Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Braz. J. Pharm. Sci. (Online) ; 58: e21010, 2022. tab, graf
Article in English | LILACS | ID: biblio-1420430

ABSTRACT

Abstract Acrylamide is a neurotoxic compound. Moreover, anakinra is an interleukin-1 (IL-1) receptor antagonist used in rheumatoid arthritis treatment. This study investigated the effect of anakinra on acrylamide-related neuropathy and neuropathic pain. Acrylamide exposure caused a significant decrease in the pain threshold; an increase in malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-α), and interleukin-1 beta (IL-1ß) levels; and a decrease in total glutathione (tGSH) values in the sciatic nerve. This indicates hyperalgesia presence, oxidative stress, and peripheral nerve tissue inflammation. Anakinra treatment significantly reduced the MDA, IL-1ß, and TNF-α levels, and increased the pain threshold and mean tGSH values. The analgesic effect of anakinra was 67.9% at the first hour, increasing to 74.9% and 76.7% at the second and third hours, respectively. The group receiving acrylamide exhibited histopathological changes (e.g., swollen and degenerated axons, hypertrophic and hyperplasic Schwann cells, and congested vessels). The use of anakinra significantly improved these morphological changes. Anakinra is concluded to reduce neuropathic pain and prevent neurotoxic effect of acrylamide on peripheral nerves due to its analgesic, antioxidant, and anti-inflammatory properties


Subject(s)
Animals , Male , Rats , Peripheral Nervous System Diseases/pathology , Acrylamide/adverse effects , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Inflammation/classification , Peripheral Nerves/abnormalities , Arthritis, Rheumatoid/pathology , Tumor Necrosis Factor-alpha/pharmacology , Pain Threshold/classification , Oxidative Stress/drug effects
2.
J Immunol ; 206(7): 1569-1575, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33547169

ABSTRACT

The IL-1 receptor antagonist, anakinra, may represent a therapeutic option for acute respiratory distress syndrome (ARDS) associated with coronavirus disease 2019 (COVID-19). In this study, COVID-19 ARDS patients admitted to the Azienda Socio Sanitaria Territoriale of Lecco, Italy, between March 5th to April 15th, 2020, and who had received anakinra off-label were retrospectively evaluated and compared with a cohort of matched controls who did not receive immunomodulatory treatment. The primary end point was survival at day 28. The population consisted of 112 patients (56 treated with anakinra and 56 controls). Survival at day 28 was obtained in 69 patients (61.6%) and was significantly higher in anakinra-treated patients than in the controls (75.0 versus 48.2%, p = 0.007). When stratified by continuous positive airway pressure support at baseline, anakinra-treated patients' survival was also significant compared with the controls (p = 0.008). Univariate analysis identified anakinra usage (odds ratio, 3.2; 95% confidence interval, 1.47-7.17) as a significant survival predictor. This was not supported by multivariate modeling. The rate of infectious-related adverse events was similar between groups. In conclusion, anakinra improved overall survival and invasive ventilation-free survival and was well tolerated in patients with ARDS associated with COVID-19.


Subject(s)
COVID-19 , Interleukin 1 Receptor Antagonist Protein/administration & dosage , Respiration, Artificial , SARS-CoV-2/immunology , Severe Acute Respiratory Syndrome , Aged , COVID-19/immunology , COVID-19/mortality , COVID-19/therapy , Disease-Free Survival , Female , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/immunology , Male , Middle Aged , Retrospective Studies , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/mortality , Severe Acute Respiratory Syndrome/therapy , Severe Acute Respiratory Syndrome/virology , Survival Rate
3.
Front Immunol ; 12: 771677, 2021.
Article in English | MEDLINE | ID: mdl-34992596

ABSTRACT

Objectives: To compare phagocytic activities of monocytes in peripheral blood mononuclear cells (PBMCs) from acute gout patients and normal subjects, examine monosodium urate monohydrate (MSU) crystal-induced IL-1ß secretion ± recombinant human proteoglycan 4 (rhPRG4) or interleukin-1 receptor antagonist (IL-1RA), and study the anti-inflammatory mechanism of rhPRG4 in MSU stimulated monocytes. Methods: Acute gout PBMCs were collected from patients in the Emergency Department and normal PBMCs were obtained from a commercial source. Monocytes in PBMCs were identified by flow cytometry. PBMCs were primed with Pam3CSK4 (1µg/mL) for 24h and phagocytic activation of monocytes was determined using fluorescently labeled latex beads. MSU (200µg/mL) stimulated IL-1ß secretion was determined by ELISA. Reactive oxygen species (ROS) generation in monocytes was determined fluorometrically. PBMCs were incubated with IL-1RA (250ng/mL) or rhPRG4 (200µg/mL) and bead phagocytosis by monocytes was determined. THP-1 monocytes were treated with MSU crystals ± rhPRG4 and cellular levels of NLRP3 protein, pro-IL-1ß, secreted IL-1ß, and activities of caspase-1 and protein phosphatase-2A (PP2A) were quantified. The peritoneal influx of inflammatory and anti-inflammatory monocytes and neutrophils in Prg4 deficient mice was studied and the impact of rhPRG4 on immune cell trafficking was assessed. Results: Enhanced phagocytic activation of gout monocytes under basal conditions (p<0.001) was associated with ROS generation and MSU stimulated IL-1ß secretion (p<0.05). rhPRG4 reduced bead phagocytosis by normal and gout monocytes compared to IL-1RA and both treatments were efficacious in reducing IL-1ß secretion (p<0.05). rhPRG4 reduced pro-IL-1ß content, caspase-1 activity, conversion of pro-IL-1ß to mature IL-1ß and restored PP2A activity in monocytes (p<0.05). PP2A inhibition reversed rhPRG4's effects on pro-IL-1ß and mature IL-1ß in MSU stimulated monocytes. Neutrophils accumulated in peritoneal cavities of Prg4 deficient mice (p<0.01) and rhPRG4 treatment reduced neutrophil accumulation and enhanced anti-inflammatory monocyte influx (p<0.05). Conclusions: MSU phagocytosis was higher in gout monocytes resulting in higher ROS and IL-1ß secretion. rhPRG4 reduced monocyte phagocytic activation to a greater extent than IL-1RA and reduced IL-1ß secretion. The anti-inflammatory activity of rhPRG4 in monocytes is partially mediated by PP2A, and in vivo, PRG4 plays a role in regulating the trafficking of immune cells into the site of a gout flare.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Gout/drug therapy , Interleukin-1beta/immunology , Proteoglycans/therapeutic use , Acetylcysteine/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents/pharmacology , Female , Free Radical Scavengers/pharmacology , Gout/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Knockout , Middle Aged , Neutrophils/drug effects , Neutrophils/immunology , Phagocytosis/drug effects , Protein Phosphatase 2/immunology , Proteoglycans/genetics , Proteoglycans/pharmacology , Reactive Oxygen Species/immunology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , THP-1 Cells , Uric Acid
5.
Arthritis Res Ther ; 21(1): 238, 2019 11 13.
Article in English | MEDLINE | ID: mdl-31722745

ABSTRACT

BACKGROUND: Most in vitro studies of potential osteoarthritis (OA) therapies have used cartilage monocultures, even though synovium is a key player in mediating joint inflammation and, thereby, cartilage degeneration. In the case of interleukin-1 (IL-1) inhibition using its receptor antagonist (IL-1Ra), like chondrocytes, synoviocytes also express IL-1 receptors that influence intra-articular IL-1 signaling and IL-1Ra efficacy. The short residence time of IL-1Ra after intra-articular injection requires the application of frequent dosing, which is clinically impractical and comes with increased risk of infection; these limitations motivate the development of effective drug delivery strategies that can maintain sustained intra-articular IL-1Ra concentrations with only a single injection. The goals of this study were to assess how the presence of synovium in IL-1-challenged cartilage-synovium co-culture impacts the time-dependent biological response of single and sustained doses of IL-1Ra, and to understand the mechanisms underlying any co-culture effects. METHODS: Bovine cartilage explants with or without synovium were treated with IL-1α followed by single or multiple doses of IL-1Ra. Effects of IL-1Ra in rescuing IL-1α-induced catabolism in cartilage monoculture and cartilage-synovium co-culture were assessed by measuring loss of glycosaminoglycans (GAGs) and collagen using DMMB (dimethyl-methylene blue) and hydroxyproline assays, respectively, nitric oxide (NO) release using Griess assay, cell viability by fluorescence staining, metabolic activity using Alamar blue, and proteoglycan biosynthesis by radiolabel incorporation. Day 2 conditioned media from mono and co-cultures were analyzed by mass spectrometry and cytokine array to identify proteins unique to co-culture that contribute to biological crosstalk. RESULTS: A single dose of IL-1Ra was ineffective, and a sustained dose was necessary to significantly suppress IL-1α-induced catabolism as observed by enhanced suppression of GAG and collagen loss, NO synthesis, rescue of chondrocyte metabolism, viability, and GAG biosynthesis rates. The synovium exhibited a protective role as the effects of single-dose IL-1Ra were significantly enhanced in cartilage-synovium co-culture and were accompanied by release of anti-catabolic factors IL-4, carbonic anhydrase-3, and matrilin-3. A total of 26 unique proteins were identified in conditioned media from co-cultures, while expression levels of many additional proteins important to cartilage homeostasis were altered in co-culture compared to monocultures; principal component analysis revealed distinct clustering between co-culture and cartilage and synovium monocultures, thereby confirming significant crosstalk. CONCLUSIONS: IL-1Ra suppresses cytokine-induced catabolism in cartilage more effectively in the presence of synovium, which was associated with endogenous production of anti-catabolic factors. Biological crosstalk between cartilage and synovium is significant; thus, their co-cultures should better model the intra-articular actions of potential OA therapeutics. Additionally, chondroprotective effects of IL-1Ra require sustained drug levels, underscoring the need for developing drug delivery strategies to enhance its joint residence time following a single intra-articular injection.


Subject(s)
Cartilage/drug effects , Chondrocytes/drug effects , Cytokines/pharmacology , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin-1alpha/pharmacology , Synovial Membrane/drug effects , Animals , Cartilage/cytology , Cartilage/metabolism , Cattle , Cells, Cultured , Chondrocytes/metabolism , Coculture Techniques/methods , Dose-Response Relationship, Drug , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Metabolism/drug effects , Metabolism/physiology , Synovial Membrane/cytology , Synovial Membrane/metabolism
7.
Bioorg Med Chem ; 27(24): 115177, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31711716

ABSTRACT

The oxytocinase subfamily of M1 aminopeptidases consists of three members, ERAP1, ERAP2 and IRAP that play several important biological roles, including key functions in the generation of antigenic peptides that drive human immune responses. They represent emerging targets for pharmacological manipulation of the immune system, albeit lack of selective inhibitors is hampering these efforts. Most of the previously explored small-molecule binders target the active site of the enzymes via strong interactions with the catalytic zinc(II) atom and, while achieving increased potency, they suffer in selectivity. Continuing our earlier efforts on weaker zinc(II) binding groups (ZBG), like the 3,4-diaminobenzoic acid derivatives (DABA), we herein synthesized and biochemically evaluated analogues of nine potentially weak ZBGs, based on differential substitutions of functionalized pyridinone- and pyridinethione-scaffolds, nicotinic-, isonicotinic-, aminobenzoic- and hydrazinobenzoic-acids. Crystallographic analysis of two analogues in complex with a metalloprotease (MMP-12) revealed unexpected binding topologies, consistent with the observed affinities. Our results suggest that the potency of the compounds as inhibitors of ERAP1, ERAP2 and IRAP is primarily driven by the occupation of active-site specificity pockets and their proper orientation within the enzymes.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Zinc/chemistry , Aminopeptidases/metabolism , Binding Sites , Drug Design , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Minor Histocompatibility Antigens/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship , Zinc/metabolism
9.
Int J Cardiol ; 270: 221-227, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-29884291

ABSTRACT

BACKGROUND: Angiotensin II (Ang II) activates components of the inflammatory cascade, which promotes hypertension and development of abdominal aortic aneurysm (AAA). This study aimed to elucidate the effects of an IL-1 receptor antagonist (IL-1Ra) and an anti-IL-1ß antibody (01BSUR) on Ang II-induced AAA. METHODS AND RESULTS: Male wild-type (WT) and IL-1Ra-deficient (IL-1Ra-/-) mice were infused with Ang II (1000 ng/kg/min) using subcutaneous osmotic pumps for 28 days. Fourteen days post-infusion, both systolic blood pressure (SBP) (Ang II-treated IL-1Ra-/-:149 ±â€¯2 vs. Ang II-treated WT:126 ±â€¯3 mm Hg, p < 0.001) and abdominal aortic width (0.94 ±â€¯0.09 vs. 0.49 ±â€¯0.03 mm, p < 0.001) were significantly higher in IL-1Ra-/- mice than in WT mice. Because 28-day infusion with Ang II in IL-1Ra-/- mice significantly increased the occurrence of fatal aortic rupture (89% vs. 6%, p < 0.0001), both types of mice were infused with Ang II for only 14 days, and histological analyses were performed at 28 days. Interestingly, AAA increased more significantly in IL-1Ra-/- mice than in WT mice (p < 0.001), although SBP did not differ at 28 days in IL-1Ra-/- and WT mice (117 ±â€¯4 vs. 115 ±â€¯3 mm Hg, p = 0.71 (after cessation of Ang II infusion)). Histological analyses showed numerous inflammatory cells around the abdominal aorta in IL-1Ra-/- mice, but not in WT mice. Finally, compared with IgG2a treatment, treatment with 01BSUR decreased Ang II-induced AAA in IL-1Ra-/- mice. CONCLUSIONS: The present study demonstrates that inhibition of IL-1ß significantly suppresses AAA formation after Ang II infusion, suggesting that suppression of IL-1ß may provide an additional strategy to protect against AAA in hypertensive patients.


Subject(s)
Angiotensin II/toxicity , Aortic Aneurysm, Abdominal/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Vasculitis/metabolism , Animals , Aortic Aneurysm, Abdominal/drug therapy , Aortic Aneurysm, Abdominal/pathology , Blood Pressure/drug effects , Blood Pressure/physiology , Heart Rate/drug effects , Heart Rate/physiology , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Vasculitis/chemically induced , Vasculitis/pathology
10.
Arthritis Res Ther ; 20(1): 101, 2018 05 30.
Article in English | MEDLINE | ID: mdl-29848388

ABSTRACT

Long-term cell cultures developed early in the twentieth century allowed identification in their supernatants of biological mediators subsequently defined as migration factors, interferons, lymphokines, monokines, cytokines and interleukins. In rheumatology, early in the 1930s, synovial cell cultures revealed two major distinct populations, i.e. synovial fibroblasts and monocyte-macrophages. Discovery of the interstitial collagenase (MMP-1) and its role in tissue destruction, such as in rheumatoid arthritis (RA), raised the question of the cellular source for this enzyme. My personal interest in the field was driven by the lack of understanding for the link between tissue destruction and immunology. This triggered our seminal contribution to the field, establishing in 1976-79 at the Arthritis Unit (Massachusetts General Hospital, with SM Krane) that a mononuclear factor (MCF, around 15 kDa) produced by stimulated macrophage, under direct contact with activated T cells, induced large amounts of collagenase and prostaglandin E2 (PGE2, a bone resorbing agent) in human synovial fibroblasts from RA patients. Our original "MCF" biological observations preceded cloning, and recombinant IL-1ß confirmed the biological activity of the purified natural IL-1. Following my return to Geneva in 1980 and searching for a high level of IL-1 in urine and serum of patients with high fever or Still's disease, to our surprise-"a finding of absence"-we found that IL-1 was masked by a factor of approximately 17 kDa and first presented this in 1984 at the Fourth International Lymphokine Workshop. In 1987, before IL-Ra cloning, my co-worker P Seckinger and I demonstrated first-time observation in cytokine biology that the mechanism was due to the inhibition of IL-1 binding the cell surface receptor, leading to the concept of IL-1 receptor antagonist (IL-1Ra). Having reported in 1985 that TNF/cachectin also induced collagenase and PGE2 in human synovial cells, we found that IL-1Ra did not block TNF-α but was due to another inhibitor. As other investigators, we confirmed that this inhibitory factor was a soluble TNF receptor. The years between the 1970s and 1990s were probably the most exciting period in the field of cytokines and cytokine antagonists; it gave rise to two concepts in the cytokine field-one of the receptor antagonist, and the other of soluble receptor antagonists.


Subject(s)
Cytokines/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/metabolism , Rheumatology/trends , Tumor Necrosis Factor-alpha/metabolism , Antirheumatic Agents/pharmacology , Cells, Cultured , Cytokines/immunology , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/immunology
11.
Immunol Lett ; 199: 44-52, 2018 07.
Article in English | MEDLINE | ID: mdl-29803636

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disease with CD4+ T cell infiltration and hyperplasia of synovial tissues leading to progressive destruction of articular cartilage. In addition to the central role of T cells in the pathogenesis of RA, recent reports have suggested that B cells also contribute to RA. To explore the effects of interleukin (IL)-17 on B cell development and response in excess IL-1 signaling, we generated IL-17 and IL-1 receptor antagonist (IL-1Ra) double-deficient mice via backcrossing IL-17 knockout (KO) and IL-1RaKO mice. We studied the effect of IL-17 deficiency on antibody-producing B cells and regulatory B cells in IL-1RaKO mice. Excess IL-1 signal increased the frequency of B220+ IgG+ cells and plasma cells. It also promoted the production of immunoglobulins in vitro. Moreover, IL-17 deficiency significantly enhanced the frequency of regulatory IL-10-producing regulatory B cells in IL-1RaKO mice. IL-17 deficiency ameliorated disease symptoms of inflammatory arthritis in IL-1RaKO mice by suppressing the frequency of plasma cells and antibody production while enhancing the frequency of IL-10-producing B cells. These findings suggest that IL-17 can trigger an inflammatory immune reaction by activating antibody-producing B cells while suppressing immune regulatory B cells in RA.


Subject(s)
Arthritis, Rheumatoid/immunology , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-17/immunology , Th17 Cells/immunology , Animals , Arthritis, Experimental/immunology , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin-10/immunology , Interleukin-17/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout
12.
Sci Rep ; 6: 21931, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26902320

ABSTRACT

It remains unclear how infantile febrile seizures (FS) enhance adult seizure susceptibility. Here we showed that the transient increase of interleukin-1ß (IL-1ß) after prolonged FS promoted adult seizure susceptibility, which was blocked by interleukin-1 receptor antagonist (IL-1Ra) within a critical time window. Postnatal administered IL-1ß alone mimicked the effect of FS on adult seizure susceptibility. IL-1R1 knockout mice were not susceptible to adult seizure after prolonged FS or IL-1ß treatment. Prolonged FS or early-life IL-1ß treatment increased the expression of cannabinoid type 1 receptor (CB1R) for over 50 days, which was blocked by IL-1Ra or was absent in IL-1R1 knockout mice. CB1R antagonist, knockdown and endocannabinoid synthesis inhibitor abolished FS or IL-1ß-enhanced seizure susceptibility. Thus, this work identifies a pathogenic role of postnatal IL-1ß/IL-1R1 pathway and subsequent prolonged prominent increase of endocannabinoid signaling in adult seizure susceptibility following prolonged FS, and highlights IL-1R1 as a potential therapeutic target for preventing the development of epilepsy after infantile FS.


Subject(s)
Aging/genetics , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/genetics , Receptor, Cannabinoid, CB1/genetics , Seizures, Febrile/genetics , Seizures/genetics , Animals , Benzoxazines/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Cyclohexanones/pharmacology , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/metabolism , Lipoprotein Lipase/antagonists & inhibitors , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mice , Mice, Knockout , Morpholines/pharmacology , Naphthalenes/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Rimonabant , Seizures/metabolism , Seizures/pathology , Seizures/physiopathology , Seizures, Febrile/metabolism , Seizures, Febrile/pathology , Seizures, Febrile/physiopathology , Signal Transduction , Tissue Culture Techniques
13.
FEBS Lett ; 590(1): 43-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26763127

ABSTRACT

Macrophages display heterogeneous phenotypes, including the classical M1 proinflammatory and the alternative M2 anti-inflammatory polarization states. The transducin-like enhancer of split-1 (TLE1) is a transcriptional corepressor whose functions in macrophages have not been studied yet. We report that TLE1 is highly expressed in human alternative macrophages in vitro and in atherosclerotic plaques as well as in adipose tissue M1/M2 mixed macrophages. TLE1 silencing in alternative macrophages decreases the expression of the M2 markers IL-1Ra and IL-10, while it exacerbates TNFα and CCL3 induction by lipopolysaccharide. Hence, TLE1 is expressed in human macrophages where it has potential anti-inflammatory and alternative phenotype promoting properties.


Subject(s)
Co-Repressor Proteins/metabolism , Macrophage Activation , Macrophages/metabolism , Repressor Proteins/metabolism , Animals , Biomarkers/metabolism , Body Mass Index , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cells, Cultured , Co-Repressor Proteins/antagonists & inhibitors , Co-Repressor Proteins/genetics , Female , Gene Expression Regulation/drug effects , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-10/antagonists & inhibitors , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-4/pharmacology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Male , Mice, 129 Strain , Obesity/blood , Obesity/immunology , Obesity/metabolism , Obesity/pathology , Plaque, Atherosclerotic/immunology , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , RNA Interference , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics
15.
J Hepatol ; 63(6): 1405-12, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26276675

ABSTRACT

BACKGROUND & AIMS: Autologous transplantation of mesenchymal stem cells (MSCs) reduces concanavalin A (Con A)-induced hepatic injury in mice. However, the mechanism is unclear and the therapeutic effect of allo-transplantation remains unknown. Our aim was to investigate the effects and mechanisms related to allo-transplantation of MSCs when used to treat Con A hepatic injury. METHODS: After Con A-induced liver injury was created in C57BL/6J mice, MSCs derived from BALB/c mice or a vehicle control was administered. RESULTS: Allo-transplantation of MSCs derived from BALB/c mice attenuated hepatic apoptosis in C57BL/6J mice that had undergone Con A-induced liver injury. MSCs increased the level of serum interleukin (IL)-10 and the phosphorylation of hepatic STAT3, but decreased the level of hepatic IFN-γ and phospho-STAT1. Notably, the administered MSCs were trapped mostly in the lungs and promoted the macrophage M2 switch, which contributed to the increased IL10 levels in the lungs and serum. Loss of the therapeutic effect was observed after knock-down of the expression of interleukin 1 receptor antagonist (IL1Ra) in the MSCs. In vitro investigation supported the hypothesis that MSCs are able to switch Con A-stimulated macrophages to the M2 phenotype, which results in an increase in IL10 production. CONCLUSIONS: Allo-transplantation of MSCs reduces Con A liver injury by increasing IL10 production through an IL1Ra dependent macrophage switch.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Interleukin 1 Receptor Antagonist Protein/metabolism , Macrophages/immunology , Mesenchymal Stem Cell Transplantation , Allografts , Animals , Apoptosis , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/toxicity , Disease Models, Animal , Gene Knockdown Techniques , Interferon-gamma/blood , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-10/blood , Interleukin-10/metabolism , Liver/immunology , Liver/metabolism , Liver/pathology , Lung/immunology , Lung/metabolism , Lung/pathology , Macrophages/metabolism , Macrophages/pathology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism
16.
Mol Pharm ; 12(7): 2517-27, 2015 Jul 06.
Article in English | MEDLINE | ID: mdl-25909959

ABSTRACT

Clofazimine (CFZ) is an FDA-approved leprostatic and anti-inflammatory drug that massively accumulates in macrophages, forming insoluble, intracellular crystal-like drug inclusions (CLDIs) during long-term oral dosing. Interestingly, when added to cells in vitro, soluble CFZ is cytotoxic because it depolarizes mitochondria and induces apoptosis. Accordingly, we hypothesized that, in vivo, macrophages detoxify CFZ by sequestering it in CLDIs. To test this hypothesis, CLDIs of CFZ-treated mice were biochemically isolated and then incubated with macrophages in vitro. The cell biological effects of phagocytosed CLDIs were compared to those of soluble CFZ. Unlike soluble CFZ, phagocytosis of CLDIs did not lead to mitochondrial destabilization or apoptosis. Rather, CLDIs altered immune signaling response pathways downstream of Toll-like receptor (TLR) ligation, leading to enhanced interleukin-1 receptor antagonist (IL-1RA) production, dampened NF-κB activation and tissue necrosis factor alpha (TNFα) production, and ultimately decreased TLR expression levels. In aggregate, our results constitute evidence that macrophages detoxify soluble CFZ by sequestering it in a biocompatible, insoluble form. The altered cellular response to TLR ligation suggests that CLDI formation may also underlie CFZ's anti-inflammatory activity.


Subject(s)
Clofazimine/pharmacology , Immunity, Innate/drug effects , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Phagocytosis/drug effects , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Line , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , NF-kappa B/immunology
17.
Oncogene ; 34(19): 2493-504, 2015 May 07.
Article in English | MEDLINE | ID: mdl-24998848

ABSTRACT

The TP53 tumor-suppressor gene is frequently mutated in human cancer. Missense mutations can add novel functions (gain-of-function, GOF) that promote tumor malignancy. Here we report that mutant (mut) p53 promotes tumor malignancy by suppressing the expression of a natural occurring anti-inflammatory cytokine, the secreted interleukin-1 receptor antagonist (sIL-1Ra, IL1RN). We show that mutp53 but not wild-type (wt) p53 suppresses the sIL-1Ra production in conditioned media of cancer cells. Moreover, mutp53, but not wtp53, binds physically the sIL-1Ra promoter and the protein-protein interaction with the transcriptional co-repressor MAFF (v-MAF musculoaponeurotic fibrosarcoma oncogene family, protein F) is required for mutp53-induced sIL-1Ra suppression. Remarkably, when exposed to IL-1 beta (IL-1ß) inflammatory stimuli, mutp53 sustains a ready-to-be-activated in vitro and in vivo cancer cells' response through the sIL-1Ra repression. Taken together, these results identify sIL-1Ra as a novel mutp53 target gene, whose suppression might be required to generate a chronic pro-inflammatory tumor microenvironment through which mutp53 promotes tumor malignancy.


Subject(s)
DNA-Binding Proteins/genetics , Inflammation/genetics , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Cell Line, Tumor , HT29 Cells , Hep G2 Cells , Humans , Inflammation/immunology , Interleukin 1 Receptor Antagonist Protein/biosynthesis , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/pharmacology , MCF-7 Cells , MafF Transcription Factor/metabolism , Mutation , Neoplasms/genetics , Neoplasms/mortality , Nuclear Proteins/metabolism , Prognosis , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , RNA, Small Interfering , Tumor Microenvironment/immunology
18.
Methods Mol Biol ; 1155: 215-22, 2014.
Article in English | MEDLINE | ID: mdl-24788185

ABSTRACT

Many anti-inflammatory agents have been exploited for the treatment of inflammatory diseases by targeting the most potent proinflammatory cytokines including tumor necrosis factor (TNF) and interleukin-1 (IL-1). Theoretically, simultaneous neutralization or blocking two important inflammatory mediators might achieve a synergistic therapeutic effect. A recombinant fusion protein, TNFR2-Fc-IL-1ra (TFI), was developed as bifunctional inflammatory inhibitor. TFI was able to strongly neutralize TNF activity and to antagonize IL-1 receptor in the cell binding inhibition assays, suggesting that TFI could be used as a bifunctional ligand with enhanced anti-inflammatory effect.


Subject(s)
Immunoglobulin Fc Fragments/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1/pharmacology , Neutralization Tests/methods , Receptors, Tumor Necrosis Factor, Type II/metabolism , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Blotting, Western , CHO Cells , Cell Proliferation/drug effects , Cells, Cultured , Cricetulus , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Immunoglobulin Fc Fragments/genetics , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/genetics , Mice , Plasmids/genetics , Receptors, Tumor Necrosis Factor, Type II/antagonists & inhibitors , Receptors, Tumor Necrosis Factor, Type II/genetics , Recombinant Fusion Proteins/genetics
19.
Expert Opin Biol Ther ; 14(7): 893-904, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24720727

ABSTRACT

INTRODUCTION: Targeted biologic immunomodulatory therapies have had a major impact in rheumatoid arthritis (RA) treatment, including tumor necrosis factor (TNF)-α inhibition, B-cell depletion, interference in T-cell costimulation and interleukin (IL)-1 and IL-6 inhibition. Along with the recognition of the importance of early, aggressive disease-modifying antirheumatic drugs (DMARDs) grounded in the use of methotrexate, the introduction of biologic DMARDs (bDMARDs) has provided significantly improved outcomes in patients with RA with a goal of true remission, or at least a state of very low disease activity, now possible in many. There are a number of methods to inhibit cytokines, cellular receptors and pathways of signal transduction that have been used thus far and are in development. In some cases, the method of target inhibition and differences in molecular construct has impacted efficacy and/or safety; whereas, in other cases, similar safety and/or efficacy signals across compounds have demonstrated class- or target-related effects. As the development of targeted therapies moves forward, it is increasingly important to understand the role of the target both in RA disease pathogenesis and normal host defense and the mechanisms of target inhibition. AREAS COVERED: This review covers the targets of therapy for biologic agents in rheumatic diseases, their molecular constructs and implications on efficacy and safety, with focus on approved treatments for RA. EXPERT OPINION: Advances in molecular biology have provided a number of different ways to impact pathobiologically relevant pathways and targets in terms of the molecular construct of individual compounds. The use of these agents have provided important mechanistic insights into disease pathogenesis, and in some cases are associated with differences in efficacy and safety among agents even with the same downstream target. As bDMARDs identify promising mechanisms, oral agents that target or specifically regulate downstream pathways are made possible.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Biological Factors/therapeutic use , Molecular Targeted Therapy , Antibodies, Monoclonal/therapeutic use , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Receptors, Interleukin-6/antagonists & inhibitors , Recombinant Fusion Proteins/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors
20.
Proc Natl Acad Sci U S A ; 110(10): 3913-8, 2013 Mar 05.
Article in English | MEDLINE | ID: mdl-23431173

ABSTRACT

IL-1 is a key inflammatory and immune mediator in many diseases, including dry-eye disease, and its inhibition is clinically efficacious in rheumatoid arthritis and cryopyrin-associated periodic syndromes. To treat ocular surface disease with a topical biotherapeutic, the uniqueness of the site necessitates consideration of the agent's size, target location, binding kinetics, and thermal stability. Here we chimerized two IL-1 receptor ligands, IL-1ß and IL-1Ra, to create an optimized receptor antagonist, EBI-005, for topical ocular administration. EBI-005 binds its target, IL-1R1, 85-fold more tightly than IL-1Ra, and this increase translates to an ∼100-fold increase in potency in vivo. EBI-005 preserves the affinity bias of IL-1Ra for IL-1R1 over the decoy receptor (IL-1R2), and, surprisingly, is also more thermally stable than either parental molecule. This rationally designed antagonist represents a unique approach to therapeutic design that can potentially be exploited for other ß-trefoil family proteins in the IL-1 and FGF families.


Subject(s)
Cytokines/antagonists & inhibitors , Drug Design , Administration, Topical , Amino Acid Sequence , Animals , Crystallography, X-Ray , Cytokines/chemistry , Drug Stability , Female , Humans , Interleukin 1 Receptor Antagonist Protein/antagonists & inhibitors , Interleukin 1 Receptor Antagonist Protein/chemistry , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/chemistry , Interleukin-1beta/genetics , Kinetics , Ligands , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Ophthalmic Solutions , Protein Conformation , Receptors, Interleukin-1 Type I/antagonists & inhibitors , Receptors, Interleukin-1 Type I/chemistry , Receptors, Interleukin-1 Type I/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Homology, Amino Acid , Static Electricity
SELECTION OF CITATIONS
SEARCH DETAIL
...