expressing tumors might be a potential therapeutic strategy for CRC patients.
Collapse
Affiliation(s)
- Qi Sun
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Fengkai Sun
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Ben Wang
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Song Liu
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Binzhou Medical College, Binzhou 256603, Shandong, China
| | - Weibo Niu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Enyu Liu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Cheng Peng
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Jiayong Wang
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Huijie Gao
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Benjia Liang
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Zhengchuan Niu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Xueqing Zou
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China; Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Public Health, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China
| | - Jun Niu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.
| |
Collapse
|
173
|
Ha H, Neamati N. Pyrimidine-based compounds modulate CXCR2-mediated signaling and receptor turnover. Mol Pharm 2014; 11:2431-41. [PMID: 24896229 DOI: 10.1021/mp500180e] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Chemokine receptor CXCR2 is expressed on various immune cells and is essential for neutrophil recruitment and angiogenesis at sites of acute and chronic inflammation caused by tissue injury or infection. Because of its role in inflammation, it has been implicated in a number of immune-mediated inflammatory diseases such as psoriasis, arthritis, COPD, cystic fibrosis, asthma, and various types of cancer. CXCR2 and its ligands are up-regulated in cancer cells as well as the tumor microenvironment, promoting tumor growth, angiogenesis, and invasiveness. Although pharmaceutical companies have pursued the development of CXCR2-specific small-molecule inhibitors as anti-inflammatory agents within the last decades, there are currently no clinically approved CXCR2 inhibitors. Using a high-throughput, cell-based assay specific for CXCR2, we screened an in-house library of structurally diverse compounds and identified a class of pyrimidine-based compounds that alter CXCR2-mediated second messenger signaling. Our lead compound, CX797, inhibited IL8-mediated cAMP signaling and receptor degradation while specifically up-regulating IL8-mediated β-arrestin-2 recruitment. CX797 also inhibited IL8-mediated cell migration. Mechanistic comparison of CX797 and a previously reported CXCR2 inhibitor, SB265610, show these two classes of compounds have a distinct mechanism of action on CXCR2.
Collapse
Affiliation(s)
- Helen Ha
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California , 1985 Zonal Avenue, Los Angeles, California 90033, United States
| | | |
Collapse
|
174
|
Rowland KJ, Diaz-Miron J, Guo J, Erwin CR, Mei J, Worthen GS, Warner BW. CXCL5 is required for angiogenesis, but not structural adaptation after small bowel resection. J Pediatr Surg 2014; 49:976-80; discussion 980. [PMID: 24888846 PMCID: PMC4044536 DOI: 10.1016/j.jpedsurg.2014.01.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 01/27/2014] [Indexed: 01/14/2023]
Abstract
PURPOSE Intestinal adaptation is the compensatory response to massive small bowel resection (SBR) and characterized by lengthening of villi and deepening of crypts, resulting in increased mucosal surface area. Previous studies have demonstrated increased villus capillary blood vessel density after SBR, suggesting a role for angiogenesis in the development of resection-induced adaptation. Since we have previously shown enhanced expression of the proangiogenic chemokine CXCL5 after SBR, the purpose of this study was to determine the effect of disrupted CXCL5 expression on intestinal adaptation. METHODS CXCL5 knockout (KO) and C57BL/6 wild type (WT) mice were subjected to either a 50% proximal SBR or sham operation. Ileal tissue was harvested on postoperative day 7. To assess for adaptation, villus height and crypt depth were measured. Submucosal capillary density was measured by CD31 immunohistochemistry. RESULTS Both CXCL5-KO and WT mice demonstrated normal structural features of adaptation. Submucosal capillary density increased in the WT but not in the KO mice following SBR. CONCLUSION CXCL5 is required for increased intestinal angiogenesis during resection-induced adaptation. Since adaptive villus growth occurs despite impaired CXCL5 expression and enhanced angiogenesis, this suggests that the growth of new blood vessels is not needed for resection-induced mucosal surface area expansion following massive SBR.
Collapse
Affiliation(s)
- Kathryn J. Rowland
- Division of Pediatric Surgery, St Louis Children’s Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jose Diaz-Miron
- Division of Pediatric Surgery, St Louis Children’s Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Jun Guo
- Division of Pediatric Surgery, St Louis Children’s Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Christopher R. Erwin
- Division of Pediatric Surgery, St Louis Children’s Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Junjie Mei
- Division of Neonatology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - G. Scott Worthen
- Division of Neonatology, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brad W. Warner
- Division of Pediatric Surgery, St Louis Children’s Hospital, Department of Surgery, Washington University School of Medicine, St Louis, MO 63110, USA
| |
Collapse
|
175
|
Immunohistochemical analysis of IL-6, IL-8/CXCR2 axis, Tyr p-STAT-3, and SOCS-3 in lymph nodes from patients with chronic lymphocytic leukemia: correlation between microvascular characteristics and prognostic significance. BIOMED RESEARCH INTERNATIONAL 2014; 2014:251479. [PMID: 24883303 PMCID: PMC4026921 DOI: 10.1155/2014/251479] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 04/04/2014] [Indexed: 02/07/2023]
Abstract
A number of studies have looked into the pathophysiological role of angiogenesis in CLL, but the results have often been inconsistent. We aimed to gain direct insight into the angiogenic process in lymph nodes involved by CLL, focusing on proangiogenic cytokines and microvessel morphometry. The tissue levels of VEGF, Th-2 cytokines IL-6 and IL-8, IL-8 receptor CXCR2, and tyrosine p-STAT-3/SOCS-3 axis modulating cytokine expression were evaluated immunohistochemically in 62 CLL/SLL cases. Microvascular characteristics were evaluated by image analysis. Results were analyzed with regard to clinicopathological characteristics. Proliferation centers (PCs) were less well vascularised compared to non-PC areas. IL-8 and CXCR2 expression was distinctly uncommon as opposed to IL-6, VEGF and SOCS-3, which were detected in the vast majority of cases. The latter two molecule expressions were more pronounced in the PCs in ∼40% of the cases. p-STAT-3 immunoreactivity was recorded in 66.67% of the cases with a predilection for PCs. Microvessel morphometry was unrelated to proangiogenic cytokines, p-STAT-3, SOCS-3, or survival. Microvascular caliber and VEGF expression were higher in Binet stage A, whereasIL-6 expression was higher in stage C. VEGF and p-STAT-3 exerted a favorable effect on progression, which remained significant in multivariate analysis, thereby constituting potential outcome predictors in CLL patients.
Collapse
|
176
|
Paige M, Wang K, Burdick M, Park S, Cha J, Jeffery E, Sherman N, Shim YM. Role of leukotriene A4 hydrolase aminopeptidase in the pathogenesis of emphysema. THE JOURNAL OF IMMUNOLOGY 2014; 192:5059-68. [PMID: 24771855 DOI: 10.4049/jimmunol.1400452] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The leukotriene A4 hydrolase (LTA4H) is a bifunctional enzyme with epoxy hydrolase and aminopeptidase activities. We hypothesize that the LTA4H aminopeptidase activity alleviates neutrophilic inflammation, which contributes to cigarette smoke (CS)-induced emphysema by clearing proline-glycine-proline (PGP), a triamino acid chemokine known to induce chemotaxis of neutrophils. To investigate the biological contributions made by the LTA4H aminopeptidase activity in CS-induced emphysema, we exposed wild-type mice to CS over 5 mo while treating them with a vehicle or a pharmaceutical agent (4MDM) that selectively augments the LTA4H aminopeptidase without affecting the bioproduction of leukotriene B4. Emphysematous phenotypes were assessed by premortem lung physiology with a small animal ventilator and by postmortem histologic morphometry. CS exposure acidified the airspaces and induced localization of the LTA4H protein into the nuclei of the epithelial cells. This resulted in accumulation of PGP in the airspaces by suppressing the LTA4H aminopeptidase activity. When the LTA4H aminopeptidase activity was selectively augmented by 4MDM, the levels of PGP in the bronchoalveolar lavage fluid and infiltration of neutrophils into the lungs were significantly reduced without affecting the levels of leukotriene B4. This protected murine lungs from CS-induced emphysematous alveolar remodeling. In conclusion, CS exposure promotes the development of CS-induced emphysema by suppressing the enzymatic activities of the LTA4H aminopeptidase in lung tissues and accumulating PGP and neutrophils in the airspaces. However, restoring the leukotriene A4 aminopeptidase activity with a pharmaceutical agent protected murine lungs from developing CS-induced emphysema.
Collapse
Affiliation(s)
- Mikell Paige
- Department of Chemistry and Biochemistry, George Mason University, Manassas, VA 22030
| | - Kan Wang
- Center for Drug Discovery, Georgetown University Medical Center, Washington, DC 20057
| | - Marie Burdick
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908; and
| | - Sunhye Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908; and
| | - Josiah Cha
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908; and
| | - Erin Jeffery
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | - Nicholas Sherman
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA 22908
| | - Y Michael Shim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, VA 22908; and
| |
Collapse
|
177
|
Stock AT, Smith JM, Carbone FR. Type I IFN suppresses Cxcr2 driven neutrophil recruitment into the sensory ganglia during viral infection. ACTA ACUST UNITED AC 2014; 211:751-9. [PMID: 24752295 PMCID: PMC4010892 DOI: 10.1084/jem.20132183] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infection induces the expression of inflammatory chemokines that recruit immune cells to the site of inflammation. Whereas tissues such as the intestine and skin express unique chemokines during homeostasis, whether different tissues express distinct chemokine profiles during inflammation remains unclear. With this in mind, we performed a comprehensive screen of the chemokines expressed by two tissues (skin and sensory ganglia) infected with a common viral pathogen (herpes simplex virus type 1). After infection, the skin and ganglia showed marked differences in their expression of the family of Cxcr2 chemokine ligands. Specifically, Cxcl1/2/3, which in turn controlled neutrophil recruitment, was up-regulated in the skin but absent from the ganglia. Within the ganglia, Cxcl2 expression and subsequent neutrophil recruitment was inhibited by type I interferon (IFN). Using a combination of bone marrow chimeras and intracellular chemokine staining, we show that type I IFN acted by directly suppressing Cxcl2 expression by monocytes, abrogating their ability to recruit neutrophils to the ganglia. Overall, our findings describe a novel role for IFN in the direct, and selective, inhibition of Cxcr2 chemokine ligands, which results in the inhibition of neutrophil recruitment to neuronal tissue.
Collapse
Affiliation(s)
- Angus T Stock
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Parkville, Victoria 3010, Australia
| | | | | |
Collapse
|
178
|
Jiang Y, Wang S, Holcomb J, Trescott L, Guan X, Hou Y, Brunzelle J, Sirinupong N, Li C, Yang Z. Crystallographic analysis of NHERF1-PLCβ3 interaction provides structural basis for CXCR2 signaling in pancreatic cancer. Biochem Biophys Res Commun 2014; 446:638-643. [PMID: 24642259 DOI: 10.1016/j.bbrc.2014.03.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 03/09/2014] [Indexed: 01/25/2023]
Abstract
The formation of CXCR2-NHERF1-PLCβ3 macromolecular complex in pancreatic cancer cells regulates CXCR2 signaling activity and plays an important role in tumor proliferation and invasion. We previously have shown that disruption of the NHERF1-mediated CXCR2-PLCβ3 interaction abolishes the CXCR2 signaling cascade and inhibits pancreatic tumor growth in vitro and in vivo. Here we report the crystal structure of the NHERF1 PDZ1 domain in complex with the C-terminal PLCβ3 sequence. The structure reveals that the PDZ1-PLCβ3 binding specificity is achieved by numerous hydrogen bonds and hydrophobic contacts with the last four PLCβ3 residues contributing to specific interactions. We also show that PLCβ3 can bind both NHERF1 PDZ1 and PDZ2 in pancreatic cancer cells, consistent with the observation that the peptide binding pockets of these PDZ domains are highly structurally conserved. This study provides an understanding of the structural basis for the PDZ-mediated NHERF1-PLCβ3 interaction that could prove valuable in selective drug design against CXCR2-related cancers.
Collapse
Affiliation(s)
- Yuanyuan Jiang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Shuo Wang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Joshua Holcomb
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Laura Trescott
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaoqing Guan
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuning Hou
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Nualpun Sirinupong
- Nutraceuticals and Functional Food Research and Development Center, Prince of Songkla University, Hat-Yai, Songkhla, Thailand
| | - Chunying Li
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
179
|
Stofas A, Levidou G, Piperi C, Adamopoulos C, Dalagiorgou G, Bamias A, Karadimou A, Lainakis GA, Papadoukakis S, Stravodimos K, Dimopoulos MA, Patsouris E, Gakiopoulou H, Korkolopoulou P. The role of CXC-chemokine receptor CXCR2 and suppressor of cytokine signaling-3 (SOCS-3) in renal cell carcinoma. BMC Cancer 2014; 14:149. [PMID: 24593195 PMCID: PMC4015755 DOI: 10.1186/1471-2407-14-149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 02/03/2014] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Chemokine receptor signaling pathways are implicated in the pathobiology of renal cell carcinoma (RCC). However, the clinical relevance of CXCR2 receptor, mediating the effects of all angiogenic chemokines, remains unclear. SOCS (suppressor of cytokine signaling)-3 is a negative regulator of cytokine-driven responses, contributing to interferon-α resistance commonly used to treat advanced RCC with limited information regarding its expression in RCC. METHODS In this study, CXCR2 and SOCS-3 were immunohistochemically investigated in 118 RCC cases in relation to interleukin (IL)-6 and (IL)-8, their downstream transducer phosphorylated (p-)STAT-3, and VEGF expression, being further correlated with microvascular characteristics, clinicopathological features and survival. In 30 cases relationships with hypoxia-inducible factors, i.e. HIF-1a, p53 and NF-κΒ (p65/RelA) were also examined. Validation of immunohistochemistry and further investigation of downstream transducers, p-JAK2 and p-c-Jun were evaluated by Western immunoblotting in 5 cases. RESULTS Both CXCR2 and IL-8 were expressed by the neoplastic cells their levels being interrelated. CXCR2 strongly correlated with the levels of HIF-1a, p53 and p65/RelA in the neoplastic cells. Although SOCS-3 was simultaneously expressed with p-STAT-3, its levels tended to show an inverse relationship with p-JAK-2 and p-c-Jun in Western blots and were positively correlated with HIF-1a, p53 and p65/p65/RelA expression. Neither CXCR2 nor SOCS-3 correlated with the extent of microvascular network. IL-8 and CXCR2 expression was associated with high grade, advanced stage and the presence/number of metastases but only CXCR2 adversely affected survival in univariate analysis. Elevated SOCS-3 expression was associated with progression, the presence/number of metastasis and shortened survival in both univariate and multivariate analysis. CONCLUSIONS Our findings implicate SOCS-3 overexpression in RCC metastasis and biologic aggressiveness advocating its therapeutic targeting. IL-8/CXCR2 signaling also contributes to the metastatic phenotype of RCC cells but appears of lesser prognostic utility. Both CXCR2 and SOCS-3 appear to be related to transcription factors induced under hypoxia.
Collapse
Affiliation(s)
- Anastasios Stofas
- First Department of Pathology, Laiko Hospital, University of Athens, Medical School, 75 Mikras Asias street, 11527 Athens, Greece
| | - Georgia Levidou
- First Department of Pathology, Laiko Hospital, University of Athens, Medical School, 75 Mikras Asias street, 11527 Athens, Greece
| | - Christina Piperi
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece
| | - Christos Adamopoulos
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece
| | - Georgia Dalagiorgou
- Department of Biological Chemistry, University of Athens, Medical School, 11527 Athens, Greece
| | - Aristotelis Bamias
- Department of Clinical Therapeutics, Alexandra General Hospital, University of Athens, Medical School, 11528 Athens, Greece
| | - Alexandra Karadimou
- Department of Clinical Therapeutics, Alexandra General Hospital, University of Athens, Medical School, 11528 Athens, Greece
| | - George A Lainakis
- Department of Clinical Therapeutics, Alexandra General Hospital, University of Athens, Medical School, 11528 Athens, Greece
| | - Stefanos Papadoukakis
- Department of Urology, Laiko Hospital, University of Athens, Medical School, 11517 Athens, Greece
| | - Konstantinos Stravodimos
- Department of Urology, Laiko Hospital, University of Athens, Medical School, 11517 Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra General Hospital, University of Athens, Medical School, 11528 Athens, Greece
| | - Efstratios Patsouris
- First Department of Pathology, Laiko Hospital, University of Athens, Medical School, 75 Mikras Asias street, 11527 Athens, Greece
| | - Hariklia Gakiopoulou
- First Department of Pathology, Laiko Hospital, University of Athens, Medical School, 75 Mikras Asias street, 11527 Athens, Greece
| | - Penelope Korkolopoulou
- First Department of Pathology, Laiko Hospital, University of Athens, Medical School, 75 Mikras Asias street, 11527 Athens, Greece
| |
Collapse
|
180
|
Ozbek C, Kestelli M, Bozok S, Ilhan G, Yurekli I, Ozpak B, Akyuz M, Bademci M. Surgical stimulation of angiogenesis. Asian Cardiovasc Thorac Ann 2014; 22:36-9. [PMID: 24585641 DOI: 10.1177/0218492312468285] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The results of surgical approaches are unsatisfactory in patients with a distal arterial bed that is ineligible for revascularization. In this retrospective study, we investigated outcomes in patients who underwent surgical interventions that are reported to induce angiogenesis. METHOD 6 patients diagnosed with thromboangiitis obliterans were included in this study. Of these 6 patients, 2 underwent femoropopliteal bypass surgery using reversed great saphenous vein as a conduit. The other 4 underwent ascending venous arterialization (bypass from the popliteal artery to the great saphenous vein at the level of medial malleolus, using reversed great saphenous vein). RESULTS During the early postoperative period, all of the reversed saphenous vein grafts were occluded. The mean postoperative ankle-brachial index increased from 0.33 to 0.83. During the postoperative period, intermittent claudication disappeared in all patients. Angiograms taken during the postoperative period showed evidence of neovascularization when compared to those taken during preoperative evaluation. CONCLUSION Wound healing is an inflammatory process that simultaneously activates angiogenesis. We propose that the improved ankle-brachial index values and neovascularization shown in our patients were associated with this inflammatory process.
Collapse
Affiliation(s)
- Cengiz Ozbek
- Department of Cardiovascular Surgery, Izmir Ataturk Training and Research Hospital, Izmir, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Roy I, Evans DB, Dwinell MB. Chemokines and chemokine receptors: update on utility and challenges for the clinician. Surgery 2014; 155:961-73. [PMID: 24856117 DOI: 10.1016/j.surg.2014.02.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Accepted: 02/05/2014] [Indexed: 11/26/2022]
Affiliation(s)
- Ishan Roy
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI
| | - Douglas B Evans
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI
| | - Michael B Dwinell
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
182
|
Caccuri F, Rueckert C, Giagulli C, Schulze K, Basta D, Zicari S, Marsico S, Cervi E, Fiorentini S, Slevin M, Guzman CA, Caruso A. HIV-1 matrix protein p17 promotes lymphangiogenesis and activates the endothelin-1/endothelin B receptor axis. Arterioscler Thromb Vasc Biol 2014; 34:846-56. [PMID: 24482377 DOI: 10.1161/atvbaha.113.302478] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE AIDS-related lymphomas are high grade and aggressively metastatic with poor prognosis. Lymphangiogenesis is essential in supporting proliferation and survival of lymphoma, as well as tumor dissemination. Data suggest that aberrant lymphangiogenesis relies on action of HIV-1 proteins rather than on a direct effect of the virus itself. HIV-1 matrix protein p17 was found to accumulate and persist in lymph nodes of patients even under highly active antiretroviral therapy. Because p17 was recently found to exert a potent proangiogenic activity by interacting with chemokine (C-X-C motif) receptors 1 and 2, we tested the prolymphangiogenic activity of the viral protein. APPROACH AND RESULTS Human primary lymph node-derived lymphatic endothelial cells were used to perform capillary-like structure formation, wound healing, spheroids, and Western blot assays after stimulation with or without p17. Here, we show that p17 promotes lymphangiogenesis by binding to chemokine (C-X-C motif) receptor-1 and chemokine (C-X-C motif) receptor-2 expressed on lymph node-derived lymphatic endothelial cells and activating the Akt/extracellular signal-regulated kinase signaling pathway. In particular, it was found to induce capillary-like structure formation, sprout formation from spheroids, and increase lymph node-derived lymphatic endothelial cells motility. The p17 lymphangiogenic activity was, in part, sustained by activation of the endothelin-1/endothelin receptor B axis. A Matrigel plug assay showed that p17 was able to promote the outgrowth of lymphatic vessels in vivo, demonstrating that p17 directly regulates lymphatic vessel formation. CONCLUSIONS Our results suggest that p17 may generate a prolymphangiogenic microenvironment and plays a role in predisposing the lymph node to lymphoma growth and metastasis. This finding offers new opportunities to identify treatment strategies in combating AIDS-related lymphomas.
Collapse
Affiliation(s)
- Francesca Caccuri
- From the Microbiology Section, Department of Molecular and Translational Medicine (F.C., C.G., D.B., S.Z., S.F., A.C.) and Section of Vascular Surgery, Department of Medical and Surgical Sciences (E.C.), University of Brescia, Brescia, Italy; Animal Models and Retroviral Vaccine Section, National Cancer Institute, National Institutes of Health, Bethesda, MD (F.C.); Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Braunschweig, Germany (C.R., K.S., C.A.G.); Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy (S.M.); and School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom (M.S.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Rentoft M, Coates PJ, Loljung L, Wilms T, Laurell G, Nylander K. Expression of CXCL10 is associated with response to radiotherapy and overall survival in squamous cell carcinoma of the tongue. Tumour Biol 2014; 35:4191-8. [PMID: 24395654 PMCID: PMC4009142 DOI: 10.1007/s13277-013-1549-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 12/12/2013] [Indexed: 12/23/2022] Open
Abstract
Five-year survival for patients with oral cancer has been disappointingly stable during the last decades, creating a demand for new biomarkers and treatment targets. Lately, much focus has been set on immunomodulation as a possible treatment or an adjuvant increasing sensitivity to conventional treatments. The objective of this study was to evaluate the prognostic importance of response to radiotherapy in tongue carcinoma patients as well as the expression of the CXC-chemokines in correlation to radiation response in the same group of tumours. Thirty-eight patients with tongue carcinoma that had received radiotherapy followed by surgery were included. The prognostic impact of pathological response to radiotherapy, N-status, T-stage, age and gender was evaluated using Cox's regression models, Kaplan-Meier survival curves and chi-square test. The expression of 23 CXC-chemokine ligands and their receptors were evaluated in all patients using microarray and qPCR and correlated with response to treatment using logistic regression. Pathological response to radiotherapy was independently associated to overall survival with a 2-year survival probability of 81% for patients showing a complete pathological response, while patients with a non-complete response only had a probability of 42% to survive for 2 years (p = 0.016). The expression of one CXC-chemokine, CXCL10, was significantly associated with response to radiotherapy and the group of patients with the highest CXCL10 expression responded, especially poorly (p = 0.01). CXCL10 is a potential marker for response to radiotherapy and overall survival in patients with squamous cell carcinoma of the tongue.
Collapse
Affiliation(s)
- Matilda Rentoft
- Department of Chemistry, Umeå University, 901 85, Umeå, Sweden,
| | | | | | | | | | | |
Collapse
|
184
|
Abstract
Chemokines are low-molecular-weight, secreted proteins that act as leukocyte-specific chemoattractants. The chemokine family has more than 40 members. Based on the position of two conserved cysteines in the N-terminal domain, chemokines can be divided into the CXC, C, CC, and CX3C subfamilies. The interaction of chemokines with their receptors mediates signaling pathways that play critical roles in cell migration, differentiation, and proliferation. The receptors for chemokines are G protein-coupled receptors (GPCRs), and thus far, seven CXC receptors have been cloned and are designated CXCR1-7. Constitutively active GPCRs are present in several human immune-mediated diseases and in tumors, and they have provided valuable information in understanding the molecular mechanism of GPCR activation. Several constitutively active CXC chemokine receptors include the V6.40A and V6.40N mutants of CXCR1; the D3.49V variant of CXCR2; the N3.35A, N3.35S, and T2.56P mutants of CXCR3; the N3.35 mutation of CXCR4; and the naturally occurring KSHV-GPCR. Here, we review the regulation of CXC chemokine receptor signaling, with a particular focus on the constitutive activation of these receptors and the implications in physiological conditions and in pathogenesis. Understanding the mechanisms behind the constitutive activation of CXC chemokine receptors may aid in pharmaceutical design and the screening of inverse agonists and allosteric modulators for the treatment of autoimmune diseases and cancers.
Collapse
Affiliation(s)
- Xinbing Han
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
185
|
McLoughlin P, Keane MP. Physiological and pathological angiogenesis in the adult pulmonary circulation. Compr Physiol 2013; 1:1473-508. [PMID: 23733650 DOI: 10.1002/cphy.c100034] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Angiogenesis occurs during growth and physiological adaptation in many systemic organs, for example, exercise-induced skeletal and cardiac muscle hypertrophy, ovulation, and tissue repair. Disordered angiogenesis contributes to chronic inflammatory disease processes and to tumor growth and metastasis. Although it was previously thought that the adult pulmonary circulation was incapable of supporting new vessel growth, over that past 10 years new data have shown that angiogenesis within this circulation occurs both during physiological adaptive processes and as part of the pathogenic mechanisms of lung diseases. Here we review the expression of vascular growth factors in the adult lung, their essential role in pulmonary vascular homeostasis and the changes in their expression that occur in response to physiological challenges and in disease. We consider the evidence for adaptive neovascularization in the pulmonary circulation in response to alveolar hypoxia and during lung growth following pneumonectomy in the adult lung. In addition, we review the role of disordered angiogenesis in specific lung diseases including idiopathic pulmonary fibrosis, acute adult distress syndrome and both primary and metastatic tumors of the lung. Finally, we examine recent experimental data showing that therapeutic enhancement of pulmonary angiogenesis has the potential to treat lung diseases characterized by vessel loss.
Collapse
Affiliation(s)
- Paul McLoughlin
- University College Dublin, School of Medicine and Medical Sciences, Conway Institute, and St. Vincent's University Hospital, Dublin, Ireland.
| | | |
Collapse
|
186
|
Abstract
Skeletal muscle is the largest organ in the body. Skeletal muscles are primarily characterized by their mechanical activity required for posture, movement, and breathing, which depends on muscle fiber contractions. However, skeletal muscle is not just a component in our locomotor system. Recent evidence has identified skeletal muscle as a secretory organ. We have suggested that cytokines and other peptides that are produced, expressed, and released by muscle fibers and exert either autocrine, paracrine, or endocrine effects should be classified as "myokines." The muscle secretome consists of several hundred secreted peptides. This finding provides a conceptual basis and a whole new paradigm for understanding how muscles communicate with other organs such as adipose tissue, liver, pancreas, bones, and brain. In addition, several myokines exert their effects within the muscle itself. Many proteins produced by skeletal muscle are dependent upon contraction. Therefore, it is likely that myokines may contribute in the mediation of the health benefits of exercise.
Collapse
Affiliation(s)
- Bente K Pedersen
- The Centre of Inflammation and Metabolism at Department of Infectious Diseases, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
187
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 PMCID: PMC3880466 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 693] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Lu G, Wu Y, Jiang Y, Wang S, Hou Y, Guan X, Brunzelle J, Sirinupong N, Sheng S, Li C, Yang Z. Structural insights into neutrophilic migration revealed by the crystal structure of the chemokine receptor CXCR2 in complex with the first PDZ domain of NHERF1. PLoS One 2013; 8:e76219. [PMID: 24098448 PMCID: PMC3788737 DOI: 10.1371/journal.pone.0076219] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/21/2013] [Indexed: 11/17/2022] Open
Abstract
Neutrophil plays an essential role in host defense against infection, but uncontrolled neutrophilic infiltration can cause inflammation and severe epithelial damage. We recently showed that CXCR2 formed a signaling complex with NHERF1 and PLC-2, and that the formation of this complex was required for intracellular calcium mobilization and neutrophilic transepithelial migration. To uncover the structural basis of the complex formation, we report here the crystal structure of the NHERF1 PDZ1 domain in complex with the C-terminal sequence of CXCR2 at 1.16 Å resolution. The structure reveals that the CXCR2 peptide binds to PDZ1 in an extended conformation with the last four residues making specific side chain interactions. Remarkably, comparison of the structure to previously studied PDZ1 domains has allowed the identification of PDZ1 ligand-specific interactions and the mechanisms that govern PDZ1 target selection diversities. In addition, we show that CXCR2 can bind both NHERF1 PDZ1 and PDZ2 in pulldown experiments, consistent with the observation that the peptide binding pockets of these two PDZ domains are highly structurally conserved. The results of this study therefore provide structural basis for the CXCR2-mediated neutrophilic migration and could have important clinical applications in the prevention and treatment of numerous neutrophil-dependent inflammatory disorders.
Collapse
Affiliation(s)
- Guorong Lu
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Yanning Wu
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Yuanyuan Jiang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Shuo Wang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Yuning Hou
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Xiaoqing Guan
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Joseph Brunzelle
- Advance Photon Source, Argonne National Laboratory, Argonne, Illinois, United States of America
| | - Nualpun Sirinupong
- Nutraceuticals and Functional Food Research and Development Center, Prince of Songkla University, Hat-Yai, Songkhla, Thailand
| | - Shijie Sheng
- Department of Pathology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Chunying Li
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Zhe Yang
- Department of Biochemistry and Molecular Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| |
Collapse
|
189
|
Choi YH, Burdick MD, Strieter BA, Mehrad B, Strieter RM. CXCR4, but not CXCR7, discriminates metastatic behavior in non-small cell lung cancer cells. Mol Cancer Res 2013; 12:38-47. [PMID: 24025971 DOI: 10.1158/1541-7786.mcr-12-0334] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
UNLABELLED Chemokines have been implicated as key contributors of non-small cell lung cancer (NSCLC) metastasis. However, the role of CXCR7, a recently discovered receptor for CXCL12 ligand, in the pathogenesis of NSCLC is unknown. To define the relative contribution of chemokine receptors to migration and metastasis, we generated human lung A549 and H157 cell lines with stable knockdown of CXCR4, CXCR7, or both. Cancer cells exhibited chemotaxis to CXCL12 that was enhanced under hypoxic conditions, associated with a parallel induction of CXCR4, but not CXCR7. Interestingly, neither knockdown cell line differed in the rate of proliferation, apoptosis, or cell adherence; however, in both cell lines, CXCL12-induced migration was abolished when CXCR4 signaling was abrogated. In contrast, inhibition of CXCR7 signaling did not alter cellular migration to CXCL12. In an in vivo heterotropic xenograft model using A549 cells, expression of CXCR4, but not CXCR7, on cancer cells was necessary for the development of metastases. In addition, cancer cells knocked down for CXCR4 (or both CXCR4 and CXCR7) produced larger and more vascular tumors as compared with wild-type or CXCR7 knockdown tumors, an effect that was attributable to cancer cell-derived CXCR4 out competing endothelial cells for available CXCL12 in the tumor microenvironment. These results indicate that CXCR4, not CXCR7, expression engages CXCL12 to mediate NSCLC metastatic behavior. IMPLICATIONS Targeting CXCR4-mediated migration and metastasis may be a viable therapeutic option in NSCLC.
Collapse
Affiliation(s)
- Young H Choi
- Division of Pulmonary and Critical Care, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908.
| | | | | | | | | |
Collapse
|
190
|
Martins-Green M, Petreaca M, Wang L. Chemokines and Their Receptors Are Key Players in the Orchestra That Regulates Wound Healing. Adv Wound Care (New Rochelle) 2013; 2:327-347. [PMID: 24587971 DOI: 10.1089/wound.2012.0380] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Indexed: 12/13/2022] Open
Abstract
SIGNIFICANCE Normal wound healing progresses through a series of overlapping phases, all of which are coordinated and regulated by a variety of molecules, including chemokines. Because these regulatory molecules play roles during the various stages of healing, alterations in their presence or function can lead to dysregulation of the wound-healing process, potentially leading to the development of chronic, nonhealing wounds. RECENT ADVANCES A discovery that chemokines participate in a variety of disease conditions has propelled the study of these proteins to a level that potentially could lead to new avenues to treat disease. Their small size, exposed termini, and the fact that their only modifications are two disulfide bonds make them excellent targets for manipulation. In addition, because they bind to G-protein-coupled receptors (GPCRs), they are highly amenable to pharmacological modulation. CRITICAL ISSUES Chemokines are multifunctional, and in many situations, their functions are highly dependent on the microenvironment. Moreover, each specific chemokine can bind to several GPCRs to stimulate the function, and both can function as monomers, homodimers, heterodimers, and even oligomers. Activation of one receptor by any single chemokine can lead to desensitization of other chemokine receptors, or even other GPCRs in the same cell, with implications for how these proteins or their receptors could be used to manipulate function. FUTURE DIRECTIONS Investment in better understanding of the functions of chemokines and their receptors in a local context can reveal new ways for therapeutic intervention. Understanding how different chemokines can activate the same receptor and vice versa could identify new possibilities for drug development based on their heterotypic interactions.
Collapse
Affiliation(s)
- Manuela Martins-Green
- Department of Cell Biology and Neuroscience, University of California, Riverside, California
| | - Melissa Petreaca
- Department of Cell Biology and Neuroscience, University of California, Riverside, California
| | - Lei Wang
- Department of Cell Biology and Neuroscience, University of California, Riverside, California
| |
Collapse
|
191
|
Kim DW, Kim WH, Kim MH, Kim CG. Novel Tc-99m labeled ELR-containing 6-mer peptides for tumor imaging in epidermoid carcinoma xenografts model: a pilot study. Ann Nucl Med 2013; 27:892-7. [PMID: 23990397 DOI: 10.1007/s12149-013-0766-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Accepted: 08/15/2013] [Indexed: 12/21/2022]
Abstract
OBJECTIVE ELR-containing peptides targeting CXCR2 could be the excellent candidate for targeting ligand of molecular tumor imaging. In this study, we had developed two ELR-containing 6-mer peptides and evaluated the diagnostic performance of Tc-99m labeled 6-mer peptides as a molecular imaging agent in murine models bearing KB epidermoid carcinoma. METHODS Peptides were synthesized using Fmoc solid phase peptide synthesis. Radiolabeling efficiency with Tc-99m was evaluated using instant thin-layer chromatography. In KB epidermoid cancer-bearing mice, gamma images had acquired and tumor-to-muscle uptake ratio was calculated. Competition and biodistribution studies had performed. RESULTS Two 6-mer peptides, ELR-ECG and ECG-ELR were successfully synthesized. After radiolabeling procedures with Tc-99m, the complex Tc-99m ELR-ECG and Tc-99m ECG-ELR were prepared in high yield. In the gamma camera imaging of murine model, Tc-99m ELR-ECG was substantially accumulated in the subcutaneously engrafted tumor and tumor uptake had been suppressed by the free ELR co-injection. However, Tc-99m ECG-ELR was minimally accumulated in the tumor. CONCLUSIONS Two ELR-containing 6-mer peptides, ELR-ECG and ECG-ELR, were developed as a molecular imaging agent to target CXCR2 of epidermoid carcinoma. Tc-99m ELR-ECG had showed significant uptake in tumor and it was good candidate for a tumor imaging.
Collapse
Affiliation(s)
- Dae-Weung Kim
- Department of Nuclear Medicine, Wonkwang University School of Medicine, 344-2 Shinyong-Dong, Iksan, Jeollabuk-do, 570-711, Republic of Korea
| | | | | | | |
Collapse
|
192
|
Rotondi M, Coperchini F, Chiovato L. CXCL8 in thyroid disease: from basic notions to potential applications in clinical practice. Cytokine Growth Factor Rev 2013; 24:539-46. [PMID: 24011840 DOI: 10.1016/j.cytogfr.2013.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 08/05/2013] [Indexed: 01/14/2023]
Abstract
CXCL8 was the first chemokine shown to be secreted by thyrocytes. Experimental data suggest that CXCL8 plays a role in thyroid homeostasis but its role in thyroid diseases remains poorly investigated. Clinical studies measuring the serum levels of CXCL8 in patients with autoimmune-thyroid-diseases reported conflicting results. Solid evidences support a role of CXCL8 as a tumor-promoting agent in several human cancers. Studies in thyroid cancer are still in their initial stage, but promising. Several evidences indicate that thyroid cancer may share with other human malignancies some of the effects of CXCL8 and highlight the possibility of using CXCL8 as a marker of aggressiveness. Basic and clinical evidences in favor or against a role for CXCL8 in thyroid diseases are discussed.
Collapse
Affiliation(s)
- Mario Rotondi
- Unit of Internal Medicine and Endocrinology, Fondazione Salvatore Maugeri I.R.C.C.S., Laboratory for Endocrine Disruptors and Chair of Endocrinology University of Pavia, Italy.
| | | | | |
Collapse
|
193
|
Al-Alwan LA, Chang Y, Mogas A, Halayko AJ, Baglole CJ, Martin JG, Rousseau S, Eidelman DH, Hamid Q. Differential roles of CXCL2 and CXCL3 and their receptors in regulating normal and asthmatic airway smooth muscle cell migration. THE JOURNAL OF IMMUNOLOGY 2013; 191:2731-41. [PMID: 23904157 DOI: 10.4049/jimmunol.1203421] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Structural cell migration plays a central role in the pathophysiology of several diseases, including asthma. Previously, we established that IL-17-induced (CXCL1, CXCL2, and CXCL3) production promoted airway smooth muscle cell (ASMC) migration, and consequently we sought to investigate the molecular mechanism of CXC-induced ASMC migration. Recombinant human CXCL1, CXCL2, and CXCL3 were used to assess migration of human primary ASMCs from normal and asthmatic subjects using a modified Boyden chamber. Neutralizing Abs or small interfering RNA (siRNA) knockdown and pharmacological inhibitors of PI3K, ERK1/2, and p38 MAPK pathways were used to investigate the receptors and the signaling pathways involved in CXC-induced ASMC migration, respectively. We established the ability of CXCL2 and CXCL3, but not CXCL1, to induce ASMC migration at the tested concentrations using normal ASMCs. We found CXCL2-induced ASMC migration to be dependent on p38 MAPK and CXCR2, whereas CXCL3-induced migration was dependent on p38 and ERK1/2 MAPK pathways via CXCR1 and CXCR2. While investigating the effect of CXCL2 and CXCL3 on asthmatic ASMC migration, we found that they induced greater migration of asthmatic ASMCs compared with normal ones. Interestingly, unlike normal ASMCs, CXCL2- and CXCL3-induced asthmatic ASMC migration was mainly mediated by the PI3K pathway through CXCR1. In conclusion, our results establish a new role of CXCR1 in ASMC migration and demonstrate the diverse mechanisms by which CXCL2 and CXCL3 mediate normal and asthmatic ASMC migration, suggesting that they may play a role in the pathogenesis of airway remodeling in asthma.
Collapse
Affiliation(s)
- Laila A Al-Alwan
- Meakins-Christie Laboratories and Respiratory Division, Department of Medicine, McGill University, Montreal, Quebec H2X 2P2, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Borsig L, Wolf MJ, Roblek M, Lorentzen A, Heikenwalder M. Inflammatory chemokines and metastasis--tracing the accessory. Oncogene 2013; 33:3217-24. [PMID: 23851506 DOI: 10.1038/onc.2013.272] [Citation(s) in RCA: 176] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 05/17/2013] [Accepted: 05/20/2013] [Indexed: 12/20/2022]
Abstract
The tumor microenvironment consists of stromal cells and leukocytes that contribute to cancer progression. Cross-talk between tumor cells and their microenvironment is facilitated by a variety of soluble factors, including growth factors and cytokines such as chemokines. Due to a wide expression of chemokine receptors on cells in the tumor microenvironment, including tumor cells, chemokines affect various processes such as leukocyte recruitment, angiogenesis, tumor cell survival, tumor cell adhesion, proliferation, vascular permeability, immune suppression, invasion and metastasis. Inflammatory chemokines are instrumental players in cancer-related inflammation and significantly contribute to numerous steps during metastasis. Recruitment of myeloid-derived cells to metastatic sites is mainly mediated by the inflammatory chemokines CCL2 and CCL5. Tumor cell homing and extravasation from the circulation to distant organs are also regulated by inflammatory chemokines. Recent experimental evidence demonstrated that besides leukocyte recruitment, tumor cell-derived CCL2 directly activated endothelial cells and together with monocytes facilitated tumor cell extravasation, in a CCL2- and CCL5-dependent manner. Furthermore, CX3CL1 expression in the bone facilitated metastasis of CX3CR1 expressing tumor cells to this site. Current findings in preclinical models strongly suggest that inflammatory chemokines have an important role during metastasis and targeting of the chemokine axis might have a therapeutic potential.
Collapse
Affiliation(s)
- L Borsig
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - M J Wolf
- Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - M Roblek
- Institute of Physiology, University of Zurich and Zurich Center for Integrative Human Physiology, Zurich, Switzerland
| | - A Lorentzen
- Institute of Virology, Technische Universität München/Helmholtz Zentrum Munich, Munich, Germany
| | - M Heikenwalder
- Institute of Virology, Technische Universität München/Helmholtz Zentrum Munich, Munich, Germany
| |
Collapse
|
195
|
Shaharuddin B, Ahmad S, Meeson A, Ali S. Concise review: immunological properties of ocular surface and importance of limbal stem cells for transplantation. Stem Cells Transl Med 2013; 2:614-24. [PMID: 23817133 DOI: 10.5966/sctm.2012-0143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cornea transplantation has been considered to be different from other solid organ transplantation because of the assumed immune-privileged state of the anterior chamber of the eye. Three major lines of thought regarding the molecular mechanisms of immune privilege in the eye are as follows: (a) anatomical, cellular, and molecular barriers in the eye; (b) anterior chamber-associated immune deviation; and (c) immunosuppressive microenvironment in the eye. However, cornea transplants suffer allograft rejection when breached by vascularization. In recent developments, cellular corneal transplantation from cultivated limbal epithelial cells has shown impressive advances as a future therapy. The limbal stem cell niche contains stem cells that promote proliferation and migration and have immunosuppressive mechanisms to protect them from immunological reactions. Limbal stem cells are also noted to display an enhanced expression of genes for the antiapoptotic proteins, a property that is imperative for the survival of transplanted tissues. Further investigation of the molecular mechanisms regulating the immune regulation of limbal stem cells is relevant in the clinical setting to promote the survival of whole corneal and limbal stem cell transplantation.
Collapse
Affiliation(s)
- Bakiah Shaharuddin
- Institute of Genetic Medicine, Newcastle University, Newcastle-Upon-Tyne, United Kingdom
| | | | | | | |
Collapse
|
196
|
Raghuwanshi SK, Smith N, Rivers EJ, Thomas AJ, Sutton N, Hu Y, Mukhopadhyay S, Chen XL, Leung T, Richardson RM. G protein-coupled receptor kinase 6 deficiency promotes angiogenesis, tumor progression, and metastasis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:5329-5336. [PMID: 23589623 PMCID: PMC3646980 DOI: 10.4049/jimmunol.1202058] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
G protein-coupled receptor kinases (GRKs) phosphorylate the activated form of G protein-coupled receptors leading to receptor desensitization and downregulation. We have recently shown that the chemokine receptor, CXCR2, couples to GRK6 to regulate cellular responses including chemotaxis, angiogenesis, and wound healing. In this study, we investigate the role of GRK6 in tumorigenesis using murine models of human lung cancer. Mice deficient in GRK6 (GRK6(-/-)) exhibited a significant increase in Lewis lung cancer growth and metastasis relative to control littermates (GRK6(+/+)). GRK6 deletion had no effect on the expression of proangiogenic chemokine or vascular endothelial growth factor, but upregulated matrix metalloproteinase (MMP)-2 and MMP-9 release, tumor-infiltrating PMNs, and microvessel density. Because β-arrestin-2-deficient (βarr2(-/-)) mice exhibited increased Lewis lung cancer growth and metastasis similar to that of GRK6(-/-), we developed a double GRK6(-/-)/βarr2(-/-) mouse model. Surprisingly, GRK6(-/-)/βarr2(-/-) mice exhibited faster tumor growth relative to GRK6(-/-) or βarr2(-/-) mice. Treatment of the mice with anti-CXCR2 Ab inhibited tumor growth in both GRK6(-/-) and GRK6(-/-)/βarr2(-/-) animals. Altogether, the results indicate that CXCR2 couples to GRK6 to regulate angiogenesis, tumor progression, and metastasis. Deletion of GRK6 increases the activity of the host CXCR2, resulting in greater PMN infiltration and MMP release in the tumor microenvironment, thereby promoting angiogenesis and metastasis. Because GRK6(-/-)/βarr2(-/-) showed greater tumor growth relative to GRK6(-/-) or βarr2(-/-) mice, the data further suggest that CXCR2 couples to different mechanisms to mediate tumor progression and metastasis.
Collapse
Affiliation(s)
- Sandeep K. Raghuwanshi
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - Nikia Smith
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - Elizabeth, J. Rivers
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - Ariel J. Thomas
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - Natalie Sutton
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - Yuhui Hu
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | | | - Xiaoxin L. Chen
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| | - TinChung Leung
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
- North Carolina Research Campus, Nutrition Research Center, 500 Laureate Way, Kannapolis, NC 28081
| | - Ricardo M. Richardson
- Julius L. Chambers Biomedical/Biotechnology Research Institute and Department of Biology, North Carolina Central University, Durham, NC 27707
| |
Collapse
|
197
|
Williamson LL, Bilbo SD. Chemokines and the hippocampus: a new perspective on hippocampal plasticity and vulnerability. Brain Behav Immun 2013; 30:186-94. [PMID: 23376170 DOI: 10.1016/j.bbi.2013.01.077] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 01/07/2013] [Accepted: 01/15/2013] [Indexed: 12/13/2022] Open
Abstract
The hippocampus is critical for several aspects of learning and memory and is unique among other cortical regions in structure, function and the potential for plasticity. This remarkable region recapitulates development throughout the lifespan with enduring neurogenesis and well-characterized plasticity. The structure and traits of the hippocampus that distinguish it from other brain regions, however, may be the same reasons that this important brain region is particularly vulnerable to insult and injury. The immune system within the brain responds to insult and injury, and the hippocampus and the immune system are extensively interconnected. Immune signaling molecules, cytokines and chemokines (chemotactic cytokines), are well known for their functions during insult or injury. They are also increasingly implicated in normal hippocampal neurogenesis (e.g., CXCR4 on newborn neurons), cellular plasticity (e.g., interleukin-6 in LTP maintenance), and learning and memory (e.g., interleukin-1β in fear conditioning). We provide evidence from the small but growing literature that neuroimmune interactions and immune signaling molecules, especially chemokines, may be a primary underlying mechanism for the coexistence of plasticity and vulnerability within the hippocampus. We also highlight the evidence that the hippocampus exhibits a remarkable resilience in response to diverse environmental events (e.g., enrichment, exercise), which all may converge onto common neuroimmune mechanisms.
Collapse
Affiliation(s)
- Lauren L Williamson
- Duke University, Genome Science Research Building 2, 210 Research Dr., Box 91050, Durham, NC 27710, United States.
| | | |
Collapse
|
198
|
Uzunoglu FG, Kolbe J, Wikman H, Güngör C, Bohn BA, Nentwich MF, Reeh M, König AM, Bockhorn M, Kutup A, Mann O, Izbicki JR, Vashist YK. VEGFR-2, CXCR-2 and PAR-1 germline polymorphisms as predictors of survival in pancreatic carcinoma. Ann Oncol 2013; 24:1282-90. [PMID: 23293110 DOI: 10.1093/annonc/mds634] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
BACKGROUND Hypoxic environment of pancreatic cancer (PC) implicates high vascular in-growth, which may be influenced by angiogenesis-related germline polymorphisms. Our purpose was to evaluate polymorphisms of vascular endothelial growth factor receptor 2 (VEGFR-2), CXC chemokine receptor 2 (CXCR-2), proteinase-activated receptor 1 (PAR-1) and endostatin (ES) as prognostic markers for disease-free (DFS) and overall survival (OS) in PC. PATIENTS AND METHODS Genotyping of 173 patients, surgically treated for PC between 2004 and 2011, was carried out by TaqMan(®) genotyping assays or polymerase chain reaction. Chi-square test, Kaplan-Meier estimator and Cox regression hazard model were used to assess the prognostic value of selected polymorphisms. RESULTS VEGFR-2 -906 T/T and PAR-1 -506 Del/Del genotypes predicted longer DFS (P = 0.003, P = 0.014) and OS (VEGFR-2 -906, P = 0.011). CXCR-2 +1208 T/T genotype was a negative predictor for DFS (P < 0.0001). Combined analysis for DFS and OS indicated that patients with the fewest number of favorable genotypes simultaneously present (VEGFR-2 -906 T/T, CXCR-2 +1208 C/T or C/C and PAR-1 -506 Del/Del) were at the highest risk for recurrence or death (P < 0.0001). CONCLUSION VEGFR-2 -906 C>T, CXCR-2 +1208 C>T and PAR-1 -506 Ins/Del polymorphisms are potential predictors for survival in PC.
Collapse
Affiliation(s)
- F G Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center of Hamburg-Eppendorf, Martinistr. 52, 20246Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
199
|
Oral H, Kanzler I, Tuchscheerer N, Curaj A, Simsekyilmaz S, Sönmez TT, Radu E, Postea O, Weber C, Schuh A, Liehn EA. CXC chemokine KC fails to induce neutrophil infiltration and neoangiogenesis in a mouse model of myocardial infarction. J Mol Cell Cardiol 2013; 60:1-7. [PMID: 23598282 DOI: 10.1016/j.yjmcc.2013.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Revised: 03/17/2013] [Accepted: 04/08/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chemokines and neutrophils, known as important players in the inflammatory cascade, also contribute to heart tissue recovery and scar formation after myocardial infarction (MI). The objective of this study was to determine the importance of ELR-containing CXC chemokine KC in neutrophil infiltration and neoangiogenesis, in a mouse model of chronic MI. METHODS AND RESULTS MI was induced in mice divided in four groups: control (untreated), anti-KC "later" (anti-KC antibody injections started 4 days after MI and then delivered every 72 hours for 3 weeks, to inhibit angiogenesis), anti-KC "earlier" (anti-KC antibody injections 1 day before and 1 day after MI, to block neutrophil infiltration), anti-KC (anti-KC antibody injections 1 day before and 1 day after MI, and then every 72 hours for 3 weeks). The efficiency of the anti-KC treatment was determined by the measurement of KC serum concentration and immunofluorescence staining, in each of the four groups. Surprisingly, we did not find any difference in neutrophil infiltration in the infarcted area between untreated and treated animals. Moreover, the heart function, infarct size, and neoangiogenesis were not different between the four groups. As expected, a comparable anti-CXCR2 treatment of mice before and after MI was able to significantly reduce neutrophil infiltration into the infarcted area and angiogenesis, but also to reduce the infarction size after long or "later" treatment. CONCLUSIONS The major finding of our study is that KC, a potent neutrophil chemoattractant and an established angiogenic factor, failed to interfere in the post-infarction inflammatory response, in wound healing and scar formation after MI. Therefore, these aspects need to be carefully taken into account when devising therapeutic strategies for myocardial infarction and ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Hasan Oral
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Sharma B, Nawandar DM, Nannuru KC, Varney ML, Singh RK. Targeting CXCR2 enhances chemotherapeutic response, inhibits mammary tumor growth, angiogenesis, and lung metastasis. Mol Cancer Ther 2013; 12:799-808. [PMID: 23468530 DOI: 10.1158/1535-7163.mct-12-0529] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Breast cancer is one of the leading causes of cancer deaths among females. Many challenges exist in the current management of advanced stage breast cancer as there are fewer recognized therapeutic strategies, often because of therapy resistance. How breast cancer cells evade chemotherapy and the underlying mechanism remains unclear. We and others have observed that malignant cells that survive initial chemo- and radiation therapy express higher levels of CXCR2 ligands, which may provide a survival benefit leading to therapy resistance. In this report, we test the hypothesis that CXCR2-dependent signaling in malignant cells may be critical for chemotherapy resistance and targeting this signaling axis may enhance the antitumor and antimetastatic activity of chemotherapeutic drugs and limit their toxicity. We used Cl66-wt, 4T1-wt, Cl66sh-CXCR2, and 4T1sh-CXCR2 cells expressing differential levels of the CXCR2 receptor to evaluate the role of targeting CXCR2 on chemotherapeutic responses. Knockdown of CXCR2 enhances paclitaxel and doxorubicin-mediated toxicity at suboptimal doses. Moreover, we observed an increase in the expression of CXCL1, a CXCR2 ligand in paclitaxel and doxorubicin-treated mammary tumor cells, which were inhibited following CXCR2 knockdown. Knockdown of CXCR2 enhanced antitumor activity of paclitaxel in an in vivo mammary tumor model. We observed significant inhibition of spontaneous lung metastases in animals bearing CXCR2 knockdown tumors and treated with paclitaxel as compared with the control group. Our data suggest the novel role of CXCR2 and its ligands in maintaining chemotherapy resistance and provide evidence that targeting CXCR2 signaling in an adjuvant setting will help circumvent chemotherapy resistance.
Collapse
Affiliation(s)
- Bhawna Sharma
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | |
Collapse
|