Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters











Publication year range
1.
Gastro Hep Adv ; 3(6): 830-841, 2024.
Article in English | MEDLINE | ID: mdl-39280905

ABSTRACT

Background and Aims: The colonic epithelium serves as both a barrier to lumenal contents and a gatekeeper of inflammatory responses. In ulcerative colitis (UC), epithelial dysfunction is a core feature, but little is known about the cellular changes that may underlie disease pathology. We therefore evaluated how the chromatin epigenetics and proteome of epithelial cells differs between health and UC. Methods: We sorted live CD326+ epithelial cells from colon biopsies of healthy control (HC) screening colonoscopy recipients and from inflamed or uninflamed colon segments of UC patients on no biologic nor immunomodulator therapy (n = 5-7 subjects per group). Cell lysates were analyzed by proteomic evaluation and nuclei were analyzed for open chromatin with assay for transposase-accessible chromatin using sequencing. Results: Proteins most highly elevated in inflamed UC biopsies relative to HC were those encoded by the HLA-DRA (P = 3.1 × 10-33) and CD74 (P = 1.6 × 10-27), genes associated with antigen presentation, and the antimicrobial dual oxidase 2 (DUOX2) (P = 3.2 × 10-28) and lipocalin-2 (P = 2.2 × 10-26) genes. Conversely, the water channel aquaporin 8 was strikingly less common with inflammation (P = 1.9 × 10-18). Assay for transposase-accessible chromatin using sequencing revealed more open chromatin around the aquaporin 8 gene in HCs (P = 2.0 × 10-2) and more around the DUOX2/DUOXA2 locus in inflamed UC colon (P = 5.7 × 10-4), suggesting an epigenetic basis for differential protein expression by epithelial cells in health and disease. Conclusion: Numerous differences exist between the proteome and chromatin of colonic epithelial cells in UC patients and HCs, some of which correlate to suggest specific epigenetic mechanisms regulating the epithelial proteome.

2.
Sci Rep ; 13(1): 8922, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37264117

ABSTRACT

The intestinal epithelium comprises diverse cell types and executes many specialized functions as the primary interface between luminal contents and internal organs. A key function provided by the epithelium is maintenance of a barrier that protects the individual from pathogens, irritating luminal contents, and the microbiota. Disruption of this barrier can lead to inflammatory disease within the intestinal mucosa, and, in more severe cases, to sepsis. Animal models to study intestinal permeability are costly and not entirely predictive of human biology. Here we present a model of human colon barrier function that integrates primary human colon stem cells into Draper's PREDICT96 microfluidic organ-on-chip platform to yield a high-throughput system appropriate to predict damage and healing of the human colon epithelial barrier. We have demonstrated pharmacologically induced barrier damage measured by both a high throughput molecular permeability assay and transepithelial resistance. Using these assays, we developed an Inflammatory Bowel Disease-relevant model through cytokine induced damage that can support studies of disease mechanisms and putative therapeutics.


Subject(s)
Colon , Inflammatory Bowel Diseases , Animals , Humans , Disease Models, Animal , Colon/metabolism , Inflammatory Bowel Diseases/metabolism , Intestines , Intestinal Mucosa/metabolism , Permeability
3.
Inflamm Bowel Dis ; 29(2): 195-206, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36356046

ABSTRACT

BACKGROUND: Patient-derived organoid (PDO) models offer potential to transform drug discovery for inflammatory bowel disease (IBD) but are limited by inconsistencies with differentiation and functional characterization. We profiled molecular and cellular features across a range of intestinal organoid models and examined differentiation and establishment of a functional epithelial barrier. METHODS: Patient-derived organoids or monolayers were generated from control or IBD patient-derived colon or ileum and were molecularly or functionally profiled. Biological or technical replicates were examined for transcriptional responses under conditions of expansion or differentiation. Cell-type composition was determined by deconvolution of cell-associated gene signatures and histological features. Differentiated control or IBD-derived monolayers were examined for establishment of transepithelial electrical resistance (TEER), loss of barrier integrity in response to a cocktail of interferon (IFN)-γ and tumor necrosis factor (TNF)-α, and prevention of cytokine-induced barrier disruption by the JAK inhibitor, tofacitinib. RESULTS: In response to differentiation media, intestinal organoids and monolayers displayed gene expression patterns consistent with maturation of epithelial cell types found in the human gut. Upon differentiation, both colon- and ileum-derived monolayers formed functional barriers, with sustained TEER. Barrier integrity was compromised by inflammatory cytokines IFN-γ and TNF-α, and damage was inhibited in a dose-dependent manner by tofacitinib. CONCLUSIONS: We describe the generation and characterization of human colonic or ileal organoid models capable of functional differentiation to mature epithelial cell types. In monolayer culture, these cells formed a robust epithelial barrier with sustained TEER and responses to pharmacological modulation. Our findings demonstrate that control and IBD patient-derived organoids possess consistent transcriptional and functional profiles that can enable development of epithelial-targeted therapies.


