Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Mol Pain ; 19: 17448069231169373, 2023.
Article in English | MEDLINE | ID: mdl-36998150

ABSTRACT

BACKGROUND: Chemokine-mediated neuroinflammation plays an important role in the pathogenesis of neuropathic pain. The chemokine CC motif ligand 7 (CCL7) and its receptor CCR2 have been reported to contribute to neuropathic pain via astrocyte-microglial interaction in the spinal cord. Whether CCL7 in the trigeminal ganglion (TG) involves in trigeminal neuropathic pain and the involved mechanism remain largely unknown. METHODS: The partial infraorbital nerve transection (pIONT) was used to induce trigeminal neuropathic pain in mice. The expression of Ccl7, Ccr1, Ccr2, and Ccr3 was examined by real-time quantitative polymerase chain reaction. The distribution of CCL7, CCR2, and CCR3 was detected by immunofluorescence double-staining. The activation of extracellular signal-regulated kinase (ERK) was examined by Western blot and immunofluorescence. The effect of CCL7 on neuronal excitability was tested by whole-cell patch clamp recording. The effect of selective antagonists for CCR1, CCR2, and CCR3 on pain hypersensitivity was checked by behavioral testing. RESULTS: Ccl7 was persistently increased in neurons of TG after pIONT, and specific inhibition of CCL7 in the TG effectively relieved pIONT-induced orofacial mechanical allodynia. Intra-TG injection of recombinant CCL7 induced mechanical allodynia and increased the phosphorylation of ERK in the TG. Incubation of CCL7 with TG neurons also dose-dependently enhanced the neuronal excitability. Furthermore, pIONT increased the expression of CCL7 receptors Ccr1, Ccr2, and Ccr3. The intra-TG injection of the specific antagonist of CCR2 or CCR3 but not of CCR1 alleviated pIONT-induced orofacial mechanical allodynia and reduced ERK activation. Immunostaining showed that CCR2 and CCR3 are expressed in TG neurons, and CCL7-induced hyperexcitability of TG neurons was decreased by antagonists of CCR2 or CCR3. CONCLUSION: CCL7 activates ERK in TG neurons via CCR2 and CCR3 to enhance neuronal excitability, which contributes to the maintenance of trigeminal neuropathic pain. CCL7-CCR2/CCR3-ERK pathway may be potential targets for treating trigeminal neuropathic pain.


Subject(s)
Chemokine CCL7 , Extracellular Signal-Regulated MAP Kinases , Neuralgia , Trigeminal Neuralgia , Animals , Mice , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Chemokine CCL7/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Hyperalgesia/metabolism , Ligands , MAP Kinase Signaling System , Neuralgia/metabolism , Trigeminal Ganglion/metabolism , Trigeminal Neuralgia/metabolism , Receptors, CCR2/metabolism , Receptors, CCR3/metabolism
2.
Int Immunopharmacol ; 99: 107975, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34293712

ABSTRACT

Microglia are immune cells of the central nervous system that mediate neuroinflammation. It is widely known that microglia-mediated inflammation in the brain contribute to the widespread tissue damage and neurological deficits in traumatic brain injury (TBI). However, the mechanisms responsible for this inflammatory response remain elusive. Here, we investigated the role of astrocyte-derived chemokine (C-C motif) ligand 7 (CCL7) in microglial-controlled inflammation following TBI. Our results demonstrated that astrocyte-derived CCL7 induced microglial activation and the release of proinflammatory mediators in the cortex and serum of rats that underwent experimental TBI. Furthermore, CCL7 knockout improved microglia-controlled inflammation, brain morphology and neurological dysfunction following TBI. In vitro, CCL7-siRNA attenuated the LPS-induced expression of pro-inflammatory markers in the co-culture of microglia and astrocytes. Collectively, our findings uncover an important role for astrocyte-derived CCL7 in promoting microglia-mediated inflammation after TBI and suggests CCL7 could serve as a potential therapeutic strategy for attenuating TBI by inhibiting microglial activation.


Subject(s)
Astrocytes/metabolism , Brain Injuries, Traumatic/pathology , Chemokine CCL7/pharmacology , Microglia/drug effects , Neuroinflammatory Diseases/pathology , Animals , Brain/pathology , Brain Injuries, Traumatic/psychology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Chemokine CCL7/metabolism , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Male , Mice , Mice, Inbred C57BL , Motor Activity , Neuroinflammatory Diseases/psychology , Primary Cell Culture , RNA, Small Interfering/pharmacology , Recognition, Psychology/drug effects
3.
Nat Commun ; 11(1): 6119, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257678

ABSTRACT

The efficacy of checkpoint immunotherapy to non-small cell lung cancer (NSCLC) largely depends on the tumor microenvironment (TME). Here, we demonstrate that CCL7 facilitates anti-PD-1 therapy for the KrasLSL-G12D/+Tp53fl/fl (KP) and the KrasLSL-G12D/+Lkb1fl/fl (KL) NSCLC mouse models by recruiting conventional DC 1 (cDC1) into the TME to promote T cell expansion. CCL7 exhibits high expression in NSCLC tumor tissues and is positively correlated with the infiltration of cDC1 in the TME and the overall survival of NSCLC patients. CCL7 deficiency impairs the infiltration of cDC1 in the TME and the subsequent expansion of CD8+ and CD4+ T cells in bronchial draining lymph nodes and TME, thereby promoting tumor development in the KP mouse model. Administration of CCL7 into lungs alone or in combination with anti-PD-1 significantly inhibits tumor development and prolongs the survival of KP and KL mice. These findings suggest that CCL7 potentially serves as a biomarker and adjuvant for checkpoint immunotherapy of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Chemokine CCL7/metabolism , Chemokine CCL7/pharmacology , Immunity , Immunotherapy/methods , Lung Neoplasms/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Chemokine CCL7/deficiency , Chemokine CCL7/genetics , Chemokines/metabolism , Disease Models, Animal , Female , Genes, ras , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53
4.
J Immunol ; 195(9): 4306-18, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26401006

ABSTRACT

West Nile virus (WNV) is a re-emerging pathogen and the leading cause of epidemic encephalitis in the United States. Inflammatory monocytes are a critical component of the cellular infiltrate found in the CNS during WNV encephalitis, although the molecular cues involved in their migration are not fully understood. In mice, we previously showed that WNV infection induces a CCR2-dependent monocytosis that precedes monocyte migration into the CNS. Currently, the relative contribution of the CCR2 ligands, chemokines CCL2 and CCL7, in directing monocyte mobilization and leukocyte migration into the CNS is unclear. In this study, we demonstrate that, although both CCL2 and CCL7 are required for efficient monocytosis and monocyte accumulation in the CNS, only CCL7 deficiency resulted in increased viral burden in the brain and enhanced mortality. The enhanced susceptibility in the absence of CCL7 was associated with the delayed migration of neutrophils and CD8(+) T cells into the CNS compared with WT or Ccl2(-/-) mice. To determine whether CCL7 reconstitution could therapeutically alter the survival outcome of WNV infection, we administered exogenous CCL7 i.v. to WNV-infected Ccl7(-/-) mice and observed a significant increase in monocytes and neutrophils, but not CD8(+) T cells, within the CNS, as well as an enhancement in survival compared with Ccl7(-/-) mice treated with a linear CCL7 control peptide. Our experiments suggest that CCL7 is an important protective signal involved in leukocyte trafficking during WNV infection, and it may have therapeutic potential for the treatment of acute viral infections of the CNS.


Subject(s)
Cell Movement , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , Leukocytosis/metabolism , Monocytes/metabolism , West Nile Fever/metabolism , Animals , Brain/metabolism , Brain/virology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL7/genetics , Chemokine CCL7/pharmacology , Chlorocebus aethiops , Encephalitis, Viral/genetics , Encephalitis, Viral/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Host-Pathogen Interactions , Leukocytosis/genetics , Mice, Inbred C57BL , Mice, Knockout , Monocytes/drug effects , Neutrophils/drug effects , Neutrophils/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , West Nile Fever/genetics , West Nile Fever/virology , West Nile virus/physiology
5.
Nan Fang Yi Ke Da Xue Xue Bao ; 33(1): 86-92, 2013 Jan.
Article in Chinese | MEDLINE | ID: mdl-23353163

ABSTRACT

OBJECTIVE: To investigate the effect of monocyte chemotactic protein-3 (MCP-3) on the expressions of intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), tissue factor (TF, and tissue factor pathway inhibitor (TFPI) and cell apoptosis in human umbilical vein endothelial cells (HUVECs). METHODS: Cultured HUVECs were treated with MCP-3 at the optimal concentration determined previously 1 h after treatments with or without MCP-3 antibody (20 ng/ml), PI3K inhibitor, or LY-294002 (5 mmol/ml). The expressions of ICAM-1, VCAM-1, TF and TFPI were analyzed using RT-PCR and Western blot after the treatments. MCP-3 mRNA and protein expressions were detected in HUVECs exposed to 50 µg/ml ox-LDL for 24 h. The cell apoptosis and caspase-3 protein production in HUVECs treated with MCP-3 or with MCP-3 plus CCR2 antagonist for 24 h and 48 h were evaluated by flow cytometry and Western blotting. RESULTS: At the optimal concentration of 0.3 ng/ml, MCP-3 treatment for 24 h caused significantly increased ICAM-1, VCAM-1, and TF expressions with lowered expression of TFPI in HUVECs (P<0.05), and such effects were significantly inhibited by the application of MCP-3 antibody, PI3K inhibitor, or LY-294002 (P<0.05). Ox-LDL exposure significantly increased the expression of MCP-3 in HUVECs (P<0.05). HUVECs showed a significantly increased apoptosis rate after treatment with MCP-3 or with MCP-3 plus CCR2 antagonist (P<0.05), and the apoptosis rate increased significantly as the treatment time prolonged (P<0.05); caspase-3 protein expression in the cells showed a similar pattern of alterations following the treatments. CONCLUSION: ox-LDL can induce MCP-3 expression in HUVECs. MCP-3 induces apoptosis of HUVECs and significantly affects the cellular function partially through the PI3K signaling pathway.


Subject(s)
Apoptosis/drug effects , Chemokine CCL7/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Intercellular Adhesion Molecule-1/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Caspase 3/metabolism , Cell Adhesion , Cells, Cultured , Chromones/pharmacology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Lipoproteins/metabolism , Lipoproteins, LDL/pharmacology , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Receptors, CCR2/antagonists & inhibitors , Signal Transduction , Thromboplastin/metabolism
6.
Endocrinology ; 150(7): 3345-52, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19342461

ABSTRACT

Cumulus cells surround the oocyte and regulate the production and assembly of the extracellular matrix (ECM) around the cumulus-oocyte complex for its timely interaction with sperm in the oviduct. We recently found that C-C chemokines such as CCL2, CCL7, and CCL9 are produced and stimulate integrin-mediated ECM assembly in the postovulatory cumulus to protect eggs and that prostaglandin E(2)-EP2 signaling in the cumulus cells facilitates fertilization by suppressing this chemokine signaling, which otherwise results in fertilization failure by preventing sperm penetration through the cumulus ECM. However, it remains unknown as to what mechanisms underlie chemokine-induced cumulus ECM assembly. Here we report that inhibition of EP2 signaling or addition of CCL7 augments RhoA activation and induces the surface accumulation of integrin and the contraction of cumulus cells. Enhanced surface accumulation of integrin then stimulates the formation and assembly of fibronectin fibrils as well as induces cumulus ECM resistance to hyaluronidase and sperm penetration. These changes in the cumulus ECM as well as cell contraction are relieved by the addition of Y27632 or blebbistatin. These results suggest that chemokines induce integrin engagement to the ECM and consequent ECM remodeling through the RhoA/Rho kinase/actomyosin pathway, making the cumulus ECM barrier resistant to sperm penetration. Based on these results, we propose that prostaglandin E(2)-EP2 signaling negatively regulates chemokine-induced Rho/ROCK signaling in cumulus cells for successful fertilization.


Subject(s)
Cumulus Cells/physiology , Receptors, Prostaglandin E/physiology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Chemokine CCL7/pharmacology , Chemokines, CC/metabolism , Cumulus Cells/ultrastructure , Extracellular Matrix/metabolism , Female , Fertilization , Humans , Male , Mice , Receptors, Prostaglandin E, EP1 Subtype , Signal Transduction , Sperm-Ovum Interactions/drug effects
7.
Eur J Immunol ; 39(4): 1118-28, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19291700

ABSTRACT

The migration of monocytes to sites of inflammation is largely determined by their response to chemokines. Although the chemokine specificities and expression patterns of chemokine receptors are well defined, it is still a matter of debate how cells integrate the messages provided by different chemokines that are concomitantly produced in physiological or pathological situations in vivo. We present evidence for one regulatory mechanism of human monocyte trafficking. Monocytes can integrate stimuli provided by inflammatory chemokines in the presence of homeostatic chemokines. In particular, migration and cell responses could occur at much lower concentrations of the CCR2 agonists, in the presence of chemokines (CCL19 and CCL21) that per se do not act on monocytes. Binding studies on CCR2(+) cells showed that CCL19 and CCL21 do not compete with the CCR2 agonist CCL2. Furthermore, the presence of CCL19 or CCL21 could influence the degradation of CCL2 and CCL7 on cells expressing the decoy receptor D6. These findings disclose a new scenario to further comprehend the complexity of chemokine-based monocyte trafficking in a vast variety of human inflammatory disorders.


Subject(s)
Cell Movement/immunology , Chemotaxis, Leukocyte/immunology , Inflammation/immunology , Monocytes/immunology , Receptors, CCR2/immunology , Receptors, CCR7/immunology , Amino Acid Sequence , Cell Movement/drug effects , Chemokine CCL19/chemistry , Chemokine CCL19/immunology , Chemokine CCL19/pharmacology , Chemokine CCL2/immunology , Chemokine CCL2/pharmacology , Chemokine CCL21/chemistry , Chemokine CCL21/immunology , Chemokine CCL21/pharmacology , Chemokine CCL7/immunology , Chemokine CCL7/pharmacology , Chemotaxis, Leukocyte/drug effects , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Glycosaminoglycans/immunology , Glycosaminoglycans/metabolism , Humans , Inflammation/metabolism , Ligands , Molecular Sequence Data , Monocytes/drug effects , Monocytes/metabolism , Phosphorylation/immunology , Protein Structure, Tertiary , Receptors, CCR10/immunology , Receptors, CCR10/metabolism , Receptors, CCR2/agonists , Receptors, CCR2/chemistry , Receptors, CCR7/agonists , Receptors, CCR7/chemistry , Chemokine Receptor D6
8.
Exp Cell Res ; 315(2): 151-61, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19038247

ABSTRACT

Recent studies have demonstrated upregulation of monocyte chemoattractant protein-3 (MCP-3/CCL7) in fibrosis and have suggested that in addition to a major role in regulating leucocyte recruitment this chemokine may also promote extracellular matrix (ECM) overproduction by fibroblasts. In the present study we explore interplay between MCP-3 and transforming growth factor beta (TGFbeta), a potent profibrotic cytokine. We demonstrate that MCP-3 promotes activation of TGFbeta signalling pathways leading to increased type I collagen secretion. In addition we show that MCP-3 gene expression is stimulated by recombinant TGFbeta1, raising the possibility for synergy between these two mediators in the fibrotic microenvironment. Comparison of downstream signalling pathways that regulate collagen gene activation by both cytokines confirms the central role of MAPK pathway activation in mediating the effects of both factors. An additive effect of these two agonists was demonstrated by comparative microarray analysis for key TGFbeta regulated transcripts including PAI-1, OSF2 and IGFBP6. Together, our results confirm cross-talk between MCP-3 and TGFbeta that may be critical in the development of fibrosis.


Subject(s)
Chemokine CCL7/metabolism , Collagen/biosynthesis , Fibroblasts/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Chemokine CCL7/genetics , Chemokine CCL7/pharmacology , Collagen/genetics , Collagen Type I/biosynthesis , Collagen Type I/genetics , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Gene Expression/drug effects , Mice , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pteridines/pharmacology , RNA, Small Interfering/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...