Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-33308119

ABSTRACT

BACKGROUND: Neonatal sepsis (NS) remains one of the leading causes of mortality among newborns. This study found the deregulated microRNA-96 (miR-96) in NS neonates, and aimed to evaluate the clinical significance of miR-96, as well as its effect on LPS-induced inflammatory response in monocytes. In addition, the relationship of interleukin-16 (IL-16) and miR-96 was investigated to understand the underlying mechanisms. METHODS: Expression of miR-96 was examined using real-time quantitative PCR. Monocytes stimulated by LPS was used to mimic excessive inflammation in the pathogenesis of NS. The enzyme-linked immunosorbent assay was applied to evaluate pro-inflammatory cytokine levels. A luciferase reporter assay was used to confirm the interaction between miR-96 and IL-16. RESULTS: Serum miR-96 expression was decreased in NS newborns and had considerable diagnostic value for NS screening. LPS inhibited miR-96 expression in monocytes, and the overexpression of miR-96 could reverse the effects of LPS on the inflammation of monocytes. IL-16 was a target gene of miR-96 and negatively correlated with miR-96 levels in NS neonates. The inhibited inflammatory responses induced by miR-96 overexpression was abolished by the elevated IL-16 in monocytes. CONCLUSION: All the data reveal that serum decreased miR-96 may serve as a candidate noninvasive biomarker for NS diagnosis. In addition, miR-96 inhibits LPS-induced inflammatory responses by targeting IL-16 in monocytes. The miR-96/IL-16 axis may provide novel therapeutic targets for NS treatment.


Subject(s)
MicroRNAs , Sepsis , Down-Regulation , Humans , Infant, Newborn , Interleukin-16/antagonists & inhibitors , Interleukin-16/genetics , Lipopolysaccharides , MicroRNAs/genetics , MicroRNAs/metabolism , Monocytes/metabolism , Sepsis/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Tumour Biol ; 43(1): 309-325, 2021.
Article in English | MEDLINE | ID: mdl-34897107

ABSTRACT

BACKGROUND: Cytokines play an important role in the immune response, angiogenesis, cell growth, and differentiation in hepatocellular carcinoma (HCC). OBJECTIVE: We performed a comprehensive study to identify tumor-related cytokines and pathways involved in HCC pathogenesis. METHODS: Cytokine production was evaluated in human HCC tissues and adjacent non-tumor tissues using an antibody-based protein array technique. We compared cytokine expression in HCC tissues with that of hepatic hemangioma (HH), liver metastasis of colorectal cancer, and noncancerous liver tissues from transplantation donors. The protein levels and localization of the candidate cytokines were analyzed by western blotting and immunohistochemistry. RESULTS: Increased expression of interleukin (IL)-1 receptor antagonist, macrophage migration inhibitory factor, and IL-16 was observed in HCC and paired adjacent non-tumor tissues compared with noncancerous livers. In addition, there were increased IL-16 levels in HCC tissues compared with HH. IL-16 treatment significantly increased cell proliferation in vitro. The expression of extracellular signal-regulated kinase (ERK)1/2 and cyclin D1 was markedly increased in cells from two HCC cell lines, Huh7 and HepG2, in a dose- and time-dependent manner. Phosphorylated to total ERK1/2 ratio was increased in Huh7 cells following IL-16 50 ng/ml, but not HepG2 cells. ERK phosphorylation have occurred earlier than protein accumulation at 48 h. Pretreatment with the ERK inhibitor, FR18024, or an anti-IL-16 antibody reduced the increase in IL-16 production in HCC cells. CONCLUSIONS: These results suggest that cell proliferation induced by IL-16 is mediated through the ERK pathway, thus, we identified a new factor associated with HCC tumor growth.


Subject(s)
Carcinoma, Hepatocellular/genetics , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-16/genetics , Liver Neoplasms/genetics , Liver/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Hemangioma/drug therapy , Hemangioma/genetics , Hemangioma/pathology , Hep G2 Cells , Humans , Interleukin-16/antagonists & inhibitors , Interleukin-16/biosynthesis , Interleukin-16/pharmacology , Liver/metabolism , Liver/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Macrophage Migration-Inhibitory Factors/genetics , Neoplasm Metastasis , Proteomics
3.
Mediators Inflamm ; 2021: 6611085, 2021.
Article in English | MEDLINE | ID: mdl-33958974

ABSTRACT

BACKGROUND: Interleukin-16 (IL-16) is an important inflammatory regulator and has been shown to have a powerful effect on the regulation of the inflammatory response. Cardiac inflammation has been reported to be closely related to doxorubicin- (DOX-) induced cardiac injury. In this study, the role of IL-16 in DOX-induced cardiac injury and the possible mechanisms were examined. METHODS: Cardiac IL-16 levels were first measured in DOX- or saline-treated mice. Additionally, mice were pretreated with the anti-IL-16-neutralizing antibody (nAb) or isotype IgG for 1 day and further administered DOX or saline for 5 days. Then, cardiac injury, cardiac M1 macrophage levels, and cardiomyocyte apoptosis were analyzed. The effects of the anti-IL-16 nAb on macrophage differentiation and cardiomyocyte apoptosis were also investigated in vitro. RESULTS: DOX administration increased IL-16 expression in cardiac macrophages compared with that of saline treatment. The anti-IL-16 nAb significantly decreased serum levels of lactate dehydrogenase (LDH), myocardial-bound creatine kinase (CK-MB), and cardiac troponin T (cTnT) and elevated cardiac function in DOX-induced mice. Treatment with the anti-IL-16 nAb also reduced p65 pathway activation, decreased M1 macrophage-related marker and cytokine expression, and protected against cardiomyocyte apoptosis in DOX-induced mice. In cell studies, the anti-IL-16 nAb also reduced DOX-induced M1 macrophage differentiation and alleviated apoptosis in cardiomyocytes cocultured with macrophages. CONCLUSIONS: The anti-IL-16 nAb protects against DOX-induced cardiac injury by reducing cardiac inflammation, and IL-16 may be a promising target to prevent DOX-related cardiac injury.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Cardiotoxicity/prevention & control , Doxorubicin/toxicity , Interleukin-16/antagonists & inhibitors , Myocarditis/drug therapy , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Interleukin-16/physiology , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology
4.
Oxid Med Cell Longev ; 2021: 6616422, 2021.
Article in English | MEDLINE | ID: mdl-33628366

ABSTRACT

Several interleukin (IL) members have been reported to participate in sepsis. In this study, the effects of IL-16 on sepsis-induced cardiac injury and dysfunction were examined, and the related mechanisms were detected. IL-16 expression in septic mice was first measured, and the results showed that both cardiac and serum IL-16 expression levels were increased in mice with sepsis induced by LPS or cecal ligation and puncture (CLP) compared with control mice. Then, IL-16 was neutralized, and the effects on lipopolysaccharide- (LPS-) induced cardiac injury were detected. The results showed that an anti-IL-16 neutralizing antibody (nAb) significantly reduced mortality and increased serum lactate dehydrogenase (LDH), creatine kinase myocardial bound (CK-MB), and cardiac troponin T (cTnT) levels while improving cardiac function in mice with LPS-induced sepsis. Neutralization of IL-16 also increased the activation of antioxidant pathways and the expression of antioxidant factors in septic mice while decreasing the activation of prooxidant pathways and the expression of prooxidants. Treatment with the anti-IL-16 nAb increased mitochondrial apoptosis-inducing factor (AIF) expression, decreased nuclear AIF and cleaved poly-ADP-ribose polymerase (PARP) expression, and decreased TUNEL-positive cell percentages in LPS-treated mice. Additionally, treatment with CPUY192018, the nuclear factor erythroid-2 related factor 2 (Nrf2) pathway, significantly increased mortality and reversed the above effects in mice treated with LPS and the anti-IL-16 nAb. Our results showed that the anti-IL-16 nAb regulates oxidative stress through the Nrf2 pathway and participates in the regulation of cardiac injury in septic mice. Neutralization of IL-16 may be a beneficial strategy for the prevention of cardiac injury and dysfunction in sepsis patients.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Interleukin-16/antagonists & inhibitors , Myocardium/pathology , NF-E2-Related Factor 2/metabolism , Sepsis/complications , Sepsis/physiopathology , Signal Transduction , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis/drug effects , Cell Line , Interleukin-16/metabolism , Lipopolysaccharides , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Sepsis/pathology , Signal Transduction/drug effects
5.
Int J Mol Sci ; 21(8)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326301

ABSTRACT

Because of lipopolysaccharide (LPS)-mediated effects on osteoclast differentiation and bone loss, periprosthetic joint infection (PJI) caused by Gram-negative bacteria increases the risk of aseptic loosening after reimplantation. Synovial fluid interleukin-16 (IL-16) expression was higher in patients with PJI than in patients without joint infection. Thus, we explored the effects of IL-16 on bone. We investigated whether IL-16 modulates osteoclast or osteoblast differentiation in vitro. An LPS-induced bone loss mice model was used to explore the possible advantages of IL-16 inhibition for the prevention of bone loss. IL-16 directly activated p38 and c-Jun N-terminal kinase (JNK)/mitogen-activated protein kinase (MAPK) signaling and increased osteoclast activation markers, including tartrate-resistant acid phosphatase (TRAP), cathepsin K, and nuclear factor of activated T cells 1 (NFATc1). IL-16 directly caused monocytes to differentiate into TRAP-positive osteoclast-like cells through NFATc1 activation dependent on JNK/MAPK signaling. Moreover, IL-16 did not alter alkaline phosphatase activity or calcium deposition during osteoblastic differentiation. Finally, IL-16 inhibition prevented LPS-induced trabecular bone loss and osteoclast activation in vivo. IL-16 directly increased osteoclast activation through the JNK/NFATc1 pathway. IL-16 inhibition could represent a new strategy for treating infection-associated bone loss.


Subject(s)
Arthritis, Infectious/metabolism , Bone Resorption/metabolism , Interleukin-16/metabolism , MAP Kinase Signaling System , Osteoclasts/metabolism , Prosthesis-Related Infections/metabolism , Synovial Fluid/metabolism , Animals , Arthritis, Infectious/etiology , Biomarkers , Cathepsin K/genetics , Cathepsin K/metabolism , Gene Expression , Immunohistochemistry , Interleukin-16/antagonists & inhibitors , Lipopolysaccharides/immunology , Mice , Models, Biological , Prosthesis-Related Infections/microbiology , RAW 264.7 Cells
6.
Clin Dermatol ; 35(4): 375-382, 2017.
Article in English | MEDLINE | ID: mdl-28709568

ABSTRACT

The pathogenesis of atopic dermatitis (AD) involves epidermal barrier dysfunction and T helper cell type 2 (Th2) lymphocyte-driven inflammation. Cytokines, such as interleukin 4 (IL-4) and IL-13, are important in this reaction. They stimulate B cells to produce immunoglobulin E, causing atopic disease. This process has been well characterized, and new therapies for AD, such as phosphodiesterase 4 (PDE-4) inhibitors, Th2-expressed chemoattractant receptor-homologous molecule antagonists, and Janus kinase inhibitors, work by antagonizing this cellular pathway. Recently, there have been many advances in treatment strategies and novel therapies for AD. This review summarizes the clinical evidence supporting the use of current and emerging topical treatments for AD, as well as their safety and efficacy profiles. Crisaborole, a novel PDE-4 inhibitor, is of particular note because phase III clinical trials were recently completed, as summarized here. It is prudent for dermatologists to be current with updates in the field because therapies are constantly changing. In addition to the academic interest, this results in improvement of patient care and advancement of the field.


Subject(s)
Dermatitis, Atopic/therapy , Administration, Topical , Boron Compounds/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Clinical Trials, Phase III as Topic , Dermatitis, Atopic/immunology , Humans , Interleukin-16/antagonists & inhibitors , Janus Kinase Inhibitors/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use
7.
J Nat Prod ; 76(11): 2120-5, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24200352

ABSTRACT

The therapeutic effect of corilagin (1) was evaluated in an acute colitis model induced by dextran sulfate sodium (DSS) in mice, and the mechanism of action was investigated in this study. Animals were challenged with 2% DSS drinking water for 5 consecutive days and then intraperitoneally treated with 1 (7.5, 15, and 30 mg/kg) daily for 7 days. It was found that 1 significantly decreased the disease activity index, inhibited the shortening of colon length, reduced colon tissue damage, and suppressed myeloperoxidase activity. Moreover, 1 greatly suppressed the secretion of TNF-α, IL-6, and IL-1ß, inhibited the degradation of IκB α, and down-regulated expression of cleaved caspase-3 and cleaved caspase-9 in colon tissues of DSS-treated mice. These findings demonstrated that 1 exerts a protective effect on DSS-induced colitis, and its underlying mechanisms are associated with inhibition of the NF-κB pathway that mitigates colon inflammatory responses and apoptosis of intestinal epithelial cells.


Subject(s)
Colitis, Ulcerative/chemically induced , Dextran Sulfate/adverse effects , Glucosides/pharmacology , Hydrolyzable Tannins/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colon/enzymology , Colon/metabolism , Drinking Water/chemistry , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Glucosides/chemistry , Interleukin-16/antagonists & inhibitors , Interleukin-16/metabolism , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Male , Mice , Mice, Inbred C57BL , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Peroxidase/antagonists & inhibitors , Peroxidase/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
Glia ; 58(14): 1649-62, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20578037

ABSTRACT

In contrast to mammals, the medicinal leech Hirudo medicinalis can completely repair its central nervous system (CNS) after injury. This invertebrate model offers unique opportunities to study the molecular and cellular basis of the CNS repair processes. When the leech CNS is injured, microglial cells migrate and accumulate at the site of lesion, a phenomenon known to be essential for the usual sprouting of injured axons. In the present study, we demonstrate that a new molecule, designated HmIL-16, having functional homologies with human interleukin-16 (IL-16), has chemotactic activity on leech microglial cells as observed using a gradient of human IL-16. Preincubation of microglial cells either with an anti-human IL-16 antibody or with anti-HmIL-16 antibody significantly reduced microglia migration induced by leech-conditioned medium. Functional homology was demonstrated further by the ability of HmIL-16 to promote human CD4+ T cell migration which was inhibited by antibody against human IL-16, an IL-16 antagonist peptide or soluble CD4. Immunohistochemistry of leech CNS indicates that HmIL-16 protein present in the neurons is rapidly transported and stored along the axonal processes to promote the recruitment of microglial cells to the injured axons. To our knowledge, this is the first identification of a functional interleukin-16 homologue in invertebrate CNS. The ability of HmIL-16 to recruit microglial cells to sites of CNS injury suggests a role for HmIL-16 in the crosstalk between neurons and microglia in the leech CNS repair.


Subject(s)
Cell Movement/physiology , Ganglia, Invertebrate/cytology , Ganglia, Invertebrate/injuries , Hirudo medicinalis/cytology , Hirudo medicinalis/physiology , Interleukin-16/physiology , Microglia/physiology , Sequence Homology, Amino Acid , Animals , Cells, Cultured , Disease Models, Animal , Ganglia, Invertebrate/physiology , Humans , Interleukin-16/antagonists & inhibitors , Microglia/cytology
9.
Clin Immunol ; 108(2): 89-94, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12921754

ABSTRACT

Langerhans-like dendritic cells (LLDC) express the high-affinity IgE receptor FcepsilonRI form that lacks the beta-chain, and may play an important role in allergic inflammation via production of IL-16. Secretion of mediators by human mast cells and basophils is mediated through FcepsilonRI and is decreased by coaggregating these receptors to the low-affinity IgG receptor, FcgammaRII. We used a recently described human Ig fusion protein (GE2), which is composed of key portions of the human gamma1 and the human epsilon heavy chains, to investigate its ability to inhibit IL-16 production from FcepsilonRI-positive Langerhans-like dendritic cells through coaggregation of FcgammaRII and FcepsilonRI. Unstimulated LLDC-derived from CD14-positive monocytes from atopic donors were shown to express FcgammaRII, an ITIM-containing receptor, but not FcepsilonRI or FcgammaRIII which are activating (ITAM) receptors. When passively sensitized with antigen-specific, human IgE and then challenged with antigen, LLDC were stimulated to produce IL-16. However, when FcepsilonRI and FcgammaRII were coaggregated with GE2, IL-16 production was significantly inhibited. Exposure of LLDCs to GE2 alone did not induce IL-16 production. Our results further extend our studies demonstrating the ability of GE2 to inhibit FcepsilonRI-mediated responses through coaggregation with FcgammaRIIB and at the same time show that human LDCC can be modulated in a fashion similar to mast cells and basophils.


Subject(s)
Interleukin-16/biosynthesis , Langerhans Cells/immunology , Receptor Aggregation , Receptors, IgE/immunology , Receptors, IgG/immunology , Cells, Cultured , Humans , Interleukin-16/antagonists & inhibitors , Langerhans Cells/metabolism , Receptor Aggregation/physiology , Receptors, IgE/biosynthesis , Receptors, IgG/biosynthesis , Signal Transduction
10.
J Immunol ; 168(2): 942-50, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777993

ABSTRACT

Thyroid-associated ophthalmopathy and dermopathy are connective tissue manifestations of Graves' disease (GD). Tissue remodeling is a prominent feature of both and is apparently driven by recruited T cells. In this study, we report that IgG isolated from patients with GD (GD-IgG) up-regulates T lymphocyte chemoattractant activity in GD-derived fibroblasts from orbit, thyroid, and several regions of skin. This chemoattractant activity, absent in fibroblasts from donors without known thyroid disease, is partially susceptible to neutralization by anti-IL-16 and anti-RANTES Abs. IL-16 is a CD4(+)-specific chemoattractant and RANTES is a C-C-type chemokine. IL-16 and RANTES protein levels, as determined by specific ELISAs, are substantially increased by GD-IgG in GD fibroblasts. Addition of the macrolide, rapamycin, to fibroblast culture medium blocked the up-regulation by GD-IgG of IL-16, implicating the FRAP/mTOR/p70(s6k) pathway in the induction of IL-16 expression. These findings suggest a specific mechanism for activation of fibroblasts in GD resulting in the recruitment of T cells. They may provide insight into a missing link between the glandular and extrathyroidal manifestations of GD.


Subject(s)
Chemotactic Factors/biosynthesis , Fibroblasts/immunology , Fibroblasts/metabolism , Graves Disease/immunology , Immunoglobulin G/pharmacology , T-Lymphocytes/immunology , Caspase 3 , Caspases/metabolism , Cells, Cultured , Chemokine CCL5/biosynthesis , Chemotactic Factors/metabolism , Chemotaxis, Leukocyte/immunology , Fibroblasts/enzymology , Humans , Immune Sera/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-16/antagonists & inhibitors , Interleukin-16/biosynthesis , Interleukin-16/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Sirolimus/pharmacology
11.
Eur Cytokine Netw ; 12(3): 478-86, 2001.
Article in English | MEDLINE | ID: mdl-11566629

ABSTRACT

Interferon alpha (IFN-alpha) is the mainstay in the treatment of chronic hepatitis C virus (HCV) infection. Interleukin-16 (IL-16) attracts CD4+ cells to sites of inflammation and plays a role in the interaction of dendritic cells, T cells and B cells. In this study, we show that IFN-alpha itself induces IL-16 secretion by peripheral blood lymphocytes (PBL) and enhances IL-16 secretion by anti-CD3 stimulated PBL. Pro-IL-16 is cleaved into its active form by caspase-3. IFN-alpha increases caspase-3 mRNA levels in activated T cells (ATC), as shown by Northern blot analysis, whereas IL-16 mRNA levels are not affected by IFN-alpha. IL-16 secretion into culture supernatants correlates tightly with intracellular caspase-3 activity in ATC. In our experiments addition of specific caspase inhibitors did not reduce the proportion of ATC undergoing Fas-mediated cell death, but completely blocked IFN-alpha-induced IL-16 secretion into culture supernatants. In conclusion, our results suggest that IFN-alpha activates caspase-3, thereby increasing secretion of IL-16, whereas IFN-alpha-enhanced Fas-mediated cell death in ATC is not caspase-dependent.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , Interferon-alpha/metabolism , Interleukin-16/biosynthesis , RNA, Messenger/metabolism , Antibodies/immunology , Antibodies/pharmacology , Apoptosis/drug effects , Blood Cells/drug effects , Blood Cells/metabolism , CD3 Complex/immunology , Caspase 3 , Caspases/drug effects , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Interferon-alpha/pharmacology , Interleukin-16/antagonists & inhibitors , Interleukin-16/metabolism , T-Lymphocytes/metabolism
12.
Eur J Pharmacol ; 383(2): 189-96, 1999 Oct 27.
Article in English | MEDLINE | ID: mdl-10585533

ABSTRACT

In this study, we examined whether peptides based on the hydrophilic Cluster of Differentiation (CD) 4-binding part of the amino acid sequence of human interleukin-16 can block interleukin-16-induced chemotaxis of murine lymphocytes in vitro. Peptide 3 was capable of inhibiting interleukin-16-induced chemotaxis of murine splenocytes in vitro. Next, we compared the effects of intra-airway administration of peptide 3 with those of antibodies to interleukin-16 on antigen-induced features in a murine model of allergic asthma. Intra-airway administration of peptide 3 largely inhibited the development of antigen-induced airway hyperresponsiveness while airway eosinophilia was not affected. Similar effects were observed after intranasal application of antibodies to interleukin-16. These results indicate that treatment with peptide 3 causes the same effects as do antibodies to interleukin-16, possibly via the inhibition of interaction between interleukin-16 and its receptor CD4. Therefore, peptide 3 could be useful as a lead compound in attempting to limit airway hyperresponsiveness via binding to CD4.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Interleukin-16/antagonists & inhibitors , Interleukin-16/pharmacology , Peptide Fragments/pharmacology , Administration, Intranasal , Airway Resistance/drug effects , Amino Acid Sequence , Animals , Anti-Asthmatic Agents/administration & dosage , Antibodies, Blocking/pharmacology , Bronchial Hyperreactivity/drug therapy , Bronchoalveolar Lavage Fluid/cytology , Cell Separation , Chemotaxis, Leukocyte/drug effects , Eosinophils/drug effects , Eosinophils/metabolism , In Vitro Techniques , Interleukin-16/administration & dosage , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Muscarinic Agonists/pharmacology , Oligopeptides/pharmacology , Ovalbumin/immunology , Peptide Fragments/administration & dosage
13.
J Immunol ; 163(4): 1827-32, 1999 Aug 15.
Article in English | MEDLINE | ID: mdl-10438915

ABSTRACT

IL-16 is a proinflammatory cytokine implicated in the pathogenesis of asthma and other conditions characterized by recruitment of CD4+ T cells to sites of disease. It is postulated that CD4 is an IL-16 receptor, although other receptors or coreceptors may exist. Among several known functions, IL-16 is a chemoattractant factor for CD4+ T cells and it inhibits MLR. We previously reported that an oligopeptide corresponding to the 16 C-terminal residues of human IL-16 inhibits chemoattractant activity. To identify functional domains with greater precision, shorter oligonucleotides containing native or mutated C-terminal IL-16 sequences were tested for IL-16 inhibition. Within the 16 C-terminal residues, the minimal peptide RRKS (corresponding to Arg106 to Ser109) was shown to mediate inhibition of IL-16 chemoattractant activity. Inhibition was lost when either arginine was substituted with alanine. Point mutations in IL-16 revealed that Arg107 is critical for chemoattractant activity, but MLR inhibition was unaffected by mutation of Arg107 or even deletion of the C-terminal tail through Arg106. Deletion of 12 or 22 N-terminal residues of IL-16 had no impact on chemoattractant activity, but MLR inhibition was reduced. Deletion of 16 C-terminal plus 12 N-terminal residues abolished both chemoattractant and MLR-inhibitory activity of IL-16. These data indicate that receptor interactions with IL-16 that activate T cell migration are not identical with those required for MLR inhibition, and suggest that both N-terminal and C-terminal domains in IL-16 participate in receptor binding or activation.


Subject(s)
Interleukin-16/chemistry , Interleukin-16/physiology , Peptide Fragments/chemistry , Peptide Fragments/physiology , Amino Acid Sequence , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Blotting, Western , Cell Migration Inhibition , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/genetics , Chemotactic Factors/physiology , Humans , Immunosuppressive Agents/pharmacology , Interleukin-16/antagonists & inhibitors , Interleukin-16/genetics , Lymphocyte Culture Test, Mixed , Molecular Sequence Data , Oligopeptides/physiology , Peptide Fragments/genetics , Peptide Fragments/immunology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Sequence Deletion , T-Lymphocytes/immunology
14.
J Biol Chem ; 274(33): 23387-95, 1999 Aug 13.
Article in English | MEDLINE | ID: mdl-10438516

ABSTRACT

Interleukin-16 (IL-16) activates CD4(+) cells, possibly by direct interaction with CD4. IL-16 structure and function are highly conserved across species, suggesting similar conservation of a putative IL-16 binding site on CD4. Comparison of the human CD4 amino acid sequence with that of several different species revealed that immunoglobulin-like domain 4 is the most conserved extracellular region. Potential interaction of this domain with IL-16 was studied by testing murine D4 sequence-based oligopeptides for inhibition of IL-16 chemoattractant activity and inhibition of IL-16 binding to CD4 in vitro. Three contiguous 12-residue D4 region peptides (designated A, B, and C) blocked IL-16 chemoattractant activity, with peptide B the most potent. Peptides A and B were synergistic for inhibition, but peptide C was not. Peptides A and B also blocked IL-16 binding to CD4 in vitro, whereas peptide C did not. CD4, in addition to its known function as a receptor for major histocompatibility complex class II, contains a binding site for IL-16 in the D4 domain. The D4 residues required for IL-16 binding overlap those previously shown to participate in CD4-CD4 dimerization following class II major histocompatibility complex binding, providing a mechanistic explanation for the known function of IL-16 to inhibit the mixed lymphocyte reaction.


Subject(s)
CD4 Antigens/metabolism , Interleukin-16/metabolism , Lymphocyte Activation , Amino Acid Sequence , Animals , Binding Sites , CD4 Antigens/chemistry , Humans , Interleukin-16/antagonists & inhibitors , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Receptors, Interleukin-2/antagonists & inhibitors , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Species Specificity
15.
J Immunol ; 162(7): 4293-9, 1999 Apr 01.
Article in English | MEDLINE | ID: mdl-10201961

ABSTRACT

T lymphocytes are a major component of the inflammatory infiltrate in rheumatoid synovitis, but their exact role in the disease process is not understood. Functional activities of synovial T cells were examined by adoptive transfer experiments in human synovium-SCID mouse chimeras. Adoptive transfer of tissue-derived autologous CD8+ T cells induced a marked reduction in the activity of lesional T cells and macrophages. Injection of CD8+, but not CD4+, T cells decreased the production of tissue IFN-gamma, IL-1beta, and TNF-alpha by >90%. The down-regulatory effect of adoptively transferred CD8+ T cells was not associated with depletion of synovial CD3+ T cells or synovial CD68+ macrophages, and it could be blocked by Abs against IL-16, a CD8+ T cell-derived cytokine. In the synovial tissue, CD8+ T cells were the major source of IL-16, a natural ligand of the CD4 molecule that can anergize CD4-expressing cells. The anti-inflammatory activity of IL-16 in rheumatoid synovitis was confirmed by treating synovium-SCID mouse chimeras with IL-16. Therapy for 14 days with recombinant human IL-16 significantly inhibited the production of IFN-gamma, IL-1beta, and TNF-alpha in the synovium. We propose that tissue-infiltrating CD8+ T cells in rheumatoid synovitis have anti-inflammatory activity that is at least partially mediated by the release of IL-16. Spontaneous production of IL-16 in synovial lesions impairs the functional activity of CD4+ T cells but is insufficient to completely abrogate their stimulation. Supplemental therapy with IL-16 may be a novel and effective treatment for rheumatoid arthritis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Arthritis, Rheumatoid/immunology , Interleukin-16/physiology , Synovitis/immunology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Rheumatoid/pathology , Arthritis, Rheumatoid/therapy , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cells, Cultured , Chronic Disease , Down-Regulation/immunology , Humans , Injections, Intraperitoneal , Interleukin-16/administration & dosage , Interleukin-16/antagonists & inhibitors , Interleukin-16/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Synovial Membrane/immunology , Synovial Membrane/metabolism , Synovial Membrane/pathology , Synovitis/pathology , Synovitis/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...