Subject(s)
Inflammatory Bowel Diseases , Intestines , Organoids , Humans , Cytokines/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Organoids/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Intestines/physiology
4.
J Immunol ; 207(12): 3070-3080, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34789557

ABSTRACT

IL-33 is a multifunctional cytokine that mediates local inflammation upon tissue damage. IL-33 is known to act on multiple cell types including group 2 innate lymphoid cells (ILC2s), Th2 cells, and mast cells to drive production of Th2 cytokines including IL-5 and IL-13. IL-33 signaling activity through transmembrane ST2L can be inhibited by soluble ST2 (sST2), which acts as a decoy receptor. Previous findings suggested that modulation of IL-13 levels in mice lacking decoy IL-13Rα2, or mice lacking IL-13, impacted responsiveness to IL-33. In this study, we used Il13 -/- mice to investigate whether IL-13 regulates IL-33 activity by modulating the transmembrane and soluble forms of ST2. In Il13 -/- mice, the effects of IL-33 administration were exacerbated relative to wild type (WT). Il13 -/- mice administered IL-33 i.p. had heightened splenomegaly, more immune cells in the peritoneum including an expanded ST2L+ ILC2 population, increased eosinophilia in the spleen and peritoneum, and reduced sST2 in the circulation and peritoneum. In the spleen, lung, and liver of mice given IL-33, gene expression of both isoforms of ST2 was increased in Il13 -/- mice relative to WT. We confirmed fibroblasts to be an IL-13-responsive cell type that can regulate IL-33 activity through production of sST2. This study elucidates the important regulatory activity that IL-13 exerts on IL-33 through induction of IL-33 decoy receptor sST2 and through modulation of ST2L+ ILC2s.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Animals , Cytokines , Immunity, Innate , Interleukin-13 , Lymphocytes/metabolism , Mice
5.
Mucosal Immunol ; 12(5): 1174-1186, 2019 09.
Article in English | MEDLINE | ID: mdl-31308480

ABSTRACT

There continues to be a major need for more effective inflammatory bowel disease (IBD) therapies. IL-13Rα2 is a decoy receptor that binds the cytokine IL-13 with high affinity and diminishes its STAT6-mediated effector functions. Previously, we found that IL-13Rα2 was necessary for IBD in mice deficient in the anti-inflammatory cytokine IL-10. Here, we tested for the first time a therapeutic antibody specifically targeting IL-13Rα2. We also used the antibody and Il13ra2-/- mice to dissect the role of IL-13Rα2 in IBD pathogenesis and recovery. Il13ra2-/- mice were modestly protected from induction of dextran sodium sulfate (DSS)-induced colitis. Following a 7-day recovery period, Il13ra2-/- mice or wild-type mice administered the IL-13Rα2-neutralizing antibody had significantly improved colon health compared to control mice. Neutralizing IL-13Rα2 to increase IL-13 bioavailability promoted resolution of IBD even if neutralization occurred only during recovery. To link our observations in mice to a large human cohort, we conducted a phenome-wide association study of a more active variant of IL-13 (R130Q) that has reduced affinity for IL-13Rα2. Human subjects carrying R130Q reported a lower risk for Crohn's disease. Our findings endorse moving anti-IL-13Rα2 into preclinical drug development with the goal of accelerating recovery and maintaining remission in Crohn's disease patients.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Inflammatory Bowel Diseases/metabolism , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/metabolism , Animals , Crohn Disease/etiology , Crohn Disease/metabolism , Crohn Disease/pathology , Dextran Sulfate/adverse effects , Disease Models, Animal , Disease Susceptibility , Eosinophils/immunology , Eosinophils/metabolism , Gain of Function Mutation , Genetic Variation , Humans , Immunity , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/pathology , Interleukin-13 Receptor alpha2 Subunit/genetics , Mice , Odds Ratio
6.
Adv Ther ; 36(4): 757-765, 2019 04.
Article in English | MEDLINE | ID: mdl-30758742

ABSTRACT

Progressive multifocal leukoencephalopathy (PML) is a rare but serious disease. Caused by the JC virus (JCV), it occurs in individuals with weakened immune systems and is a potential adverse reaction for certain immunomodulatory drugs. The PML Consortium was created to find better methods to predict, prevent, and treat PML. The Consortium brought together the pharmaceutical industry with academic, regulatory, and patient communities to advance research and dialogue on PML through a not-for-profit, collaborative approach involving a grant program, scientific workshops and conferences, and disease awareness efforts. Over nearly a decade, the Consortium contributed to the PML and JCV fields by advancing research, scientific exchange, and awareness of PML. In addition to advancing knowledge and helping to build cross-sector consensus on research priorities, the Consortium's grant program filled a funding gap and brought new investigators into PML and JCV research. Additionally, the Consortium's workshops and conferences created platforms for exchange that drove dialogue on knowledge gaps and future research directions. The Consortium also contributed to the scientific knowledge base with two literature reviews, one on PML treatment studies and a second on T cell deficiencies as a risk factor for PML and the brain as a site for conversion of harmless JCV into a pathogenic virus. Finally, the Consortium addressed a significant information gap with its disease awareness website for healthcare professionals, patients, and caregivers. Beyond its impact on the PML and JCV fields, the PML Consortium is important because it provides a precedent for how the pharmaceutical industry, academic researchers, patient organizations, and government can work together to address rare diseases, in particular rare adverse events. This kind of collaboration could be replicated to speed progress in addressing other rare diseases and adverse events, with significant potential benefits for the scientific, medical, and patient communities. FUNDING: PML Consortium (PML Consortium, Washington, DC).


Subject(s)
Drug-Related Side Effects and Adverse Reactions/prevention & control , Leukoencephalopathy, Progressive Multifocal , Organizations, Nonprofit , Humans , Intersectoral Collaboration , JC Virus , Leukoencephalopathy, Progressive Multifocal/therapy , Leukoencephalopathy, Progressive Multifocal/virology , Research , Stakeholder Participation
7.
Drug Metab Dispos ; 46(11): 1581-1587, 2018 11.
Article in English | MEDLINE | ID: mdl-30126862

ABSTRACT

Intestinal disposition of small molecules involves interplay of drug metabolizing enzymes (DMEs), transporters, and host-microbiome interactions, which has spurred the development of in vitro intestinal models derived from primary tissue sources. Such models have been bioengineered from intestinal crypts, mucosal extracts, induced pluripotent stem cell (iPSC)-derived organoids, and human intestinal tissue. This minireview discusses the utility and limitations of these human-derived models in support of small molecule drug metabolism and disposition. Enteroids from human intestinal crypts, organoids derived from iPSCs using growth factors or small molecule compounds, and enterocytes extracted from mucosal scrapings show key absorptive cell morphology while are limited in quantitative applications due to the lack of accessibility to the apical compartment, the lack of monolayers, or low expression of key DMEs, transporters, and nuclear hormone receptors. Despite morphogenesis to epithelial cells, similar challenges have been reported by more advanced technologies that have explored the impact of flow and mechanical stretch on proliferation and differentiation of Caco-2 cells. Most recently, bioengineered human intestinal epithelial or ileal cells have overcome many of the challenges, as the DME and transporter expression pattern resembles that of native intestinal tissue. Engineering advances may improve such models to support longer-term applications and meet end-user needs. Biochemical characterization and transcriptomic, proteomic, and functional endpoints of emerging novel intestinal models, when referenced to native human tissue, can provide greater confidence and increased utility in drug discovery and development.


Subject(s)
Biological Transport/physiology , Inactivation, Metabolic/physiology , Intestines/physiology , Small Molecule Libraries/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Humans , Membrane Transport Proteins/metabolism
8.
J Immunol ; 200(4): 1347-1359, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29305434

ABSTRACT

In obesity, IL-13 overcomes insulin resistance by promoting anti-inflammatory macrophage differentiation in adipose tissue. Endogenous IL-13 levels can be modulated by the IL-13 decoy receptor, IL-13Rα2, which inactivates and depletes the cytokine. In this study, we show that IL-13Rα2 is markedly elevated in adipose tissues of obese mice. Mice deficient in IL-13Rα2 had high expression of IL-13 response markers in adipose tissue, consistent with increased IL-13 activity at baseline. Moreover, exposure to the type 2 cytokine-inducing alarmin, IL-33, enhanced serum and tissue IL-13 concentrations and elevated tissue eosinophils, macrophages, and type 2 innate lymphoid cells. IL-33 also reduced body weight, fat mass, and fasting blood glucose levels. Strikingly, however, the IL-33-induced protection was greater in IL-13Rα2-deficient mice compared with wild-type littermates, and these changes were largely attenuated in mice lacking IL-13. Although IL-33 administration improved the metabolic profile in the context of a high fat diet, it also resulted in diarrhea and perianal irritation, which was enhanced in the IL-13Rα2-deficient mice. Weight loss in this group was associated with reduced food intake, which was likely related to the gastrointestinal effects. These findings outline both potentially advantageous and deleterious effects of a type 2-skewed immune response under conditions of metabolic stress, and identify IL-13Rα2 as a critical checkpoint in adipose tissues that limits the protective effects of the IL-33/IL-13 axis in obesity.


