Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 176
Filter
1.
Radiother Oncol ; 194: 110194, 2024 May.
Article in English | MEDLINE | ID: mdl-38447871

ABSTRACT

High precision, image-guided radiotherapy (RT) has increased the therapeutic ratio, enabling higher tumor and lower normal tissue doses, leading to improved patient outcomes. Nevertheless, some patients remain at risk of developing serious side effects.In many clinical situations, the radiation tolerance of normal tissues close to the target volume limits the dose that can safely be delivered and thus the potential for tumor control and cure. This is particularly so in patients being re-treated for tumor progression or a second primary tumor within a previous irradiated volume, scenarios that are becoming more frequent in clinical practice.Various normal tissue 'radioprotective' drugs with the potential to reduce side effects have been studied previously. Unfortunately, most have failed to impact clinical practice because of lack of therapeutic efficacy, concern about concurrent tumor protection or excessive drug-related toxicity. This review highlights the evidence indicating that targeting the CXCL12/CXCR4 pathway can mitigate acute and late RT-induced injury and reduce treatment side effects in a manner that overcomes these previous translational challenges. Pre-clinical studies involving a broad range of normal tissues commonly affected in clinical practice, including skin, lung, the gastrointestinal tract and brain, have shown that CXCL12 signalling is upregulated by RT and attracts CXCR4-expressing inflammatory cells that exacerbate acute tissue injury and late fibrosis. These studies also provide convincing evidence that inhibition of CXCL12/CXCR4 signalling during or after RT can reduce or prevent RT side effects, warranting further evaluation in clinical studies. Greater dialogue with the pharmaceutical industry is needed to prioritize the development and availability of CXCL12/CXCR4 inhibitors for future RT studies.


Subject(s)
Chemokine CXCL12 , Neoplasms , Radiation Injuries , Radiation-Protective Agents , Signal Transduction , Animals , Humans , Chemokine CXCL12/metabolism , Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Radiation Tolerance/drug effects , Radiation-Protective Agents/pharmacology , Radiation-Protective Agents/therapeutic use , Radiotherapy, Image-Guided/methods , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Signal Transduction/drug effects , Chemokines, CXC/antagonists & inhibitors
2.
Int Immunopharmacol ; 97: 107681, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33932697

ABSTRACT

Cancer is a leading cause of death worldwide and imposes a substantial financial burden. Therefore, it is essential to develop cost-effective approaches to inhibit tumor growth and development. The imbalance of cytokines and chemokines play an important role among different mechanisms involved in cancer development. One of the strongly conserved chemokines that is constitutively expressed in skin epithelia is the chemokine CXCL14. As a member of the CXC subfamily of chemokines, CXCL14 is responsible for the infiltration of immune cells, maturation of dendritic cells, upregulation of major histocompatibility complex (MHC)-I expression, and cell mobilization. Moreover, dysregulation of CXCL14 in several cancers has been identified by several studies. Depending on the type or origin of the tumor and components of the tumor microenvironment, CXCL14 plays a conflicting role in cancer. Although fibroblast-derived CXCL14 has a tumor-supportive role, epithelial-derived CXCL14 mainly inhibits tumor progression. Hence, this review will elucidate what is known on the mechanisms of CXCL14 and its therapeutic approaches in tumor treatment. CXCL14 is a promising approach for cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Chemokines, CXC/metabolism , Neoplasms/immunology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/agonists , Biomarkers, Tumor/analysis , Biomarkers, Tumor/antagonists & inhibitors , Chemokines, CXC/agonists , Chemokines, CXC/analysis , Chemokines, CXC/antagonists & inhibitors , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/genetics , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Up-Regulation
3.
Eur Rev Med Pharmacol Sci ; 23(9): 3821-3828, 2019 May.
Article in English | MEDLINE | ID: mdl-31115009

ABSTRACT

OBJECTIVE: Emerging evidence has indicated that microRNAs (miRNAs) play crucial roles in regulating cancer carcinogenesis; however, its role in oral squamous cell carcinoma (OSCC) remains largely unknown. Our work was aimed to investigate the role of miR-4513 in regulating OSCC cells behaviors. MATERIALS AND METHODS: MiR-4513 expression in OSCC cells was analyzed by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). Cell proliferation, migration, invasion, and apoptosis were analyzed by the cell counting kit-8 (CCK-8) assay, wound-healing assay, transwell invasion assay, and flow cytometry, respectively. The connections of miR-4513 and CXC ligand 17 (CXCL17) were analyzed by luciferase reporter assay and Western blot assay. RESULTS: MiR-4513 expression was found elevated in the OSCC cell lines. The downregulation of miR-4513 expression inhibits cell proliferation, migration, invasion and, at the same time, promotes apoptosis. Furthermore, we validated CXCL17 as a direct target of miR-4513. Knocking down the expression of CXCL17, inhibited the effects of miR-4513 on OSCC cell behaviors. CONCLUSIONS: Our results suggested the oncogenic role of miR-4513 in OSCC, and therefore it might be used as a target for the OSCC treatment.


Subject(s)
Apoptosis , Cell Proliferation , Chemokines, CXC/metabolism , MicroRNAs/metabolism , Antagomirs/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Mutagenesis , RNA Interference , RNA, Small Interfering/metabolism
4.
Circ Res ; 124(8): 1253-1265, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30739581

ABSTRACT

RATIONALE: Regeneration of denuded or injured endothelium is an important component of vascular injury response. Cell-cell communication between endothelial cells and smooth muscle cells (SMCs) plays a critical role not only in vascular homeostasis but also in disease. We have previously demonstrated that PKCδ (protein kinase C-delta) regulates multiple components of vascular injury response including apoptosis of SMCs and production of chemokines, thus is an attractive candidate for a role in SMC-endothelial cells communication. OBJECTIVE: To test whether PKCδ-mediated paracrine functions of SMCs influence reendothelialization in rodent models of arterial injury. METHODS AND RESULTS: Femoral artery wire injury was performed in SMC-conditional Prkcd knockout mice, and carotid angioplasty was conducted in rats receiving transient Prkcd knockdown or overexpression. SMC-specific knockout of Prkcd impaired reendothelialization, reflected by a smaller Evans blue-excluding area in the knockout compared with the wild-type controls. A similar impediment to reendothelialization was observed in rats with SMC-specific knockdown of Prkcd. In contrast, SMC-specific gene transfer of Prkcd accelerated reendothelialization. In vitro, medium conditioned by AdPKCδ-infected SMCs increased endothelial wound closure without affecting their proliferation. A polymerase chain reaction-based array analysis identified Cxcl1 and Cxcl7 among others as PKCδ-mediated chemokines produced by SMCs. Mechanistically, we postulated that PKCδ regulates Cxcl7 expression through STAT3 (signal transducer and activator of transcription 3) as knockdown of STAT3 abolished Cxcl7 expression. The role of CXCL7 in SMC-endothelial cells communication was demonstrated by blocking CXCL7 or its receptor CXCR2, both significantly inhibited endothelial wound closure. Furthermore, insertion of a Cxcl7 cDNA in the lentiviral vector that carries a Prkcd shRNA overcame the adverse effects of Prkcd knockdown on reendothelialization. CONCLUSIONS: SMCs promote reendothelialization in a PKCδ-dependent paracrine mechanism, likely through CXCL7-mediated recruitment of endothelial cells from uninjured endothelium.


Subject(s)
Endothelial Cells/metabolism , Myocytes, Smooth Muscle/metabolism , Paracrine Communication/physiology , Protein Kinase C-delta/metabolism , Regeneration/genetics , Vascular System Injuries/metabolism , Animals , Apoptosis/physiology , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Chemokine CXCL1/biosynthesis , Chemokines/biosynthesis , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Femoral Artery/injuries , Gene Knockout Techniques , Mice , Mice, Transgenic , Protein Kinase C-delta/genetics , Receptors, Interleukin-8B/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Vascular System Injuries/physiopathology , Wound Healing
5.
World J Gastroenterol ; 24(42): 4738-4749, 2018 Nov 14.
Article in English | MEDLINE | ID: mdl-30479461

ABSTRACT

Colorectal cancer (CRC) is the third most common cancer in men and the second most common cancer in women, worldwide. In the early stages of the disease, biomarkers predicting early relapse would improve survival rates. In metastatic patients, the use of predictive biomarkers could potentially result in more personalized treatments and better outcomes. The CXC family of chemokines (CXCL1 to 17) are small (8 to 10 kDa) secreted proteins that attract neutrophils and lymphocytes. These chemokines signal through chemokine receptors (CXCR) 1 to 8. Several studies have reported that these chemokines and receptors have a role in either the promotion or inhibition of cancer, depending on their capacity to suppress or stimulate the action of the immune system, respectively. In general terms, activation of the CXCR1/CXCR2 pathway or the CXCR4/CXCR7 pathway is associated with tumor aggressiveness and poor prognosis; therefore, the specific inhibition of these receptors is a possible therapeutic strategy. On the other hand, the lesser known CXCR3 and CXCR5 axes are generally considered to be tumor suppressor signaling pathways, and their stimulation has been suggested as a way to fight cancer. These pathways have been studied in tumor tissues (using immunohistochemistry or measuring mRNA levels) or serum [using enzyme-linked immuno sorbent assay (ELISA) or multiplexing techniques], among other sample types. Common variants in genes encoding for the CXC chemokines have also been investigated as possible biomarkers of the disease. This review summarizes the most recent findings on the role of CXC chemokines and their receptors in CRC and discusses their possible value as prognostic or predictive biomarkers as well as the possibility of targeting them as a therapeutic strategy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Chemokines, CXC/metabolism , Colorectal Neoplasms/pathology , Neoplasm Recurrence, Local/diagnosis , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/immunology , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/immunology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Humans , Prognosis , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/immunology , Receptors, CXCR/metabolism , Signal Transduction/drug effects , Survival Rate
6.
Cell Physiol Biochem ; 45(5): 1731-1746, 2018.
Article in English | MEDLINE | ID: mdl-29495008

ABSTRACT

BACKGROUND/AIMS: Breast cancer is a common cause of cancer mortality throughout the world. The cross-talk between cancer cells and interstitial cells exerts significant effects on neoplasia and tumor development and is modulated in part by chemokines. CXC is one of four chemokine families involved in mediating survival, angiogenesis, and immunosensitization by chemoattracting leukocytes, and it incentivizes tumor cell growth, invasion and metastasis in the tumor microenvironment. However, the differential expression profiles and prognostic values of these chemokines remains to be elucidated. METHODS: In this study, we compared transcriptional CXC chemokines and survival data of patients with breast carcinoma (BC) using the ONCOMINE dataset, Kaplan-Meier Plotter, TCGA and cBioPortal. RESULTS: We discovered increased mRNA levels for CXCL8/10/11/16/17, whereas mRNA expression of CXCL1/2/3/4/5/6/7/12/14 was lower in BC patients compared to non-tumor tissues. Kaplan-Meier plots revealed that high mRNA levels of CXCL1/2/3/4/5/6/7/12/14 correlate with relapse-free survival (RFS) in all types of BC patients. Conversely, high CXCL8/10/11 predicted worse RFS in BC patients. Significantly, high transcription levels of CXCL9/12/13/14 conferred an overall survival (OS) advantage in BC patients, while high levels of CXCL8 demonstrated shorter OS in all BC sufferers. CONCLUSIONS: Integrative bioinformatics analysis suggests that CXCL8/12/14 are potential suitable targets for precision therapy in BC patients compared to other CXC chemokines.


Subject(s)
Chemokines, CXC/metabolism , Computational Biology/methods , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Chemokine CXCL9/genetics , Chemokine CXCL9/metabolism , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Databases, Factual , Disease-Free Survival , Female , Gene Regulatory Networks , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Kaplan-Meier Estimate , Prognosis , RNA, Messenger/metabolism , Tumor Microenvironment
7.
Eur J Pharmacol ; 823: 1-10, 2018 Mar 15.
Article in English | MEDLINE | ID: mdl-29378189

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease that is characterized by uncontrolled joint inflammation and destruction of bone and cartilage. Previous studies have shown that C-X-C motif chemokine 10 (CXCL10) has important roles in RA development and that blocking CXCL10 expression effectively inhibits arthritis progression in animal models. However, clinical study using anti-CXCL10 monoclonal antibody (MDX-1100) to block CXCL10 expression in patients with RA did not show significant effectiveness. Therefore, we turned our attention to C-X-C motif chemokine receptor 3 (CXCR3), which is a receptor for CXCL9, CXCL10, and CXCL11, to treat RA. In the present study, administration of JN-2, our newly developed CXCR3 antagonist, ameliorated the progression of arthritis in a collagen-induced arthritis animal model. JN-2 also inhibited CXCR3-induced cell migration and pro-inflammatory cytokine expression of bone marrow-derived macrophages and CD4+ T cells in vitro. In addition, we found that CXCL10 formed an auto-amplification loop through activation of NFκB. Furthermore, Phosphorylation of p65 at serine 536 played an important role in the auto-amplification of CXCL10. Overall, the present results demonstrated that JN-2 decreased inflammation by inhibiting CXCR3-enhanced cell migration and pro-inflammatory cytokine expression, which then ameliorated arthritis progression.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Chemokines, CXC/antagonists & inhibitors , Disease Progression , Oxazoles/pharmacology , Animals , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cell Movement/drug effects , Chemokine CXCL10/metabolism , Cytokines/biosynthesis , Disease Models, Animal , Male , Mice , Oxazoles/therapeutic use , Transcription Factor RelA/metabolism
9.
mBio ; 7(3)2016 05 03.
Article in English | MEDLINE | ID: mdl-27143385

ABSTRACT

UNLABELLED: High-risk human papillomaviruses (HPVs) are causally associated with multiple human cancers. Previous studies have shown that the HPV oncoprotein E7 induces immune suppression; however, the underlying mechanisms remain unknown. To understand the mechanisms by which HPV deregulates host immune responses in the tumor microenvironment, we analyzed gene expression changes of all known chemokines and their receptors using our global gene expression data sets from human HPV-positive and -negative head/neck cancer and cervical tissue specimens in different disease stages. We report that, while many proinflammatory chemokines increase expression throughout cancer progression, CXCL14 is dramatically downregulated in HPV-positive cancers. HPV suppression of CXCL14 is dependent on E7 and associated with DNA hypermethylation in the CXCL14 promoter. Using in vivo mouse models, we revealed that restoration of Cxcl14 expression in HPV-positive mouse oropharyngeal carcinoma cells clears tumors in immunocompetent syngeneic mice, but not in Rag1-deficient mice. Further, Cxcl14 reexpression significantly increases natural killer (NK), CD4(+) T, and CD8(+) T cell infiltration into the tumor-draining lymph nodes in vivo In vitro transwell migration assays show that Cxcl14 reexpression induces chemotaxis of NK, CD4(+) T, and CD8(+) T cells. These results suggest that CXCL14 downregulation by HPV plays an important role in suppression of antitumor immune responses. Our findings provide a new mechanistic understanding of virus-induced immune evasion that contributes to cancer progression. IMPORTANCE: Human papillomaviruses (HPVs) are causally associated with more than 5% of all human cancers. During decades of cancer progression, HPV persists, evading host surveillance. However, little is known about the immune evasion mechanisms driven by HPV. Here we report that the chemokine CXCL14 is significantly downregulated in HPV-positive head/neck and cervical cancers. Using patient tissue specimens and cultured keratinocytes, we found that CXCL14 downregulation is linked to CXCL14 promoter hypermethylation induced by the HPV oncoprotein E7. Restoration of Cxcl14 expression in HPV-positive cancer cells clears tumors in immunocompetent syngeneic mice, but not in immunodeficient mice. Mice with Cxcl14 reexpression show dramatically increased natural killer and T cells in the tumor-draining lymph nodes. These results suggest that epigenetic downregulation of CXCL14 by HPV plays an important role in suppressing antitumor immune responses. Our findings may offer novel insights to develop preventive and therapeutic tools for restoring antitumor immune responses in HPV-infected individuals.


Subject(s)
Chemokines, CXC/antagonists & inhibitors , Down-Regulation , Epigenesis, Genetic , Host-Pathogen Interactions , Immune Evasion , Papillomaviridae/pathogenicity , Papillomavirus Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , DNA Methylation , Disease Models, Animal , Female , Head and Neck Neoplasms/pathology , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Mice , Papillomavirus E7 Proteins/metabolism , Promoter Regions, Genetic , Uterine Cervical Neoplasms/pathology
10.
J Virol ; 90(7): 3366-84, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26764004

ABSTRACT

UNLABELLED: Previous studies in our laboratory have identified equine CXCL16 (EqCXCL16) to be a candidate molecule and possible cell entry receptor for equine arteritis virus (EAV). In horses, the CXCL16 gene is located on equine chromosome 11 (ECA11) and encodes a glycosylated, type I transmembrane protein with 247 amino acids. Stable transfection of HEK-293T cells with plasmid DNA carrying EqCXCL16 (HEK-EqCXCL16 cells) increased the proportion of the cell population permissive to EAV infection from <3% to almost 100%. The increase in permissiveness was blocked either by transfection of HEK-EqCXCL16 cells with small interfering RNAs (siRNAs) directed against EqCXCL16 or by pretreatment with guinea pig polyclonal antibody against EqCXCL16 protein (Gp anti-EqCXCL16 pAb). Furthermore, using a virus overlay protein-binding assay (VOPBA) in combination with far-Western blotting, gradient-purified EAV particles were shown to bind directly to the EqCXCL16 protein in vitro. The binding of biotinylated virulent EAV strain Bucyrus at 4°C was significantly higher in HEK-EqCXCL16 cells than nontransfected HEK-293T cells. Finally, the results demonstrated that EAV preferentially infects subpopulations of horse CD14(+) monocytes expressing EqCXCL16 and that infection of these cells is significantly reduced by pretreatment with Gp anti-EqCXCL16 pAb. The collective data from this study provide confirmatory evidence that the transmembrane form of EqCXCL16 likely plays a major role in EAV host cell entry processes, possibly acting as a primary receptor molecule for this virus. IMPORTANCE: Outbreaks of EVA can be a source of significant economic loss for the equine industry from high rates of abortion in pregnant mares, death in young foals, establishment of the carrier state in stallions, and trade restrictions imposed by various countries. Similar to other arteriviruses, EAV primarily targets cells of the monocyte/macrophage lineage, which, when infected, are believed to play a critical role in EVA pathogenesis. To this point, however, the host-specified molecules involved in EAV binding and entry into monocytes/macrophages have not been identified. Identification of the cellular receptors for EAV may provide insights to design antivirals and better prophylactic reagents. In this study, we have demonstrated that EqCXCL16 acts as an EAV entry receptor in EAV-susceptible cells, equine monocytes. These findings represent a significant advance in our understanding of the fundamental mechanisms associated with the entry of EAV into susceptible cells.


Subject(s)
Chemokines, CXC/physiology , Equartevirus/physiology , Host Specificity/genetics , Receptors, Virus/genetics , Virus Internalization , Amino Acid Sequence , Animals , Antibodies, Viral/immunology , Arterivirus Infections/virology , Base Sequence , Cell Line , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Cricetinae , Equartevirus/genetics , Guinea Pigs , HEK293 Cells , Horse Diseases/virology , Horses , Humans , RNA Interference , RNA, Small Interfering/genetics , Rabbits , Receptors, Virus/metabolism , Sequence Analysis, DNA , Virus Attachment
11.
Neurosci Lett ; 606: 117-22, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26320021

ABSTRACT

Inflammation-related cerebral damage mediated by infiltrating neutrophils following reperfusion plays a role in reperfusion-induced brain damage subsequent to a stroke event. The ELR-CXC family of chemokines are CXCR1 and CXCR2 agonists that are known to drive neutrophil migration and activation. The present study demonstrated the benefit of anti-inflammatory therapy in the treatment of ischemic stroke with the administration of the competitive ELR-CXC chemokine antagonist, CXCL8(3-72)K11R/G31P (G31P). Male Sprague-Dawley rats were anaesthetized, and the middle cerebral artery (MCA) was occluded for 30 min followed by 5.5 h of reperfusion. Pretreatment with G31P resulted in a significant, dose-dependent (approximately 61-72%) decrease in infarct volumes compared to vehicle-treated animals, but neuroprotection was also observed when G31P (0.5 mg/kg) was administered 1 or 3 h following the start of reperfusion. The neuroprotection observed following the administration of this competitive CXCR1/CXCR2 antagonist may present therapeutic opportunities for addressing reperfusion-induced inflammatory damage in patients presenting with transient ischemic episodes.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Chemokines, CXC/antagonists & inhibitors , Interleukin-8/therapeutic use , Neuroprotective Agents/therapeutic use , Peptide Fragments/therapeutic use , Stroke/drug therapy , Animals , Anti-Inflammatory Agents/pharmacology , Brain/blood supply , Brain/drug effects , Brain/pathology , Brain Infarction/drug therapy , Brain Infarction/pathology , Brain Ischemia/etiology , Brain Ischemia/immunology , Brain Ischemia/pathology , Chemokines, CXC/immunology , Infarction, Middle Cerebral Artery/complications , Interleukin-8/pharmacology , Male , Neuroprotective Agents/pharmacology , Peptide Fragments/pharmacology , Rats, Sprague-Dawley , Stroke/etiology , Stroke/immunology , Stroke/pathology
12.
Pulm Pharmacol Ther ; 31: 55-62, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25681618

ABSTRACT

Inhalation of agricultural occupational dusts from swine confinement facilities can result in lung inflammation. The innate immune response to organic barn dusts results in production of a number of pro-inflammatory factors in the lungs of barn workers such as cytokines, chemokines, and an influx of neutrophils. Many of these inflammatory factors are influenced by the chemokine CXCL8/IL-8 (KC or MIP-2 in mice). Previously, we have demonstrated that an endotoxin-independent component of swine barn dust extract (SBE) elevates lung chemokines in a protein kinase C (PKC)-dependent manner resulting in the significant formation of lung inflammatory cell infiltrates in a mouse model of SBE injury. In this study we test the ability of a CXCR1/CXCR2 antagonist, CXCL8(3-74)K11R/G31P (G31P) to block many of the features of lung-inflammation in response to challenge with SBE in an established mouse exposure system. Injection of G31P concurrent with SBE nasal instillation over a course of 3 weeks significantly reduced neutrophil accumulation in the lungs of barn dust exposed animals compared to those given SBE alone. There was a similar reduction in pro-inflammatory cytokines and chemokines IL-6, KC, and MIP-2 in SBE plus G31P-treated mice. In addition to excreted products, the receptors ICAM-1, CXCR1, and CXCR2, which all were elevated with SBE exposure, were also decreased with G31P treatment. SBE activation of PKCα and PKCε was reduced as well with G31P treatment. Thus, G31P was found to be highly effective at reducing several features of lung inflammation in mice exposed to barn dust extracts.


Subject(s)
Chemokines, CXC/antagonists & inhibitors , Inflammation/physiopathology , Interleukin-8/pharmacology , Peptide Fragments/pharmacology , Animal Husbandry , Animals , Bronchoalveolar Lavage Fluid/immunology , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Dust , Inflammation/immunology , Inflammation Mediators/metabolism , Mice , Occupational Diseases/physiopathology , Protein Kinase C/metabolism , Swine
13.
Biomol Concepts ; 5(2): 167-73, 2014 May.
Article in English | MEDLINE | ID: mdl-25372750

ABSTRACT

CXCL12 and CXCL14 are evolutionarily conserved members of the CXC-type chemokine family. CXCL12 binds specifically to the G-protein-coupled receptor CXCR4 to induce the migration of primordial germ cells, hematopoietic stem cells, and inflammation-associated immune cells. In addition, CXCL12-CXCR4 signaling is often enhanced in malignant tumor cells and facilitates increased proliferation as well as metastasis. Although macrophage migration inhibitory factor and extracellular ubiquitin interact with CXCR4 as agonistic factors, CXCL12 was believed to be the sole chemokine ligand for CXCR4. However, a very recent report revealed that CXCL14 binds to CXCR4 with high affinity and efficiently inhibits CXCL12-mediated chemotaxis of hematopoietic progenitor and leukemia-derived cells. CXCL14 does not directly cross-compete with CXCL12 for the CXCR4 binding but instead inactivates CXCR4 via receptor internalization. Because both CXCL12 and CXCL14 are expressed during embryogenesis and brain development in mice, these two chemokines could function in an interactive fashion. We propose that the CXCL14 gene has been conserved from fish to man due to its role in fine-tuning the strength of CXCL12-mediated signal transduction. In addition to its biological implications, the above finding will be important for designing anti-cancer compounds targeting the CXCL12-CXCR4 signaling axis. In fact, a stabilized dimeric peptide containing the C-terminal 51-77 amino acid residues of CXCL14 has been shown to have stronger CXCL12 antagonistic activity than full-length CXCL14.


Subject(s)
Chemokine CXCL12/metabolism , Chemokines, CXC/metabolism , Receptors, CXCR4/metabolism , Animals , Chemokine CXCL12/antagonists & inhibitors , Chemokines, CXC/antagonists & inhibitors , Chemotaxis , Homeostasis , Humans , Neoplasms/metabolism , Protein Binding , Signal Transduction
14.
PLoS One ; 9(6): e99056, 2014.
Article in English | MEDLINE | ID: mdl-24897301

ABSTRACT

Despite advances in early diagnosis and multimodality therapy for cancers, most of lung cancer patients have been locally advanced or metastatic at the time of diagnosis, suggesting the highly progressive characteristic of lung cancer cells. The mechanisms underling invasiveness and metastasis of lung cancer are yet to be elucidated. In the present study, immunohistochemistry was performed to detect the expression of CXCL16-CXCR6 in human lung cancer tissues. It was demonstrated that similar to CXCL12 and CXCR4, CXCL16 and CXCR6 were also coexpressed in human primary lung cancer tissues. After confirming the functional existence of CXCL16 and CXCR6 protein in A549, 95D and H292 cells by ELSA and flow cytometry analysis, we further explored the significance of CXCL16-CXCR6 axis in the biological functions of lung cancer cell lines in vitro. It was found that CXCL16 had no effects on the PCNA (proliferating cell nuclear antigen) expression of A549, 95D and H292 cells. However, both exogenous CXCL16 and CM (conditioned medium from A549, 95D or H292) significantly improved the in vitro viability and invasion of three lung cancer cell lines. The neutralizing antibody to CXCL16 or down-regulation of CXCR6 was able to inhibit the increased viability and invasiveness of A549, 95D and H292 cells stimulated by CXCL16 or CM. Our results imply that CXCL16-CXCR6 axis is involved in the regulation of viability and invasion rather than PCNA expression of lung caner cells, which opens the door for better understanding the mechanisms of lung tumor progression and metastasis.


Subject(s)
Adenocarcinoma, Bronchiolo-Alveolar/pathology , Carcinoma, Adenosquamous/pathology , Carcinoma, Squamous Cell/pathology , Cell Movement , Chemokines, CXC/metabolism , Lung Neoplasms/pathology , Receptors, Chemokine/metabolism , Receptors, Scavenger/metabolism , Receptors, Virus/metabolism , Adenocarcinoma, Bronchiolo-Alveolar/genetics , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Apoptosis , Blotting, Western , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Proliferation , Chemokine CXCL16 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoenzyme Techniques , In Vitro Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Neoplasm Invasiveness , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR6 , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/genetics , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/genetics , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
15.
Curr Cancer Drug Targets ; 14(3): 294-309, 2014.
Article in English | MEDLINE | ID: mdl-24605943

ABSTRACT

Secreted proteins are an attractive minefield for cancer drug targets. An iTRAQ-based tandem mass spectrometry approach was employed to relatively quantify proteins in the secretomes of four isogenic breast cancer cell lines with increasing metastatic potential. CXCL3 was found to be upregulated in aggressive cancer cells. SiRNA and antibody neutralization studies supported a role of CXCL3 in metastatic processes. Meta-analysis of the mRNA level of CXCL3 in 1881 breast tumors supported a role of CXCL3 in clinical breast cancer. Our results support a functional role of CXCL3 in breast cancer metastasis and as a viable target for cancer therapy.


Subject(s)
Breast Neoplasms/pathology , Chemokines, CXC/antagonists & inhibitors , Neoplasm Metastasis , Breast Neoplasms/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/physiology , Female , Humans , RNA, Messenger/genetics
16.
Oncol Rep ; 31(4): 1599-604, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24482222

ABSTRACT

Cisplatin (cis-diamminedichloroplatinum) is one of the most commonly used agents for the chemotherapy of various types of cancers, but its use is limited by its dose-dependent side-effects (e.g., nephrotoxicity). The ELR-CXC chemokines are potent tumor growth, metastatic and angiogenic factors and can foster tumor resistance to chemotherapeutic agents. They are also potent proinflammatory agents. The aim of the present study was to evaluate the added effects of combining cisplatin chemotherapy with ELR-CXC chemokine antagonism in a mouse H22 hepatoma cancer cell model. The mice were injected with tumor cells and were then treated with cisplatin (12.5 or 2 mg/kg doses), either alone or together with the chemokine antagonist CXCL8(3-72)K11R/G31P (G31P) (50 µg/kg). At varying time-points renal function was examined using blood urea nitrogen (BUN) and serum creatinine (SCr) as read-outs for the toxic effects of cisplatin, while tumor growth and metastasis were assessed as endpoints. High-dose cisplatin therapy reduced the tumor burden by 52%, while co-delivery of G31P further augmented the tumor growth-suppressive effects of this dose of cisplatin to 71%; G31P by itself and low-dose cisplatin reduced the tumor burden by 19 and 39%, respectively. G31P also reduced the nephrotoxic effects of high-dose cisplatin to the effects observed in the low-dose cisplatin-treated animals. These data confirm the beneficial effects of combined cisplatin chemotherapy and ELR-CXC chemokine anatagonism in the context of both tumor progression and adverse side-effects.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chemokines, CXC/antagonists & inhibitors , Cisplatin/adverse effects , Neoplasms, Experimental/pathology , Animals , Carcinoma, Hepatocellular/pathology , Cisplatin/administration & dosage , Disease Models, Animal , Disease Progression , Female , Immunohistochemistry , Kidney/drug effects , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
17.
Curr Pharm Des ; 20(12): 1971-9, 2014.
Article in English | MEDLINE | ID: mdl-23844733

ABSTRACT

Chemokines are a family of chemotactic cytokines that play an essential role in leukocyte trafficking. Upregulation of both CC and CXC chemokines is a hallmark of the inflammatory and reparative response following myocardial infarction. Release of danger signals from dying cells and damaged extracellular matrix activates innate immune pathways that stimulate chemokine synthesis. Cytokineand chemokine-driven adhesive interactions between endothelial cells and leukocytes mediate extravasation of immune cells into the infarct. CXC chemokines (such as interleukin-8) are bound to glycosaminoglycans on the endothelial surface and activate captured neutrophils, inducing expression of integrins. CC chemokines (such as monocyte chemoattractant protein (MCP)-1) mediate recruitment of proinflammatory and phagocytotic mononuclear cells into the infarct. CC Chemokines may also regulate late infiltration of the healing infarct with inhibitory leukocytes that suppress inflammation and restrain the post-infarction immune response. Non-hematopoietic cells are also targeted by chemokines; in healing infarcts, the CXC chemokine Interferon-γ inducible Protein (IP)-10 exerts antifibrotic actions, inhibiting fibroblast migration. Another member of the CXC subfamily, Stromal cell-derived Factor (SDF)-1, may protect the infarcted heart by activating pro-survival signaling in cardiomyocytes, while exerting angiogenic actions through chemotaxis of endothelial progenitors. Several members of the chemokine family may be promising therapeutic targets to attenuate adverse remodeling in patients with myocardial infarction.


Subject(s)
Chemokines/metabolism , Drug Delivery Systems/trends , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Animals , Chemokine CXCL12/administration & dosage , Chemokines/antagonists & inhibitors , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/metabolism , Humans
18.
Endocr J ; 60(2): 137-47, 2013.
Article in English | MEDLINE | ID: mdl-23037942

ABSTRACT

Muscle contractile activity functions as a potent stimulus for acute interleukin (IL)-6 expression in working skeletal muscles. Recently, we established an "in vitro contraction model" using highly-developed contractile C2C12 myotubes by applying electric pulse stimulation (EPS). Herein, we characterize the effects of EPS-evoked contraction on IL-6 expression in contractile C2C12 myotubes. Both secretion and mRNA expression of IL-6 were significantly up-regulated by EPS in a frequency-dependent manner in contracting myotubes during a 24-h period, and the response was blunted by cyclosporine A, a calcineurin inhibitor. Longer time (~12h) was required for the induction of IL-6 after the initiation of EPS as compared to that of other contraction-inducible CXC chemokines such as CXCL1/KC, which were induced in less than 3 hours. Furthermore, these acute inducible CXC chemokines exhibited no autocrine effect on IL-6 expression. Importantly, contraction-dependent IL-6 up-regulation was markedly suppressed in the presence of high levels of glucose along with increased glycogen accumulations. Experimental manipulation of intracellular glycogen contents by modulating available glucose or pyruvate during a certain EPS period further established the suppressive effect of glycogen accumulations on contraction-induced IL-6 up-regulation, which appeared to be independent of calcineurin activity. We also document that EPS-evoked contractile activity improved insulin-responsiveness in terms of intracellular glycogen accumulations. Taken together, these data provide important insights into the regulation of IL-6 expression in response to contractile activity of muscle cells, which is difficult to examine using in vivo experimental techniques. Our present results thus expand the usefulness of our "in vitro contraction model".


Subject(s)
Glycogen/metabolism , Insulin Resistance , Interleukin-6/metabolism , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Up-Regulation , Animals , Calcineurin/metabolism , Calcineurin Inhibitors , Calcium Signaling/drug effects , Cell Line , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/metabolism , Electric Stimulation , Glucose/metabolism , Hyperglycemia/metabolism , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Immunosuppressive Agents/pharmacology , Insulin/metabolism , Insulin/pharmacology , Interleukin-6/genetics , Kinetics , Mice , Muscle Contraction/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/immunology , RNA, Messenger/metabolism , Up-Regulation/drug effects
19.
Am J Physiol Cell Physiol ; 303(9): C991-9, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22954799

ABSTRACT

Eryptosis, the suicidal erythrocyte death, leads to cell shrinkage and cell membrane scrambling with phosphatidylserine exposure at the cell surface. Eryptotic erythrocytes adhere to the vascular wall by binding of phosphatidylserine to the CXC chemokine ligand 16 (CXCL16). Stimulators of eryptosis include increased cytosolic Ca(2+) activity, energy depletion, and activation of ceramide-producing sphingomyelinase. The present study explored whether sphingomyelinase triggers erythrocyte adhesion to endothelial cells. To this end, human erythrocytes were exposed for 6 h to bacterial sphingomyelinase (1-10 mU/ml) and phosphatidylserine exposure was estimated from fluorescent annexin-V-binding, cell volume from forward scatter in FACS-analysis, erythrocyte adhesion to human umbilical vein endothelial cells (HUVEC) from trapping of labeled erythrocytes in a flow chamber under flow conditions at arterial shear rates, and CXCL16 protein abundance utilizing Western blotting and FACS analysis of fluorescent antibody binding. As a result, sphingomyelinase (≥1 mU/ml) triggered cell shrinkage, phosphatidylserine exposure and erythrocyte adhesion to HUVEC, effects blunted by Ca(2+) removal. Adhesion was significantly blunted by phosphatidylserine-coating annexin-V (5 µl/ml), following addition of neutralizing antibodies against endothelial CXCL16 (4 µg/ml) and following silencing of the CXCL16 gene with small interfering RNA. Pretreatment of HUVEC with sphingomyelinase upregulated CXCL16 protein abundance. Six hours pretreatment of HUVEC with sphingomyelinase (10 mU/ml) or C6-ceramide (50 µM) augmented erythrocyte adhesion following a 30-min treatment with Ca(2+) ionophore ionomycin (1 µM) or following energy depletion by 48-h glucose removal. Thus exposure to sphingomyelinase or C6-ceramide triggers eryptosis followed by phosphatidylserine- and CXCL16-sensitive adhesion of eryptotic erythrocytes to HUVEC.


Subject(s)
Apoptosis/drug effects , Erythrocytes/drug effects , Sphingomyelin Phosphodiesterase/pharmacology , Annexin A5/physiology , Antibodies, Neutralizing/pharmacology , Apoptosis/physiology , Calcium/pharmacology , Calcium Ionophores/pharmacology , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Size , Cells, Cultured , Ceramides/pharmacology , Chemokine CXCL16 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Chemokines, CXC/physiology , Erythrocytes/physiology , Gene Silencing , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Ionomycin/pharmacology , Phosphatidylserines/physiology , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/genetics , Receptors, Scavenger/physiology
20.
Am J Physiol Cell Physiol ; 302(4): C644-51, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22173866

ABSTRACT

Suicidal death of erythrocytes, or eryptosis, is characterized by cell shrinkage and cell membrane scrambling leading to phosphatidylserine exposure at the cell surface. Eryptosis is triggered by increase of cytosolic Ca2+ activity, which may result from treatment with the Ca2+ ionophore ionomycin or from energy depletion by removal of glucose. The present study tested the hypothesis that phosphatidylserine exposure at the erythrocyte surface fosters adherence to endothelial cells of the vascular wall under flow conditions at arterial shear rates and that binding of eryptotic cells to endothelial cells is mediated by the transmembrane CXC chemokine ligand 16 (CXCL16). To this end, human erythrocytes were exposed to energy depletion (for 48 h) or treated with the Ca2+ ionophore ionomycin (1 µM for 30 min). Phosphatidylserine exposure was quantified utilizing annexin-V binding, cell volume was estimated from forward scatter in FACS analysis, and erythrocyte adhesion to human vascular endothelial cells (HUVEC) was determined in a flow chamber model. As a result, both, ionomycin and glucose depletion, triggered eryptosis and enhanced the percentage of erythrocytes adhering to HUVEC under flow conditions at arterial shear rates. The adhesion was significantly blunted in the presence of erythrocyte phosphatidylserine-coating annexin-V (5 µl/ml), of a neutralizing antibody against endothelial CXCL16 (4 µg/ml), and following silencing of endothelial CXCL16 with small interfering RNA. The present observations demonstrate that eryptotic erythrocytes adhere to endothelial cells of the vascular wall in part by interaction of phosphatidylserine exposed at the erythrocyte surface with endothelial CXCL16.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Chemokines, CXC/metabolism , Endothelium, Vascular/metabolism , Erythrocytes/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Phosphatidylserines/metabolism , Receptors, Scavenger/metabolism , Annexin A5/metabolism , Antibodies/metabolism , Calcium Ionophores/metabolism , Cell Adhesion , Cell Death/physiology , Cell Size , Chemokine CXCL16 , Chemokines, CXC/antagonists & inhibitors , Chemokines, CXC/genetics , Cytosol/metabolism , Endothelium, Vascular/cytology , Erythrocyte Count , Erythrocytes/cytology , Gene Silencing , Glucose/deficiency , Human Umbilical Vein Endothelial Cells/cytology , Humans , Ionomycin/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/genetics , Rheology
SELECTION OF CITATIONS
SEARCH DETAIL
...