Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Arthritis Rheumatol ; 73(12): 2206-2218, 2021 12.
Article in English | MEDLINE | ID: mdl-34423919

ABSTRACT

OBJECTIVE: To investigate the role of PF-06650833, a highly potent and selective small-molecule inhibitor of interleukin-1-associated kinase 4 (IRAK4), in autoimmune pathophysiology in vitro, in vivo, and in the clinical setting. METHODS: Rheumatoid arthritis (RA) inflammatory pathophysiology was modeled in vitro through 1) stimulation of primary human macrophages with anti-citrullinated protein antibody immune complexes (ICs), 2) RA fibroblast-like synoviocyte (FLS) cultures stimulated with Toll-like receptor (TLR) ligands, as well as 3) additional human primary cell cocultures exposed to inflammatory stimuli. Systemic lupus erythematosus (SLE) pathophysiology was simulated in human neutrophils, dendritic cells, B cells, and peripheral blood mononuclear cells stimulated with TLR ligands and SLE patient ICs. PF-06650833 was evaluated in vivo in the rat collagen-induced arthritis (CIA) model and the mouse pristane-induced and MRL/lpr models of lupus. Finally, RNA sequencing data generated with whole blood samples from a phase I multiple-ascending-dose clinical trial of PF-06650833 were used to test in vivo human pharmacology. RESULTS: In vitro, PF-06650833 inhibited human primary cell inflammatory responses to physiologically relevant stimuli generated with RA and SLE patient plasma. In vivo, PF-06650833 reduced circulating autoantibody levels in the pristane-induced and MRL/lpr murine models of lupus and protected against CIA in rats. In a phase I clinical trial (NCT02485769), PF-06650833 demonstrated in vivo pharmacologic action pertinent to SLE by reducing whole blood interferon gene signature expression in healthy volunteers. CONCLUSION: These data demonstrate that inhibition of IRAK4 kinase activity can reduce levels of inflammation markers in humans and provide confidence in the rationale for clinical development of IRAK4 inhibitors for rheumatologic indications.


Subject(s)
Arthritis, Experimental/drug therapy , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Isoquinolines/therapeutic use , Lactams/therapeutic use , Macrophages/drug effects , Rheumatic Diseases/drug therapy , Synoviocytes/drug effects , Animals , Arthritis, Experimental/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Humans , Inflammation/drug therapy , Inflammation/immunology , Isoquinolines/pharmacology , Lactams/pharmacology , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Mice , Rats , Rheumatic Diseases/immunology , Synoviocytes/immunology
3.
J Immunol Methods ; 467: 19-28, 2019 04.
Article in English | MEDLINE | ID: mdl-30735689

ABSTRACT

The NLRP3 inflammasome is a complex multimeric signaling apparatus that regulates production of the pro-inflammatory cytokine IL-1ß. To overcome both the variability among primary immune cells and the limitations of genetic manipulation of differentiated human or murine macrophages, we developed a simplified, reliable and relevant cell-based model for studying the NLRP3 inflammasome using the undifferentiated human myelomonocytic cell line THP1. Undifferentiated THP1 cells constitutively express NLRP3, and NLRP3 inflammasome activation occurred in response to canonical NLRP3 activation stimuli including nigericin, ATP, and urea crystals, culminating in pro-IL-1ß cleavage, extracellular release of mature IL-1ß, and pyroptosis. We used this THP1 cell system to investigate potential targets of the potent, NLRP3 inflammasome selective inhibitor CP-456,773. We optimized a viral shRNA transduction method for gene expression knockdown (KD), and the KD of NLRP3 itself eliminated inflammasome activation and IL-1ß production. NLRP3 inflammasome activation and CP-453,773 pharmacology were not altered in ABCb7- or ABCb10-deficient THP1 cells, eliminating these gene products as candidate pharmacological targets of CP-453,773. For ABCb10, we confirmed our results using CRISPR/CAS9-mediated ABCb10 knockout (KO) THP1 sub-lines. In summary, undifferentiated THP1 cells are fully competent for activation of the NLRP3 inflammasome and production of IL-1ß, without differentiation into macrophages, and we describe optimized KD and KO methodologies to manipulate gene expression in these cells. As an example of the utility of undifferentiated THP1 cells for investigations into the biology of the NLRP3 inflammasome, we have used this cell system to rule out ABCb7 and ABCb10 as potential targets of the NLRP3 inflammasome inhibitor CP-453,773.


Subject(s)
Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfonylurea Compounds/pharmacology , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , HEK293 Cells , Humans , Inflammasomes/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nigericin/antagonists & inhibitors , Nigericin/pharmacology , Uric Acid/antagonists & inhibitors , Uric Acid/pharmacology
4.
J Immunol ; 197(6): 2421-33, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27521339

ABSTRACT

A critical component of innate immune response to infection and tissue damage is the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome, and this pathway and its activation products have been implicated in the pathophysiology of a variety of diseases. NLRP3 inflammasome activation leads to the cleavage of pro-IL-1ß and pro-IL-18, as well as the subsequent release of biologically active IL-1ß, IL-18, and other soluble mediators of inflammation. In this study, we further define the pharmacology of the previously reported NLRP3 inflammasome-selective, IL-1ß processing inhibitor CP-456,773 (also known as MCC950), and we demonstrate its efficacy in two in vivo models of inflammation. Specifically, we show that in human and mouse innate immune cells CP-456,773 is an inhibitor of the cellular release of IL-1ß, IL-1α, and IL-18, that CP-456,773 prevents inflammasome activation induced by disease-relevant soluble and crystalline NLRP3 stimuli, and that CP-456,773 inhibits R848- and imiquimod-induced IL-1ß release. In mice, CP-456,773 demonstrates potent inhibition of the release of proinflammatory cytokines following acute i.p. challenge with LPS plus ATP in a manner that is proportional to the free/unbound concentrations of the drug, thereby establishing an in vivo pharmacokinetic/pharmacodynamic model for CP-456,773. Furthermore, CP-456,773 reduces ear swelling in an imiquimod cream-induced mouse model of skin inflammation, and it reduces airway inflammation in mice following acute challenge with house dust mite extract. These data implicate the NLRP3 inflammasome in the pathogenesis of dermal and airway inflammation, and they highlight the utility of CP-456,773 for interrogating the contribution of the NLRP3 inflammasome and its outputs in preclinical models of inflammation and disease.


Subject(s)
Dermatitis/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammasomes/antagonists & inhibitors , Inflammation/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pneumonia/drug therapy , Pneumonia/immunology , Sulfones/pharmacology , Animals , Cytokines/antagonists & inhibitors , Cytokines/immunology , Dermatitis/immunology , Dermatitis/physiopathology , Disease Models, Animal , Furans , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Immunity, Innate/drug effects , Indenes , Inflammation/drug therapy , Inflammation/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/metabolism , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Mice , Pneumonia/physiopathology , Signal Transduction , Sulfonamides , Sulfones/administration & dosage , Sulfones/therapeutic use
5.
Am J Respir Cell Mol Biol ; 46(1): 71-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21836154

ABSTRACT

The expression of acidic mammalian chitinase (AMCase) is associated with Th2-driven respiratory disorders. To investigate the potentially pathological role of AMCase in allergic airway disease (AAD), we sensitized and challenged mice with ovalbumin or a combination of house dust mite (HDM) plus cockroach allergen. These mice were treated or not treated with small molecule inhibitors of AMCase, which significantly reduced allergen-induced chitinolytic activity in the airways, but exerted no apparent effect on pulmonary inflammation per se. Transgenic and AMCase-deficient mice were also submitted to protocols of allergen sensitization and challenge, yet we found little or no difference in the pattern of AAD between mutant mice and wild-type (WT) control mice. In a separate model, where mice were challenged only with intratracheal instillations of HDM without adjuvant, total bronchoalveolar lavage (BAL) cellularity, inflammatory infiltrates in lung tissues, and lung mechanics remained comparable between AMCase-deficient mice and WT control mice. However BAL neutrophil and lymphocyte counts were significantly increased in AMCase-deficient mice, whereas concentrations in BAL of IL-13 were significantly decreased compared with WT control mice. These results indicate that, although exposure to allergen stimulates the expression of AMCase and increased chitinolytic activity in murine airways, the overexpression or inhibition of AMCase exerts only a subtle impact on AAD. Conversely, the increased numbers of neutrophils and lymphocytes in BAL and the decreased concentrations of IL-13 in AMCase-deficient mice challenged intratracheally with HDM indicate that AMCase contributes to the Th1/Th2 balance in the lungs. This finding may be of particular relevance to patients with asthma and increased airway neutrophilia.


Subject(s)
Asthma/enzymology , Chitinases/antagonists & inhibitors , Hypersensitivity/enzymology , Allergens/immunology , Animals , Asthma/genetics , Asthma/immunology , Bronchoalveolar Lavage Fluid/immunology , Chitinases/deficiency , Chitinases/genetics , Chitinases/immunology , Female , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Interleukin-13/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Neutrophils/immunology , Th1 Cells/immunology , Th2 Cells/immunology
6.
Ann Allergy Asthma Immunol ; 98(1): 83-8, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17225725

ABSTRACT

BACKGROUND: Several studies indicate that selectin-mediated leukocyte migration may depend on the types of initiating inflammatory stimuli or on the vascular beds involved in the inflammatory response. Thus, targeting selectin interactions to treat inflammation may have variable effects depending on the site and origin of the inflammatory response. OBJECTIVE: To address whether selectin-mediated leukocyte recruitment is stimulus or tissue dependent. METHODS: We examined pulmonary and cutaneous allergic inflammatory responses and silica-induced nonallergic lung inflammation and fibrosis in wild-type and P- and E-selectin-deficient (P/E-/-) double knockout mice. Allergen-sensitized wild-type and P/E-/- double knockout mice were challenged either intradermally or via the airways to induce allergic responses in the skin or lung, respectively. Other animals were subjected to intranasal silica administration to induce a nonallergic lung inflammatory/fibrotic response. RESULTS: The P/E-/- mice exhibited significantly reduced allergic inflammation in the skin and lung. Allergic late-phase ear swelling and allergic lung airway hyperresponsiveness were also significantly attenuated in the P/E-/- mice compared with identically treated wild-type animals. In contrast, pulmonary inflammation and fibrosis induced by intranasal administration of silica particles resulted in a more severe phenotype in the P/E-/- mice. CONCLUSIONS: Selectin interactions drive allergic inflammation in the lung and skin. Silica-induced pulmonary inflammation and fibrosis, however, was more pronounced in the absence of selectin interactions, suggesting that selectin-mediated leukocyte migration may depend on the types of initiating inflammatory stimuli.


Subject(s)
E-Selectin/metabolism , Endothelium, Vascular/metabolism , Hypersensitivity/immunology , Inflammation/metabolism , P-Selectin/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Chemotaxis, Leukocyte/immunology , Cytokines/immunology , Cytokines/metabolism , E-Selectin/immunology , Endothelium, Vascular/immunology , Hypersensitivity/metabolism , Inflammation/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Ovalbumin/toxicity , P-Selectin/metabolism , Silicon Dioxide/toxicity , Skin/immunology
7.
J Allergy Clin Immunol ; 115(2): 309-15, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15696086

ABSTRACT

BACKGROUND: T(H)2-mediated allergic asthma is characterized by eosinophilia, mucus overproduction, and airway hyperresponsiveness (AHR). Although it is clear that T(H)2 cells and their cytokines play an important role in AHR, the roles of T(H)1 cells and neutrophils in AHR are controversial. OBJECTIVE: We sought to determine the roles of T(H)1 cells and neutrophils in AHR. METHODS: Ovalbumin-specific CD4(+) T cells were purified from DO11.10 mice, differentiated into T(H)1 cells, and injected into naive BALB/c, IL-4RalphaKO, or IL-8RKO mice. After ovalbumin antigen challenge, cytokine mRNA levels in lung samples, as well as inflammatory cell types and numbers in bronchoalveolar lavage fluid (BALF), were determined. AHR was assessed by measuring resistance in tracheostomized mice and enhanced pause in freely moving mice. RESULTS: T(H)1 cells induced AHR as robust as T(H)2 cells. They also induced lung inflammation dominated by neutrophils. Neither AHR nor inflammation were reduced when T(H)1 cells were transferred into IL-4RalphaKO mice. When IL-8RKO mice were used as recipients of T(H)1 cells, neutrophilia was greatly reduced, but the AHR was as strong as that seen in wild-type mice. On the other hand, dexamethasone treatment had no effect on neutrophilia but has significantly reduced AHR. Reduction in AHR was accompanied by a reduction in the numbers of lymphocytes and macrophages in BALF. CONCLUSIONS: T(H)1 cells can induce strong AHR independent of IL-4 and IL-13. The AHR is associated with the presence of lymphocytes and macrophages, but not neutrophils, in BALF. Our results point to a pathway whereby T(H)1 cells mediate AHR independent of neutrophilic inflammation.


Subject(s)
Bronchial Hyperreactivity/physiopathology , Bronchitis/pathology , Neutrophils/pathology , Th1 Cells/immunology , Adoptive Transfer , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Cytokines/genetics , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Interleukin-13/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mucus/metabolism , Ovalbumin/pharmacology , RNA, Messenger/metabolism , Receptors, Interleukin-8A/deficiency , Severity of Illness Index , Th2 Cells/immunology
8.
J Orthop Res ; 20(5): 1050-9, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12382973

ABSTRACT

The purpose of this study was to characterize the retention kinetics of recombinant human bone morphogenetic protein-2 (rhBMP-2) applied to two calcium-based delivery matrices. Biphasic calcium phosphate (BCP) and a composite containing BCP in an absorbable collagen sponge (BCP/ACS) were evaluated using a spinal fusion model in rabbits. rhBMP-2 labeled with radioactive iodine (125I) was used as a tracer to assess in vivo retention of rhBMP-2 in the presence of these materials (nine animals per material studied). Over a 36 day study period, animals were assessed for the following: percent administered dose retained at the implant site as measured by scintigraphic imaging (counting) with a gamma camera (all animals), radiography of the implant site (all animals), radioactivity in blood and plasma (all animals), and radioactivity in the urine and feces (three animals for each material). Radioactivity data were corrected for the decay of 125I and the attenuation between the implant in vivo and the gamma camera. Differences observed between the two materials for the area under the retention vs. time profile (AUC; 988%*day for BCP vs. 1070%*day for BCP/ACS, p = 0.57) and the mean residence time (MRT; 10.2 days for BCP vs. 7.6 days for BCP/ACS, p = 0.06) were not statistically significant. Initial retention/incorporation of rhBMP-2 was slightly higher for rhBMP-2/BCP/ACS than for rhBMP-2/BCP (96.8% vs. 86.0%, p < 0.05). Animals receiving rhBMP-2/BCP showed a longer terminal retention half-life (t1/2) than did those receiving rhBMP-2/BCP/ACS (7.5 vs. 4.5 days, p < 0.05). The urinary radioactivity recovery data supported the data obtained by scintigraphy. Over the 36 day collection period, essentially complete recovery of radioactivity (dose) in urine was observed for rhBMP-2/BCP and rhBMP-2/BCP/ACS and the majority of the radioactivity (approximately 95%) was soluble in trichloroacetic acid, suggesting extensive catabolism of rhBMP-2 before renal excretion. Fecal recovery of radioactivity was low, approximately 2-3%. In conclusion, rhBMP-2 was retained at the implant site when delivered with either BCP or BCP/ACS based on mean residence time and area under the retention curve vs. time profile. Use of these matrices resulted in detectable rhBMP-2 levels at the surgical site for over a week in contrast to data reported with several other matrices that lasted less time. Systemic catabolism and elimination of the rhBMP-2 was extensive and systemic presence of the protein was negligible.


Subject(s)
Bone Morphogenetic Proteins/pharmacokinetics , Spinal Fusion/methods , Transforming Growth Factor beta , Animals , Area Under Curve , Bone Morphogenetic Protein 2 , Bone Substitutes/metabolism , Calcium Phosphates/metabolism , Collagen/metabolism , Drug Carriers/metabolism , Female , Half-Life , Humans , Iodine Radioisotopes , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/metabolism , Lumbar Vertebrae/surgery , Models, Animal , Rabbits , Radiography , Recombinant Proteins/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...