Subject(s)
Interleukin-13 Receptor alpha2 Subunit/metabolism , Interleukin-13/metabolism , Interleukin-33/metabolism , Obesity/immunology , Obesity/metabolism , Adipose Tissue/immunology , Adipose Tissue/metabolism , Animals , Humans , Interleukin-13/immunology , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-33/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
9.
MAbs ; 8(5): 983-90, 2016 07.
Article in English | MEDLINE | ID: mdl-27049478

ABSTRACT

IMA-638 and IMA-026 are humanized IgG1 monoclonal antibodies (mAbs) that target non-overlapping epitopes of IL-13. Separate first-in-human single ascending dose studies were conducted for each mAb. These studies had similar study designs, but mild to moderate asthmatics were recruited for the IMA-638 study and healthy subjects were recruited for the IMA-026 study. IMA-638 and IMA-026 showed similar pharmacokinetic (PK) profiles, but very different total IL-13 (free and drug bound IL-13) profiles; free IL13 was not measured. IMA-026 treatment induced a dose-dependent accumulation of total IL-13, while IMA-638 treatment led to a much smaller accumulation without any clear dose-response. To understand the differences between the two total IL-13 profiles and to predict the free IL-13 profiles for each mAb treatment, a mechanistic PK/pharmacodynamic model was developed. PK-related parameters were first fit to the mean PK profiles of each mAb separately; thereafter, the target-related parameters were fit to both total IL-13 profiles simultaneously. The IL-13 degradation rate was assumed to be the same for asthma patients and healthy subjects. The model suggests that an approximately 100× faster elimination of IL-13-IMA-638 complex than IL-13-IMA-026 complex could be responsible for the differences observed in total IL-13 profiles for the two mAbs. Furthermore, the model predicts that IMA-638 administration results in greater and more prolonged free IL-13 inhibition than equivalent dosing of IMA-026 despite similar binding KD and PK profile. In conclusion, joint modeling of two similar molecules provided mechanistic insight that the elimination rate of mAb-target complex can regulate the degree of free target inhibition.


Subject(s)
Anti-Asthmatic Agents/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Asthma/drug therapy , Interleukin-13/antagonists & inhibitors , Models, Theoretical , Adult , Female , Humans , Immunoglobulin G/pharmacology , Male
10.
Immunology ; 143(3): 416-27, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24831554

ABSTRACT

Interleukin-4 (IL-4) and IL-13 are critical drivers of immune activation and inflammation in ulcerative colitis, asthma and other diseases. Because these cytokines may have redundant function, dual targeting holds promise for achieving greater efficacy. We have recently described a bifunctional therapeutic targeting IL-4 and IL-13 developed on a novel protein scaffold, generated by combining specific binding domains in an optimal configuration using appropriate linker regions. In the current study, the bifunctional IL-4/IL-13 antagonist was evaluated in the murine oxazolone-induced colitis model, which produces disease with features of ulcerative colitis. The bifunctional IL-4/IL-13 antagonist reduced body weight loss throughout the 7-day course of the model, and ameliorated the increased colon weight and decreased colon length that accompany disease. Colon tissue gene expression was modulated in accordance with the treatment effect. Concentrations of serum amyloid P were elevated in proportion to disease severity, making it an effective biomarker. Serum concentrations of the bifunctional IL-4/IL-13 antagonist were inversely proportional to disease severity, colon tissue expression of pro-inflammatory genes, and serum amyloid P concentration. Taken together, these results define a panel of biomarkers signifying engagement of the IL-4/IL-13 pathway, confirm the T helper type 2 nature of disease in this model, and demonstrate the effectiveness of dual cytokine blockade.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colitis, Ulcerative/metabolism , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/pharmacology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Disease Models, Animal , Gene Expression Regulation/drug effects , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Mice , Oxazolone/adverse effects , Recombinant Fusion Proteins/administration & dosage , Serum Amyloid A Protein/metabolism , Serum Amyloid P-Component/metabolism , Severity of Illness Index
11.
Am J Respir Cell Mol Biol ; 49(1): 37-46, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23449738

ABSTRACT

IL-4 and IL-13 comprise promising targets for therapeutic interventions in asthma and other Th2-associated diseases, but agents targeting either IL-4 or IL-13 alone have shown limited efficacy in human clinical studies. Because these cytokines may involve redundant function, dual targeting holds promise for achieving greater efficacy. We describe a bifunctional therapeutic targeting IL-4 and IL-13, developed by a combination of specific binding domains. IL-4-targeted and IL-13-targeted single chain variable fragments were joined in an optimal configuration, using appropriate linker regions on a novel protein scaffold. The bifunctional IL-4/IL-13 antagonist displayed high affinity for both cytokines. It was a potent and efficient neutralizer of both murine IL-4 and murine IL-13 bioactivity in cytokine-responsive Ba/F3 cells, and exhibited a half-life of approximately 4.7 days in mice. In a murine model of ovalbumin-induced ear swelling, the bifunctional molecule blocked both the IL-4/IL-13-dependent early-phase response and the IL-4-dependent late-phase response. In the ovalbumin-induced lung inflammation model, the bifunctional IL-4/IL-13 antagonist reduced the IL-4-dependent rise in serum IgE titers, and reduced IL-13-dependent airway hyperresponsiveness, lung inflammation, mucin gene expression, and serum chitinase responses. Taken together, these findings demonstrate the effective dual blockade of IL-4 and IL-13 with a single agent, which resulted in the modulation of a more extensive range of endpoints than could be achieved by targeting either cytokine alone.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Immunoglobulin E/immunology , Interleukin-13/antagonists & inhibitors , Interleukin-4/antagonists & inhibitors , Pneumonia/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/immunology , Binding Sites , Bronchial Hyperreactivity/drug therapy , Bronchial Hyperreactivity/immunology , CHO Cells , Cricetinae , Cross-Linking Reagents/chemistry , Ear/physiopathology , Female , Half-Life , Interleukin-13 Receptor alpha2 Subunit/immunology , Interleukin-13 Receptor alpha2 Subunit/metabolism , Mice , Mice, Inbred C57BL , Molecular Conformation , Neutralization Tests , Ovalbumin/adverse effects , Ovalbumin/immunology , Pneumonia/immunology , Protein Binding , Protein Interaction Domains and Motifs , Single-Chain Antibodies/metabolism
12.
J Immunol ; 187(1): 561-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21622864

ABSTRACT

Human studies using Abs to two different, nonoverlapping epitopes of IL-13 suggested that epitope specificity can have a clinically significant impact on clearance of IL-13. We propose that Ab modulation of IL-13 interaction with IL-13Rα2 underlies this effect. Two Abs were administered to healthy subjects and mild asthmatics in separate dose-ranging studies and allergen-challenge studies. IMA-638 allows IL-13 interaction with IL-13Rα1 or IL-13Rα2 but blocks recruitment of IL-4Rα to the IL-13/IL-13Rα1 complex, whereas IMA-026 competes with IL-13 interaction with IL-13Rα1 and IL-13Rα2. We found ∼10-fold higher circulating titer of captured IL-13 in subjects treated with IMA-026 compared with those administered IMA-638. To understand how this difference could be related to epitope, we asked whether either Ab affects IL-13 internalization through cell surface IL-13Rα2. Humans inducibly express cell surface IL-13Rα2 but lack the soluble form that regulates IL-13 responses in mice. Cells with high IL-13Rα2 expression rapidly and efficiently depleted extracellular IL-13, and this activity persisted in the presence of IMA-638 but not IMA-026. The potency and efficiency of this clearance pathway suggest that cell surface IL-13Rα2 acts as a scavenger for IL-13. These findings could have important implications for the design and characterization of IL-13 antagonists.


Subject(s)
Interleukin-13 Receptor alpha2 Subunit/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Isoantibodies/physiology , Receptors, Scavenger/metabolism , Animals , Antibody-Dependent Cell Cytotoxicity/immunology , Dose-Response Relationship, Immunologic , Drug Delivery Systems , Extracellular Space/immunology , Extracellular Space/metabolism , HT29 Cells , Humans , Interleukin-13/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/antagonists & inhibitors , Interleukin-13 Receptor alpha2 Subunit/biosynthesis , Macaca fascicularis , Mice , Mice, Inbred BALB C , Receptors, Scavenger/antagonists & inhibitors , Receptors, Scavenger/physiology
13.
Rheumatology (Oxford) ; 50(6): 1033-44, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21258049

ABSTRACT

OBJECTIVES: To characterize the in vitro binding and effector function properties of CD20-directed small modular immunopharmaceutical (SMIP) 2LM20-4, and to compare its in vivo B-cell depletion activity with the mutated 2LM20-4 P331S [no in vitro complement-dependent cytotoxicity (CDC)] and rituximab in cynomolgus monkeys. METHODS: Direct binding is examined in flow cytometry, confocal microscopy, scatchard and lipid raft assays. Effector function assays include CDC and Fc-mediated cellular toxicity. In the 6-month-long in vivo B-cell depletion study, single i.v. dosages of 1 or 10 mg/kg of anti-CD20 proteins were administered to monkeys and B-cell counts were monitored in peripheral blood, bone marrow and lymph nodes. RESULTS: 2LM20-4 has lower saturation binding to human primary B cells and recruits fewer CD20 molecules into lipid rafts compared with rituximab; however, it induces higher in vitro CDC. In competitive binding, 2LM20-4 only partially displaces rituximab, suggesting that it binds to a fraction of CD20 molecules within certain locations of the plasma membrane as compared with rituximab. In monkeys, 2LM20-4 had more sustained B-cell depletion activity than rituximab in peripheral blood and had significantly more profound and sustained activity than 2LM20-4 P331S and rituximab in the lymph nodes. CONCLUSIONS: SMIP 2LM20-4, which binds to a fraction of CD20 molecules as compared with rituximab, has more potent in vitro CDC, and more potent and sustained B-cell depletion activity in cynomolgus monkeys. Our work has considerable clinical relevance since it provides novel insights related to the emerging B-cell depletion therapies in autoimmune diseases.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD20/drug effects , Antigens, CD20/immunology , Single-Chain Antibodies/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Binding Sites , Cells, Cultured , Disease Models, Animal , Humans , Immunologic Factors/pharmacology , In Vitro Techniques , Linear Models , Macaca fascicularis , Random Allocation , Rituximab , Sensitivity and Specificity
14.
Am J Respir Crit Care Med ; 183(8): 1007-14, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21057005

ABSTRACT

RATIONALE: Extensive evidence in animal models supports a role for IL-13 in the pathobiology of asthma. IMA-638 and IMA-026 are fully humanized IgG(1) antibodies that bind to different epitopes and neutralize IL-13 bioactivity. OBJECTIVES: We hypothesized that anti-IL-13 treatment would inhibit allergen-induced late-phase asthmatic responses, airway hyperresponsiveness, and inflammation in subjects with asthma. METHODS: Fifty-six subjects with mild, atopic asthma were recruited for two double-blind, randomized, placebo-controlled, parallel group trials to compare IMA-638 and IMA-026 IL-13 antibody treatments with placebo treatment. Drug was administered on Days 1 and 8, and allergen challenges were performed on Days 14 and 35. The primary outcome variable was the late-phase area under the curve (AUC), and secondary outcome variables were the early- and late-phase maximum percent fall in FEV(1), early AUC, allergen-induced shift in airway hyperresponsiveness, and sputum eosinophils. MEASUREMENTS AND MAIN RESULTS: The treatment difference with IMA-638 on Day 14 was -19.1 FEV(1) × hour (95% confidence interval: -36.2, -1.9) for the allergen-induced early AUC and -23.8 FEV(1) × hour (95% confidence interval: -46.4, -1.2) for the late AUC (both P < 0.05), but this effect was lost by Day 35. Treatment with IMA-026 did not attenuate the asthmatic responses on Day 14 or Day 35. There was no effect of either antibody on allergen-induced airway hyperresponsiveness or sputum eosinophils. The frequency of adverse events after administration of the IL-13 antibodies was similar to placebo. CONCLUSIONS: IL-13 has a role in allergen-induced airway responses in humans. Further study is required to determine whether anti-IL-13 monoclonal antibodies will be beneficial clinically.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Asthma/drug therapy , Interleukin-13/antagonists & inhibitors , Adolescent , Adult , Allergens/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Double-Blind Method , Female , Forced Expiratory Volume , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Immunoglobulin G/therapeutic use , Interleukin-13/blood , Interleukin-13/immunology , Male , Middle Aged , Sputum/cytology , Sputum/immunology , Treatment Outcome , Young Adult
15.
Assay Drug Dev Technol ; 8(1): 73-84, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20085454

ABSTRACT

The chemokine receptor CXCR5 is predominantly expressed on mature B cells and follicular T-helper cells. CXCR5 and its ligand CXCL13 participate in ectopic germinal center formation at the inflammatory sites of multiple immune diseases such as rheumatoid arthritis, multiple sclerosis, and Sjogren's syndrome. Therefore, disrupting CXCL13-induced chemotaxis may be a fruitful approach for developing therapeutics in treating these diseases. Cells undergo cytoskeletal rearrangement prior to chemotaxis, and therefore actin polymerization can be used as a surrogate readout more proximal to chemokine receptor activation than chemotaxis. Conventionally, actin polymerization is measured by fluorescence microscopy or flow cytometry, which are either of low throughput or in need of special instruments. We developed a 96-well actin polymerization assay that can process 1,000 to 1,500 samples a day. This assay uses a standard laboratory fluorescence microplate reader as the detection instrument and was optimized for various experimental conditions such as cell density, actin filament staining reagent, staining buffer, and cell culture conditions. We demonstrate that this actin polymerization assay in 96-well format exhibits the expected pharmacology for human CXCR5 and is suitable as a primary functional assay to screen neutralizing scFv in crude bacterial peri-preps and a secondary assay for small compound collections.


Subject(s)
Actins/metabolism , Chemokine CXCL13/pharmacology , Animals , Cell Count , Cell Line , Flow Cytometry , Fluorescence , Humans , Mice , Microscopy, Confocal , Temperature
16.
Inflamm Res ; 59(3): 207-18, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19763788

ABSTRACT

OBJECTIVE: Mast cell and basophil activation contributes to inflammation, bronchoconstriction, and airway hyperresponsiveness in asthma. Because IL-33 expression is inflammation inducible, we investigated IL-33-mediated effects in concert with both IgE-mediated and IgE-independent stimulation. METHODS: Because the HMC-1 mast cell line can be activated by GPCR and RTK signaling, we studied the effects of IL-33 on these pathways. The IL-33- and SCF-stimulated HMC-1 cells were co-cultured with human lung fibroblasts and airway smooth muscle cells in a collagen gel contraction assay. IL-33 effects on IgE-mediated activation were studied in primary mast cells and basophils. RESULT: IL-33 synergized with adenosine, C5a, SCF, and NGF receptor activation. IL-33-stimulated and SCF-stimulated HMC-1 cells demonstrated enhanced collagen gel contraction when cultured with fibroblasts or smooth muscle cells. IL-33 also synergized with IgE receptor activation of primary human mast cells and basophils. CONCLUSION: IL-33 amplifies inflammation in both IgE-independent and IgE-dependent responses.


Subject(s)
Basophils/drug effects , Basophils/metabolism , Immunoglobulin E/metabolism , Interleukins/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Basophils/cytology , Cell Line , Chemokines/metabolism , Coculture Techniques , Collagen/metabolism , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Histamine/metabolism , Humans , Interleukin-33 , MAP Kinase Kinase 4/metabolism , Mast Cells/cytology , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Receptors, Purinergic P1/metabolism , Stem Cell Factor/pharmacology
17.
J Immunol Methods ; 350(1-2): 161-70, 2009 Oct 31.
Article in English | MEDLINE | ID: mdl-19732777

ABSTRACT

IL-13 is a Th2 cytokine that has been shown to be an important mediator of airway inflammation contributing to asthma lesions. Given its proposed role in asthma, measurements of this cytokine in serum may provide insights into disease mechanisms, progression and pharmacodynamic effects of IL-13 targeted therapeutics. However, current commercially available ELISA immunoassays are frequently unable to detect baseline concentrations of IL-13 in serum from healthy individuals, which are below the limit of detection. Here we describe the use of the novel microparticle-based Erenna IL-13 human immunoassay (Singulex, Inc.), which utilizes proprietary antibodies and single molecule counting technology, to quantify IL-13 from 100 microL of serum from apparently healthy subjects and clinically defined symptomatic and asymptomatic asthma subjects. The lower limit of quantification of the Erenna assay was validated at 0.07 pg/mL and the assay detected baseline concentrations of IL-13 in 98% of serum samples tested. The calibration curve showed good precision over the entire linear range of 0.07-50 pg/mL, with inter-assay imprecision <10% CV except at the lowest concentration tested (<15%). The intra- and inter-assay imprecision of spiked serum samples containing three different IL-13 concentrations (2, 8, and 25 pg/mL) ranged from 2.2-2.4% and 6.1-6.8%, respectively. Using the Erenna IL-13 assay, we observe that serum IL-13 concentrations range from <0.07-1.02 pg/mL in apparently healthy subjects (N=60) with similar ranges in asymptomatic (0.07-0.66 pg/mL, N=26) and symptomatic (<0.07-1.26 pg/mL, N=96) asthma subjects. The Erenna immunoassay improved sensitivity by over two full logs compared to previous ELISA methods, while using smaller sample volumes. In addition, the Erenna assay reliably measured IL-13 in endogenous and spiked human serum samples that were not quantifiable using other methods. Taken together, these results show that this novel assay offers a significant improvement over previous methods for high-sensitive quantitative measurement of IL-13 in human serum samples obtained from both apparently healthy and asthmatic subjects, and can be used in future clinical studies to accurately measure concentrations of this cytokine prior to and following drug therapy in human serum.


Subject(s)
Asthma/blood , Interleukin-13/blood , Reagent Kits, Diagnostic , Calibration , Enzyme-Linked Immunosorbent Assay/methods , Fluorescent Antibody Technique, Direct/methods , Fluorescent Antibody Technique, Direct/standards , Humans , Interleukin-13/analysis , Sensitivity and Specificity
18.
J Immunol ; 183(3): 1739-50, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19592653

ABSTRACT

In asthma, mast cells infiltrate the airway smooth muscle cell layer and secrete proinflammatory and profibrotic agents that contribute to airway remodeling. To study the effects of mast cell activation on smooth muscle cell-dependent matrix contraction, we developed coculture systems of human airway smooth muscle cells (HASM) with primary human mast cells derived from circulating progenitors or with the HMC-1 human mast cell line. Activation of primary human mast cells by IgE receptor cross-linking or activation of HMC-1 cells with C5a stimulated contraction of HASM-embedded collagen gels. Contractile activity could be transferred with conditioned medium from activated mast cells, implicating involvement of soluble factors. Cytokines and proteases are among the agents released by activated mast cells that may promote a contractile response. Both IL-13 and IL-6 enhanced contraction in this model and the activity of IL-13 was ablated under conditions leading to expression of the inhibitory receptor IL-13Ralpha2 on HASM. In addition to cytokines, matrix metalloproteinases (MMPs), and serine proteases induced matrix contraction. Inhibitor studies suggested that, although IL-13 could contribute to contraction driven by mast cell activation, MMPs were critical mediators of the response. Both MMP-1 and MMP-2 were strongly expressed in this system. Serine proteases also contributed to contraction induced by mast cell-activating agents and IL-13, most likely by mediating the proteolytic activation of MMPs. Hypercontractility is a hallmark of smooth muscle cells in the asthmatic lung. Our findings define novel mechanisms whereby mast cells may modulate HASM-driven contractile responses.


Subject(s)
Mast Cells/physiology , Muscle Contraction , Myocytes, Smooth Muscle/physiology , Paracrine Communication , Respiratory System/cytology , Coculture Techniques , Collagen , Cytokines/physiology , Extracellular Matrix/enzymology , Humans , Interleukin-13/physiology , Interleukin-6/physiology , Metalloproteases/physiology , Muscle, Smooth , Serine Endopeptidases/physiology
19.
Cytokine ; 46(1): 111-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19237298

ABSTRACT

IL-13 is known to affect many processes that contribute to an asthmatic phenotype, including inflammation, fibrosis, and mucus production. Members of the aquaporin (AQP) family of transmembrane water channels are targets of regulation in models of lung injury and inflammation. Therefore, we examined AQP mRNA and protein expression in allergen and IL-13-induced mouse models of asthma. Lungs from ovalbumin sensitized and ovalbumin challenged (OVA/OVA) and IL-13 treated mice showed airway thickening, increased mucus production, and pulmonary eosinophilia. Pulmonary function tests showed a significant increase in methacholine-induced airway hyperreactivity in OVA/OVA and IL-13-treated mice as compared with controls. Quantitative PCR analysis revealed differential regulation of AQPs in these two models. AQP1 and AQP4 mRNA expression was downregulated in the OVA/OVA model, but not in the IL-13 model. AQP5 mRNA was reduced in both models, whereas AQP3 was upregulated only in the IL-13 model. Western analysis showed that diminished expression of an apically localized aquaporin, (AQP5), and concomitant upregulation of a basolateral aquaporin (AQP3 or AQP4) are characteristic features of both inducible asthma models. These results demonstrate that aquaporins are common targets of gene expression in both allergen and IL-13 induced mouse models of asthma.


Subject(s)
Aquaporin 5/biosynthesis , Aquaporin 5/metabolism , Asthma/metabolism , Gene Expression Regulation , Interleukin-13/biosynthesis , Animals , Aquaporin 1/biosynthesis , Aquaporin 4/biosynthesis , Bronchoconstrictor Agents/pharmacology , Disease Models, Animal , Female , Inflammation , Lung/metabolism , Methacholine Chloride/pharmacology , Mice , Mice, Inbred BALB C , Models, Biological
20.
Am J Physiol Lung Cell Mol Physiol ; 296(2): L236-47, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19060229

ABSTRACT

Mast cell-fibroblast interactions may contribute to fibrosis in asthma and other disease states. Fibroblast contraction is known to be stimulated by coculture with the human mast cell line, HMC-1, or by mast cell-derived agents. Matrix metalloproteinases (MMPs) can also mediate contraction, but the MMP-dependence of mast cell-induced fibroblast contractility is not established, and the consequences of mast cell activation within the coculture system have not been fully explored. We demonstrate that activation of primary human mast cells (pHMC) with IgE receptor cross-linking, or activation of HMC-1 with C5a, enhanced contractility of human lung fibroblasts in a three-dimensional collagen lattice system. This enhanced contractility was inhibited by the pan-MMP antagonist, batimastat, and was transferrable in the conditioned medium of activated mast cells. Exogenously added MMPs promoted gel contraction by mediating the proteolytic activation of latent transforming growth factor-beta (TGF-beta). Consistent with this, fibroblast contraction induced by mast cell activation was enhanced by addition of excess latent TGF-beta to the cultures. Batimastat inhibited this response, suggesting that MMPs capable of activating latent TGF-beta were released following mast cell activation in coculture with fibroblasts. Collagen production was also stimulated by activated mast cells in an MMP-dependent manner. MMP-2 and MMP-3 content of the gels increased in the presence of activated mast cells, and inhibition of these enzymes blocked the contractile response. These findings demonstrate the MMP dependence of mast cell-induced fibroblast contraction and collagen production.


Subject(s)
Collagen Type I/metabolism , Fibroblasts/metabolism , Mast Cells/physiology , Matrix Metalloproteinases/metabolism , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/pharmacology , Enzyme-Linked Immunosorbent Assay , Fibroblasts/drug effects , Humans , Immunoglobulin E/metabolism , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Protease Inhibitors/pharmacology , Thiophenes/pharmacology , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL