Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Cell Mol Life Sci ; 79(3): 155, 2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35218410

ABSTRACT

Cellular senescence is closely related to tissue aging including bone. Bone homeostasis is maintained by the tight balance between bone-forming osteoblasts and bone-resorbing osteoclasts, but it undergoes deregulation with age, causing age-associated osteoporosis, a main cause of which is osteoblast dysfunction. Oxidative stress caused by the accumulation of reactive oxygen species (ROS) in bone tissues with aging can accelerate osteoblast senescence and dysfunction. However, the regulatory mechanism that controls the ROS-induced senescence of osteoblasts is poorly understood. Here, we identified Peptidyl arginine deiminase 2 (PADI2), a post-translational modifying enzyme, as a regulator of ROS-accelerated senescence of osteoblasts via RNA-sequencing and further functional validations. PADI2 downregulation by treatment with H2O2 or its siRNA promoted cellular senescence and suppressed osteoblast differentiation. CCL2, 5, and 7 known as the elements of the senescence-associated secretory phenotype (SASP) which is a secretome including proinflammatory cytokines and chemokines emitted by senescent cells and a representative feature of senescence, were upregulated by H2O2 treatment or Padi2 knockdown. Furthermore, blocking these SASP factors with neutralizing antibodies or siRNAs alleviated the senescence and dysfunction of osteoblasts induced by H2O2 treatment or Padi2 knockdown. The elevated production of these SASP factors was mediated by the activation of NFκB signaling pathway. The inhibition of NFκB using the pharmacological inhibitor or siRNA effectively relieved H2O2 treatment- or Padi2 knockdown-induced senescence and osteoblast dysfunction. Together, our study for the first time uncover the role of PADI2 in ROS-accelerated cellular senescence of osteoblasts and provide new mechanistic and therapeutic insights into excessive ROS-promoted cellular senescence and aging-related bone diseases.


Subject(s)
Cellular Senescence/drug effects , Chemokines, CC/metabolism , Hydrogen Peroxide/pharmacology , NF-kappa B/metabolism , Protein-Arginine Deiminase Type 2/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CCL7/antagonists & inhibitors , Chemokine CCL7/genetics , Chemokine CCL7/metabolism , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/genetics , DNA Damage/drug effects , Down-Regulation/drug effects , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Protein-Arginine Deiminase Type 2/antagonists & inhibitors , Protein-Arginine Deiminase Type 2/genetics , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
2.
J Cell Mol Med ; 25(14): 6558-6572, 2021 07.
Article in English | MEDLINE | ID: mdl-34041839

ABSTRACT

The antiviral treatment efficacy varies among chronic hepatitis B (CHB) patients and the underlying mechanism is unclear. An integrated bioinformatics analysis was performed to investigate the host factors that affect the therapeutic responsiveness in CHB patients. Four GEO data sets (GSE54747, GSE27555, GSE66698 and GSE66699) were downloaded from the Gene Expression Omnibus (GEO) database and analysed to identify differentially expressed genes(DEGs). Enrichment analyses of the DEGs were conducted using the DAVID database. Immune cell infiltration characteristics were analysed by CIBERSORT. Upstream miRNAs and lncRNAs of hub DEGs were identified by miRWalk 3.0 and miRNet in combination with the MNDR platform. As a result, seventy-seven overlapping DEGs and 15 hub genes were identified including CCL5, CXCL9, MYH2, CXCR4, CD74, CCL4, HLA-DRB1, ACTA1, CD69, CXCL10, HLA-DRB5, HLA-DQB1, CXCL13, STAT1 and CKM. The enrichment analyses revealed that the DEGs were mainly enriched in immune response and chemokine signalling pathways. Investigation of immune cell infiltration in liver samples suggested significantly different infiltration between responders and non-responders, mainly characterized by higher proportions of CD8+ T cells and activated NK cells in non-responders. The prediction of upstream miRNAs and lncRNAs led to the identification of a potential mRNA-miRNA-lncRNA regulatory network composed of 2 lncRNAs (H19 and GAS5) and 5 miRNAs (hsa-mir-106b-5p, hsa-mir-17-5p, hsa-mir-20a-5p, hsa-mir-6720-5p and hsa-mir-93-5p) targeting CCL5 mRNA. In conclusion, our study suggested that host genetic factors could affect therapeutic responsiveness in CHB patients. The antiviral process might be associated with the chemokine-mediated immune response and immune cell infiltration in the liver microenvironment.


Subject(s)
Hepatitis B virus/genetics , Hepatitis B/drug therapy , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Biomarkers/chemistry , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Hepatitis B/genetics , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B virus/drug effects , Hepatitis B virus/pathogenicity , Humans , Protein Interaction Maps/genetics , Signal Transduction/genetics
3.
J Biol Chem ; 295(42): 14367-14378, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32817341

ABSTRACT

Ticks, as blood-sucking parasites, have developed a complex strategy to evade and suppress host immune responses during feeding. The crucial part of this strategy is expression of a broad family of salivary proteins, called Evasins, to neutralize chemokines responsible for cell trafficking and recruitment. However, structural information about Evasins is still scarce, and little is known about the structural determinants of their binding mechanism to chemokines. Here, we studied the structurally uncharacterized Evasin-4, which neutralizes a broad range of CC-motif chemokines, including the chemokine CC-motif ligand 5 (CCL5) involved in atherogenesis. Crystal structures of Evasin-4 and E66S CCL5, an obligatory dimeric variant of CCL5, were determined to a resolution of 1.3-1.8 Å. The Evasin-4 crystal structure revealed an L-shaped architecture formed by an N- and C-terminal subdomain consisting of eight ß-strands and an α-helix that adopts a substantially different position compared with closely related Evasin-1. Further investigation into E66S CCL5-Evasin-4 complex formation with NMR spectroscopy showed that residues of the N terminus are involved in binding to CCL5. The peptide derived from the N-terminal region of Evasin-4 possessed nanomolar affinity to CCL5 and inhibited CCL5 activity in monocyte migration assays. This suggests that Evasin-4 derivatives could be used as a starting point for the development of anti-inflammatory drugs.


Subject(s)
Chemokine CCL5/antagonists & inhibitors , Salivary Proteins and Peptides/chemistry , Ticks/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Movement/drug effects , Chemokine CCL5/metabolism , Crystallography, X-Ray , Humans , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Salivary Proteins and Peptides/genetics , Salivary Proteins and Peptides/metabolism
4.
J Immunol ; 200(7): 2291-2303, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29445009

ABSTRACT

Binge/moderate alcohol suppresses TLR4-MyD88 proinflammatory cytokines; however, alcohol's effects on TLR-TRIF signaling, especially after in vivo exposure in humans, are unclear. We performed a comparative analysis of the TLR4-MyD88, TLR4-TRIF, and TLR3-TRIF pathways in human monocytes following binge alcohol exposure. Mechanistic regulation of TLR-TRIF signaling by binge alcohol was evaluated by analyzing IRF3 and TBK1, upstream regulator protein phosphatase 1 (PP1), and immunoregulatory stress proteins HspA1A and XBP-1 in alcohol-treated human and mouse monocytes/macrophages. Two approaches for alcohol exposure were used: in vivo exposure of primary monocytes in binge alcohol-consuming human volunteers or in vitro exposure of human monocytes/murine macrophages to physiological alcohol concentrations (25-50 mM ethanol), followed by LPS (TLR4) or polyinosinic-polycytidylic acid (TLR3) stimulation ex vivo. In vivo and in vitro binge alcohol exposure significantly inhibited the TLR4-MyD88 cytokines TNF-α and IL-6, as well as the TLR4-TRIF cytokines/chemokines IFN-ß, IP-10, and RANTES, in human monocytes, but not TLR3-TRIF-induced cytokines/chemokines, as detected by quantitative PCR and ELISA. Mechanistic analyses revealed TBK-1-independent inhibition of the TLR4-TRIF effector IRF3 in alcohol-treated macrophages. Although stress protein XBP-1, which is known to regulate IRF3-mediated IFN-ß induction, was not affected by alcohol, HspA1A was induced by in vivo alcohol in human monocytes. Alcohol-induced HspA1A was required for inhibition of TLR4-MyD88 signaling but not TLR4-TRIF cytokines in macrophages. In contrast, inhibition of PP1 prevented alcohol-mediated TLR4-TRIF tolerance in macrophages. Collectively, our results demonstrate that in vivo and in vitro binge alcohol exposure in humans suppresses TLR4-MyD88 and TLR4-TRIF, but not TLR3-TRIF, responses. Whereas alcohol-mediated effects on the PP1-IRF3 axis inhibit the TLR4-TRIF pathway, HspA1A selectively suppresses the TLR4-MyD88 pathway in monocytes/macrophages.


Subject(s)
Adaptor Proteins, Vesicular Transport/antagonists & inhibitors , Binge Drinking/pathology , Ethanol/toxicity , Macrophages/immunology , Monocytes/immunology , Myeloid Differentiation Factor 88/antagonists & inhibitors , Toll-Like Receptor 3/antagonists & inhibitors , Toll-Like Receptor 4/antagonists & inhibitors , Adolescent , Adult , Animals , Cell Line , Chemokine CCL5/antagonists & inhibitors , Chemokine CXCL10/antagonists & inhibitors , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Inflammation/pathology , Interferon-beta/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides/immunology , Macrophages/drug effects , Male , Mice , Middle Aged , Monocytes/drug effects , Poly I-C/immunology , RAW 264.7 Cells , Receptors, Neuropeptide Y/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , X-Box Binding Protein 1/drug effects , Young Adult
5.
Int J Mol Sci ; 21(19)2020 Oct 05.
Article in English | MEDLINE | ID: mdl-33027922

ABSTRACT

Chrysin (5,7-dihydroxyflavone) is a natural polyphenolic compound that induces an anti-inflammatory response. In this study, we investigated the molecular mechanism underlying the chrysin-induced suppression of C-C motif chemokine ligand 5 (CCL5) gene expression in atopic dermatitis (AD)-like inflammatory microenvironment. We showed that chrysin inhibited CCL5 expression at the transcriptional level through the suppression of nuclear factor kappa B (NF-κB) in the inflammatory environment. Chrysin could bind to the ATP-binding pocket of the inhibitor of κB (IκB) kinase (IKK) and, subsequently, prevent IκB degradation and NF-κB activation. The clinical efficacy of chrysin in targeting IKK was evaluated in 2,4-dinitrochlorobenzene-induced skin lesions in BALB/c mice. Our results suggested that chrysin prevented CCL5 expression by targeting IKK to reduce the infiltration of mast cells to the inflammatory sites and at least partially attenuate the inflammatory responses. These findings suggested that chrysin might be useful as a platform for the design and synthesis of small-molecule IKK-targeting drugs for the treatment of chronic inflammatory diseases, such as AD.


Subject(s)
Chemokine CCL5/genetics , Dermatitis, Atopic/genetics , Flavonoids/pharmacology , I-kappa B Kinase/genetics , Inflammation/drug therapy , Animals , Cellular Microenvironment/drug effects , Cellular Microenvironment/genetics , Chemokine CCL5/antagonists & inhibitors , Dermatitis, Atopic/pathology , Flavonoids/chemistry , Humans , Inflammation/genetics , Inflammation/pathology , Mast Cells/drug effects , Mast Cells/metabolism , Mice , NF-kappa B/genetics , Tumor Necrosis Factor-alpha
6.
Breast Cancer Res ; 20(1): 54, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29898755

ABSTRACT

BACKGROUND: Metastatic triple-negative breast cancer (TNBC) is a heterogeneous and incurable disease. Numerous studies have been conducted to seek molecular targets to treat TNBC effectively, but chemotherapy is still the main choice for patients with TNBC. We have previously presented evidence of the important roles of interleukin-6 (IL-6) and chemokine (C-C motif) ligand 5 (CCL5) in TNBC tumor growth and metastasis. These experiments highlighted the importance of the crosstalk between cancer cells and stromal lymphatic endothelial cells (LECs) in tumor growth and metastasis. METHODS: We examined the viability and migration of MDA-MB-231-LN, SUM149, and SUM159 cells co-cultured with LECs when treated with maraviroc (CCR5 inhibitor) and tocilizumab (anti-IL-6 receptor antibody). To assess the anti-tumor effects of the combination of these two drugs in an athymic nude mouse model, MDA-MB-231-LN cells were implanted in the mammary fat pad and maraviroc (8 mg/kg, orally daily) and cMR16-1 (murine surrogate of the anti-IL-6R antibody, 10 mg/kg, IP, 3 days a week) were administrated for 5 weeks and effects on tumor growth and thoracic metastasis were measured. RESULTS: In this study, we used maraviroc and tocilizumab to confirm that IL-6 and CCL5 signaling are key pathways promoting TNBC cell proliferation and migration. Further, in a xenograft mouse model, we showed that tumor growth was dramatically inhibited by cMR16-1, the mouse version of the anti-IL6R antibody. The combination of maraviroc and cMR16-1 caused significant reduction of TNBC tumor growth compared to the single agents. Significantly, the combination of maraviroc and cMR16-1 abrogated thoracic metastasis. CONCLUSION: Taken together, these findings show that IL-6 and CCL5 signaling, which promote crosstalk between TNBC and lymphatic vessels, are key enhancers of TNBC tumor growth and metastasis. Furthermore, these results demonstrate that a drug combination inhibiting these pathways may be a promising therapy for TNBC patients.


Subject(s)
Chemokine CCL5/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chemokine CCL5/genetics , Female , Humans , Interleukin-6/genetics , Maraviroc/administration & dosage , Mice , Neoplasm Metastasis , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Cell Physiol Biochem ; 47(2): 707-720, 2018.
Article in English | MEDLINE | ID: mdl-29794461

ABSTRACT

BACKGROUND/AIMS: Hyperlipidemia induces dysfunction in the smooth muscle cells (SMCs) of the blood vessels, and the vascular remodeling that ensues is a key proatherogenic factor contributing to cardiovascular events. Chemokines and chemokine receptors play crucial roles in vascular remodeling. Here, we examined whether the hyperlipidemia-derived chemokine CCL5 and its receptor CCR5 influence vascular SMC proliferation, phenotypic switching, and explored the underlying mechanisms. METHODS: Thoracoabdominal aorta were isolated from wild-type, CCL5 and CCR5 double-knockout mice (CCL5-/-CCR5-/-) fed a high-fat diet (HFD) for 12 weeks. Expression of the contractile, synthetic, and proliferation markers were assayed using immunohistochemical and western blotting. The effects of CCL5 and palmitic acid on cultured SMC proliferation and phenotypic modulation were evaluated using flow cytometry, bromodeoxyuridine (BrdU), and western blotting. RESULTS: Wild-type mice fed an HFD showed markedly increased total cholesterol, triglyceride, and CCL5 serum levels, as well as significantly increased CCL5 and CCR5 expression in the thoracoabdominal aorta vs. normal-diet-fed controls. HFD-fed CCL5-/-CCR5-/- mice showed significantly decreased expression of the synthetic phenotype marker osteopontin and the proliferation marker proliferating cell nuclear antigen, and increased expression of the contractile phenotype marker smooth muscle α-actin in the thoracoabdominal aorta vs. wild-type HFD-fed mice. Human aorta-derived SMCs stimulated with palmitic acid showed significantly increased expression of CCL5, CCR5, and synthetic phenotype markers, as well as increased proliferation. CCL5-treated SMCs showed increased cell cycle regulatory protein expression, paralleling increased synthetic and decreased contractile phenotype marker expression. Inhibition of CCR5 activity by the specific antagonist maraviroc or its expression using small interfering RNA significantly inhibited human aortic SMC proliferation and synthetic phenotype formation. Therefore, CCL5 induces SMC proliferation and phenotypic switching from a contractile to synthetic phenotype via CCR5. CCL5-mediated SMC stimulation activated ERK1/2, Akt/p70S6K, p38 MAPK, and NF-κB signaling. NF-κB inhibition significantly reduced CCR5 expression along with CCR5-induced SMC proliferation and synthetic phenotype formation. CONCLUSIONS: Hyperlipidemia-induced CCL5/CCR5 axis activation serves as a pivotal mediator of vascular remodeling, indicating that CCL5 and CCR5 are key chemokine-related factors in atherogenesis. SMC proliferation and synthetic phenotype transformation attenuation by CCR5 pharmacological inhibition may offer a new approach to treatment or prevention of atherosclerotic diseases associated with hyperlipidemia.


Subject(s)
Cell Proliferation , Chemokine CCL5/genetics , Receptors, CCR5/genetics , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , Cell Line , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/metabolism , Diet, High-Fat , Humans , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Osteopontin/metabolism , Phenotype , Proliferating Cell Nuclear Antigen/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, CCR5/metabolism
8.
Lipids Health Dis ; 17(1): 182, 2018 Jul 31.
Article in English | MEDLINE | ID: mdl-30064511

ABSTRACT

BACKGROUND: Atherosclerosis (AS) is mainly responsible for cardiovascular diseases. The present study investigated whether Lipingshu capsule (LPS), whose ingredients are present in health food stores, has beneficial effect on AS. METHODS: C57BL/6 J mice were given a low fat rodent diet and assigned as control group (CON). ApoE-/- mice were placed on high fat rodent diet and randomly separated into high fat diet (HFD) group and HFD + LPS group whose animals were given 0.9 g/kg.BW LPS daily for 10 weeks. Atherosclerotic lesions in aorta and aortic root were evaluated. Serum lipids and multiple cytokine were measured. RESULTS: ApoE-/- mice fed with high fat diet had serious aortic lesions, whereas LPS markedly decreased plaque area of the total aorta and of the aortic root. LPS recovered the serum lipid profiles by substantially reducing TC, LDL-C, TG and Ox-LDL contents. Multi-cytokine analysis revealed greater serum levels of IL-1α, IL-1ß, IL-6, IFN-γ, GMCSF, RANTES and TNF-α induced by high fat diet slumped with LPS treatment. CONCLUSION: LPS reduces atherosclerotic lesions and thus alleviates AS by lipid profile modulation and inflammation inhibition.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/drug therapy , Cardiovascular Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Plaque, Atherosclerotic/drug therapy , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Capsules , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/biosynthesis , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Diet, High-Fat/adverse effects , Disease Models, Animal , Humans , Lipoproteins, LDL/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Plant Extracts/chemistry , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/etiology , Plaque, Atherosclerotic/pathology , Treatment Outcome , Triglycerides/blood , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis
9.
J Biol Chem ; 291(29): 15267-81, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27226559

ABSTRACT

Parkinson disease (PD) is second only to Alzheimer disease as the most common human neurodegenerative disorder. Despite intense investigation, no interdictive therapy is available for PD. Recent studies indicate that both innate and adaptive immune processes are active in PD. Accordingly, we found a rapid increase in RANTES (regulated on activation normal T cell expressed and secreted) and eotaxin, chemokines that are involved in T cell trafficking, in vivo in the substantia nigra pars compacta and the serum of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. RANTES and eotaxin were also up-regulated in the substantia nigra pars compacta of post-mortem PD brains as compared with age-matched controls. Therefore, we investigated whether neutralization of RANTES and eotaxin could protect against nigrostriatal degeneration in MPTP-intoxicated mice. Interestingly, after peripheral administration, functional blocking antibodies against RANTES and eotaxin reduced the infiltration of CD4(+) and CD8(+) T cells into the nigra, attenuated nigral expression of proinflammatory molecules, and suppressed nigral activation of glial cells. These findings paralleled dopaminergic neuronal protection, normalized striatal neurotransmitters, and improved motor functions in MPTP-intoxicated mice. Therefore, we conclude that attenuation of the chemokine-dependent adaptive immune response may be of therapeutic benefit for PD patients.


Subject(s)
Chemokine CCL11/antagonists & inhibitors , Chemokine CCL5/antagonists & inhibitors , Dopaminergic Neurons/immunology , Parkinsonian Disorders/therapy , Adaptive Immunity , Aged , Aged, 80 and over , Animals , Antibodies, Blocking/administration & dosage , Brain/immunology , Brain/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Chemokine CCL11/biosynthesis , Chemokine CCL11/immunology , Chemokine CCL5/biosynthesis , Chemokine CCL5/immunology , Disease Models, Animal , Dopaminergic Neurons/pathology , Humans , Immunosuppression Therapy , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Middle Aged , Parkinsonian Disorders/immunology , Parkinsonian Disorders/pathology , Substantia Nigra/immunology , Substantia Nigra/pathology
10.
J Hepatol ; 66(4): 743-753, 2017 04.
Article in English | MEDLINE | ID: mdl-28011329

ABSTRACT

BACKGROUND & AIMS: During liver inflammation, triggering fibrogenesis and carcinogenesis immune cells play a pivotal role. In the present study we investigated the role of CCL5 in human and in murine models of chronic liver inflammation leading to hepatocellular carcinoma (HCC) development. METHODS: CCL5 expression and its receptors were studied in well-defined patients with chronic liver disease (CLD) and in two murine inflammation based HCC models. The role of CCL5 in inflammation, fibrosis, tumor initiation and progression was analyzed in different cell populations of NEMOΔhepa/CCL5-/- animals and after bone marrow transplantation (BMT). For therapeutic intervention Evasin-4 was injected for 24h or 8weeks. RESULTS: In CLD patients, CCL5 and its receptor CCR5 are overexpressed - an observation confirmed in the Mdr2-/- and NEMOΔhepa model. CCL5 deletion in NEMOΔhepa mice diminished hepatocyte apoptosis, compensatory proliferation and fibrogenesis due to reduced immune cell infiltration. Especially, CD45+/Ly6G+ granulocytes, CD45+/CD11b+/Gr1.1+/F4/80+ pro-inflammatory monocytes, CD4+ and CD8+ T cells were decreased. One year old NEMOΔhepa/CCL5-/- mice displayed smaller and less malignant tumors, characterized by reduced proliferative capacity and less pronounced angiogenesis. We identified hematopoietic cells as the main source of CCL5, while CCL5 deficiency did not sensitise NEMOΔhepa hepatocytes towards TNFα induced apoptosis. Finally, therapeutic intervention with Evasin-4 over a period of 8weeks ameliorated liver disease progression. CONCLUSION: We identified an important role of CCL5 in human and functionally in mice with disease progression, especially HCC development. A novel approach to inhibit CCL5 in vivo thus appears encouraging for patients with CLD. LAY SUMMARY: Our present study identifies the essential role of the chemoattractive cytokine CCL5 for liver disease progression and especially hepatocellular carcinoma development in men and mice. Finally, the inhibition of CCL5 appears to be encouraging for therapy of human chronic liver disease.


Subject(s)
Carcinoma, Hepatocellular/immunology , Chemokine CCL5/metabolism , Hepatitis, Chronic/immunology , Liver Neoplasms/immunology , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/genetics , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/deficiency , Chemokine CCL5/genetics , Disease Progression , Hematopoiesis/immunology , Hepatitis, Chronic/complications , Hepatitis, Chronic/genetics , Humans , Liver Cirrhosis/etiology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Liver Neoplasms/etiology , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/etiology , Liver Neoplasms, Experimental/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, CCR5/metabolism
11.
FASEB J ; 29(12): 4853-65, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26246404

ABSTRACT

The neuroinflammation associated with multiple sclerosis involves activation of astrocytes that secrete and respond to inflammatory mediators such as IL-1. IL-1 stimulates expression of many chemokines, including C-C motif ligand (CCL) 5, that recruit immune cells, but it also stimulates sphingosine kinase-1, an enzyme that generates sphingosine-1-phosphate (S1P), a bioactive lipid mediator essential for inflammation. We found that whereas S1P promotes IL-1-induced expression of IL-6, it inhibits IL-1-induced CCL5 expression in astrocytes. This inhibition is mediated by the S1P receptor (S1PR)-2 via an inhibitory G-dependent mechanism. Consistent with this surprising finding, infiltration of macrophages into sites of inflammation increased significantly in S1PR2(-/-) animals. However, activation of NF-κB, IFN regulatory factor-1, and MAPKs, all of which regulate CCL5 expression in response to IL-1, was not diminished by the S1P in astrocytes. Instead, S1PR2 stimulated inositol 1,4,5-trisphosphate-dependent Ca(++) release and Elk-1 phosphorylation and enhanced c-Fos expression. In our study, IL-1 induced the IFNß production that supports CCL5 expression. An intriguing finding was that S1P induced c-Fos-inhibited CCL5 directly and also indirectly through inhibition of the IFN-ß amplification loop. We propose that in addition to S1PR1, which promotes inflammation, S1PR2 mediates opposing inhibitory functions that limit CCL5 expression and diminish the recruitment of immune cells.


Subject(s)
Chemokine CCL5/antagonists & inhibitors , Interferon-beta/metabolism , Interleukin-1/antagonists & inhibitors , Lysophospholipids/physiology , Proto-Oncogene Proteins c-fos/metabolism , Sphingosine/analogs & derivatives , Animals , Cells, Cultured , Humans , Interferon Regulatory Factor-1/biosynthesis , Interferon-beta/biosynthesis , Ligands , Mice , Mice, Knockout , Phosphorylation , Protein Kinases/metabolism , STAT1 Transcription Factor/metabolism , STAT2 Transcription Factor/metabolism , Sphingosine/physiology
12.
Carcinogenesis ; 36(1): 104-14, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25330803

ABSTRACT

Chemokines modulate angiogenesis and metastasis that dictate cancer development in tumor microenvironment. Osteosarcoma is the most frequent bone tumor and is characterized by a high metastatic potential. Chemokine CCL5 (previously called RANTES) has been reported to facilitate tumor progression and metastasis. However, the crosstalk between chemokine CCL5 and vascular endothelial growth factor (VEGF) as well as tumor angiogenesis in human osteosarcoma microenvironment has not been well explored. In this study, we found that CCL5 increased VEGF expression and production in human osteosarcoma cells. The conditioned medium (CM) from CCL5-treated osteosarcoma cells significantly induced tube formation and migration of human endothelial progenitor cells. Pretreatment of cells with CCR5 antibody or transfection with CCR5 specific siRNA blocked CCL5-induced VEGF expression and angiogenesis. CCL5/CCR5 axis demonstrably activated protein kinase Cδ (PKCδ), c-Src and hypoxia-inducible factor-1 alpha (HIF-1α) signaling cascades to induce VEGF-dependent angiogenesis. Furthermore, knockdown of CCL5 suppressed VEGF expression and attenuated osteosarcoma CM-induced angiogenesis in vitro and in vivo. CCL5 knockdown dramatically abolished tumor growth and angiogenesis in the osteosarcoma xenograft animal model. Importantly, we demonstrated that the expression of CCL5 and VEGF were correlated with tumor stage according the immunohistochemistry analysis of human osteosarcoma tissues. Taken together, our findings provide evidence that CCL5/CCR5 axis promotes VEGF-dependent tumor angiogenesis in human osteosarcoma microenvironment through PKCδ/c-Src/HIF-1α signaling pathway. CCL5 may represent a potential therapeutic target against human osteosarcoma.


Subject(s)
Bone Neoplasms/blood supply , Chemokine CCL5/metabolism , Neovascularization, Pathologic/metabolism , Osteosarcoma/blood supply , Receptors, CCR5/metabolism , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Animals , Blotting, Western , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Cell Proliferation , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chick Embryo , Chorioallantoic Membrane/metabolism , Chorioallantoic Membrane/pathology , Chromatin Immunoprecipitation , Culture Media, Conditioned/pharmacology , Flow Cytometry , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Nude , Osteosarcoma/metabolism , Osteosarcoma/pathology , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptors, CCR5/chemistry , Receptors, CCR5/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tissue Array Analysis , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics , Xenograft Model Antitumor Assays
13.
Am J Physiol Gastrointest Liver Physiol ; 308(7): G613-24, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25617348

ABSTRACT

Grb2-associated binder 1 (Gab1) adaptor protein amplifies signals downstream of a broad range of growth factors/receptor tyrosine kinases. Although these signals are implicated in liver fibrogenesis, the role of Gab1 remains unclear. To elucidate the role of Gab1, liver fibrosis was examined in hepatocyte-specific Gab1-conditional knockout (Gab1CKO) mice upon bile duct ligation (BDL). Gab1CKO mice developed exacerbated liver fibrosis with activation of hepatic myofibroblasts after BDL compared with control mice. The antifibrotic role of hepatocyte Gab1 was further confirmed by another well-established mouse model of liver fibrosis using chronic injections of carbon tetrachloride. After BDL, Gab1CKO mice also displayed exacerbated liver injury, decreased hepatocyte proliferation, and enhanced liver inflammation. Furthermore, cDNA microarray analysis was used to investigate the potential molecular mechanisms of the Gab1-mediated signal in liver fibrosis, and the fibrosis-promoting factor chemokine (C-C motif) ligand 5 (Ccl5) was identified as upregulated in the livers of Gab1CKO mice following BDL. Interestingly, in vitro studies using primary hepatocytes isolated from control and Gab1CKO mice revealed that the loss of Gab1 resulted in increased hepatocyte CCL5 synthesis upon lipopolysaccharide stimulation. Finally, pharmacological antagonism of CCL5 reduced BDL-induced liver fibrosis in Gab1CKO mice. In conclusion, our results demonstrate that hepatocyte Gab1 is required for liver fibrosis and that hepatocyte CCL5 could be an important contributor to this process. Thus, we present a novel antifibrotic function of hepatocyte Gab1 in liver fibrogenesis.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Hepatocytes/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Phosphoproteins/deficiency , Adaptor Proteins, Signal Transducing , Animals , Carbon Tetrachloride , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/pathology , Liver Cirrhosis, Experimental/prevention & control , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Phosphoproteins/genetics , Signal Transduction , Up-Regulation
14.
N Engl J Med ; 367(2): 135-45, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22784116

ABSTRACT

BACKGROUND: Graft-versus-host disease (GVHD) is a major barrier to successful allogeneic hematopoietic stem-cell transplantation (HSCT). The chemokine receptor CCR5 appears to play a role in alloreactivity. We tested whether CCR5 blockade would be safe and limit GVHD in humans. METHODS: We tested the in vitro effect of the CCR5 antagonist maraviroc on lymphocyte function and chemotaxis. We then enrolled 38 high-risk patients in a single-group phase 1 and 2 study of reduced-intensity allogeneic HSCT that combined maraviroc with standard GVHD prophylaxis. RESULTS: Maraviroc inhibited CCR5 internalization and lymphocyte chemotaxis in vitro without impairing T-cell function or formation of hematopoietic-cell colonies. In 35 patients who could be evaluated, the cumulative incidence rate (±SE) of grade II to IV acute GVHD was low at 14.7±6.2% on day 100 and 23.6±7.4% on day 180. Acute liver and gut GVHD were not observed before day 100 and remained uncommon before day 180, resulting in a low cumulative incidence of grade III or IV GVHD on day 180 (5.9±4.1%). The 1-year rate of death that was not preceded by disease relapse was 11.7±5.6% without excessive rates of relapse or infection. Serum from patients receiving maraviroc prevented CCR5 internalization by CCL5 and blocked T-cell chemotaxis in vitro, providing evidence of antichemotactic activity. CONCLUSIONS: In this study, inhibition of lymphocyte trafficking was a specific and potentially effective new strategy to prevent visceral acute GVHD. (Funded by Pfizer and others; ClinicalTrials.gov number, NCT00948753.).


Subject(s)
CCR5 Receptor Antagonists , Chemotaxis, Leukocyte/drug effects , Cyclohexanes/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes/drug effects , Triazoles/therapeutic use , Adult , Aged , Chemokine CCL3/antagonists & inhibitors , Chemokine CCL5/antagonists & inhibitors , Cyclohexanes/adverse effects , Cyclohexanes/pharmacology , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Hematologic Neoplasms/therapy , Humans , Kaplan-Meier Estimate , Male , Maraviroc , Middle Aged , T-Lymphocytes/physiology , Transplantation, Homologous , Triazoles/adverse effects , Triazoles/pharmacology , Young Adult
15.
J Neuroinflammation ; 11: 146, 2014 Aug 21.
Article in English | MEDLINE | ID: mdl-25182681

ABSTRACT

BACKGROUND: Infection of rabies virus (RABV) causes central nervous system (CNS) dysfunction and results in high mortality in human and animals. However, it is still unclear whether and how CNS inflammation and immune response contribute to RABV infection. METHODS: Suckling mice were intracerebrally infected with attenuated RABV aG and CTN strains, followed by examination of chemokine or cytokine production, inflammatory cell infiltration and neuron apoptosis in the brain. Furthermore, the suckling mice and adult mice that were intracerebrally infected with aG and the adult mice that were intramuscularly infected with street RABV HN10 were treated with CCL5 antagonist (Met-CCL5) daily beginning on day 2 postinfection. The survival rates and inflammation responses in the CNS of these mice were analyzed. RESULTS: Excessive CCL5 in the CNS was associated with CNS dysfunction, inflammation, and macrophage or lymphocyte infiltration after attenuated or street RABV infection. Administration of exogenous CCL5 induced excessive infiltration of immune cells into the CNS and enhanced inflammatory chemokine and cytokine production. Met-CCL5 treatment significantly prolonged survival time of the suckling mice inoculated with aG and adult mice infected with aG and HN10. CONCLUSIONS: These results suggest that CCL5 in the CNS is a key regulator involved in inducing rabies encephalomyelitis. Furthermore, treatment with the CCL5 antagonist Met-CCL5 prolongs survival time of the mice infected with attenuated or street RABVs, which might represent a novel therapeutic strategy to ameliorate RABV infection.


Subject(s)
Central Nervous System Viral Diseases/therapy , Chemokine CCL5/antagonists & inhibitors , Immunotherapy/methods , Rabies/therapy , Animals , Blotting, Western , Brain/pathology , Brain/virology , Central Nervous System Viral Diseases/immunology , Flow Cytometry , Mice , Rabies/immunology , Real-Time Polymerase Chain Reaction
16.
Cytokine ; 68(1): 40-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24656930

ABSTRACT

An anti-inflammatory cytokine, interleukin-10 (IL-10) exerts inhibitory effects on vascular inflammation. Chemokines promote vascular inflammation and play a pathogenic role in the development and maintenance of hypertension. However, chemokine CCL5 has down-regulatory effects on angiotensin II (Ang II)-induced hypertensive mediators. In the present study, IL-10 increased CCL5 expression and attenuated Ang II-induced CCL5 inhibition significantly in vascular smooth muscle cells (VSMCs) from spontaneously hypertensive rats (SHR), whereas TGF-ß had no effect on CCL5 expression or Ang II-induced CCL5 inhibition. Increased CCL5 expression due to IL-10 was mediated mainly through AT2 R activation. Additionally, IL-10 increased activation of AMP-activated protein kinase (AMPK), which further mediated the up-regulatory effect of IL-10 on CCL5 expression. Attenuation of Ang II-induced CCL5 inhibition by IL-10 was associated with suppression of NF-кB activation, and IL-10 inhibited both Ang II-induced IкB-α and IкB-ß degradation in SHR VSMCs. Moreover, IL-10 partially mediated the inhibitory effects of CCL5 on Ang II-induced 12-lipoxygenase (LO) and endothelin (ET)-1 expression in SHR VSMCs. Taken together, this study provides novel evidence that IL-10 plays an up-regulatory role in the anti-hypertensive activity of CCL5 in SHR VSMCs.


Subject(s)
Chemokine CCL5/metabolism , Interleukin-10/physiology , Up-Regulation/physiology , Adenylate Kinase/metabolism , Angiotensin II/pharmacology , Animals , Cells, Cultured , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Electrophoretic Mobility Shift Assay , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , RNA, Small Interfering , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Real-Time Polymerase Chain Reaction
17.
Arterioscler Thromb Vasc Biol ; 33(4): 718-26, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23288157

ABSTRACT

OBJECTIVE: Macrophages are critical contributors to abdominal aortic aneurysm (AAA) disease. We examined the ability of MKEY, a peptide inhibitor of CXCL4-CCL5 interaction, to influence AAA progression in murine models. APPROACH AND RESULTS: AAAs were created in 10-week-old male C57BL/6J mice by transient infrarenal aortic porcine pancreatic elastase infusion. Mice were treated with MKEY via intravenous injection either (1) before porcine pancreatic elastase infusion or (2) after aneurysm initiation. Immunostaining demonstrated CCL5 and CCR5 expression on aneurysmal aortae and mural monocytes/macrophages, respectively. MKEY treatment partially inhibited migration of adaptively transferred leukocytes into aneurysmal aortae in recipient mice. Although all vehicle-pretreated mice developed AAAs, aneurysms formed in only 60% (3/5) and 14% (1/7) of mice pretreated with MKEY at 10 and 20 mg/kg, respectively. MKEY pretreatment reduced aortic diameter enlargement, preserved medial elastin fibers and smooth muscle cells, and attenuated mural macrophage infiltration, angiogenesis, and aortic metalloproteinase 2 and 9 expression after porcine pancreatic elastase infusion. MKEY initiated after porcine pancreatic elastase infusion also stabilized or reduced enlargement of existing AAAs. Finally, MKEY treatment was effective in limiting AAA formation after angiotensin II infusion in apolipoprotein E-deficient mice. CONCLUSIONS: MKEY suppresses AAA formation and progression in 2 complementary experimental models. Peptide inhibition of CXCL4-CCL5 interactions may represent a viable translational strategy to limit progression of human AAA disease.


