Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Life Sci ; 240: 117071, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31783051

ABSTRACT

AIMS: AML (Acute myeloid leukemia) is characterized as a heterogeneous cancer. Chemokines play fundamental roles in the onset, progression cellular, migration, survival and improvement of AML therapy outcomes. The CCR5 receptors together with their ligands have indirect effects on the progression of cancer. In the present study, we have decided to investigate the impact of chemotherapy on the expression of CCR5 and its related ligands (CCL5, CCL4 and CCL3). MAIN METHODS: In this study, peripheral blood and bone marrow specimens were collected prior and post the first stage of (7 + 3) chemotherapy from 25 AML-M4/M5 patients. The expression of CCR by Lymphocytes in peripheral blood was examined by flow cytometry and QRT-PCR. The serum levels of chemokines were measured by ELISA. KEY FINDINGS: There was not observed leukemic blast cells in peripheral blood smear at post first stage of chemotherapy. We found that the expression of CCR5 was attenuated in patients post the first stage of chemotherapy and the healthy control subjects. We have also observed that the serum levels of chemokines were elevated in AML patients prior to chemotherapy. Although in post-chemotherapy stage, only CCL3 was found to reach to the baseline level, CCL5 and CCL4 have not returned to the basal level and were significantly higher than healthy control subjects. SIGNIFICANCE: The current chemotherapy protocol was not able to completely inhibit CCL5 and CCL4. In conclusion, our findings in harmony with previous studies suggest that inhibition of chemokines along with chemotherapy in AML patients may aid therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemokine CCL3/drug effects , Chemokine CCL4/drug effects , Chemokine CCL5/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Monocytes/pathology , Receptors, CCR5/drug effects , Adult , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Lineage , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , Chemokine CCL5/biosynthesis , Chemokines/blood , Disease Progression , Female , Humans , Leukemia, Myeloid, Acute/pathology , Leukocyte Count , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Middle Aged , Receptors, CCR5/biosynthesis
2.
Int J Mol Sci ; 20(18)2019 09 19.
Article in English | MEDLINE | ID: mdl-31546972

ABSTRACT

Chronic low-grade inflammation, also known as metabolic inflammation, is a hallmark of obesity and parallels with the presence of elevated circulatory levels of free fatty acids and inflammatory cytokines/chemokines. CCL4/MIP-1ß chemokine plays a key role in the adipose tissue monocyte recruitment. Increased circulatory levels of TNF-α, palmitate and CCL4 are co-expressed in obesity. We asked if the TNF-α/palmitate could interact cooperatively to augment the CCL4 production in human monocytic cells and macrophages. THP-1 cells/primary macrophages were co-treated with TNF-α/palmitate and CCL4 mRNA/protein expression was assessed using qRT-PCR/ELISA. TLR4 siRNA, a TLR4 receptor-blocking antibody, XBlue™-defMyD cells and pathway inhibitors were used to decipher the signaling mechanisms. We found that TNF-α/palmitate co-stimulation augmented the CCL4 expression in monocytic cells and macrophages compared to controls (p < 0.05). TLR4 suppression or neutralization abrogated the CCL4 expression in monocytic cells. Notably, CCL4 cooperative induction in monocytic cells was: (1) Markedly less in MyD88-deficient cells, (2) IRF3 independent, (3) clathrin dependent and (4) associated with the signaling mechanism involving ERK1/2, c-Jun, JNK and NF-κB. In conclusion, TNF-α/palmitate co-stimulation promotes the CCL4 expression in human monocytic cells through the mechanism involving a TLR4-MyD88 axis and MAPK/NF-κB pathways. These findings unravel a novel mechanism of the cooperative induction of CCL4 by TNF-α and palmitate which could be relevant to metabolic inflammation.


Subject(s)
Chemokine CCL4/biosynthesis , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Monocytes/metabolism , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Palmitic Acid/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Monocytes/cytology , THP-1 Cells
3.
Cell Physiol Biochem ; 52(4): 908-921, 2019.
Article in English | MEDLINE | ID: mdl-30964608

ABSTRACT

BACKGROUND/AIMS: Increased circulatory levels of both TNF-α and CCL4/MIP-1ß are found in metabolic diseases. However, it is unclear whether TNF-α which is a signature proinflammatory cytokine involved in metabolic inflammation, can induce/promote the expression of CCL4. METHODS: THP-1 human monocytic cells and THP-1-derived macrophages were stimulated with TNF-α and LPS-treatment as a positive control. CCL4 mRNA/protein expression was measured using qRT-PCR/ELISA, respectively. Stress-activated protein kinases (SAPK)/ c-Jun N-terminal kinase (JNK) activity was determined using the assay kit. Mechanistic pathways were studied using anti-TNFR1/2 antibodies, pharmacological inhibitors, siRNAs, and NF-κB/AP-1 reporter-expressing THP-1-XBlue cells. Phosphorylation of signaling molecules was assessed by Western blotting. RESULTS: TNF-α induces CCL4 expression at mRNA and protein levels, in both THP-1 monocytic cells and macrophages (P<0.05). TNF-α-driven CCL4 production was markedly abrogated by pre-treatment with anti-TNFR1/2 neutralizing antibodies. TNF-α treatment induced phosphorylation of SAPK/JNK, c-Jun, and NF-κB. Genetic and/or pharmacologic inhibition of SAPK/JNK and NF-κB pathways suppressed the TNF-α-induced CCL4 expression (P<0.05). NF-κB/AP-1 activity was found to be significantly increased in TNFα-treated SEAP reporter-expressing monocytic cells. CONCLUSION: These data suggest that TNF-α drives CCL4 expression in THP-1 monocytic cells/macrophages via the activation of SAPK/JNK and NF-κB pathways. The findings may provide new insights into understanding the regulatory role of TNF-α in augmenting CCL4 expression during inflammatory conditions.


Subject(s)
Chemokine CCL4/biosynthesis , Gene Expression Regulation , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Monocytes/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chemokine CCL4/genetics , Humans , MAP Kinase Kinase 4/genetics , Monocytes/cytology , NF-kappa B/genetics , THP-1 Cells , Tumor Necrosis Factor-alpha/genetics
4.
Eur J Immunol ; 48(1): 194-203, 2018 01.
Article in English | MEDLINE | ID: mdl-28850672

ABSTRACT

Rheumatoid arthritis (RA) is associated with abnormal B cell-functions implicating antibody-dependent and -independent mechanisms. B cells have emerged as important cytokine-producing cells, and cytokines are well-known drivers of RA pathogenesis. To identify novel cytokine-mediated B-cell functions in RA, we comprehensively analysed the capacity of B cells from RA patients with an inadequate response to disease modifying anti-rheumatic drugs to produce cytokines in comparison with healthy donors (HD). RA B cells displayed a constitutively higher production of the pathogenic factors interleukin (IL)-8 and Gro-α, while their production of several cytokines upon activation via the B cell receptor for antigen (BCR) was broadly suppressed, including a loss of the expression of the protective factor TRAIL, compared to HD B cells. These defects were partly erased after treatment with the IL-6-signalling inhibitor tocilizumab, indicating that abnormal IL-6 signalling contributed to these abnormalities. Noteworthy, the clinical response of individual patients to tocilizumab therapy could be predicted using the amounts of MIP-1ß and ß-NGF produced by these patients' B cells before treatment. Taken together, our study highlights hitherto unknown abnormal B-cell functions in RA patients, which are related to the unbalanced cytokine network, and are potentially relevant for RA pathogenesis and treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/pathology , B-Lymphocytes/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Arthritis, Rheumatoid/immunology , Chemokine CCL4/biosynthesis , Chemokine CXCL1/biosynthesis , Humans , Interleukin-8/biosynthesis , Nerve Growth Factor/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/biosynthesis
5.
Cytokine ; 96: 102-106, 2017 08.
Article in English | MEDLINE | ID: mdl-28390264

ABSTRACT

Cardiovascular disease (CVD) is the leading cause of death in the industrialized world. Aging is the most predictive risk factor for CVD and is associated with arterial inflammation which contributes to increased CVD risk. Although age-related arterial inflammation has been described in both humans and animals, only a limited number of inflammatory mediators, cytokines and chemokines have been identified. In this investigation we sought to determine whether lifespan extending interventions, including crowded litter early life nutrient deprivation (CL), traditional lifelong caloric restriction (CR) and lifelong Rapamycin treatment (Rap) would attenuate age-related arterial inflammation using multi analyte profiling. Aortas from Young (4-6months), Old (22months), Old CL, Old CR and Old Rap mice were homogenized and cytokine concentrations were assessed using Luminex Multi Analyte Profiling. Chemokines involved in immune cell recruitment, such as CCL2, CXCL9, CXCL10, GMCSF and MCSF, were increased in Old vs. Young (p<0.05). The age-related increase of CXCL10 was prevented by CR (p<0.05 vs. Old). MSCF concentrations were lower in aortas of Rap treated mice (p<0.05 vs. Old). Interleukins (IL), IL-1α, IL-1ß and IL-10, were also greater in Old vs. Young mice (p<0.05). These data demonstrate selected lifespan extending interventions can prevent or limit age-related increases in selected aortic chemokines.


Subject(s)
Aging , Arteries/immunology , Chemokine CXCL10/blood , Macrophage Colony-Stimulating Factor/blood , Animals , Caloric Restriction , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Chemokine CCL2/biosynthesis , Chemokine CCL4/biosynthesis , Chemokine CXCL10/immunology , Chemokines/biosynthesis , Chemokines/immunology , Cytokines/biosynthesis , Cytokines/immunology , Early Medical Intervention , Interleukin-10/biosynthesis , Interleukin-1beta/blood , Interleukins/biosynthesis , Interleukins/immunology , Macrophage Colony-Stimulating Factor/immunology , Male , Mice , Sirolimus/therapeutic use
6.
J Acquir Immune Defic Syndr ; 75(3): 338-344, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28418988

ABSTRACT

BACKGROUND: Immunological nonresponse represents the Achilles heel in the combination antiretroviral therapy (cART) effectiveness, and increases risk of clinical events and death. CD8 T cells play a crucial role in controlling HIV replication, and polyfunctional HIV-specific CD8 T cells have been associated with nonprogressive HIV infection. However, the possible role of polyfunctional CD8 T cells in predicting posttreatment immune reconstitution has not yet been explored. The aim of this study was to identify functional markers predictive of immunological response to cART in chronic HIV-infected patients. METHODS: A cohort of chronic HIV-infected individuals naive to cART were enrolled in the ALPHA study. CD4/CD8 T-cell subsets, their differentiation/activation, as well as susceptibility to apoptosis were analyzed before and after 12 months of cART. Moreover, CD8 T cells polyfunctional response after HIV antigenic stimulation was also assessed. RESULTS: Results showed a significant correlation between worse CD4 T-cell restoration and low frequency of naive CD4 T cells, high frequency of effector memory CD4 T cells, and high susceptibility to apoptosis of CD4 T cells all before cART. Moreover, CD8 functional subsets expressing total C-C motif chemokine ligand 4 (CCL-4) or in combination with CD107a and interferon gamma (IFNγ) were negatively associated with immune reconstitution. CONCLUSIONS: In conclusion, our study shows that a more differentiated phenotype of CD4 T cells and CCL-4-producing CD8 T cells could represent valuable predictors of worse immune reconstitution. These parameters may be used as tools for identifying patients at risk of immunological failure during cART and eventually represent the basis for innovative therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokine CCL4/biosynthesis , Chemokine CCL4/immunology , HIV Infections/immunology , HIV-1/immunology , Adult , Anti-HIV Agents/therapeutic use , CD8-Positive T-Lymphocytes/metabolism , Chronic Disease , Female , HIV Infections/drug therapy , HIV Infections/physiopathology , Humans , Immunophenotyping , Lymphocyte Activation , Male , Middle Aged , Treatment Outcome , Viral Load , Young Adult
7.
PLoS One ; 11(9): e0163140, 2016.
Article in English | MEDLINE | ID: mdl-27632174

ABSTRACT

We reported the expression of angiopoietin Tie2 receptor on human neutrophils and the capacity of angiopoietins (Ang1 and Ang2) to induce pro-inflammatory activities, such as platelet-activating factor synthesis, ß2-integrin activation and neutrophil migration. Recently, we observed differential effects between both angiopoietins, namely, the capacity of Ang1, but not Ang2, to promote rapid interleukin-8 synthesis and release, as well as neutrophil viability. Herein, we addressed whether Ang1 and/or Ang2 could modulate the synthesis and release of macrophage inflammatory protein-1ß (MIP-1ß) by neutrophils. Neutrophils were isolated from blood of healthy volunteers; intracellular and extracellular MIP-1ß protein concentrations were assessed by ELISA. After 24 hours, the basal intracellular and extracellular MIP-1ß protein concentrations were ≈500 and 100 pg/106 neutrophils, respectively. Treatment with Ang1 (10 nM) increased neutrophil intracellular and extracellular MIP-1ß concentrations by 310 and 388% respectively. Pretreatment with PI3K (LY294002), p38 MAPK (SB203580) and MEK (U0126) inhibitors completely inhibited Ang1-mediated increase of MIP-1ß intracellular and extracellular protein levels. Pretreatment with NF-κB complex inhibitors, namely Bay11-7085 and IKK inhibitor VII or with a transcription inhibitor (actinomycin D) and protein synthesis inhibitor (cycloheximide), did also abrogate Ang1-mediated increase of MIP-1ß intracellular and extracellular protein levels. We validated by RT-qPCR analyses the effect of Ang1 on the induction of MIP-1ß mRNA levels. Our study is the first one to report Ang1 capacity to induce MIP-1ß gene expression, protein synthesis and release from neutrophils, and that these effects are mediated by PI3K, p38 MAPK and MEK activation and downstream NF-κB activation.


Subject(s)
Angiopoietin-1/physiology , Chemokine CCL4/biosynthesis , Inflammation Mediators/physiology , Neutrophils/metabolism , Angiopoietin-1/antagonists & inhibitors , Chemokine CCL4/metabolism , Enzyme Inhibitors/pharmacology , Humans , NF-kappa B/metabolism , Neutrophils/drug effects
8.
J Clin Invest ; 126(3): 1039-51, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26854929

ABSTRACT

T regulatory cells (Tregs) control immune homeostasis by preventing inappropriate responses to self and nonharmful foreign antigens. Tregs use multiple mechanisms to control immune responses, all of which require these cells to be near their targets of suppression; however, it is not known how Treg-to-target proximity is controlled. Here, we found that Tregs attract CD4+ and CD8+ T cells by producing chemokines. Specifically, Tregs produced both CCL3 and CCL4 in response to stimulation, and production of these chemokines was critical for migration of target T cells, as Tregs from Ccl3-/- mice, which are also deficient for CCL4 production, did not promote migration. Moreover, CCR5 expression by target T cells was required for migration of these cells to supernatants conditioned by Tregs. Tregs deficient for expression of CCL3 and CCL4 were impaired in their ability to suppress experimental autoimmune encephalomyelitis or islet allograft rejection in murine models. Moreover, Tregs from subjects with established type 1 diabetes were impaired in their ability to produce CCL3 and CCL4. Together, these results demonstrate a previously unappreciated facet of Treg function and suggest that chemokine secretion by Tregs is a fundamental aspect of their therapeutic effect in autoimmunity and transplantation.


Subject(s)
Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , T-Lymphocytes, Regulatory/physiology , Adolescent , Adoptive Transfer , Animals , Cell Proliferation , Cells, Cultured , Chemokine CCL3/metabolism , Chemokine CCL4/metabolism , Chemotaxis, Leukocyte , Child , Child, Preschool , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Female , Humans , Infant , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR5/physiology
9.
Peptides ; 69: 133-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25963277

ABSTRACT

Macrophages are an important source of pro-inflammatory and pro-angiogenic factors, which can promote pathological processes involving inflammation and angiogenesis. This study investigated the effects of Apelin on macrophages under both normal and hypoxic conditions. Under normal culture conditions, Apelin down-regulated the mRNA expression levels of monocyte chemotactic protein 1 (MCP1), monocyte chemotactic protein 3 (MCP3), macrophage inflammatory protein 1 (MIP1α, MIP1ß), vascular endothelial growth factor A (VEGFA), Angiopoietin 2 (Ang2) and tumor necrosis factor α (TNFα). The supernatant concentrations of MCP1, MCP3, MIP1α, MIP1ß, macrophage inflammatory protein 2 (MIP2) and TNFα proteins were significantly decreased in the Apelin treated group. Hypoxia induced profound up-regulations of the angiogenic, chemokine, and inflammatory factors at both the mRNA and protein levels. Apelin suppressed the hypoxia-induced increases in MCP1, MCP3, MIP2, MIP1ß and TNFα expression. The underlying mechanism of Apelin inhibit inflammation is regulating NF-κB/JNK signal pathway. Additionally, Apelin can protect macrophages from apoptosis and can enhance cell migration during hypoxia. And cleaved Caspase9/3 pathways were involved in Apelin inhibiting RAW264.7 apoptosis. In conclusion, we showed the effect of Apelin on RAW264.7 macrophage under normal and hypoxic condition, which could further influence the angiogenesis and inflammation process that promoted by macrophages.


Subject(s)
Adipokines/metabolism , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Macrophages/metabolism , Peptides/metabolism , Adipokines/administration & dosage , Adipokines/genetics , Angiopoietin-2/biosynthesis , Animals , Apelin , Cell Movement/genetics , Chemokine CCL2/biosynthesis , Chemokine CCL2/genetics , Chemokine CCL4/biosynthesis , Chemokine CCL4/genetics , Gene Expression Regulation/drug effects , Inflammation/genetics , Inflammation/pathology , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/genetics , Macrophages/drug effects , Macrophages/pathology , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Peptides/administration & dosage , Peptides/chemical synthesis , Tumor Necrosis Factor-alpha/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis
10.
Immunobiology ; 220(9): 1093-100, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26001731

ABSTRACT

Mast cells are crucial elements of the innate immune response. They reside in tissues that are commonly exposed to the external environment, such as the skin and mucosae, where they can rapidly detect the presence of pathogens and mount a potent inflammatory response that recruits other cellular effectors of the immune response. The contribution of mast cells to the immune response to viruses, bacteria, protozoa and multicellular parasites is well established, but there is scarce information about the role of these cells in fungal infections. In this study, we analyzed if mast cells are activated by Candida albicans and if the C-type lectin receptor Dectin-1 is involved in its recognition. We found that both yeasts and hyphae of C. albicans-induced mast cell degranulation and production of TNF-α, IL-6, IL-10, CCL3 and CCL4, while only yeasts were able to induce IL-1ß. Mast cells also produced ROS after stimulation with both dimorphic phases of C. albicans. When mast cells were activated with yeasts and hyphae, they showed decreased expression of IκBα and increased presence of phosphorylated Syk. Blockade of the receptor Dectin-1, but not Toll-like receptor 2, decreased TNF-α production by mast cell in response to C. albicans. These results indicate that mast cells are capable of sensing the two phases of C. albicans, and suggest that mast cells participate as an early inductor of inflammation during the early innate immune response to this fungus.


Subject(s)
Candida albicans/immunology , Cell Degranulation/immunology , Inflammation/immunology , Lectins, C-Type/immunology , Mast Cells/immunology , Animals , Cells, Cultured , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , Hyphae/immunology , I-kappa B Kinase/metabolism , Interleukin-10/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Male , Phosphorylation/immunology , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Syk Kinase , Tumor Necrosis Factor-alpha/biosynthesis , Yeasts/immunology
11.
Int Arch Allergy Immunol ; 166(3): 225-30, 2015.
Article in English | MEDLINE | ID: mdl-25924652

ABSTRACT

BACKGROUND: Natural killer (NK) cells have been detected in the lesional skin of patients with inflammatory skin diseases, where high levels of histamine are also present. Therefore, we investigated the effect of histamine, in particular via the histamine H4 receptor (H4R), on gene expression levels in human NK cells. METHODS: Comprehensive microarray-based mRNA expression profiling was performed to assess the gene expression levels in human NK cells in response to H4R stimulation in an unbiased approach. The expression of selected cytokines and chemokines was quantified by real-time PCR and enzyme-linked immunosorbent assay. RESULTS: The microarray analysis identified only few genes which were differentially regulated upon H4R stimulation. In follow-up studies, a significant upregulation of CCL3 and CCL4 at the mRNA level and in addition for CCL3 also at the protein level via the H4R was observed. CONCLUSION: The elevated expression levels of chemokines in response to H4R stimulation might foster the inflammation in allergic skin diseases and characterize the H4R as a promising therapeutic target.


Subject(s)
Cytokines/biosynthesis , Dermatitis, Atopic/immunology , Killer Cells, Natural/immunology , RNA, Messenger/biosynthesis , Receptors, G-Protein-Coupled/immunology , Receptors, Histamine/immunology , CD4-Positive T-Lymphocytes/immunology , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , Humans , Inflammation/immunology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Receptors, Histamine H4
12.
Mol Ther ; 23(7): 1160-1168, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25896249

ABSTRACT

Metachromatic leukodystrophy (MLD) is a lysosomal storage disease caused by a functional deficiency of the lysosomal enzyme arylsulfatase A. The prevailing late-infantile variant of MLD is characterized by widespread and progressive demyelination of the central nervous system (CNS) causing death during childhood. In order to gain insight into the pathomechanism of the disease and to identify novel therapeutic targets, we analyzed neuroinflammation in two mouse models reproducing a mild, nondemyelinating, and a more severe, demyelinating, variant of MLD, respectively. Microgliosis and upregulation of cytokine/chemokine levels were clearly more pronounced in the demyelinating model. The analysis of the temporal cytokine/chemokine profiles revealed that the onset of demyelination is preceded by a sustained elevation of the macrophage inflammatory protein (MIP)-1α followed by an upregulation of MIP-1ß, monocyte chemotactic protein (MCP)-1, and several interleukins. The tumor necrosis factor (TNF)-α remains unchanged. Treatment of the demyelinating mouse model with the nonsteroidal anti-inflammatory drug simvastatin reduced neuroinflammation, improved the swimming performance and ataxic gait, and retarded demyelination of the spinal cord. Our data suggest that neuroinflammation is causative for demyelination in MLD mice and that anti-inflammatory treatment might be a novel therapeutic option to improve the CNS function of MLD patients.


Subject(s)
Central Nervous System/drug effects , Inflammation/drug therapy , Leukodystrophy, Metachromatic/drug therapy , Simvastatin/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Central Nervous System/physiopathology , Chemokine CCL2/biosynthesis , Chemokine CCL4/biosynthesis , Demyelinating Diseases/drug therapy , Demyelinating Diseases/pathology , Disease Models, Animal , Humans , Inflammation/physiopathology , Leukodystrophy, Metachromatic/immunology , Mice , Spinal Cord/drug effects , Spinal Cord/pathology , Tumor Necrosis Factor-alpha/biosynthesis
13.
Tumour Biol ; 36(8): 5881-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25744730

ABSTRACT

Both intra-tumor macrophage and T-box transcription factor Brachyury (T) have been proved to play important roles in tumor progression and metastasis. However, it is still unknown whether T could regulate the infiltration of macrophages. Here, we report that the Brachyury expression in human lung tumors was inversely correlated with the infiltration of macrophages. Brachyury suppressed the capability of human lung cancer cells to attract macrophages. Using PCR array, we found that Brachyury inhibited expression of several chemokines, including CCL2, CCL4, and CXCL10. Accordingly, knockdown of CCL2 and CCL4 in lung cancer cells suppressed macrophage invasion under coculture conditions. Furthermore, we found that Brachyury expression was inversely correlated with CCL2 and CCL4 expression in human lung tumors. Taken together, our findings shed light on the novel role of Brachyury in regulation of macrophage infiltration.


Subject(s)
Chemokine CCL2/biosynthesis , Chemokine CCL4/biosynthesis , Fetal Proteins/genetics , Lung Neoplasms/genetics , T-Box Domain Proteins/genetics , Carcinoma, Squamous Cell , Cell Line, Tumor , Cell Movement/genetics , Chemokine CCL2/genetics , Chemokine CCL4/genetics , Chemokine CXCL10/biosynthesis , Gene Expression Regulation, Neoplastic , Genetic Vectors , Humans , Lung Neoplasms/pathology , Macrophages/pathology , Neoplasm Invasiveness/genetics
14.
Infect Immun ; 83(4): 1406-17, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25644000

ABSTRACT

Gamma interferon (IFN-γ) drives antiparasite responses and immunopathology during infection with Plasmodium species. Immunity-related GTPases (IRGs) are a class of IFN-γ-dependent proteins that are essential for cell autonomous immunity to numerous intracellular pathogens. However, it is currently unknown whether IRGs modulate responses during malaria. We have used the Plasmodium berghei ANKA (PbA) model in which mice develop experimental cerebral malaria (ECM) to study the roles of IRGM1 and IRGM3 in immunopathology. Induction of mRNA for Irgm1 and Irgm3 was found in the brains and spleens of infected mice at times of peak IFN-γ production. Irgm3-/- but not Irgm1-/- mice were completely protected from the development of ECM, and this protection was associated with the decreased induction of inflammatory cytokines, as well as decreased recruitment and activation of CD8+ T cells within the brain. Although antigen-specific proliferation of transferred CD8+ T cells was not diminished compared to that of wild-type recipients following PbA infection, T cells transferred into Irgm3-/- recipients showed a striking impairment of effector differentiation. Decreased induction of several inflammatory cytokines and chemokines (interleukin-6, CCL2, CCL3, and CCL4), as well as enhanced mRNA expression of type-I IFNs, was found in the spleens of Irgm3-/- mice at day 4 postinfection. Together, these data suggest that protection from ECM pathology in Irgm3-/- mice occurs due to impaired generation of CD8+ effector function. This defect is nonintrinsic to CD8+ T cells. Instead, diminished T cell responses most likely result from defective initiation of inflammatory responses in myeloid cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , GTP Phosphohydrolases/immunology , Malaria, Cerebral/immunology , Plasmodium berghei/immunology , Adoptive Transfer , Animals , Antigens, Protozoan/immunology , Brain/immunology , Brain/parasitology , Brain/pathology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/transplantation , Cell Proliferation/genetics , Chemokine CCL2/biosynthesis , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , GTP Phosphohydrolases/genetics , GTP-Binding Proteins/genetics , GTP-Binding Proteins/immunology , Inflammation/genetics , Inflammation/immunology , Interferon Type I/biosynthesis , Interferon-gamma/immunology , Interleukin-6/biosynthesis , Malaria, Cerebral/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics
15.
Aging Cell ; 14(2): 200-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25620312

ABSTRACT

Chemokines play a pivotal role in regulating the immune response through a tightly controlled expression. Elevated levels of inflammatory chemokines commonly occur with aging but the mechanism underlying this age-associated change is not fully understood. Here, we report the role of microRNA-125b (miR-125b) in regulating inflammatory CC chemokine 4 (CCL4) expression in human immune cells and its altered expression with aging. We first analyzed the mRNA level of CCL4 in eight different types of immune cells including CD4 and CD8 T-cell subsets (naïve, central and effector memory), B cells and monocytes in blood from both young (≤42 years) and old (≥70 years) adults. We observed that monocytes and naïve CD8 T cells expressed higher levels of CCL4 and exhibited an age-related increase in CCL4. We then found the level of miR-125b was inversely correlated with the level of CCL4 in these cells, and the level of miR-125b was reduced in monocytes and naïve CD8 T cells of the old compared to the young adults. Knock-down of miR-125b by shRNA in monocytes and naïve CD8 T cells led to an increase of CCL4 protein, whereas enhanced miR-125b expression by transfection in naïve CD8 T cells resulted in a reduction of the CCL4 mRNA and protein in response to stimulation. Finally, we demonstrated that miR-125b action requires the 'seed' sequence in 3'UTR of CCL4. Together these findings demonstrated that miR-125b is a negative regulator of CCL4 and its reduction is partially responsible for the age-related increase of CCL4.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cellular Senescence/immunology , Chemokine CCL4/biosynthesis , MicroRNAs/immunology , Monocytes/immunology , 3' Untranslated Regions , Age Factors , Aged , Chemokine CCL4/genetics , Chemokine CCL4/immunology , Humans , Lymphocyte Activation , MicroRNAs/biosynthesis , MicroRNAs/genetics
16.
J Immunol ; 193(9): 4507-14, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25261480

ABSTRACT

Vizantin has immunostimulating properties and anticancer activity. In this study, we investigated the molecular mechanism of immune activation by vizantin. THP-1 cells treated with small interfering RNA for TLR-4 abolished vizantin-induced macrophage activation processes such as chemokine release. In addition, compared with wild-type mice, the release of MIP-1ß induced by vizantin in vivo was significantly decreased in TLR-4 knockout mice, but not in TLR-2 knockout mice. Vizantin induced the release of IL-8 when HEK293T cells were transiently cotransfected with TLR-4 and MD-2, but not when they were transfected with TLR-4 or MD-2 alone or with TLR-2 or TLR-2/MD-2. A dipyrromethene boron difluoride-conjugated vizantin colocalized with TLR-4/MD-2, but not with TLR-4 or MD-2 alone. A pull-down assay with vizantin-coated magnetic beads showed that vizantin bound to TLR-4/MD-2 in extracts from HEK293T cells expressing both TLR-4 and MD-2. Furthermore, vizantin blocked the LPS-induced release of TNF-α and IL-1ß and inhibited death in mice. We also performed in silico docking simulation analysis of vizantin and MD-2 based on the structure of MD-2 complexed with the LPS antagonist E5564; the results suggested that vizantin could bind to the active pocket of MD-2. Our observations show that vizantin specifically binds to the TLR-4/MD-2 complex and that the vizantin receptor is identical to the LPS receptor. We conclude that vizantin could be an effective adjuvant and a therapeutic agent in the treatment of infectious diseases and the endotoxin shock caused by LPS.


Subject(s)
Endotoxins/immunology , Glycolipids/pharmacology , Immunity/drug effects , Lymphocyte Antigen 96/metabolism , Trehalose/analogs & derivatives , Animals , Chemokine CCL4/biosynthesis , Cytokines/biosynthesis , Gene Expression , Glycolipids/metabolism , HEK293 Cells , Humans , Immunity/genetics , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/genetics , Macrophages/chemistry , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Models, Molecular , Protein Binding , Protein Conformation , Protein Transport , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Trehalose/metabolism , Trehalose/pharmacology
17.
Sci Rep ; 4: 5845, 2014 Aug 18.
Article in English | MEDLINE | ID: mdl-25132382

ABSTRACT

Plasmacytoid dendritic cells (pDC) expressing FcγRIIa are antigen-presenting cells able to link innate and adaptive immunity and producing various cytokines and chemokines. Although highly restricted, they are able to replicate HIV-1. We determined the activity of anti-HIV-1 neutralizing antibodies (NAb) and non-neutralizing inhibitory antibodies (NNIAb) on the infection of primary pDC by HIV-1 primary isolates and analyzed cytokines and chemokines production. Neutralization assay was performed with primary pDC in the presence of serial antibodies (Ab) concentrations. In parallel, we measured the release of cytokines and chemokines by ELISA and CBA Flex assay. We found that NAb, but not NNIAb, inhibit HIV-1 replication in pDC. This inhibitory activity was lower than that detected for myeloid dendritic cells (mDC) infection and independent of FcγRIIa expressed on pDC. Despite the complete protection, IFN-α production was detected in the supernatant of pDC treated with NAb VRC01, 4E10, PGT121, 10-1074, 10E8, or polyclonal IgG44 but not with NAb b12. Production of MIP-1α, MIP-1ß, IL-6, and TNF-α by pDC was also maintained in the presence of 4E10, b12 and VRC01. These findings suggest that pDC can be protected from HIV-1 infection by both NAb and IFN-α release triggered by the innate immune response during infection.


Subject(s)
Antibodies, Neutralizing/immunology , Dendritic Cells/virology , HIV Antibodies/immunology , HIV-1/immunology , Receptors, IgG/biosynthesis , Cells, Cultured , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , Dendritic Cells/immunology , HIV Infections/immunology , HIV Infections/prevention & control , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interleukin-6/biosynthesis , Neutralization Tests , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Virus Replication/immunology
18.
J Immunol ; 192(4): 1361-71, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24453247

ABSTRACT

The myeloid C-type lectin receptor Dectin-2 directs the generation of Th2 and Th17 immune responses to the house dust mite Dermatophagoides farinae through the generation of cysteinyl leukotrienes and proinflammatory cytokines, respectively, but a role for Dectin-2 in effector phase responses has not been described. In this study, we demonstrate that administration of the Dectin-2 mAb solely at the time of D. farinae challenge abrogated eosinophilic and neutrophilic inflammation in the bronchoalveolar lavage fluid and Th1, Th2, and Th17 inflammation in the lung of previously sensitized mice. Furthermore, Dectin-2 null mice (Clec4n(-/-)) sensitized with the adoptive transfer of D. farinae-pulsed wild-type (WT) bone marrow-derived dendritic cells (DCs) also had less D. farinae-elicited pulmonary inflammation, supporting an effector function for Dectin-2. The protection from pulmonary inflammation seen with the Dectin-2 mAb or in Clec4n(-/-) mice was associated with little or no reduction in lung-draining lymph node cells or their cytokine production and with no reduction in serum IgE. WT and Clec4n(-/-) mice recipients, sensitized with D. farinae-pulsed WT bone marrow-derived DCs, had comparable levels of D. farinae-elicited IL-6, IL-23, TNF-α, and cysteinyl leukotrienes in the lung. By contrast, D. farinae-elicited CCL4 and CCL8 production from pulmonary CD11c(+)CD11b(+)Ly6C(+) and CD11c(+)CD11b(+)Ly6C(-)CD64(+) monocyte-derived DCs was reduced in Clec4n(-/-) recipients. Addition of CCL8 at the time of D. farinae challenge abrogated the protection from eosinophilic, neutrophilic, and Th2 pulmonary inflammation seen in Clec4n(-/-) recipients. Taken together, these results reveal that Dectin-2 regulates monocyte-derived DC function in the pulmonary microenvironment at D. farinae challenge to promote the local inflammatory response.


Subject(s)
Dendritic Cells/immunology , Dermatophagoides farinae/immunology , Lectins, C-Type/immunology , Pneumonia/immunology , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Antigens, Dermatophagoides/immunology , Antigens, Ly/metabolism , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Chemokine CCL4/biosynthesis , Chemokine CCL4/metabolism , Chemokine CCL8/biosynthesis , Chemokine CCL8/metabolism , Cysteine/immunology , Dendritic Cells/transplantation , Eosinophils/immunology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Interleukin-23/metabolism , Interleukin-6/metabolism , Lectins, C-Type/deficiency , Lectins, C-Type/genetics , Leukotrienes/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Receptors, IgG/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism
19.
J Virol ; 87(16): 9041-52, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23760235

ABSTRACT

Epstein-Barr virus (EBV) alters the regulation and expression of a variety of cytokines in its host cells to modulate host immune surveillance and facilitate viral persistence. Using cytokine antibody arrays, we found that, in addition to the cytokines reported previously, two chemotactic cytokines, CCL3 and CCL4, were induced in EBV-infected B cells and were expressed at high levels in all EBV-immortalized lymphoblastoid cell lines (LCLs). Furthermore, EBV latent membrane protein 1 (LMP1)-mediated Jun N-terminal protein kinase activation was responsible for upregulation of CCL3 and CCL4. Inhibition of CCL3 and CCL4 in LCLs using a short hairpin RNA approach or by neutralizing antibodies suppressed cell proliferation and caused apoptosis, indicating that autocrine CCL3 and CCL4 are required for LCL survival and growth. Importantly, significant amounts of CCL3 were detected in EBV-positive plasma from immunocompromised patients, suggesting that EBV modulates this chemokine in vivo. This study reveals the regulatory mechanism and a novel function of CCL3 and CCL4 in EBV-infected B cells. CCL3 might be useful as a therapeutic target in EBV-associated lymphoproliferative diseases and malignancies.


Subject(s)
B-Lymphocytes/virology , Cell Proliferation , Chemokine CCL3/biosynthesis , Chemokine CCL4/biosynthesis , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions , Viral Matrix Proteins/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/physiology , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction
20.
Proc Natl Acad Sci U S A ; 110(27): E2480-9, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23610399

ABSTRACT

High surface expression of programmed death 1 (PD-1) is associated with T-cell exhaustion; however, the relationship between PD-1 expression and T-cell dysfunction has not been delineated. We developed a model to study PD-1 signaling in primary human T cells to study how PD-1 expression affected T-cell function. By determining the number of T-cell receptor/peptide-MHC complexes needed to initiate a Ca(2+) flux, we found that PD-1 ligation dramatically shifts the dose-response curve, making T cells much less sensitive to T-cell receptor-generated signals. Importantly, other T-cell functions were differentially sensitive to PD-1 expression. We observed that high levels of PD-1 expression were required to inhibit macrophage inflammatory protein 1 beta production, lower levels were required to block cytotoxicity and IFN-γ production, and very low levels of PD-1 expression could inhibit TNF-α and IL-2 production as well as T-cell expansion. These findings provide insight into the role of PD-1 expression in enforcing T-cell exhaustion and the therapeutic potential of PD-1 blockade.


Subject(s)
Programmed Cell Death 1 Receptor/physiology , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Calcium Signaling/immunology , Cells, Cultured , Chemokine CCL4/biosynthesis , Cytotoxicity, Immunologic , HIV-1/immunology , Humans , Interferon-gamma/biosynthesis , Interleukin-2/metabolism , Models, Immunological , Programmed Cell Death 1 Receptor/chemistry , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/physiology , T-Lymphocytes/cytology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL