Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Metab ; 53: 101322, 2021 11.
Article in English | MEDLINE | ID: mdl-34416393

ABSTRACT

OBJECTIVE: It was reported that chemerin as an adipocyte-secreted protein could regulate bone resorption and bone formation. However, the specific molecular and gene mechanism of the chemerin role is unclear. The aim of this study is to evaluate the role of chemerin in bone metabolism. METHODS: In the present study, we investigated the effects of chemerin on bone remodeling in rarres2 knockout (Rarres2-/-) mice and examined the role of chemerin as a determinant of osteoblast and osteoclast differentiation in Mc3t3-E1 and Raw264.7 cell lines. RESULTS: The results showed that the bone mineral density and volume score, trabecular thickness, weight and bone formation marker BALP increased, but Tb.Sp and bone resorption marker TRACP-5b decreased in Rarres2-/- mice. Furthermore, the mRNA and protein expression of biomarkers of osteoblasts (ß-catenin, RANKL and OPG) significantly increased, but those of osteoclasts (CTSK and RANK) decreased in Rarres2-/- mice. In vitro, chemerin markedly suppressed ß-catenin and OPG, but increased RANKL, CTSK and RANK expression. Moreover, knockdown of chemerin using RNA interference enhanced osteoblastogenesis genes and inhibited osteoclastogenesis genes in Mc3t3-E1 and Raw264.7 cells. CONCLUSIONS: Taken together, these data suggest an inhibitive effect of chemerin on osteoblast differentiation and proliferation through inhibition of Wnt/ß-catenin signaling, as well as a stimulative effect of chemerin on osteoclast differentiation and proliferation via activation of RANK signaling. The maintenance of a low chemerin level may be a strategy for the prevention and treatment of osteoporosis.


Subject(s)
Bone Remodeling , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chemokines/deficiency , Female , Intercellular Signaling Peptides and Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism
2.
Int Immunol ; 31(11): 715-728, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31081901

ABSTRACT

Innate-like B-1a cells are an important cell population for production of natural IgM and interleukin-10 (IL-10), and act as the first line against pathogens. We determined that CMTM7 is essential for B-1a cell development. Following Cmtm7 (CKLF-like MARVEL transmembrane domain-containing 7) knockout, B-1a cell numbers decreased markedly in all investigated tissues. Using a bone marrow and fetal liver adoptive transfer model and conditional knockout mice, we showed that the reduction of B-1a cells resulted from B-cell-intrinsic defects. Because of B-1a cell loss, Cmtm7-deficient mice produced less IgM and IL-10, and were more susceptible to microbial sepsis. Self-renewal and homeostasis of mature B-1a cells in Cmtm7-/- mice were not impaired, suggesting the effect of Cmtm7 on B-1a cell development. Further investigations demonstrated that the function of Cmtm7 in B-1a cell development occurred at the specific transitional B-1a (TrB-1a) stage. Cmtm7 deficiency resulted in a slow proliferation and high cell death rate of TrB-1a cells. Thus, Cmtm7 controls B-1a cell development at the transitional stage.


Subject(s)
Chemokines/immunology , MARVEL Domain-Containing Proteins/immunology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , B-Lymphocyte Subsets/immunology , Cell Death , Cell Proliferation , Chemokines/deficiency , MARVEL Domain-Containing Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/immunology
3.
Immunology ; 157(3): 248-256, 2019 07.
Article in English | MEDLINE | ID: mdl-31063220

ABSTRACT

Down-regulated chemerin expression has been reported to correlate with poor prognosis of several types of cancer including melanoma. All-trans retinoic acid (atRA) is a potent inducer of chemerin, and we previously reported that atRA inhibited murine melanoma growth through enhancement of anti-tumor T-cell immunity. Here, we aimed to investigate whether loss of endogenous chemerin accelerated melanoma growth and whether chemerin was involved in the melanoma-inhibitory effect of atRA. We demonstrated that chemerin was constitutively expressed in the skin, which was down-regulated during murine melanoma growth. Rarres2-/- mice, which are deficient in chemerin, exhibited aggravated tumor growth and impaired tumor-infiltrating natural killer (NK) cells that express CMKLR1, the functional receptor of chemerin. Topical treatment with atRA up-regulated skin chemerin expression, which was primarily derived from dermal cells. Moreover, atRA treatment significantly enhanced tumor-infiltrating NK cells, which was completely abrogated in Rarres2-/- mice and Cmklr1-/- mice, suggesting a dependency of NK cell recruitment on the chemerin-CMKLR1 axis in melanoma. Despite comparable melanoma growth detected in wild-type mice and Cmklr1-/- mice, lack of CMKLR1 partially abrogated the melanoma-inhibitory effect of atRA. This may be due to the inability to enhance tumor-infiltrating NK cells in Cmklr1-/- mice following atRA treatment. Collectively, our study suggests that down-regulation of chemerin could be a strategy used by cancers such as melanoma to impair anti-tumor NK cell immunity and identifies a new anti-tumor mechanism of atRA by up-regulating chemerin to enhance CMKLR1-dependent NK cell recruitment.


Subject(s)
Antineoplastic Agents/pharmacology , Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Killer Cells, Natural/drug effects , Melanoma, Experimental/drug therapy , Receptors, G-Protein-Coupled/metabolism , Skin Neoplasms/drug therapy , Tretinoin/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokines/deficiency , Chemokines/genetics , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Receptors, Chemokine , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Tumor Burden/drug effects , Tumor Escape , Tumor Microenvironment
4.
Arterioscler Thromb Vasc Biol ; 39(4): 685-693, 2019 04.
Article in English | MEDLINE | ID: mdl-30786742

ABSTRACT

Objective- Expression of the chemokine-like receptor ChemR23 (chemerin receptor 23) has been specifically attributed to plasmacytoid dendritic cells (pDCs) and macrophages and ChemR23 has been suggested to mediate an inflammatory immune response in these cells. Because chemokine receptors are important in perpetuating chronic inflammation, we aimed to establish the role of ChemR23-deficiency on macrophages and pDCs in atherosclerosis. Approach and Results- ChemR23-knockout/knockin mice expressing eGFP (enhanced green fluorescent protein) were generated and after crossing with apolipoprotein E-deficient ( Apoe-/- ChemR23 e/e) animals were fed a western-type diet for 4 and 12 weeks. Apoe-/- ChemR23 e/e mice displayed reduced lesion formation and reduced leukocyte adhesion to the vessel wall after 4 weeks, as well as diminished plaque growth, a decreased number of lesional macrophages with an increased proportion of M2 cells and a less inflammatory lesion composition after 12 weeks of western-type diet feeding. Hematopoietic ChemR23-deficiency similarly reduced atherosclerosis. Additional experiments revealed that ChemR23-deficiency induces an alternatively activated macrophage phenotype, an increased cholesterol efflux and a systemic reduction in pDC frequencies. Consequently, expression of the pDC marker SiglecH in atherosclerotic plaques of Apoe-/- ChemR23 e/e mice was declined. ChemR23-knockout pDCs also exhibited a reduced migratory capacity and decreased CCR (CC-type chemokine receptor)7 expression. Finally, adoptive transfer of sorted wild-type and knockout pDCs into Apoe-/- recipient mice revealed reduced accumulation of ChemR23-deficient pDCs in atherosclerotic lesions. Conclusions- Hematopoietic ChemR23-deficiency increases the proportion of alternatively activated M2 macrophages in atherosclerotic lesions and attenuates pDC homing to lymphatic organs and recruitment to atherosclerotic lesions, which synergistically restricts atherosclerotic plaque formation and progression.


Subject(s)
Atherosclerosis/metabolism , Chemokines/physiology , Dendritic Cells/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Macrophages/metabolism , Animals , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Cell Adhesion , Chemokines/deficiency , Chemokines/genetics , Cholesterol/metabolism , Diet, Western/adverse effects , Disease Progression , Female , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, Reporter , Inflammation , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Receptors, CCR7/metabolism
5.
J Interferon Cytokine Res ; 38(9): 363-369, 2018 09.
Article in English | MEDLINE | ID: mdl-30230981

ABSTRACT

Inflammation is mediated by cytokines and chemokines, which are considered targets of inflammatory diseases. Mounting evidence has demonstrated the anti-inflammatory benefits of metformin. However, the underlying mechanisms are not completely understood. In this study, we aim to elucidate the regulatory effects of metformin on chemokine expression and the possible mechanisms using RAW264.7 cells, a mouse macrophage cell line, as a model. First, we treated the cells with lipopolysaccharide (LPS), and found that the expression of CXCL10 and CXCL11 was markedly induced in a dose- and time-dependent fashion concurrent with the inhibition of AMPK activity. Then, we treated the cells with metformin, and analyzed the expression of CCL2, CXCL10, and CXCL11 by quantitative real-time polymerase chain reaction (PCR). We observed that metformin prevented the stimulating effect of LPS on these chemokines as well as IL-1 and IL-6. Second, the inhibitory effects of metformin on LPS-induced chemokine expression were diminished by Compound C, a chemical inhibitor of AMPK. Finally, we investigated whether the NF-κB signaling pathway is regulated by metformin in this setting. Our results showed that metformin inhibited the phosphorylation of I-κBα and p65 while it activated AMPK. Therefore, the results suggest that metformin inhibits LPS-induced chemokine expression through the AMPK and NF-κB signaling pathways.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Chemokines/biosynthesis , Chemokines/deficiency , Metformin/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Animals , Chemokines/genetics , Chemokines/metabolism , Gene Expression Profiling , Mice , RAW 264.7 Cells
6.
J Pharmacol Exp Ther ; 365(2): 212-218, 2018 05.
Article in English | MEDLINE | ID: mdl-29467310

ABSTRACT

Chemerin is an inflammatory adipokine positively associated with hypertension and obesity. The majority of chemerin derives from the liver and adipose tissue, however, their individual contributions to blood pressure are unknown. We began studying chemerin in the normal rat using antisense oligonucleotides (ASO) with whole-body activity (Gen 2.5 chemerin ASO) or liver-restricted activity (GalNAc chemerin ASO). We hypothesized that in normotensive male Sprague-Dawley rats, circulating chemerin is predominately liver-derived and regulates blood pressure. A dosing study of the Gen 2.5 chemerin ASO (with a scrambled control ASO) supported 25 mg/kg as the appropriate dose. GalNAc chemerin ASO was also assessed and used at 10 mg/kg. Radiotelemetry monitored mean arterial pressure (MAP) for a 1-week baseline and weekly subcutaneous ASO injections for 4 weeks. Two days after the final injection, animals were euthanized for tissue reverse transcription-polymerase chain reaction and chemerin Western analysis. Gen 2.5 chemerin ASO treatments reduced chemerin mRNA and protein in liver, retroperitoneal fat (RP), and mesenteric perivascular adipose tissue (mPVAT), as well as reducing protein in plasma. GalNAc chemerin ASO treatments reduced chemerin mRNA and protein in liver and chemerin protein in plasma but had no effect on expression in RP fat or mPVAT. Gen 2.5 chemerin ASO treatment reduced MAP compared with control ASO but was unchanged in animals receiving the GalNAc chemerin ASO. Although circulating chemerin is liver-derived, it does not play a major role in blood pressure regulation. Local effects of chemerin from fat may explain this discrepancy and support chemerin's association with hypertension and obesity.


Subject(s)
Blood Pressure/genetics , Chemokines/deficiency , Chemokines/genetics , Gene Knockdown Techniques , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Liver/metabolism , Oligonucleotides, Antisense/genetics , Animals , Male , Rats , Rats, Sprague-Dawley , Time Factors
7.
Elife ; 52016 10 20.
Article in English | MEDLINE | ID: mdl-27763263

ABSTRACT

Integrin activation is crucial for the regulation of leukocyte rolling, adhesion and trans-vessel migration during inflammation and occurs by engagement of myeloid cells through factors presented by inflamed vessels. However, endothelial-dependent mechanisms of myeloid cell recruitment are not fully understood. Here we show using an autoperfused flow chamber assay of whole blood neutrophils and intravital microscopy of the inflamed cremaster muscle that CD95 mediates leukocyte slow rolling, adhesion and transmigration upon binding of CD95-ligand (CD95L) that is presented by endothelial cells. In myeloid cells, CD95 triggers activation of Syk-Btk/PLCγ2/Rap1 signaling that ultimately leads to integrin activation. Excitingly, CD95-deficient myeloid cells exhibit impaired bacterial clearance in an animal model of sepsis induced by cecal ligation and puncture (CLP). Our data identify the cellular and molecular mechanisms underlying the chemoattractant effect of endothelial cell-derived CD95L in induction of neutrophil recruitment and support the use of therapeutic inhibition of CD95's activity in inflammatory diseases.


Subject(s)
Cell Adhesion , Chemokines/metabolism , Endothelial Cells/chemistry , Fas Ligand Protein/metabolism , Locomotion , Neutrophils/drug effects , Neutrophils/physiology , Abdominal Muscles/pathology , Animals , Bacterial Infections/immunology , Cell Movement , Chemokines/deficiency , Disease Models, Animal , Fas Ligand Protein/deficiency , Mice, Inbred C57BL , Microscopy , Myositis/pathology , Sepsis/immunology
8.
Adv Healthc Mater ; 5(6): 730-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26853449

ABSTRACT

The aim is to evaluate the effect of modifying poly[(l-lactide)-co-(ε-caprolactone)] scaffolds (PLCL) with nanodiamonds (nDP) or with nDP+physisorbed BMP-2 (nDP+BMP-2) on in vivo host tissue response and degradation. The scaffolds are implanted subcutaneously in Balb/c mice and retrieved after 1, 8, and 27 weeks. Molecular weight analysis shows that modified scaffolds degrade faster than the unmodified. Gene analysis at week 1 shows highest expression of proinflammatory markers around nDP scaffolds; although the presence of inflammatory cells and foreign body giant cells is more prominent around the PLCL. Tissue regeneration markers are highly expressed in the nDP+BMP-2 scaffolds at week 8. A fibrous capsule is detectable by week 8, thinnest around nDP scaffolds and at week 27 thickest around PLCL scaffolds. mRNA levels of ALP, COL1α2, and ANGPT1 are significantly upregulating in the nDP+BMP-2 scaffolds at week 1 with ectopic bone seen at week 8. Even when almost 90% of the scaffold is degraded at week 27, nDP are observable at implantation areas without adverse effects. In conclusion, modifying PLCL scaffolds with nDP does not aggravate the host response and physisorbed BMP-2 delivery attenuates inflammation while lowering the dose of BMP-2 to a relatively safe and economical level.


Subject(s)
Bone Morphogenetic Protein 2/chemistry , Nanodiamonds/chemistry , Polyesters/chemistry , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biocompatible Materials/pharmacology , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration/physiology , Bone and Bones/diagnostic imaging , Bone and Bones/physiology , Chemokines/deficiency , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Female , Injections, Subcutaneous , Mice , Mice, Inbred BALB C , Neovascularization, Physiologic , Prostheses and Implants , Skin/metabolism , Skin/pathology , Up-Regulation/drug effects , X-Ray Microtomography
9.
J Immunol ; 177(3): 1817-24, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16849492

ABSTRACT

Influenza A virus pneumonia is characterized by severe lung injury and high mortality. Early infection elicits a strong recruitment of monocytes from the peripheral blood across the endo-/epithelial barrier into the alveolar air space. However, it is currently unclear which of the infected resident lung cell populations, alveolar epithelial cells or alveolar macrophages, elicit monocyte recruitment during influenza A virus infection. In the current study, we investigated whether influenza A virus infection of primary alveolar epithelial cells and resident alveolar macrophages would elicit a basal-to-apical monocyte transepithelial migration in vitro. We found that infection of alveolar epithelial cells with the mouse-adapted influenza A virus strain PR/8 strongly induced the release of monocyte chemoattractants CCL2 and CCL5 followed by a strong monocyte transepithelial migration, and this monocytic response was strictly dependent on monocyte CCR2 but not CCR5 chemokine receptor expression. Analysis of the adhesion molecule pathways demonstrated a role of ICAM-1, VCAM-1, integrin-associated protein (CD47), and junctional adhesion molecule-c on the epithelial cell surface interacting with monocyte beta(1) and beta(2) integrins and integrin-associated protein in the monocyte transmigration process. Importantly, addition of influenza A virus-infected alveolar macrophages further enhanced monocyte transmigration across virus-infected epithelium in a TNF-alpha-dependent manner. Collectively, the data show an active role for virus-infected alveolar epithelium in the regulation of CCL2/CCR2-dependent monocyte transepithelial migration during influenza infection that is essentially dependent on both classical beta(1) and beta(2) integrins but also junctional adhesion molecule pathways.


Subject(s)
Cell Adhesion Molecules/physiology , Cell Movement/immunology , Chemokines/physiology , Epithelial Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Monocytes/immunology , Pulmonary Alveoli/cytology , Pulmonary Alveoli/immunology , Animals , Cell Adhesion Molecules/biosynthesis , Cell Communication/immunology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Chemokines/deficiency , Chemokines/genetics , Epithelial Cells/chemistry , Epithelial Cells/virology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/chemistry , Monocytes/virology , Nucleocapsid Proteins , Nucleoproteins/analysis , Pulmonary Alveoli/virology , RNA-Binding Proteins/analysis , Receptors, CCR2 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Chemokine/physiology , Up-Regulation/immunology , Viral Core Proteins/analysis
10.
J Immunol ; 173(10): 6161-8, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15528353

ABSTRACT

The lymphotoxin (LT) beta receptor plays a critical role in secondary lymphoid organogenesis and the classical and alternative NF-kappaB pathways have been implicated in this process. IKKalpha is a key molecule for the activation of the alternative NF-kappaB pathway. However, its precise role and target genes in secondary lymphoid organogenesis remain unknown, particularly with regard to high endothelial venules (HEV). In this study, we show that IKKalpha(AA) mutant mice, who lack inducible kinase activity, have hypocellular lymph nodes (LN) and nasal-associated lymphoid (NALT) tissue characterized by marked defects in microarchitecture and HEV. In addition, IKKalpha(AA) LNs showed reduced lymphoid chemokine CCL19, CCL21, and CXCL13 expression. IKKalpha(AA) LN- and NALT-HEV were abnormal in appearance with reduced expression of peripheral node addressin (PNAd) explained by a severe reduction in the HEV-associated proteins, glycosylation-dependent cell adhesion molecule 1 (GlyCAM-1), and high endothelial cell sulfotransferase, a PNAd-generating enzyme that is a target of LTalphabeta. In this study, analysis of LTbeta(-/-) mice identifies GlyCAM-1 as another LTbeta-dependent gene. In contrast, TNFRI(-/-) mice, which lose classical NF-kappaB pathway activity but retain alternative NF-kappaB pathway activity, showed relatively normal GlyCAM-1 and HEC-6ST expression in LN-HEV. In addition, in this communication, it is demonstrated that LTbetaR is prominently expressed on LN- and NALT-HEV. Thus, these data reveal a critical role for IKKalpha in LN and NALT development, identify GlyCAM-1 and high endothelial cell sulfotransferase as new IKKalpha-dependent target genes, and suggest that LTbetaR signaling on HEV can regulate HEV-specific gene expression.


Subject(s)
Chemokines/biosynthesis , Chemokines/genetics , Endothelium, Lymphatic/metabolism , Gene Expression Regulation, Developmental/immunology , Lymph Nodes/enzymology , Nasal Mucosa/enzymology , Protein Serine-Threonine Kinases/physiology , Protein Subunits/physiology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Chemokines/deficiency , Endothelium, Lymphatic/immunology , Endothelium, Lymphatic/pathology , Enzyme Activation/genetics , Enzyme Activation/immunology , I-kappa B Kinase , Ligands , Lymph Nodes/growth & development , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoid Tissue/enzymology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Lymphotoxin beta Receptor , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Nasal Mucosa/growth & development , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Subunits/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Receptors, Tumor Necrosis Factor/physiology
11.
J Allergy Clin Immunol ; 111(2): 227-42; quiz 243, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12589338

ABSTRACT

The asthmatic response is characterized by elevated production of IgE, cytokines, chemokines, mucus hypersecretion, air-way obstruction, eosinophilia, and enhanced airway hyperreactivity to spasmogens. Clinical and experimental investigations have demonstrated a strong correlation between the presence of CD4+ TH2 cells, eosinophils, and disease severity, suggesting an integral role for these cells in the pathophysiology of asthma. TH2 cells are thought to induce asthma through the secretion of an array of cytokines (IL-4, -5, -9 -1),-13, -25) that activate inflammatory and residential effector pathways both directly and indirectly. In particular, IL-4 and IL-13 are produced at elevated levels in the asthmatic lung and are thought to be central regulators of many of the hallmark features of the disease. The potency of IL-13 in promoting airway hyperreactivity and mucus hypersecretion and the ability of IL-13 blockade to abrogate several critical aspects of experimental asthma have led to the view that this is a critical cytokine in disease pathogenesis. Extensive studies have also demonstrated a central role for chemokines in orchestrating multiple aspects of the asthmatic response. Chemokines are potent leukocyte chemoattractants, cellular activating factors, and histamine-releasing factors, which makes them particularly important in the pathogenesis of allergic inflammation. In particular, the eotaxin subfamily of chemokines and their receptor CC chemokine receptor 3 have emerged as central regulators of the asthmatic response. Recent studies have provided an integrated mechanism by which to explain the coordinate interaction between IL-13 and chemokines in the pathogenesis of asthma. In this regard, chemokines and IL-13 are attractive new therapeutic targets for the treatment of allergic disease. This article focuses on recently emerging data pertaining to the importance of chemokines, especially eotaxins, in promoting IL-13-associated allergic lung responses, as well as the potential for pharmacologically targeting these pathways.


Subject(s)
Asthma/immunology , Chemokines/metabolism , Interleukin-13/metabolism , Animals , Asthma/genetics , Chemokines/deficiency , Chemokines/genetics , Eosinophils/immunology , Humans , Hypersensitivity/immunology , Interleukin-4/metabolism , Mice , Mice, Knockout , Models, Immunological , Polymorphism, Genetic , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Signal Transduction , Th2 Cells/immunology
12.
Curr Top Microbiol Immunol ; 265: 23-48, 2002.
Article in English | MEDLINE | ID: mdl-12014193

ABSTRACT

Cytokines and chemokines are potent biologic response molecules that play a key role in cellular communication in physiologic and pathophysiologic states. An understanding of the actions and roles of these molecules in CNS biology has been greatly facilitated by molecular genetic approaches that permit the targeted manipulation of gene expression in an intact organism. Studies in promoter-driven transgenic mice with CNS production of a number of cytokines or chemokines have demonstrated that these factors can directly induce a spectrum of cellular alterations often resulting in pronounced neurological disease (Table 1). Thus, these factors, in addition to initiating and maintaining immunoinflammatory responses, can be direct mediators of CNS injury. The neuropathological outcomes in the transgenic mice often recapitulate those reported in human neurological disorders such as MS, neurological diseases associated with AIDS and Alzheimer's disease, pointing to the importance of these animal models to our understanding of the role of cytokines and chemokines in these human disorders. Despite problems of timing and tissue specificity as well as some inconsistencies in the findings from different groups, knockout mice have begun to provide insights that are altering our view of the contribution made by individual cytokines to immunoinflammatory responses in the brain. For example, IL-6 and TNF were originally viewed as having minor and major proinflammatory contributions, respectively, in EAE, but now, based on findings from knockout mice, the opposite seems true. Studies in transgenic and knockout mice now offer strong evidence that, in addition to being mediators of damage, cytokines can have beneficial functions, e.g. the antiviral functions of the IFNs or the trophic and/or neuroprotective actions of some cytokines such as IL-6 and TNF. Clearly, studies in mutant mice, as summarized here, will continue to provide important insights into the nature of cytokine and chemokine actions in the CNS and will offer the possibility that we may identify new targets for effective therapeutic intervention in neuroinflammatory disorders.


Subject(s)
Central Nervous System/immunology , Chemokines/immunology , Cytokines/immunology , Animals , Animals, Genetically Modified , Central Nervous System/pathology , Chemokines/deficiency , Chemokines/genetics , Cytokines/deficiency , Cytokines/genetics , Interleukins/immunology , Mice , Mice, Knockout
13.
Atherosclerosis ; 159(2): 253-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11730804

ABSTRACT

This paper reviews published studies since 1995 dealing with many atherogenic mechanisms where exogenous heparin was beneficial. In these areas endogenous heparin deficiency is likely to be harmful. Mechanisms included inflammatory factors, lower endogenous plasma heparin levels, lipoprotein lipase, chemokines, APOE e4, lipoprotein(a), among others. Demonstrated reduction of heparan sulfate proteoglycans (HSPG) and of endogenous plasma heparin was reviewed.


Subject(s)
Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Heparin/deficiency , Heparin/metabolism , Animals , Chemokines/deficiency , Chemokines/metabolism , Haplorhini , Humans , Lipoprotein Lipase/deficiency , Lipoprotein Lipase/metabolism , Peptidyl-Dipeptidase A/deficiency , Peptidyl-Dipeptidase A/metabolism , Phospholipases A/deficiency , Phospholipases A/metabolism , Prognosis , Risk Factors , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...