201
|
Hertzer KM, Donald GW, Hines OJ. CXCR2: a target for pancreatic cancer treatment? Expert Opin Ther Targets 2013; 17:667-80. [PMID: 23425074 DOI: 10.1517/14728222.2013.772137] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Pancreatic cancer, a leading cause of cancer deaths worldwide, is very aggressive and has minimally effective treatment options. For those who have no surgical options, medical treatments are limited. The chemokine receptor CXCR2 has become the subject of much interest recently because of multiple studies indicating its involvement in cancer and inflammatory conditions. Research now indicates that CXCR2 and its ligands are intimately involved in tumor regulation and growth and that inhibition of its function shows promising results in multiple cancer types, including pancreatic cancer. AREAS COVERED In this study, the authors review basic molecular and structural details of CXCR2, as well as the known functions of CXCR2 and several of its ligands in inflammation and cancer biology with specific attention to pancreatic cancer. Then the future possibilities and questions remaining for pharmacological intervention against CXCR2 in pancreatic cancer are explored. EXPERT OPINION Many current inhibitory strategies already exist for targeting CXCR2 in vitro as well as in vivo. Clinically speaking, CXCR2 is an exciting potential target for pancreatic cancer; however, CXCR2 is functionally important for multiple processes and therapeutic options would benefit from further work toward understanding of these roles as well as structural and target specificity.
Collapse
Affiliation(s)
- Kathleen M Hertzer
- Hirshberg Translational Pancreatic Cancer Research Laboratory, David Geffen School of Medicine at UCLA, Department of Surgery , 675 Charles E Young Drive, MRL 2535, Los Angeles, CA 90095 , USA
| | | | | |
Collapse
|
202
|
An important role of blood and lymphatic vessels in inflammation and allergy. J Allergy (Cairo) 2013; 2013:672381. [PMID: 23431319 PMCID: PMC3574757 DOI: 10.1155/2013/672381] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Revised: 12/04/2012] [Accepted: 12/19/2012] [Indexed: 01/24/2023] Open
Abstract
Angiogenesis and lymphangiogenesis, the growth of new vessels from preexisting ones, have received increasing interest due to their role in tumor growth and metastatic spread. However, vascular remodeling, associated with vascular hyperpermeability, is also a key feature of many chronic inflammatory diseases including asthma, atopic dermatitis, psoriasis, and rheumatoid arthritis. The major drivers of angiogenesis and lymphangiogenesis are vascular endothelial growth factor- (VEGF-)A and VEGF-C, activating specific VEGF receptors on the lymphatic and blood vascular endothelium. Recent experimental studies found potent anti-inflammatory responses after targeted inhibition of activated blood vessels in models of chronic inflammatory diseases. Importantly, our recent results indicate that specific activation of lymphatic vessels reduces both acute and chronic skin inflammation. Thus, antiangiogenic and prolymphangiogenic therapies might represent a new approach to treat chronic inflammatory disorders, including those due to chronic allergic inflammation.
Collapse
|
203
|
Mohit E, Rafati S. Chemokine-based immunotherapy: delivery systems and combination therapies. Immunotherapy 2013; 4:807-40. [PMID: 22947009 DOI: 10.2217/imt.12.72] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
A major role of chemokines is to mediate leukocyte migration through interaction with G-protein-coupled receptors. Various delivery systems have been developed to utilize the chemokine properties for combating disease. Viral and mutant viral vectors expressing chemokines, genetically modified dendritic cells with chemokine or chemokine receptors, engineered chemokine-expressing tumor cells and pDNA encoding chemokines are among these methods. Another approach for inducing a targeted immune response is fusion of a targeting antibody or antibody fragment to a chemokine. In addition, chemokines induce more effective antitumor immunity when used as adjuvants. In this regard, chemokines are codelivered along with antigens or fused as a targeting unit with antigenic moieties. In this review, several chemokines with their role in inducing immune response against different diseases are discussed, with a major emphasis on cancer.
Collapse
Affiliation(s)
- Elham Mohit
- Molecular Immunology & Vaccine Research Lab, Pasteur Institute of Iran, Tehran 13164, Iran
| | | |
Collapse
|
204
|
Ahmadi Z, Arababadi MK, Hassanshahi G. CXCL10 Activities, Biological Structure, and Source Along with Its Significant Role Played in Pathophysiology of Type I Diabetes Mellitus. Inflammation 2012; 36:364-71. [DOI: 10.1007/s10753-012-9555-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
205
|
Abstract
Leukocyte recruitment to sites of infection or tissue damage plays a crucial role for the innate immune response. Chemokine-dependent signaling in immune cells is a very important mechanism leading to integrin activation and leukocyte recruitment. CXC chemokine receptor 2 (CXCR2) is a prominent chemokine receptor on neutrophils. During the last years, several studies were performed investigating the role of CXCR2 in different diseases. Until now, many CXCR2 inhibitors are tested in animal models and clinical trials and promising results were obtained. This review gives an overview of the structure of CXCR2 and the signaling pathways that are activated following CXCR2 stimulation. We discuss in detail the role of this chemokine receptor in different disease models including acute lung injury, COPD, sepsis, and ischemia-reperfusion-injury. Furthermore, this review summarizes the results of clinical trials which used CXCR2 inhibitors.
Collapse
Affiliation(s)
- Anika Stadtmann
- Department of Anaesthesiology, Intensive Care and Pain Medicine, University of Münster Münster, Germany
| | | |
Collapse
|
206
|
HIV-1 matrix protein p17 promotes angiogenesis via chemokine receptors CXCR1 and CXCR2. Proc Natl Acad Sci U S A 2012; 109:14580-5. [PMID: 22904195 DOI: 10.1073/pnas.1206605109] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Vascular diseases supported by aberrant angiogenesis have increased incidence in HIV-1-infected patients. Several data suggest that endothelium dysfunction relies on action of HIV-1 proteins rather than on a direct effect of the virus itself. The HIV-1 matrix protein p17 is known to deregulate the biological activity of different immune cells. Recently, p17 was found to mimic IL-8 chemokine activity by binding to the IL-8 receptor CXCR1. Here we show that p17 binds with high affinity to CXCR2, a CXCR1-related receptor, and promotes the formation of capillary-like structures on human endothelial cells (ECs) by interacting with both CXCR1 and CXCR2 expressed on the EC surface. ERK signaling via Akt was defined as the pathway responsible for p17-induced tube formation. Ex vivo and in vivo experimental models confirmed the provasculogenic activity of p17, which was comparable to that induced by VEGF-A. The hypothesis of a major role for p17 in HIV-1-induced aberrant angiogenesis is enforced by the finding that p17 is detected, as a single protein, in blood vessels of HIV-1-patients and in particular in the nucleus of ECs. Localization of p17 in the nucleus of ECs was evidenced also in in vitro experiments, suggesting the internalization of exogenous p17 in ECs by mechanisms of receptor-mediated endocytosis. Recognizing p17 interaction with CXCR1 and CXCR2 as the key event in sustaining EC aberrant angiogenesis could help us to identify new treatment strategies in combating AIDS-related vascular diseases.
Collapse
|
207
|
Yates-Binder CC, Rodgers M, Jaynes J, Wells A, Bodnar RJ, Turner T. An IP-10 (CXCL10)-derived peptide inhibits angiogenesis. PLoS One 2012; 7:e40812. [PMID: 22815829 PMCID: PMC3397949 DOI: 10.1371/journal.pone.0040812] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Accepted: 06/15/2012] [Indexed: 12/30/2022] Open
Abstract
Angiogenesis plays a critical role in processes such as organ development, wound healing, and tumor growth. It requires well-orchestrated integration of soluble and matrix factors and timely recognition of such signals to regulate this process. Previous work has shown that newly forming vessels express the chemokine receptor CXC receptor 3 (CXCR3) and, activation by its ligand IP-10 (CXCL10), both inhibits development of new vasculature and causes regression of newly formed vessels. To identify and develop new therapeutic agents to limit or reverse pathological angiogenesis, we identified a 21 amino acid fragment of IP-10, spanning the α-helical domain residues 77–98, that mimic the actions of the whole IP-10 molecule on endothelial cells. Treatment of the endothelial cells with the 22 amino acid fragment referred to as IP-10p significantly inhibited VEGF-induced endothelial motility and tube formation in vitro, properties critical for angiogenesis. Using a Matrigel plug assay in vivo, we demonstrate that IP-10p both prevented vessel formation and induced involution of nascent vessels. CXCR3 neutralizing antibody was able to block the inhibitory effects of the IP-10p, demonstrating specificity of the peptide. Inhibition of endothelial function by IP-10p was similar to that described for IP-10, secondary to CXCR3-mediated increase in cAMP production, activation of PKA inhibiting cell migration, and inhibition of VEGF-mediated m-calpain activation. IP-10p provides a novel therapeutic agent that inhibits endothelial cell function thus, allowing for the modulation of angiogenesis.
Collapse
Affiliation(s)
- Cecelia C. Yates-Binder
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama, United States of America
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (CCYB); (RJB)
| | - Margaret Rodgers
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Jesse Jaynes
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama, United States of America
| | - Alan Wells
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama, United States of America
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
| | - Richard J. Bodnar
- Departments of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States of America
- * E-mail: (CCYB); (RJB)
| | - Timothy Turner
- Tuskegee University, Center for Cancer Research, Tuskegee, Alabama, United States of America
| |
Collapse
|
208
|
Salim PH, Jobim M, Bredemeier M, Chies JAB, Brenol JCT, Jobim LF, Xavier RM. Combined effects of CXCL8 and CXCR2 gene polymorphisms on susceptibility to systemic sclerosis. Cytokine 2012; 60:473-7. [PMID: 22763041 DOI: 10.1016/j.cyto.2012.05.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 05/26/2012] [Accepted: 05/26/2012] [Indexed: 01/05/2023]
Abstract
UNLABELLED A previous study suggested that the CXCR2 (+1208) TT genotype was associated with increased risk of systemic sclerosis (SSc). In the present study, we investigated the influence of variation in the CXCL8 and CXCR2 genes on susceptibility to SSc and combined the variant alleles of these genes to analyze their effects on SSc. METHODS One fifty one patients with SSc and 147 healthy bone marrow donors were enrolled in a case-control study. Blood was collected for DNA extraction; typing of CXCL8 (-251) T/A and CXCR2 (+1208) T/C genes was made by polymerase chain reaction with sequence specific primers (PCR-SSP), followed by agarose gel electrophoresis. RESULTS The CXCR2-TC genotype was significantly less frequent in patients (23.8% versus 55.1% in controls; P<0.001, OR=0.26, 95%CI=0.15-0.43), whereas the CXCR2-CC genotype was significantly more frequent (44.4% versus 22.4% in controls; P<0.001, OR=2.76, 95%CI, 1.62-4.72). When CXCR2 and CXCL8 combinations were analyzed, the presence of CXCR2 T in the absence of CXCL8 A (CXCR2 T+/CXCL8 A-) was more frequent in patients than in controls (34.5% versus 3.5%; P<0.001, OR=14.50, 95%CI=5.04-41.40). However, CXCR2 TT and CXCL8 A were significantly more common in controls (100%) than in patients (58.3%) (P<0.001). Likewise, the presence of CXCR2 TC and CXCL8 A was more frequent in controls (95.1%) than in patients (75%) (P=0.004). Furthermore, the CXCR2-CC genotype in CXCL8 A was more frequent in patients (59.7% versus 0% in controls; P<0.001, adjusted OR=98.67, 95%CI=6.04-1610.8). In patients, a high frequency was observed in combination with the CXCL8 TA and AA genotypes (P<0.001; OR=28.92), whereas in controls, there was a high frequency of combination with CXCL8 T (P<0.001; OR=0.03) and TT (P<0.001; OR=0.01). CONCLUSIONS These findings suggest a protective role of CXCL8 (-251) A in the CXCR2 (+1208) TT and TC genotypes and an increased risk of CXCL8 (-251) A in association with the CXCR2 (+1208) CC genotype in SSc patients.
Collapse
Affiliation(s)
- Patricia Hartstein Salim
- Graduate Program in Medical Sciences, Universidade Federal do Rio Grande do Sul. Porto Alegre, RS, Brazil.
| | | | | | | | | | | | | |
Collapse
|
209
|
Zhu Q, Han X, Peng J, Qin H, Wang Y. The role of CXC chemokines and their receptors in the progression and treatment of tumors. J Mol Histol 2012; 43:699-713. [PMID: 22752457 DOI: 10.1007/s10735-012-9435-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/21/2012] [Indexed: 12/25/2022]
Abstract
Chemokines are a class of functional chemotactic peptides that contribute to a number of tumor-related processes. They are functionally defined as soluble factors that are able to control the directional migration of leukocytes, in particular, during infection and inflammation. It appears, however, that the biological effects mediated by chemokines are far more complex, and virtually all cells, including many tumor cell types, can express chemokines and chemokine receptors. A growing body of evidence indicates that they also contribute to a number of tumor-related processes, such as tumor cell growth, angiogenesis/angiostasis, local invasion, and mediate organ-specific metastases of cancer. The CXC chemokine class is a subfamily of a large family of chemokines. During the occurrence and development of tumor cells, this chemokine class is often accompanied by a series of molecular and biological changes. The CXC chemokine subfamily is closely related to the body's immune response to tumors and biological behaviors of tumors. In this paper, CXC chemokines and their role in the progression and treatment of tumors will be reviewed.
Collapse
Affiliation(s)
- Qingchao Zhu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, 200233, People's Republic of China.
| | | | | | | | | |
Collapse
|
210
|
Schraufstatter IU, Zhao M, Khaldoyanidi SK, Discipio RG. The chemokine CCL18 causes maturation of cultured monocytes to macrophages in the M2 spectrum. Immunology 2012; 135:287-98. [PMID: 22117697 DOI: 10.1111/j.1365-2567.2011.03541.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The observation that human monocytes cultured in the presence of the chemokine CCL18 showed increased survival, led us to profile cytokine expression in CCL18-stimulated versus control cultures. CCL18 caused significantly increased expression of chemokines (CXCL8, CCL2, CCL3 and CCL22), interleukin-10 (IL-10) and platelet-derived growth factor, but no up-regulation of M1 cytokines IL-1β or IL-12. CCL18-stimulated monocytes matured into cells with morphological resemblance to IL-4-stimulated macrophages, and expressed the monocyte marker CD14 as well the M2 macrophage markers CD206 and 15-lipoxygenase, but no mature dendritic cell markers (CD80, CD83 or CD86). Functionally, CCL18-stimulated macrophages showed a high capacity for unspecific phagocytosis and for pinocytosis, which was not associated with an oxidative burst. These findings suggest that CCL18-activated macrophages stand at the cross-roads between inflammation and its resolution. The chemokines that are produced in response to CCL18 are angiogenic and attract various leucocyte populations, which sustain inflammation. However, the capacity of these cells to remove cellular debris without causing oxidative damage and the production of the anti-inflammatory IL-10 will initiate termination of the inflammatory response. In summary, CCL18 induces an M2 spectrum macrophage phenotype in the absence of IL-4.
Collapse
|
211
|
Tyagi P, Killinger K, Tyagi V, Nirmal J, Chancellor M, Peters KM. Urinary chemokines as noninvasive predictors of ulcerative interstitial cystitis. J Urol 2012; 187:2243-8. [PMID: 22503040 DOI: 10.1016/j.juro.2012.01.034] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Indexed: 01/14/2023]
Abstract
PURPOSE Based on basic research findings an increase in chemokines and cytokines (CXCL-1 and 10, nerve growth factor and interleukin-6) is considered responsible for inflammation and afferent sensitization. In this cross-sectional study we tested the hypothesis that select chemokines are increased in the urine of patients with ulcerative and nonulcerative interstitial cystitis/painful bladder syndrome. MATERIALS AND METHODS Midstream urinary specimens were collected from 10 patients with ulcerative and nonulcerative interstitial cystitis/painful bladder syndrome, respectively, and from 10 asymptomatic controls. Urinary levels of 7 cytokines were measured by a human cytokine/chemokine assay. Nerve growth factor was measured by enzyme-linked immunosorbent assay. RESULTS Urinary levels of most chemokines/cytokines were tenfold to 100-fold lower in asymptomatic controls vs patients with ulcerative and nonulcerative interstitial cystitis/painful bladder syndrome. Univariate comparison of 8 tested proteins in the ulcerative vs nonulcerative groups revealed a significant fivefold to twentyfold increase in CXCL-10 and 1, interleukin-6 and nerve growth factor (ANOVA p<0.001). CONCLUSIONS Differential expression of chemokines in ulcerative and nonulcerative subtypes of interstitial cystitis/painful bladder syndrome suggests differences in paracrine signaling between the 2 entities.
Collapse
Affiliation(s)
- Pradeep Tyagi
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
212
|
Bertini R, Barcelos LS, Beccari AR, Cavalieri B, Moriconi A, Bizzarri C, Di Benedetto P, Di Giacinto C, Gloaguen I, Galliera E, Corsi MM, Russo RC, Andrade SP, Cesta MC, Nano G, Aramini A, Cutrin JC, Locati M, Allegretti M, Teixeira MM. Receptor binding mode and pharmacological characterization of a potent and selective dual CXCR1/CXCR2 non-competitive allosteric inhibitor. Br J Pharmacol 2012; 165:436-54. [PMID: 21718305 DOI: 10.1111/j.1476-5381.2011.01566.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND AND PURPOSE DF 2156A is a new dual inhibitor of IL-8 receptors CXCR1 and CXCR2 with an optimal pharmacokinetic profile. We characterized its binding mode, molecular mechanism of action and selectivity, and evaluated its therapeutic potential. EXPERIMENTAL APPROACH The binding mode, molecular mechanism of action and selectivity were investigated using chemotaxis of L1.2 transfectants and human leucocytes, in addition to radioligand and [(35) S]-GTPγS binding approaches. The therapeutic potential of DF 2156A was evaluated in acute (liver ischaemia and reperfusion) and chronic (sponge-induced angiogenesis) experimental models of inflammation. KEY RESULTS A network of polar interactions stabilized by a direct ionic bond between DF 2156A and Lys(99) on CXCR1 and the non-conserved residue Asp(293) on CXCR2 are the key determinants of DF 2156A binding. DF 2156A acted as a non-competitive allosteric inhibitor blocking the signal transduction leading to chemotaxis without altering the binding affinity of natural ligands. DF 2156A effectively and selectively inhibited CXCR1/CXCR2-mediated chemotaxis of L1.2 transfectants and leucocytes. In a murine model of sponge-induced angiogenesis, DF 2156A reduced leucocyte influx, TNF-α production and neovessel formation. In vitro, DF 2156A prevented proliferation, migration and capillary-like organization of HUVECs in response to human IL-8. In a rat model of liver ischaemia and reperfusion (I/R) injury, DF 2156A decreased PMN and monocyte-macrophage infiltration and associated hepatocellular injury. CONCLUSION AND IMPLICATIONS DF 2156A is a non-competitive allosteric inhibitor of both IL-8 receptors CXCR1 and CXCR2. It prevented experimental angiogenesis and hepatic I/R injury in vivo and, therefore, has therapeutic potential for acute and chronic inflammatory diseases.
Collapse
|
213
|
Abstract
During the past decade, skeletal muscle has been identified as a secretory organ. Accordingly, we have suggested that cytokines and other peptides that are produced, expressed and released by muscle fibres and exert either autocrine, paracrine or endocrine effects should be classified as myokines. The finding that the muscle secretome consists of several hundred secreted peptides 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. However, some myokines exert their effects within the muscle itself. Thus, myostatin, LIF, IL-6 and IL-7 are involved in muscle hypertrophy and myogenesis, whereas BDNF and IL-6 are involved in AMPK-mediated fat oxidation. IL-6 also appears to have systemic effects on the liver, adipose tissue and the immune system, and mediates crosstalk between intestinal L cells and pancreatic islets. Other myokines include the osteogenic factors IGF-1 and FGF-2; FSTL-1, which improves the endothelial function of the vascular system; and the PGC-1α-dependent myokine irisin, which drives brown-fat-like development. Studies in the past few years suggest the existence of yet unidentified factors, secreted from muscle cells, which may influence cancer cell growth and pancreas function. Many proteins produced by skeletal muscle are dependent upon contraction; therefore, physical inactivity probably leads to an altered myokine response, which could provide a potential mechanism for the association between sedentary behaviour and many chronic diseases.
Collapse
Affiliation(s)
- Bente K Pedersen
- The Centre of Inflammation and Metabolism, Department of Infectious Diseases and CMRC, Rigshospitalet, Section 7641, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 9, DK-2100, Copenhagen, Denmark.
| | | |
Collapse
|
214
|
Pedersen L, Olsen CH, Pedersen BK, Hojman P. Muscle-derived expression of the chemokine CXCL1 attenuates diet-induced obesity and improves fatty acid oxidation in the muscle. Am J Physiol Endocrinol Metab 2012; 302:E831-40. [PMID: 22275756 DOI: 10.1152/ajpendo.00339.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serum levels and muscle expression of the chemokine CXCL1 increase markedly in response to exercise in mice. Because several studies have established muscle-derived factors as important contributors of metabolic effects of exercise, this study aimed at investigating the effect of increased expression of muscle-derived CXCL1 on systemic and intramuscular metabolic parameters, with focus on fatty acid oxidation and oxidative metabolism in skeletal muscle. By overexpression of CXCL1 in the tibialis cranialis muscle in mice, significant elevations in muscle and serum CXCL1 within a physiological range were obtained. At 3 mo of high-fat feeding, visceral and subcutaneous fat mass were 32.4 (P < 0.01) and 22.4% (P < 0.05) lower, respectively, in CXCL1-overexpressing mice compared with control mice. Also, chow-fed CXCL-transfected mice had 35.4% (P < 0.05) lower visceral fat mass and 33.4% (P < 0.05) lower subcutaneous fat mass compared with chow-fed control mice. These reductions in accumulation of adipose tissue were accompanied by improved glucose tolerance and insulin sensitivity. Furthermore, in CXCL1-transfected muscles, muscular ex vivo fatty acid oxidation was significantly enhanced compared with control muscles (chow fed: 2.2-fold, P < 0.05; high-fat fed: 2-fold, P < 0.05) and also showed increased expression levels of major fatty acid oxidation genes (CD36, CPT I, and HADH). Finally, CXCL1 expression was associated with increased muscle mRNA expression of VEGF and CD31, suggesting a role for CXCL1 in muscle angiogenesis. In conclusion, our data show that overexpression of CXCL1 within a physiological range attenuates diet-induced obesity, likely mediated through a CXCL1-induced improvement of fatty acid oxidation and oxidative capacity in skeletal muscle tissue.
Collapse
Affiliation(s)
- Line Pedersen
- Centre of Inflammation and Metabolism, Department of Infectious Diseases, Rigshospitalet, Copenhagen, Denmark
| | | | | | | |
Collapse
|
215
|
Castor MGM, Pinho V, Teixeira MM. The role of chemokines in mediating graft versus host disease: opportunities for novel therapeutics. Front Pharmacol 2012; 3:23. [PMID: 22375119 PMCID: PMC3285883 DOI: 10.3389/fphar.2012.00023] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 02/08/2012] [Indexed: 11/25/2022] Open
Abstract
Bone marrow transplantation (BMT) is the current therapy of choice for several malignancies and severe autoimmune diseases. Graft versus host disease (GVHD) is the major complication associated with BMT. T lymphocytes and other leukocytes migrate into target organs during GVHD, become activated and mediate tissue damage. Chemokines are well known inducers of leukocyte trafficking and activation and contribute to the pathogenesis of GVHD. Here, we review the major animal models used to study GVHD and the role of chemokines in mediating tissue damage in these models. The role of these molecules in promoting potential beneficial effects of the graft, especially graft versus leukemia, is also discussed. Finally, the various pharmacological strategies to block the chemokine system or downstream signaling events in the context of GVHD are discussed.
Collapse
Affiliation(s)
- Marina G M Castor
- Immunopharmacology, Department of Immunology and Biochemistry, Institute of Biological Sciences, Universidade Federal de Minas Gerais Belo Horizonte, Brazil
| | | | | |
Collapse
|
216
|
Han L, Jiang B, Wu H, Wang X, Tang X, Huang J, Zhu J. High expression of CXCR2 is associated with tumorigenesis, progression, and prognosis of laryngeal squamous cell carcinoma. Med Oncol 2012; 29:2466-72. [PMID: 22274915 DOI: 10.1007/s12032-011-0152-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 12/21/2011] [Indexed: 12/27/2022]
Abstract
The laryngeal squamous cell carcinoma (LSCC) is one of the most common cancers threatening people's life. CXC-chemokine receptor type 2 (CXCR2) was reported to play critical roles in angiogenesis, tumorigenesis, and metastasis of several cancers such as colon cancer, melanoma, lung cancer, and so on. However, the expression of CXCR2 in LSCC and its association with clinical characters of LSCC remain unclear. Quantitative real-time reverse transcription-PCR and immunohistochemistry were used, respectively, to analyze the mRNA level and protein level of CXCR2 in 109 cases of LSCC tissues and 28 cases of tumor-adjacent normal tissues. The expression of CXCR2 in LSCC was significantly higher than that in tumor-adjacent tissues. Moreover, the expression level of CXCR2 protein in LSCC was significantly related to lymph node metastasis (P=.022), histopathological grade (P=.038), and 5 years' survival (P=.007). Cox regression analysis revealed that CXCR2 expression (P=.031), as well as lymphatic metastasis (P=.026) and TNM classification (P=.042), is an independent prognostic marker of LSCC. High expression of CXCR2 is also associated with short survival of LSCC patients. Our data indicate that the expression of CXCR2 is associated with the development and progression of LSCC. CXCR2 expression may serve as an independent prognostic marker for LSCC patients.
Collapse
Affiliation(s)
- Liang Han
- Department of Head and Neck Surgery, Nantong Tuomor Hospital, Nantong, Jiangsu, China
| | | | | | | | | | | | | |
Collapse
|
217
|
Mukaida N, Baba T. Chemokines in tumor development and progression. Exp Cell Res 2012; 318:95-102. [PMID: 22036649 DOI: 10.1016/j.yexcr.2011.10.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 10/14/2011] [Accepted: 10/14/2011] [Indexed: 12/17/2022]
Abstract
Chemokines were originally identified as mediators of the inflammatory process and regulators of leukocyte trafficking. Subsequent studies revealed their essential roles in leukocyte physiology and pathology. Moreover, chemokines have profound effects on other types of cells associated with the inflammatory response, such as endothelial cells and fibroblasts. Thus, chemokines are crucial for cancer-related inflammation, which can promote tumor development and progression. Increasing evidence points to the vital effects of several chemokines on the proliferative and invasive properties of tumor cells. The wide range of activities of chemokines in tumorigenesis highlights their roles in tumor development and progression.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| | | |
Collapse
|
218
|
Ghasemi H, Ghazanfari T, Yaraee R, Faghihzadeh S, Hassan ZM. Roles of IL-8 in Ocular Inflammations: A Review. Ocul Immunol Inflamm 2011; 19:401-12. [DOI: 10.3109/09273948.2011.618902] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
219
|
Epidermal growth factor receptor reactivation induced by E-prostanoid-3 receptor- and tumor necrosis factor-alpha-converting enzyme-dependent feedback exaggerates interleukin-8 production in airway cancer (NCI-H292) cells. Exp Cell Res 2011; 317:2650-60. [DOI: 10.1016/j.yexcr.2011.08.023] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 08/11/2011] [Accepted: 08/30/2011] [Indexed: 01/31/2023]
|
220
|
Schauer IG, Rowley DR. The functional role of reactive stroma in benign prostatic hyperplasia. Differentiation 2011; 82:200-10. [PMID: 21664759 PMCID: PMC3179838 DOI: 10.1016/j.diff.2011.05.007] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 05/03/2011] [Accepted: 05/16/2011] [Indexed: 12/19/2022]
Abstract
The human prostate gland is one of the only internal organs that continue to enlarge throughout adulthood. The specific mechanisms that regulate this growth, as well as the pathological changes leading to the phenotype observed in the disease benign prostatic hyperplasia (BPH), are essentially unknown. Recent studies and their associated findings have made clear that many complex alterations occur, involving persistent and chronic inflammation, circulating hormonal level deregulation, and aberrant wound repair processes. BPH has been etiologically characterized as a progressive, albeit discontinuous, hyperplasia of both the glandular epithelial and the stromal cell compartments coordinately yielding an expansion of the prostate gland and clinical symptoms. Interestingly, the inflammatory and repair responses observed in BPH are also key components of general wound repair in post-natal tissues. These responses include altered expression of chemokines, cytokines, matrix remodeling factors, chronic inflammatory processes, altered immune surveillance and recognition, as well as the formation of a prototypical 'reactive' stroma, which is similar to that observed across various fibroplasias and malignancies of a variety of tissue sites. Stromal tissue, both embryonic mesenchyme and adult reactive stroma myofibroblasts, has been shown to exert potent and functional regulatory control over epithelial proliferation and differentiation as well as immunoresponsive modulation. Thus, the functional biology of a reactive stroma, within the context of an adult disease typified by epithelial and stromal aberrant hyperplasia, is critical to understand within the context of prostate disease and beyond. The mechanisms that regulate reactive stroma biology in BPH represent targets of opportunity for new therapeutic approaches that may extend to other tissue contexts. Accordingly, this review seeks to address the dissection of important factors, signaling pathways, genes, and other regulatory components that mediate the interplay between epithelium and stromal responses in BPH.
Collapse
Affiliation(s)
- Isaiah G Schauer
- Department of Molecular and Cellular Biology, One Baylor Plaza, Jewish Research Institute, Baylor College of Medicine, 325D, mailstop BCM130, Houston, TX 77030, USA.
| | | |
Collapse
|
221
|
Singh S, Wu S, Varney M, Singh AP, Singh RK. CXCR1 and CXCR2 silencing modulates CXCL8-dependent endothelial cell proliferation, migration and capillary-like structure formation. Microvasc Res 2011; 82:318-25. [PMID: 21749879 PMCID: PMC3215896 DOI: 10.1016/j.mvr.2011.06.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Revised: 05/30/2011] [Accepted: 06/23/2011] [Indexed: 12/17/2022]
Abstract
CXCR1 and CXCR2 are receptors for angiogenic ELR+CXC chemokines and are differentially expressed on endothelial cells; however, their functional significance in angiogenesis remains unclear. In this study, we determined the functional significance of these receptors in modulating endothelial cell phenotype by knocking-down the expression of CXCR1 and/or CXCR2 in human microvascular endothelial cells (HMEC-1) using short-hairpin RNA (shRNA). Cell proliferation, migration, invasion and capillary-like structure (CLS) formation were analyzed. Our data demonstrate that knock-down of CXCR1 and/or CXCR2 expression inhibited endothelial cell proliferation, survival, migration, invasion and CLS formation. Additionally, we examined the mechanism of CXCL8-dependent CXCR1 and/or CXCR2 mediated phenotypic changes by evaluating ERK phosphorylation and cytoskeletal rearrangement and observed inhibition of ERK phosphorylation and cytoskeletal rearrangement in HMEC-1-shCXCR1, HMEC-1-shCXCR2 and HMEC-1-shCXCR1/2 cells. Together, these data demonstrate that CXCR1 and CXCR2 expression plays a critical role in regulating multiple biological activities in human microvascular endothelial cells.
Collapse
Affiliation(s)
- Seema Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.
| | | | | | | | | |
Collapse
|
222
|
Zeng Y, Shen Y, Huang XL, Liu XJ, Liu XH. Roles of mechanical force and CXCR1/CXCR2 in shear-stress-induced endothelial cell migration. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2011; 41:13-25. [PMID: 21989491 DOI: 10.1007/s00249-011-0752-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 09/13/2011] [Indexed: 02/05/2023]
Abstract
We previously demonstrated that CXCR1 and CXCR2 are novel mechanosensors mediating laminar shear-stress-induced endothelial cell (EC) migration (Zeng et al. in Cytokine 53:42-51, 2011). In the present study, an analytical model was proposed to further analyze the underlying mechanisms, assuming the mechanical force (MF) and mechanosensor-mediated biochemical reactions induce cell migration together. Shear stress can regulate both mechanosensor-mediated migration in the flow direction (Ms-M(FD)) and mechanosensor-mediated migration toward a wound (Ms-M(W)). Next, the migration distance, the roles of MF-induced cell migration (MF-M), and the mobilization mechanisms of mechanosensors were analyzed. The results demonstrated that MF-M plays an important role in 15.27 dyn/cm(2) shear-stress-induced EC migration but is far weaker than Ms-M(W) at 5.56 dyn/cm(2). Our findings also indicated that CXCR2 played a primary role, in synergy with CXCR1. The Ms-M(FD) was primarily mediated by the synergistic effect of CXCR1 and CXCR2. In Ms-M(W), when shear stress was beyond a certain threshold, the synergistic effect of CXCR1 and CXCR2 was enhanced, and the effect of CXCR1 was inhibited. Therefore, the retarding of EC migration and wound closure capacity under low shear flow was related to the low magnitude of shear stress, which may contribute to atherogenesis and many other vascular diseases.
Collapse
Affiliation(s)
- Ye Zeng
- Laboratory of Cardiovascular Diseases, West China Hospital, Sichuan University, No.17 Renmin Nanlu 3 Duan, Chengdu, People's Republic of China
| | | | | | | | | |
Collapse
|
223
|
Imaoka H, Punia N, Irshad A, Ying S, Corrigan CJ, Howie K, O'Byrne PM, Gauvreau GM, Sehmi R. Lung homing of endothelial progenitor cells in humans with asthma after allergen challenge. Am J Respir Crit Care Med 2011; 184:771-8. [PMID: 21719753 DOI: 10.1164/rccm.201102-0272oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
RATIONALE Increased bronchial vascularity is a feature of asthma that can contribute to airflow obstruction and progressive decline in lung function. Angiogenesis is associated with the lung homing and in situ differentiation of endothelial progenitor cells (EPC) in mouse models of asthma. We have previously shown that inhibiting allergen (Ag)-induced recruitment of EPC in sensitized mice attenuated increased bronchial vascularity and development of airway hyperresponsiveness. OBJECTIVES We investigated the accumulation of EPC and formation of new blood vessels in the lungs of human subjects with asthma after Ag inhalation challenge. METHODS Consenting patients with mild atopic asthma (n = 13) with FEV1 ≥ 70%, methacholine PC20 ≤ 16 mg/ml, and a dual response to Ag were recruited. Sputum levels of EPC were determined by multigating flow cytometry, and lung vascularity was enumerated by immunostaining with von Willebrand factor. MEASUREMENTS AND MAIN RESULTS Sputum levels of EPC were determined by multigating flow cytometry and lung vascularity was enumerated by immunostaining with von Willebrand factor. There was a significant increase in sputum EPC levels 24 hours post Ag but not diluent challenge. Similarly, a significant increase in the number and diameter of blood vessels in lung biopsy tissue 24 hours post Ag was observed. In vitro culture of EPC demonstrated the capacity of these cells to differentiate into mature endothelial cells and form tubelike vessel structures. In sputum supernatants, there was a significant increase in CXCR2 agonists, IL-8, and Gro-α 24 hours post Ag. Only Gro-α stimulated a significant EPC migrational response in vitro. CONCLUSIONS Our data suggest that increased lung homing of EPC may promote bronchial vascularity in allergic asthmatic responses and that the recruitment of these progenitors maybe orchestrated by CXCR2 chemokines.
Collapse
Affiliation(s)
- Haruki Imaoka
- Asthma Research Group, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
224
|
Kiefer F, Siekmann AF. The role of chemokines and their receptors in angiogenesis. Cell Mol Life Sci 2011; 68:2811-30. [PMID: 21479594 PMCID: PMC11115067 DOI: 10.1007/s00018-011-0677-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 03/09/2011] [Accepted: 03/22/2011] [Indexed: 12/21/2022]
Abstract
Chemokines are a vertebrate-specific group of small molecules that regulate cell migration and behaviour in diverse contexts. So far, around 50 chemokines have been identified in humans, which bind to 18 different chemokine receptors. These are members of the seven-transmembrane receptor family. Initially, chemokines were identified as modulators of the immune response. Subsequently, they were also shown to regulate cell migration during embryonic development. Here, we discuss the influence of chemokines and their receptors on angiogenesis, or the formation of new blood vessels. We highlight recent advances in our understanding of how chemokine signalling might directly influence endothelial cell migration. We furthermore examine the contributions of chemokine signalling in immune cells during this process. Finally, we explore possible implications for disease settings, such as chronic inflammation and tumour progression.
Collapse
Affiliation(s)
- Friedemann Kiefer
- Max Planck Institute for Molecular Biomedicine, Roentgenstr. 20, 48149 Muenster, Germany
| | - Arndt F. Siekmann
- Max Planck Institute for Molecular Biomedicine, Roentgenstr. 20, 48149 Muenster, Germany
| |
Collapse
|
225
|
Piperi C, Samaras V, Levidou G, Kavantzas N, Boviatsis E, Petraki K, Grivas A, Barbatis C, Varsos V, Patsouris E, Korkolopoulou P. Prognostic significance of IL-8-STAT-3 pathway in astrocytomas: Correlation with IL-6, VEGF and microvessel morphometry. Cytokine 2011; 55:387-95. [DOI: 10.1016/j.cyto.2011.05.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 05/16/2011] [Indexed: 01/07/2023]
|
226
|
Kyriakakis E, Cavallari M, Pfaff D, Fabbro D, Mestan J, Philippova M, De Libero G, Erne P, Resink TJ. IL-8-mediated angiogenic responses of endothelial cells to lipid antigen activation of iNKT cells depend on EGFR transactivation. J Leukoc Biol 2011; 90:929-39. [PMID: 21807744 DOI: 10.1189/jlb.0211097] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
iNKT cells are a unique T cell subset, which is CD1d-restricted and specific for glycolipid antigens. In advanced atherosclerotic plaques, focal collections of inflammatory cells correlate with areas of intraplaque neovascularization. We reported recently that iNKT cells might facilitate intraplaque neovascularization by enhancing EC migration and sprouting in an IL-8-dependent manner. This study investigated the participating effector mechanisms. In ECs, CM, derived from antigen-stimulated human iNKT cells (CM+), induced up-regulation of IL-8R CXCR2 and the phosphorylation of EGFR and of multiple intracellular signaling effectors, including FAK, Src, Erk, Jnk, p38-MAPK, and STAT1 and -3. We found that a cascade of events, which were IL-8-dependent and involved EGFR activation, was responsible for signaling through FAK and Src kinases and necessary for acquisition of angiogenic morphology, migration in a two-dimensional wound assay, and sprout outgrowth in a three-dimensional model of angiogenesis in vitro. The data support that IL-8-dependent activation of angiogenic behavior in ECs, in response to activated iNKT, involves CXCR2, transactivation of EGFR, and subsequent FAK/Src signaling. We found too that activated iNKT increased VEGFR2 expression in ECs. Functional studies confirmed that EGF is the motogenic-enhancing factor in CM+ and is necessary, together with an exogenous source of VEGF, for iNKT-promoted sprout formation. EGFR inhibition may represent a novel therapeutic modality aimed at plaque stabilization through control of neovascularization within developing atherosclerotic plaques.
Collapse
|
227
|
Farkas L, Gauldie J, Voelkel NF, Kolb M. Pulmonary Hypertension and Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2011; 45:1-15. [DOI: 10.1165/rcmb.2010-0365tr] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
228
|
Mukaida N, Sasakki SI, Popivanova BK. Tumor Necrosis Factor (TNF) and Chemokines in Colitis-Associated Cancer. Cancers (Basel) 2011; 3:2811-2826. [PMID: 24212934 PMCID: PMC3759172 DOI: 10.3390/cancers3032811] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 06/17/2011] [Accepted: 06/21/2011] [Indexed: 12/13/2022] Open
Abstract
The connection between inflammation and tumorigenesis has been well established, based on a great deal of supporting evidence obtained from epidemiological, pharmacological, and genetic studies. One representative example is inflammatory bowel disease, because it is an important risk factor for the development of colon cancer. Moreover, intratumoral infiltration of inflammatory cells suggests the involvement of inflammatory responses also in other forms of sporadic as well as heritable colon cancer. Inflammatory responses and tumorigenesis activate similar sets of transcription factors such as NF-kB, Stat3, and hypoxia inducible factor and eventually enhances the expression of inflammatory cytokines including tumor necrosis factor (TNF) and chemokines. The expression of TNF and chemokines is aberrantly expressed in a mouse model of colitis-associated carcinogenesis as well as in inflammatory bowel disease and colon cancer in humans. Here, after summarizing the presumed actions of TNF and chemokines in tumor biology, we will discuss the potential roles of TNF and chemokines in chronic inflammation-associated colon cancer in mice.
Collapse
Affiliation(s)
- Naofumi Mukaida
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; E-Mail: (S.S.)
| | - So-ichiro Sasakki
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; E-Mail: (S.S.)
| | - Boryana K. Popivanova
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan; E-Mail: (S.S.)
- Present Address, Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; E-Mail:
| |
Collapse
|
229
|
Mihara K, Wijkmans J. Low Molecular Weight CXCR2 Antagonists as Promising Therapeutics. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2011. [DOI: 10.1002/9783527631995.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
230
|
Abstract
Chemokines and their receptors play a key role in development and homeostasis as well as in the pathogenesis of tumors and autoimmune diseases. Chemokines are involved in the implantation of the early conceptus, the migration of subsets of cells during embryonic development, and the overall growth of the embryo. Chemokines also have an important role in the development and maintenance of innate and adaptive immunity. In addition, they play a significant role in wound healing and angiogenesis. When the physiological role of chemokines is subverted or chronically amplified, disease often follows. Chemokines are involved in the pathobiology of chronic inflammation, tumorigenesis and metastasis, as well as autoimmune diseases. This article reviews the role of chemokines and their receptors in normal and disease processes and the potential for using chemokine antagonists for appropriate targeted therapy.
Collapse
Affiliation(s)
- Dayanidhi Raman
- Department of Cancer Biology, Vanderbilt University, School of Medicine, Nashville, TN 37232
| | - Tammy Sobolik-Delmaire
- Department of Cancer Biology, Vanderbilt University, School of Medicine, Nashville, TN 37232
| | - Ann Richmond
- Department of Cancer Biology, Vanderbilt University, School of Medicine, Nashville, TN 37232
- VA Medical Center; Nashville, TN 37232
| |
Collapse
|
231
|
Keeley EC, Mehrad B, Strieter RM. Chemokines as mediators of tumor angiogenesis and neovascularization. Exp Cell Res 2011; 317:685-90. [PMID: 21040721 PMCID: PMC3073599 DOI: 10.1016/j.yexcr.2010.10.020] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 10/25/2010] [Indexed: 12/11/2022]
Abstract
Chemokines are a superfamily of structurally homologous heparin-binding proteins that influence tumor growth and metastasis. Several members of the CXC and CC chemokine families are potent inducers of neovascularization, whereas a subset of the CXC chemokines are potent inhibitors. In this paper, we review the current literature regarding the role of chemokines as mediators of tumor angiogenesis and neovascularization.
Collapse
Affiliation(s)
- Ellen C. Keeley
- Department of Medicine, Division of Cardiology, University of Virginia, Charlottesville, Virginia
| | - Borna Mehrad
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| | - Robert M. Strieter
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
232
|
Li A, King J, Moro A, Sugi MD, Dawson DW, Kaplan J, Li G, Lu X, Strieter RM, Burdick M, Go VLW, Reber HA, Eibl G, Hines OJ. Overexpression of CXCL5 is associated with poor survival in patients with pancreatic cancer. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1340-9. [PMID: 21356384 PMCID: PMC3069811 DOI: 10.1016/j.ajpath.2010.11.058] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 10/20/2010] [Accepted: 11/09/2010] [Indexed: 01/12/2023]
Abstract
Epithelial neutrophil-activating peptide-78 (CXCL5), a member of the CXC chemokine family, has been shown to be involved in angiogenesis, tumor growth, and metastasis. The objective of this study was to determine the relationship between CXCL5 expression and tumor progression in human pancreatic cancer and to elucidate the mechanism underlying CXCL5-mediated tumor angiogenesis and cancer growth. We report herein that CXCL5 is overexpressed in human pancreatic cancer compared with paired normal pancreas tissue. Overexpression of CXCL5 is significantly correlated with poorer tumor differentiation, advanced clinical stage, and shorter patient survival. Patients with pancreatic cancer and CXCL5 overexpression who underwent resection of cancer had a mean survival time 25.5 months shorter than that of patients who did not overexpress CXCL5. Blockade of CXCL5 or its receptor CXCR2 by small-interfering RNA knockdown or antibody neutralization attenuated human pancreatic cancer growth in a nude mouse model. Finally, we demonstrated that CXCL5 mediates pancreatic cancer-derived angiogenesis through activation of several signaling pathways, including protein kinase B (Akt), extracellular signal-regulated kinase (ERK), and signal transducer and activator of transcription (STAT) in human endothelial cells. These data suggest that CXCL5 is an important mediator of tumor-derived angiogenesis and that it may serve as a survival factor for pancreatic cancer. Blockade of either CXCL5 or CXCR2 may be a critical adjunct antiangiogenic therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Aihua Li
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - Jonathan King
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - Aune Moro
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - Mark D. Sugi
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - David W. Dawson
- Department of Pathology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Jeffrey Kaplan
- Department of Pathology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Gang Li
- Department of Biostatistics, School of Public Health, University of California at Los Angeles, Los Angeles, California
| | - Xuyang Lu
- Department of Biostatistics, School of Public Health, University of California at Los Angeles, Los Angeles, California
| | - Robert M. Strieter
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Marie Burdick
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Vay Liang W. Go
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Howard A. Reber
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - Guido Eibl
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| | - O. Joe Hines
- Department of Surgery, Hirshberg Laboratories for Pancreatic Cancer Research, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
233
|
Elson JK, Beebe-Dimmer JL, Morgenstern H, Chilkuri M, Blanchard J, Lentsch AB. The Duffy Antigen/Receptor for Chemokines (DARC) and prostate-cancer risk among Jamaican men. J Immigr Minor Health 2011; 13:36-41. [PMID: 20596779 PMCID: PMC3017736 DOI: 10.1007/s10903-010-9330-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As an evolutionary response to prevent malaria infection, most Africans do not express the Duffy Antigen/Receptor for Chemokines (DARC) on their red blood cells. Results from experimental studies suggest that DARC expression inhibits prostate-tumor growth. We tested the hypothesis that men of African descent who lack DARC expression are at increased risk of prostate cancer. A case-control study involving 81 age-matched pairs was conducted in Jamaica. Participants were interviewed to collect data, and they donated blood for determination of DARC expression. Logistic regression was used to estimate associations with prostate cancer and aggressive disease. Little or no association was observed between erythrocyte DARC expression and prostate cancer or between DARC expression and aggressive disease. These associations changed little when adjusting for other potential confounders. Our results do not support an effect of erythrocyte DARC expression on the risk or progression of prostate cancer in men of African descent.
Collapse
Affiliation(s)
- Joshua K. Elson
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jennifer L. Beebe-Dimmer
- Karmanos Cancer Institute and Department of Internal Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hal Morgenstern
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, School of Public Health and Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | | | - John Blanchard
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| | - Alex B. Lentsch
- Department of Surgery, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
234
|
Varney ML, Singh S, Li A, Mayer-Ezell R, Bond R, Singh RK. Small molecule antagonists for CXCR2 and CXCR1 inhibit human colon cancer liver metastases. Cancer Lett 2011; 300:180-8. [PMID: 21035946 PMCID: PMC2994987 DOI: 10.1016/j.canlet.2010.10.004] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 09/28/2010] [Accepted: 10/04/2010] [Indexed: 12/12/2022]
Abstract
CXCR1 and CXCR2 are G-protein coupled receptors, that have been shown to play important role in tumor growth and metastasis, and are prime targets for the development of novel therapeutics. Here, we report that targeting CXCR2 and CXCR1 activity using orally active small molecule antagonist (SCH-527123, SCH-479833) inhibits human colon cancer liver metastasis mediated by decreased neovascularization and enhanced malignant cell apoptosis. There were no differences in primary tumor growth. These studies demonstrate the important role of CXCR2/1 in colon cancer metastasis and that inhibition of CXCR2 and CXCR1, small molecule antagonists provides a novel therapeutic strategy.
Collapse
Affiliation(s)
- Michelle L. Varney
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Seema Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Aihua Li
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | | | - Richard Bond
- Schering-Plough Research Institute, Kenilworth, NJ
| | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
235
|
Baird AM, Gray SG, O'Byrne KJ. Epigenetics underpinning the regulation of the CXC (ELR+) chemokines in non-small cell lung cancer. PLoS One 2011; 6:e14593. [PMID: 21298036 PMCID: PMC3029265 DOI: 10.1371/journal.pone.0014593] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 01/02/2011] [Indexed: 12/21/2022] Open
Abstract
Background Angiogenesis may play a role in the pathogenesis of Non-Small Cell Lung cancer (NSCLC). The CXC (ELR+) chemokine family are powerful promoters of the angiogenic response. Methods The expression of the CXC (ELR+) family members (CXCL1-3/GROα-γ, CXCL8/IL-8, CXCR1/2) was examined in a series of resected fresh frozen NSCLC tumours. Additionally, the expression and epigenetic regulation of these chemokines was examined in normal bronchial epithelial and NSCLC cell lines. Results Overall, expression of the chemokine ligands (CXCL1, 2, 8) and their receptors (CXCR1/2) were down regulated in tumour samples compared with normal, with the exception of CXCL3. CXCL8 and CXCR1/2 were found to be epigenetically regulated by histone post-translational modifications. Recombinant CXCL8 did not stimulate cell growth in either a normal bronchial epithelial or a squamous carcinoma cell line (SKMES-1). However, an increase was observed at 72 hours post treatment in an adenocarcinoma cell line. Conclusions CXC (ELR+) chemokines are dysregulated in NSCLC. The balance of these chemokines may be critical in the tumour microenvironment and requires further elucidation. It remains to be seen if epigenetic targeting of these pathways is a viable therapeutic option in lung cancer treatment.
Collapse
MESH Headings
- Biopsy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line
- Cell Line, Tumor
- Chemokine CXCL1
- Chemokine CXCL2
- Chemokines, CXC/analysis
- Chemokines, CXC/genetics
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Interleukin-8
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Neovascularization, Pathologic/genetics
- Protein Processing, Post-Translational
- Receptors, Interleukin-8A
- Receptors, Interleukin-8B
Collapse
Affiliation(s)
- Anne-Marie Baird
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Steven G. Gray
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | - Kenneth J. O'Byrne
- Thoracic Oncology Research Group, Institute of Molecular Medicine, Trinity College, Dublin, Ireland
- HOPE Directorate, St. James's Hospital, Dublin, Ireland
- * E-mail:
| |
Collapse
|
236
|
Pedersen L, Pilegaard H, Hansen J, Brandt C, Adser H, Hidalgo J, Olesen J, Pedersen BK, Hojman P. Exercise-induced liver chemokine CXCL-1 expression is linked to muscle-derived interleukin-6 expression. J Physiol 2011; 589:1409-20. [PMID: 21224226 DOI: 10.1113/jphysiol.2010.200733] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The chemokine CXC ligand-1 (CXCL-1) is a small cytokine that elicits effects by signalling through the chemokine receptor CXCR2. CXCL-1 has neutrophil chemoattractant activity, is involved in the processes of angiogenesis, inflammation and wound healing, and may possess neuroprotective effects. The aim of this study was to unravel the mechanisms whereby CXCL-1 is regulated by exercise inmice. After a single bout of exercise, CXCL-1 protein increased in serum(2.4-fold), and CXCL-1 mRNA in muscle (6.5-fold) and liver (41-fold). These increases in CXCL-1 were preceded by increases in serum interleukin-6 (IL-6) and muscle IL-6 mRNA. In contrast, exercise-induced regulation of liver CXCL-1 mRNA expression was completely blunted in IL-6 knockout mice. Based on these findings, we examined the possible existence of a muscle-to-liver axis by overexpressing IL-6 in muscles. This resulted in increases in serum CXCL-1 (5-fold) and liver CXCL-1 mRNA expression (24-fold) compared with control. Because IL-6 expression and release are known to be augmented during exercise in glycogen-depleted animals, CXCL-1 and IL-6 expression were examined after exercise in overnight-fasted mice.We found that fasting significantly augmented serum CXCL-1, and CXCL-1 expression in liver and muscle. Taken together, these data indicate that liver is the main source of serum CXCL-1 during exercise in mice, and that the CXCL-1 expression in the liver is regulated by muscle-derived IL-6.
Collapse
Affiliation(s)
- Line Pedersen
- Centre of Inflammation and Metabolism at Department of Infectious Diseases and Copenhagen Muscle Research Centre, Rigshospitalet and Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
Chemokines are a large group of small cytokines known for their chemotactic ability to regulate the recruitment of leukocytes to sites of inflammation. This occurs through the binding of chemokines to their receptors located on the leukocyte that results in cellular changes such as actin rearrangement and cell shape, which allow for the migration of the leukocyte. In addition to regulating leukocyte function, it is now becoming apparent that other nonhematopoetic cells, such as smooth muscle cells and endothelial cells, can also be regulated by chemokines. Studies within the past 10 years has demonstrated the presence of various chemokine receptors on endothelial cells as well as the ability of chemokines to activate these receptors resulting in various cellular responses including migration, proliferation, and cellular activation. The purpose of this review is to highlight the research that has been done to date demonstrating the important role for chemokines in regulating endothelial function during various inflammatory conditions associated with angiogenesis, homeostasis, and leukocyte transmigration. This review will focus specifically on the role of the endothelium in mediating chemokine effects associated with wound healing, atherosclerosis, and autoimmune diseases, conditions where leukocyte recruitment and angiogenesis play a major role. Recent progress in the development and implementation of therapeutics agents against these small molecules, or their receptors, will also be addressed.
Collapse
Affiliation(s)
- Cecilia L Speyer
- Department of Surgery, Wayne State University School of Medicine, Detroit, MI 48201, United States.
| | | |
Collapse
|
238
|
Abstract
Chemokines are a family of small heparin-binding proteins, mostly known for their role in inflammation and immune surveillance, which have emerged as important regulators of angiogenesis. Chemokines influence angiogenesis either through recruitment of pro-angiogenic immune cells and endothelial progenitors to the neo-vascular niche or via direct regulation of endothelial function downstream of activation of G-protein coupled chemokine receptors. The dual function of chemokines in regulating immune response and angiogenesis confers a central role in modulating the tissue microenvironment. Therefore, chemokines may constitute attractive targets for therapeutic intervention in several pathological disorders. This review will summarize the current understanding of the role of chemokines in angiogenesis, and give an overview of angiostatic and angiogenic chemokines and their crosstalk with other angiogenic factors.
Collapse
Affiliation(s)
- Anna Dimberg
- Department of Genetics and Pathology, Uppsala University, Rudbeck Laboratory, 75185 Uppsala, Sweden.
| |
Collapse
|
239
|
McMellen ME, Wakeman D, Erwin CR, Guo J, Warner BW. Epidermal growth factor receptor signaling modulates chemokine (CXC) ligand 5 expression and is associated with villus angiogenesis after small bowel resection. Surgery 2010; 148:364-70. [PMID: 20471049 PMCID: PMC2917230 DOI: 10.1016/j.surg.2010.03.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 03/25/2010] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adaptive villus growth after a massive small bowel resection (SBR) is an important response to the loss of intestinal surface area and is regulated via epidermal growth factor receptor (EGFR) signaling. Increased levels of the proangiogenic chemokine ligand 5 (CXCL5) have been found within the adapting bowel in which angiogenesis is increased. We sought to determine whether CXCL5 was expressed specifically in the villus mesenchymal zone (area of increased blood vessel growth) and whether this expression was affected by EGF. METHODS C57BL/6J mice were subjected to sham operation (bowel transaction with reanastomosis) or 50% proximal SBR. The remnant intestine was harvested, and the villus lamina propria was isolated by laser capture microdissection. The expression of CXCL5 messenger RNA (mRNA) was analyzed using real-time polymerase chain reaction (RT-PCR). Furthermore, CXCL5 mRNA levels were determined in EGF-stimulated human umbilical vein endothelial cells (HUVECs). RESULTS A 2.39-fold increase (P < .05) in CXCL5 mRNA occurred in the lamina propria after SBR. In addition, villus height was found to be related directly to the degree of CXCL5 mRNA (R(2) = 0.97) expression. HUVECs treated with EGF demonstrated a 9-fold increase in CXCL5 mRNA expression. CONCLUSION The villus growth observed in resection-induced adaptation is associated with increased expression of the chemokine CXCL5 within the lamina propria. Because EGF enhances CXCL5 expression directly in endothelial cells, EGFR-directed proangiogenic gene expression may be a critical mechanism for adaptive ileal villus growth.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Cells, Cultured
- Chemokine CXCL5/genetics
- Chemokine CXCL5/metabolism
- Epidermal Growth Factor/genetics
- Epidermal Growth Factor/metabolism
- ErbB Receptors/metabolism
- Gene Expression
- Humans
- Intestinal Mucosa/blood supply
- Intestinal Mucosa/metabolism
- Intestine, Small/blood supply
- Intestine, Small/metabolism
- Intestine, Small/surgery
- Male
- Mesoderm/blood supply
- Mesoderm/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neovascularization, Physiologic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Mark E. McMellen
- Division of Pediatric Surgery, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| | - Derek Wakeman
- Division of Pediatric Surgery, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| | - Christopher R. Erwin
- Division of Pediatric Surgery, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| | - Jun Guo
- Division of Pediatric Surgery, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| | - Brad W. Warner
- Division of Pediatric Surgery, St. Louis Children's Hospital, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
240
|
Flad HD, Brandt E. Platelet-derived chemokines: pathophysiology and therapeutic aspects. Cell Mol Life Sci 2010; 67:2363-86. [PMID: 20213276 PMCID: PMC11115602 DOI: 10.1007/s00018-010-0306-x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 01/28/2010] [Accepted: 02/05/2010] [Indexed: 02/05/2023]
Abstract
The identification of chemokines in blood platelets has strengthened our view of these cells as participants in immune host defense. Platelet chemokines representing prestored and rapidly releasable proteins may play a major role as first-line inflammatory mediators. This is evident from their capability to recruit early inflammatory cells such as neutrophil granulocytes and monocytes and even to exhibit direct antimicrobial activity. However, insight is growing that platelet chemokines may be also long-term regulators, e.g., by activating T lymphocytes, by modulating the formation of endothelium and even thrombocytopoiesis itself. This review deals with the individual and cooperative functionality of platelet chemokines, as well as their potential as a basis for therapeutic intervention in the pathology of inflammation, infection, allergy and tumors. Within this context, therapeutic strategies based on the use of antibodies, modified chemokines, chemokine-binding proteins and chemokine receptor antagonists as well as first clinical studies will be addressed.
Collapse
Affiliation(s)
- Hans-Dieter Flad
- Department of Immunology and Cell Biology, Research Center Borstel, Borstel, Germany.
| | | |
Collapse
|
241
|
Snoussi K, Mahfoudh W, Bouaouina N, Fekih M, Khairi H, Helal AN, Chouchane L. Combined effects of IL-8 and CXCR2 gene polymorphisms on breast cancer susceptibility and aggressiveness. BMC Cancer 2010; 10:283. [PMID: 20540789 PMCID: PMC2895614 DOI: 10.1186/1471-2407-10-283] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2009] [Accepted: 06/12/2010] [Indexed: 02/05/2023] Open
Abstract
Background Interleukin-8 (IL-8/CXCL-8) is a prototype of the ELR+CXC chemokines that play an important role in the promotion and progression of many human cancers including breast cancer. We have recently showed the implication of polymorphism (-251) T/A of IL-8 gene in the susceptibility and prognosis of breast carcinoma. IL-8 acts through its CXCR1 and CXCR2 receptors. CXCR2, expressed on the endothelial cells, is the receptor involved in mediating the angiogenic effects of ELR+CXC chemokines and in particular IL-8. In the current study, we investigated the susceptibility and prognostic implications of the genetic variation in CXCR2 in breast carcinoma. We also confirmed the implication of IL-8 (-251) T/A polymorphism in a larger cohort. Finally, we combined the IL-8 and CXCR2 variant alleles and analyzed their effects in breast cancer risk and prognosis. Methods We used the allele-specific polymerase chain reaction to characterize the variation of IL-8 and CXCR2 for 409 unrelated Tunisian patients with breast carcinoma and 301 healthy control subjects. To estimate the relative risks, Odds ratios and 95% confidence intervals were calculated using unconditional logistic regression after adjusting for the known risk factors for breast cancer. Associations of the genetic marker with the rates of breast carcinoma-specific overall survival and disease-free survival were assessed using univariate and multivariate analyses. Results A highly significant association was found between the homozygous CXCR2 (+ 1208) TT genotype (adjusted OR = 2.89; P = 0.008) and breast carcinoma. A significantly increased risk of breast carcinoma was associated with IL-8 (-251) A allele (adjusted OR = 1.86; P = 0.001). The presence of two higher risk genotypes (the TA and TT in IL-8, and the TT in CXCR2) significantly increased the risk of developing breast carcinoma (adjusted OR = 4.15; P = 0.0004). The CXCR2 (+ 1208) T allele manifested a significant association with an aggressive phenotype of breast carcinoma as defined by a large tumor size, a high histological grade, and auxiliary's lymph node metastasis. A significant association between the IL-8 (-251) A allele and the aggressive form of breast carcinoma was also found. Moreover, the presence of the IL-8 (-251) A and/or the CXCR2 (+ 1208) T allele showed a significant association with a decreased overall survival and disease-free survival in breast carcinoma patients. Conclusion Our results indicated that the polymorphisms in IL-8 and CXCR2 genes are associated with increased breast cancer risk, as well as disease progress, supporting our hypothesis for IL-8 and ELR+CXC chemokine receptor (CXCR2) involvement in breast cancer pathogenesis.
Collapse
Affiliation(s)
- Kaouther Snoussi
- Laboratoire d'Immuno-Oncologie Moléculaire, Faculté de Médecine de Monastir, Université de Monastir, Monastir 5019, Tunisia.
| | | | | | | | | | | | | |
Collapse
|
242
|
Lin CS, He PJ, Hsu WT, Wu MS, Wu CJ, Shen HW, Hwang CH, Lai YK, Tsai NM, Liao KW. Helicobacter pylori-derived Heat shock protein 60 enhances angiogenesis via a CXCR2-mediated signaling pathway. Biochem Biophys Res Commun 2010; 397:283-9. [PMID: 20580690 DOI: 10.1016/j.bbrc.2010.05.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Accepted: 05/19/2010] [Indexed: 12/18/2022]
Abstract
Helicobacter pylori is a potent carcinogen associated with gastric cancer malignancy. Recently, H. pylori Heat shock protein 60 (HpHSP60) has been reported to promote cancer development by inducing chronic inflammation and promoting tumor cell migration. This study demonstrates a role for HpHSP60 in angiogenesis, a necessary precursor to tumor growth. We showed that HpHSP60 enhanced cell migration and tube formation, but not cell proliferation, in human umbilical vein endothelial cells (HUVECs). HpHSP60 also indirectly promoted HUVEC proliferation when HUVECs were co-cultured with supernatants collected from HpHSP60-treated AGS or THP-1 cells. The angiogenic array showed that HpHSP60 dramatically induced THP-1 cells and HUVECs to produce the chemotactic factors IL-8 and GRO. Inhibition of CXCR2, the receptor for IL-8 and GRO, or downstream PLCbeta2/Ca2+-mediated signaling, significantly abolished HpHSP60-induced tube formation. In contrast, suppression of MAP K or PI3 K signaling did not affect HpHSP60-mediated tubulogenesis. These data suggest that HpHSP60 enhances angiogenesis via CXCR2/PLCbeta2/Ca2+ signal transduction in endothelial cells.
Collapse
Affiliation(s)
- Chen-Si Lin
- Department of Biological Science and Technology, National Chiao-Tung University, Hsin-Chu, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Doll D, Keller L, Maak M, Boulesteix AL, Siewert JR, Holzmann B, Janssen KP. Differential expression of the chemokines GRO-2, GRO-3, and interleukin-8 in colon cancer and their impact on metastatic disease and survival. Int J Colorectal Dis 2010; 25:573-81. [PMID: 20162422 DOI: 10.1007/s00384-010-0901-1] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/13/2010] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Chemotactic cytokines play a role in angiogenesis and attraction of immune cells. However, their contribution to tumor formation remains incompletely understood. In a previous transcriptome study, we identified a family of structurally related chemokines of the CXC-family to be specifically up-regulated in colorectal cancer. The aim of the present study was to investigate the regulation of their expression in colon cancer cells and to test the hypothesis that altered CXC-chemokine expression is related to critical clinical parameters, such as survival or metastasis formation. MATERIALS AND METHODS Expression levels of interleukin-8 (CXCL-8) and growth-related oncogenes 2 and 3 (GRO-2/CXCL-2 and GRO-3/CXCL-3) were quantified using qRT-PCR in 97 patients with completely resected colon carcinoma and correlated with clinical parameters. Moreover, 16 samples of normal mucosa, nine samples of benign adenoma, and 11 samples of liver metastasis were analyzed. Next, the regulation of chemokine expression in response to various stimuli was tested in colon cancer cell lines (HT29, HCT116, CaCO2). RESULTS Expression of GRO-2, GRO-3, and IL-8 was significantly increased in colon cancer as compared to normal colon tissue. Expression of GRO-2 and GRO-3 was already enhanced in premalignant adenomas, and GRO-3 was significantly down-regulated in liver metastasis as compared to the primary tumor. Importantly, expression of GRO-3 was significantly higher in patients with local versus systemic disease. Moreover, IL-8 expression was significantly associated to overall post-operative survival. Finally, all chemokines were strongly induced by IL-1alpha in the colon cancer cell lines tested, indicating a potential link to inflammatory processes. CONCLUSION In accordance with earlier findings, we report here a significantly increased expression of GRO-2, GRO-3, and IL-8 in colon carcinoma as compared to normal tissue. Furthermore, GRO-3 was related to metastasis formation, and IL-8 was associated with survival, suggesting a potential predictive power of these markers.
Collapse
Affiliation(s)
- Dietrich Doll
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, München, Germany.
| | | | | | | | | | | | | |
Collapse
|
244
|
Raman D, Sai J, Neel NF, Chew CS, Richmond A. LIM and SH3 protein-1 modulates CXCR2-mediated cell migration. PLoS One 2010; 5:e10050. [PMID: 20419088 PMCID: PMC2856662 DOI: 10.1371/journal.pone.0010050] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2010] [Accepted: 03/16/2010] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The chemokine receptor CXCR2 plays a pivotal role in migration of neutrophils, macrophages and endothelial cells, modulating several biological responses such as angiogenesis, wound healing and acute inflammation. CXCR2 is also involved in pathogenesis of chronic inflammation, sepsis and atherosclerosis. The ability of CXCR2 to associate with a variety of proteins dynamically is responsible for its effects on directed cell migration or chemotaxis. The dynamic network of such CXCR2 binding proteins is termed as "CXCR2 chemosynapse". Proteomic analysis of proteins that co-immunoprecipitated with CXCR2 in neutrophil-like dHL-60 cells revealed a novel protein, LIM and SH3 protein 1 (LASP-1), binds CXCR2 under both basal and ligand activated conditions. LASP-1 is an actin binding cytoskeletal protein, involved in the cell migration. METHODOLOGY/PRINCIPAL FINDINGS We demonstrate that CXCR2 and LASP-1 co-immunoprecipitate and co-localize at the leading edge of migrating cells. The LIM domain of LASP-1 directly binds to the carboxy-terminal domain (CTD) of CXCR2. Moreover, LASP-1 also directly binds the CTD of CXCR1, CXCR3 and CXCR4. Using a site-directed and deletion mutagenesis approach, Iso323-Leu324 of the conserved LKIL motif on CXCR2-CTD was identified as the binding site for LASP-1. Interruption of the interaction between CXCR2-CTD and LIM domain of LASP-1 by dominant negative and knock down approaches inhibited CXCR2-mediated chemotaxis. Analysis for the mechanism for inhibition of CXCR2-mediated chemotaxis indicated that LASP-1/CXCR2 interaction is essential for cell motility and focal adhesion turnover involving activation of Src, paxillin, PAK1, p130CAS and ERK1/2. CONCLUSIONS/SIGNIFICANCE We demonstrate here for the first time that LASP-1 is a key component of the "CXCR2 chemosynapse" and LASP-1 interaction with CXCR2 is critical for CXCR2-mediated chemotaxis. Furthermore, LASP-1 also directly binds the CTD of CXCR1, CXCR3 and CXCR4, suggesting that LASP-1 is a general mediator of CXC chemokine mediated chemotaxis. Thus, LASP-1 may serve as a new link coordinating the flow of information between chemokine receptors and nascent focal adhesions, especially at the leading edge. Thus the association between the chemokine receptors and LASP-1 suggests to the presence of a CXC chemokine receptor-LASP-1 pro-migratory module in cells governing the cell migration.
Collapse
Affiliation(s)
- Dayanidhi Raman
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Jiqing Sai
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Nicole F. Neel
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Catherine S. Chew
- Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia, United States of America
| | - Ann Richmond
- Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Veterans Affairs, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
245
|
Galzi JL, Hachet-Haas M, Bonnet D, Daubeuf F, Lecat S, Hibert M, Haiech J, Frossard N. Neutralizing endogenous chemokines with small molecules. Principles and potential therapeutic applications. Pharmacol Ther 2010; 126:39-55. [PMID: 20117133 PMCID: PMC7112609 DOI: 10.1016/j.pharmthera.2009.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 12/24/2009] [Indexed: 02/08/2023]
Abstract
Regulation of cellular responses to external stimuli such as hormones, neurotransmitters, or cytokines is achieved through the control of all steps of the complex cascade starting with synthesis, going through maturation steps, release, distribution, degradation and/or uptake of the signalling molecule interacting with the target protein. One possible way of regulation, referred to as scavenging or neutralization of the ligand, has been increasingly studied, especially for small protein ligands. It shows innovative potential in chemical biology approaches as well as in disease treatment. Neutralization of protein ligands, as for example cytokines or chemokines can lead to the validation of signalling pathways under physiological or pathophysiological conditions, and in certain cases, to the development of therapeutic molecules now used in autoimmune diseases, chronic inflammation and cancer treatment. This review explores the field of ligand neutralization and tries to determine to what extent small chemical molecules could substitute for neutralizing antibodies in therapeutic approaches.
Collapse
Affiliation(s)
- Jean-Luc Galzi
- IREBS, FRE3211, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, 67412 Illkirch, France.
| | | | | | | | | | | | | | | |
Collapse
|
246
|
Onori P, Franchitto A, Mancinelli R, Carpino G, Alvaro D, Francis H, Alpini G, Gaudio E. Polycystic liver diseases. Dig Liver Dis 2010; 42:261-71. [PMID: 20138815 PMCID: PMC2894157 DOI: 10.1016/j.dld.2010.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Revised: 01/07/2010] [Accepted: 01/07/2010] [Indexed: 12/11/2022]
Abstract
Polycystic liver diseases (PCLDs) are genetic disorders with heterogeneous etiologies and a range of phenotypic presentations. PCLD exhibits both autosomal or recessive dominant pattern of inheritance and is characterized by the progressive development of multiple cysts, isolated or associated with polycystic kidney disease, that appear more extensive in women. Cholangiocytes have primary cilia, functionally important organelles (act as mechanosensors) that are involved in both normal developmental and pathological processes. The absence of polycystin-1, 2, and fibrocystin/polyductin, normally localized to primary cilia, represent a potential mechanism leading to cyst formation, associated with increased cell proliferation and apoptosis, enhanced fluid secretion, abnormal cell-matrix interactions, and alterations in cell polarity. Proliferative and secretive activities of cystic epithelium can be regulated by estrogens either directly or by synergizing growth factors including nerve growth factor, IGF1, FSH and VEGF. The abnormalities of primary cilia and the sensitivity to proliferative effects of estrogens and different growth factors in PCLD cystic epithelium provide the morpho-functional basis for future treatment targets, based on the possible modulation of the formation and progression of hepatic cysts.
Collapse
Affiliation(s)
- P. Onori
- Experimental Medicine, University of L'Aquila, L'Aquila, Italy
| | - A. Franchitto
- Dept Human Anatomy, University of Rome “La Sapienza”, Rome, Italy
| | - R. Mancinelli
- Dept Human Anatomy, University of Rome “La Sapienza”, Rome, Italy
| | - G. Carpino
- Dept Health Science, University of Rome “Foro Italico”, Italy
| | - D. Alvaro
- Gastroenterology, Polo Pontino, University of Rome “La Sapienza”, Rome, Italy
| | - H. Francis
- Research, Central Texas Veterans Health Care System, USA
| | - G. Alpini
- Research, Central Texas Veterans Health Care System, USA
- Scott & White Digestive Disease Research Center, Texas A&M Health Science Center, College of Medicine, USA
| | - E. Gaudio
- Dept Human Anatomy, University of Rome “La Sapienza”, Rome, Italy
| |
Collapse
|
247
|
Abstract
IMPORTANCE OF THE FIELD The incidence of malignant melanoma is increasing throughout the world and is currently rising faster than any other cancer in men and second only to lung cancer in women. Current strategies focused on systemic therapy for treatment of melanoma have shown no effect on survival. Therefore there is a pressing need for developing novel targeted therapeutics. AREAS COVERED IN THIS REVIEW Our goal is to provide an overview regarding targeting CXCR1/2 in malignant melanoma, the rationale behind these approaches and the future perspective. WHAT THE READER WILL GAIN This review illustrates our current understanding of CXCR1/2 receptor in melanoma progression and metastasis. We describe approaches that are being developed to block CXCR1/2 activation, including low-molecular-weight antagonists, modified chemokines and antibodies directed against ligands and receptors. TAKE HOME MESSAGE The chemokine receptors CXCR1 and CXCR2 and their ligands play an important role in the pathogenesis of malignant melanoma. Recent reports demonstrated that CXCR1 is constitutively expressed in all melanoma cases irrespective of stage and grade, however, CXCR2 expression was restricted to aggressive melanoma tumors,. Furthermore, modulation of CXCR1/2 expression and/or activity has been shown to regulate malignant melanoma growth, angiogenesis and metastasis, suggesting CXCR1/2 targeting as a novel therapeutic approach for malignant melanoma.
Collapse
Affiliation(s)
- Bhawna Sharma
- University of Nebraska Medical Center, Department of Pathology and Microbiology, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - Seema Singh
- University of South Alabama, Mitchell Cancer Institute, Department of Oncologic Sciences, Mobile, AL, USA
| | - Michelle L Varney
- University of Nebraska Medical Center, Department of Pathology and Microbiology, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, USA
| | - Rakesh K Singh
- University of Nebraska Medical Center, Department of Pathology and Microbiology, 985900 Nebraska Medical Center, Omaha, NE 68198-5900, USA
| |
Collapse
|
248
|
Lüttichau HR. The cytomegalovirus UL146 gene product vCXCL1 targets both CXCR1 and CXCR2 as an agonist. J Biol Chem 2010; 285:9137-46. [PMID: 20044480 PMCID: PMC2838333 DOI: 10.1074/jbc.m109.002774] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Large DNA viruses, such as herpesvirus and poxvirus, encode proteins that target and exploit the chemokine system of their host. UL146 and UL147 in the cytomegalovirus (CMV) genome encode the two CXC chemokines vCXCL1 and vCXCL2. In this study, vCXCL1 was probed against a panel of the 18 classified human chemokine receptors. In calcium mobilization assays vCXCL1 acted as an agonist on both CXCR1 and CXCR2 but did not activate or block any of the other 16 chemokine receptors. vCXCL1 was characterized and compared with CXCL1/GROalpha, CXCL2/GRObeta, CXCL3/GROgamma, CXCL5/ENA-78, CXCL6/GCP-2, CXCL7/NAP-2 and CXCL8/IL-8 in competition binding, calcium mobilization, inositol triphosphate turnover, and chemotaxis assays using CXCR1- and CXCR2-expressing Chinese hamster ovary, 300.19, COS7, and L1.2 cells. The affinities of vCXCL1 for the CXCR1 and CXCR2 receptors were 44 and 5.6 nm, respectively, as determined in competition binding against radioactively labeled CXCL8. In calcium mobilization, phosphatidylinositol turnover, and chemotaxis assays, vCXCL1 acted as a highly efficacious activator of both receptors, with a rather low potency for the CXCR1 receptor but comparable with CXCL5 and CXCL7. It is suggested that CMV uses the UL146 gene product expressed in infected endothelial cells to attract neutrophils by activating their CXCR1 and CXCR2 receptors, whereby neutrophils can act as carriers of the virus to uninfected endothelial cells. In that way a lasting pool of CMV-infected endothelial cells could be maintained.
Collapse
Affiliation(s)
- Hans R Lüttichau
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
249
|
Singh S, Singh AP, Sharma B, Owen LB, Singh RK. CXCL8 and its cognate receptors in melanoma progression and metastasis. Future Oncol 2010; 6:111-6. [PMID: 20021212 DOI: 10.2217/fon.09.128] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The incidence of melanoma is rising at an alarming rate and we are still awaiting an effective treatment for this malignancy. In its early stage, melanoma can be cured by surgical removal, but once metastasis has occurred there is no effective treatment. Recent findings have suggested multiple functional implications of CXCL8 and its cognate receptors, CXCR1 and CXCR2, in melanoma pathogenesis, thus underscoring their importance as targets for cancer therapy. This review provides an update on the roles of CXCL8 and its receptors in melanoma progression and metastasis.
Collapse
Affiliation(s)
- Seema Singh
- Department of Oncologic Sciences, Mitchell Cancer Institute, University of South Alabama, AL, USA
| | | | | | | | | |
Collapse
|
250
|
Gerber PA, Hippe A, Buhren BA, Müller A, Homey B. Chemokines in tumor-associated angiogenesis. Biol Chem 2010; 390:1213-23. [PMID: 19804363 DOI: 10.1515/bc.2009.144] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor growth is dependent on several key factors. Apart from immune escape and an efficient blockade of apoptotic signals, tumors require oxygen and nutrients to grow past a diameter of 2 microm. Therefore, it is of vital importance for the tumor to facilitate tumor-associated angiogenesis, e.g., the de novo formation of new blood vessels. In addition to established and key angiogenic factors, such as vascular endothelial growth factor, chemokines, a superfamily of cytokine-like proteins that bind to seven transmembrane-spanning G-protein-coupled receptors, have been associated with angiogenesis under homeostatic conditions. Chemokines were initially identified as key factors that control the directional migration of leukocytes, stem cells and cancer cells in vitro and which critically regulate their trafficking in vivo. Recently their role in establishing a favorable microenvironment for tumor-associated angiogenesis, a process that requires complex bidirectional interactions of the tumor and associated vessels, has been the focus of research. Chemokine-promoted angiogenesis not only facilitates tumor growth by supplying nutrients and oxygen but it is also a prerequisite to tumor metastasis. Hence, the pharmacologic control of tumor angiogenesis presents a promising strategy for novel anticancer therapeutics. Here, we discuss the current pathogenetic concepts of tumor-associated angiogenesis in the context of chemokines and their receptors and highlight promising therapeutic strategies.
Collapse
Affiliation(s)
- Peter Arne Gerber
- Department of Dermatology, University Hospital Düsseldorf, D-40225 Düsseldorf, Germany
| | | | | | | | | |
Collapse
|