Subject(s)
Aorta, Abdominal/drug effects , Aortic Aneurysm, Abdominal/prevention & control , Chemokine CCL5/antagonists & inhibitors , Oligopeptides/pharmacology , Platelet Factor 4/antagonists & inhibitors , Angiotensin II , Animals , Aorta, Abdominal/immunology , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/immunology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Cells, Cultured , Chemokine CCL5/metabolism , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Disease Progression , Injections, Intravenous , Leukocytes/drug effects , Leukocytes/immunology , Macrophages/drug effects , Macrophages/immunology , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Oligopeptides/administration & dosage , Pancreatic Elastase , Platelet Factor 4/metabolism , Receptors, CCR5/metabolism , Time Factors
18.
J Immunol ; 189(7): 3480-9, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22942430

ABSTRACT

IL-4 is one of the main cytokines produced during Th2-inducing pathologies. This cytokine has been shown to affect a number of immune processes such as Th differentiation and innate immune responses. However, the impact of IL-4 on CD8 T cell responses remains unclear. In this study, we analyzed the effects of IL-4 on global gene expression profiles of Ag-induced memory CD8 T cells in the mouse. Gene ontology analysis of this signature revealed that IL-4 regulated most importantly genes associated with immune responses. Moreover, this IL-4 signature overlapped with the set of genes preferentially expressed by memory CD8 T cells over naive CD8 T cells. In particular, IL-4 downregulated in vitro and in vivo in a STAT6-dependent manner the memory-specific expression of NKG2D, thereby increasing the activation threshold of memory CD8 T cells. Furthermore, IL-4 impaired activation of memory cells as well as their differentiation into effector cells. This phenomenon could have an important clinical relevance as patients affected by Th2 pathologies such as parasitic infections or atopic dermatitis often suffer from viral-induced complications possibly linked to inefficient CD8 T cell responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Down-Regulation/immunology , Immunologic Memory , Interleukin-4/physiology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Animals , Cells, Cultured , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/metabolism , Down-Regulation/genetics , Humans , Immunity, Innate/genetics , Immunologic Memory/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/genetics , STAT6 Transcription Factor/physiology
19.
Part Fibre Toxicol ; 11: 66, 2014 Dec 14.
Article in English | MEDLINE | ID: mdl-25497303

ABSTRACT

BACKGROUND: Airborne exposure to nanomaterials from unintended occupational or environmental exposures or as a consequence of product use may lead to adverse health effects. Numerous studies have focused on single-walled carbon nanotubes (SWCNTs) and their ability to cause pulmonary injury related to fibrosis, and cancer; however few studies have addressed their impact on infectious agents, particularly viruses that are known for causing severe disease. Here we have demonstrated the ability of pristine SWCNTs of diverse electronic structure to increase the susceptibility of small airway epithelial cells (SAEC) to pandemic influenza A H1N1 infection and discerned potential mechanisms of action driving this response. METHODS: Small airway epithelial cells (SAEC) were exposed to three types of SWCNTs with varying electronic structure (SG65, SG76, CG200) followed by infection with A/Mexico/4108/2009 (pH1N1). Cells were then assayed for viral infectivity by immunofluorescence and viral titers. We quantified mRNA and protein levels of targets involved in inflammation and anti-viral activity (INFß1, IL-8, RANTES/CCL5, IFIT2, IFIT3, ST3GAL4, ST6GAL1, IL-10), localized sialic acid receptors, and assessed mitochondrial function. Hyperspectral imaging analysis was performed to map the SWCNTs and virus particles in fixed SAEC preparations. We additionally performed characterization analysis to monitor SWCNT aggregate size and structure under biological conditions using dynamic light scattering (DLS), static light scattering (SLS). RESULTS: Based on data from viral titer and immunofluorescence assays, we report that pre-treatment of SAEC with SWCNTs significantly enhances viral infectivity that is not dependent on SWCNT electronic structure and aggregate size within the range of 106 nm - 243 nm. We further provide evidence to support that this noted effect on infectivity is not likely due to direct interaction of the virus and nanoparticles, but rather a combination of suppression of pro-inflammatory (RANTES) and anti-viral (IFIT2, IFIT3) gene/protein expression, impaired mitochondrial function and modulation of viral receptors by SWCNTs. CONCLUSIONS: Results of this work reveal the potential for SWCNTs to increase susceptibility to viral infections as a mechanism of adverse effect. These data highlight the importance of investigating the ability of carbon-nanomaterials to modulate the immune system, including impacts on anti-viral mechanisms in lung cells, thereby increasing susceptibility to infectious agents.


Subject(s)
Air Pollutants/toxicity , Bronchi/virology , Immunity, Innate/drug effects , Influenza A Virus, H1N1 Subtype/pathogenicity , Nanotubes, Carbon/toxicity , Respiratory Mucosa/virology , Air Pollutants/chemistry , Apoptosis Regulatory Proteins , Bronchi/cytology , Bronchi/immunology , Cell Survival/drug effects , Cells, Cultured , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation, Viral/drug effects , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/immunology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/drug effects , Mitochondria/immunology , Mitochondria/metabolism , Nanotubes, Carbon/chemistry , Particle Size , Proteins/antagonists & inhibitors , Proteins/genetics , Proteins/metabolism , RNA-Binding Proteins , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology , Time Factors
20.
J Virol ; 86(18): 10218-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22787219

ABSTRACT

CCR5 is the major HIV-1 entry coreceptor. RANTES/CCL5 analogs are more potent inhibitors of infection than native chemokines; one class activates and internalizes CCR5, one neither activates nor internalizes, and a third partially internalizes without activation. Here we show that mutations in CCR5 transmembrane domains differentially impact the activity of these three inhibitor classes, suggesting that the transmembrane region of CCR5, a key interaction site for inhibitors, is a sensitive molecular switch, modulating receptor activity.


Subject(s)
Chemokine CCL5/physiology , Mutation , Receptors, CCR5/genetics , Receptors, CCR5/physiology , Amino Acid Sequence , Amino Acid Substitution , Anti-HIV Agents/pharmacology , Cell Line , Chemokine CCL5/agonists , Chemokine CCL5/antagonists & inhibitors , Chemokine CCL5/genetics , HIV-1/pathogenicity , HIV-1/physiology , Host-Pathogen Interactions , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Receptors, CCR5/chemistry , Signal Transduction , Virus Internalization/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL