Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Crohns Colitis ; 15(6): 938-949, 2021 Jun 22.
Article in English | MEDLINE | ID: mdl-33599720

ABSTRACT

BACKGROUND AND AIMS: Ontamalimab, a fully-human monoclonal antibody targeting MAdCAM-1, induced remission in patients with moderate-to-severe ulcerative colitis [UC] in the TURANDOT study. We aimed to assess long-term safety, tolerability, and efficacy of ontamalimab in TURANDOT II. METHODS: TURANDOT II was a phase 2, multicentre, open-label [OL] study in patients with moderate-to-severe UC who completed TURANDOT on placebo or ontamalimab (NCT01771809). Patients were randomised to 75 mg or 225 mg ontamalimab every 4 weeks for 72 weeks [OL1]. The dosage could be increased to 225 mg from Week 8 at the investigator's discretion. All patients then received 75 mg every 4 weeks for 72 weeks [OL2], followed by 6-month safety follow-up. The primary objective was safety, measured by adverse events [AEs], serious AEs [SAEs], and AEs leading to withdrawal. Mucosal healing [MH; centrally read endoscopy] was assessed. RESULTS: Of 330 patients, 180 completed OL1; 94 escalated to 225 mg; 127 completed OL2. Overall, 36.1% experienced drug-related AEs. The most common SAE [10.0%] was worsening/ongoing UC; 5.5% of patients had serious infections, the most common being gastroenteritis [0.9%]. One death and four cancers [all unrelated to ontamalimab] occurred. No PML [progressive multifocal leukoencephalopathy]/lymphoproliferative disorders occurred. Geometric mean high-sensitivity C-reactive protein [hsCRP] and faecal calprotectin decreased across OL1 in both dose groups. The proportion of patients assigned to placebo in TURANDOT achieving MH increased from 8.8% [6/68] at baseline to 35.3% at Week 16 [24/68; non-responder imputation]. The corresponding increase in the ontamalimab group was from 23.3% [61/262] to 26.7% [70/262]. CONCLUSIONS: Ontamalimab was well tolerated up to 144 weeks in patients with moderate-to-severe UC, with good safety and efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized , Cell Adhesion Molecules/antagonists & inhibitors , Colitis, Ulcerative , Drug Monitoring , Mucoproteins/antagonists & inhibitors , Adult , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , C-Reactive Protein/analysis , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Dose-Response Relationship, Immunologic , Drug Monitoring/methods , Drug Monitoring/statistics & numerical data , Endoscopy, Gastrointestinal/methods , Endoscopy, Gastrointestinal/statistics & numerical data , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Humans , Leukocyte L1 Antigen Complex/analysis , Male , Treatment Outcome
2.
J Hepatol ; 73(5): 1013-1022, 2020 11.
Article in English | MEDLINE | ID: mdl-32540177

ABSTRACT

BACKGROUND & AIMS: The heterodimeric integrin receptor α4ß7 regulates CD4 T cell recruitment to inflamed tissues, but its role in the pathogenesis of non-alcoholic steatohepatitis (NASH) is unknown. Herein, we examined the role of α4ß7-mediated recruitment of CD4 T cells to the intestine and liver in NASH. METHODS: Male littermate F11r+/+ (control) and junctional adhesion molecule A knockout F11r-/- mice were fed a normal diet or a western diet (WD) for 8 weeks. Liver and intestinal tissues were analyzed by histology, quantitative reverse transcription PCR (qRT-PCR), 16s rRNA sequencing and flow cytometry. Colonic mucosa-associated microbiota were analyzed using 16s rRNA sequencing. Liver biopsies from patients with NASH were analyzed by confocal imaging and qRT-PCR. RESULTS: WD-fed knockout mice developed NASH and had increased hepatic and intestinal α4ß7+ CD4 T cells relative to control mice who developed mild hepatic steatosis. The increase in α4ß7+ CD4 T cells was associated with markedly higher expression of the α4ß7 ligand mucosal addressin cell adhesion molecule 1 (MAdCAM-1) in the colonic mucosa and livers of WD-fed knockout mice. Elevated MAdCAM-1 expression correlated with increased mucosa-associated Proteobacteria in the WD-fed knockout mice. Antibiotics reduced MAdCAM-1 expression indicating that the diet-altered microbiota promoted colonic and hepatic MAdCAM-1 expression. α4ß7 blockade in WD-fed knockout mice significantly decreased α4ß7+ CD4 T cell recruitment to the intestine and liver, attenuated hepatic inflammation and fibrosis, and improved metabolic indices. MAdCAM-1 blockade also reduced hepatic inflammation and fibrosis in WD-fed knockout mice. Hepatic MAdCAM-1 expression was elevated in patients with NASH and correlated with higher expression of α4 and ß7 integrins. CONCLUSIONS: These findings establish α4ß7/MAdCAM-1 as a critical axis regulating NASH development through colonic and hepatic CD4 T cell recruitment. LAY SUMMARY: Non-alcoholic steatohepatitis (NASH) is an advanced and progressive form of non-alcoholic fatty liver disease (NAFLD), and despite its growing incidence no therapies currently exist to halt NAFLD progression. Herein, we show that blocking integrin receptor α4ß7-mediated recruitment of CD4 T cells to the intestine and liver not only attenuates hepatic inflammation and fibrosis, but also improves metabolic derangements associated with NASH. These findings provide evidence for the potential therapeutic application of α4ß7 antibody in the treatment of human NASH.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Diet, Western/adverse effects , Integrins/metabolism , Intestinal Mucosa/immunology , Liver/immunology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/deficiency , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Humans , Integrins/antagonists & inhibitors , Integrins/immunology , Liver/pathology , Male , Mice , Mice, Knockout , Mucoproteins/antagonists & inhibitors , Mucoproteins/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , RNA, Ribosomal, 16S/genetics , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics
3.
J Clin Pharmacol ; 60(7): 903-914, 2020 07.
Article in English | MEDLINE | ID: mdl-32119128

ABSTRACT

Ontamalimab (SHP647) is a fully human, immunoglobulin G2 , antihuman mucosal addressin cell adhesion molecule-1 (MAdCAM-1) monoclonal antibody being developed for the treatment of ulcerative colitis (UC) and Crohn's disease (CD). A population pharmacokinetic/pharmacodynamic (PK/PD) analysis was conducted using clinical phase 2 study data to evaluate the PK and PD of ontamalimab following subcutaneous administrations of 7.5, 22.5, 75, and 225 mg every 4 weeks in patients with moderate to severe UC or CD. A total of 440 patients with UC (n = 249; 56.6%) or CD (n = 191; 43.4%) were included in the analysis. A 2-compartment model with parallel linear and nonlinear elimination adequately characterized concentration-time profiles of ontamalimab. The apparent clearance and volume of distribution were 0.0127 L/h (0.305 L/day) and 6.53 L, respectively. Apparent clearance and volume of distribution were mainly dependent on baseline albumin and body weight, respectively. No differences in the PK properties of ontamalimab were observed between patients with UC or CD. The presence of antidrug antibodies did not impact the PK of ontamalimab. Nonlinear elimination occurred at very low concentrations and was unlikely to contribute to the elimination half-life under steady-state conditions. A linear PK/PD model described the relationship between ontamalimab and free MAdCAM-1. Minimum concentrations of ontamalimab at steady state following 75 mg every 4 weeks were associated with >95% suppression of circulating free MAdCAM-1. The PK/PD properties characterized support phase 3 testing in UC and CD.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Gastrointestinal Agents/pharmacology , Mucoproteins/antagonists & inhibitors , Adolescent , Adult , Aged , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/blood , Body Weight , C-Reactive Protein/metabolism , Cell Adhesion Molecules/blood , Clinical Trials, Phase II as Topic , Colitis, Ulcerative/blood , Crohn Disease/blood , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/blood , Humans , Leukocyte L1 Antigen Complex/metabolism , Male , Middle Aged , Models, Biological , Mucoproteins/blood , Serum Albumin/metabolism , Young Adult
4.
Cancer Lett ; 449: 125-134, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30685412

ABSTRACT

Increased drug resistance and acute side effects on normal organs are the major disadvantages of traditional cancer chemotherapy and radiotherapy. This has increased the focus on targeted therapeutic strategies such as monoclonal antibody-based cancer therapies. The major advantage of antibody-based therapies is the specific inhibition of cancer-related targets, with reduced off-target side effects. Anterior gradient-2 (AGR2) is a prometastatic and proangiogenic tumor marker that is overexpressed in multiple cancers. Therefore, anti-AGR2 antibodies may be potential therapeutic agents for treating different cancers. In the present study, we examined a novel anti-AGR2 monoclonal antibody mAb18A4 and found that this antibody inhibited lung cancer progression and metastasis without exerting any adverse side effects on the major organs and blood in mice. Moreover, we found that mAb18A4 activated p53 pathway and attenuated ERK1/2-MAPK pathway. Furthermore, mAb18A4-treated cancer cell lines showed attenuated proliferation and colony formation, enhanced apoptosis, increased p53 expression, and reduced phosphorylated ERK1/2 expression. Treatment with mAb18A4 significantly reduced tumor size and suppressed tumor metastasis in and increased the survival of different xenograft tumor models. In addition, mAb18A4 potently suppressed AGR2-induced angiogenesis. Results of pharmacokinetic and toxicological analyses confirmed the safety of mAb18A4 as an antitumor treatment.


Subject(s)
Antineoplastic Agents, Immunological/administration & dosage , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mucoproteins/antagonists & inhibitors , Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , A549 Cells , Animals , Antineoplastic Agents, Immunological/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/metabolism , Mice , Signal Transduction/drug effects , Up-Regulation , Xenograft Model Antitumor Assays
5.
J Crohns Colitis ; 12(suppl_2): S633-S640, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-30137311

ABSTRACT

Studies in the 1990s using animal models of intestinal inflammation delineated the crucial molecules involved in leukocyte attraction and retention to the inflamed gut and associated lymphoid tissues. The first drug targeting leukocyte trafficking tested in inflammatory bowel diseases was the anti-ICAM-1 antisense oligonucleotide alicaforsen, showing only modest efficacy. Subsequently, the anti-α4 monoclonal antibody natalizumab proved efficacious for induction and maintenance of remission in Crohn's disease, but was associated with progressive multifocal leukoencephalopathy due to its ability to interfere with both α4ß1 and α4ß7 function. Later developments in this area took advantage of the fairly selective expression of MAdCAM-1 in the digestive organs, showing that vedolizumab, a more specific monoclonal antibody selectively blocking MAdCAM-1 binding to integrin α4ß7, was efficacious for induction and maintenance of remission in ulcerative colitis and Crohn's disease, and it was not associated with neurological complications. Currently, other drugs targeting the ß7 subunit, immunoglobulin superfamily molecules expressed on the endothelium, as well as blockade of lymphocyte recirculation in lymph nodes through modulation of sphingosine 1-phosphate receptors are under development. The potential use and risks of combined anti-trafficking therapy will be examined in this review.


Subject(s)
Chemotaxis, Leukocyte/drug effects , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Integrins/antagonists & inhibitors , Leukocytes/physiology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Adhesion Molecules , Cell Communication/drug effects , Endothelial Cells/physiology , Fingolimod Hydrochloride/therapeutic use , Gastrointestinal Agents/pharmacology , Gastrointestinal Tract/immunology , Humans , Immunoglobulins , Immunosuppressive Agents/therapeutic use , Indans/therapeutic use , Intercellular Adhesion Molecule-1 , Lysophospholipids/agonists , Mucoproteins/antagonists & inhibitors , Natalizumab/therapeutic use , Oxadiazoles/therapeutic use , Phosphorothioate Oligonucleotides/therapeutic use , Receptors, Lysosphingolipid/agonists , Sphingosine/agonists , Sphingosine/analogs & derivatives
6.
J Crohns Colitis ; 12(suppl_2): S653-S668, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-29767705

ABSTRACT

Integrins are cell surface receptors with bidirectional signalling capabilities that can bind to adhesion molecules in order to mediate homing of leukocytes to peripheral tissues. Gut-selective leukocyte homing is facilitated by interactions between α4ß7 and its ligand, mucosal addressin cellular adhesion molecule-1 [MAdCAM-1], while retention of lymphocytes in mucosal tissues is mediated by αEß7 binding to its ligand E-cadherin. Therapies targeting gut-selective trafficking have shown efficacy in inflammatory bowel disease [IBD], confirming the importance of leukocyte trafficking in disease pathobiology. This review will provide an overview of integrin structure, function and signalling, and highlight the role that these molecules play in leukocyte homing and retention. Anti-integrin therapeutics, including gut-selective antibodies against the ß7 integrin subunit [etrolizumab] and the α4ß7 integrin heterodimer [vedolizumab and abrilumab], and the non-gut selective anti-α4 integrin [natalizumab], will be discussed, as well as novel targeting approaches using small molecules.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Integrins/metabolism , Lymphocytes/metabolism , Natalizumab/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Cell Adhesion Molecules , Cell Movement/drug effects , Gastrointestinal Tract/metabolism , Humans , Immunoglobulins/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Integrins/antagonists & inhibitors , Lymphocytes/immunology , Molecular Targeted Therapy , Mucoproteins/antagonists & inhibitors , Mucoproteins/metabolism
7.
J Crohns Colitis ; 12(suppl_2): S669-S677, 2018 Aug 22.
Article in English | MEDLINE | ID: mdl-29757363

ABSTRACT

Specific blockade of the endothelial ligands intercellular adhesion molecule-1 [ICAM-1] and mucosal addressin cell adhesion molecule [MAdCAM] involved in leukocyte recruitment to the site of inflammation as therapeutic targets in inflammatory bowel disease [IBD] has been recognized from their overexpression in the inflamed mucosa and successful intervention based on these ligands in preclinical animal models. Interventions to target ICAM-1 in human IBD are confined to the ICAM-1 anti-sense oligonucleotide alicaforsen. While results with parenteral formulations of alicaforsen in Crohn's disease have largely been negative, efficacy signals derived from studies with an enema formulation in ulcerative colitis and pouchitis are promising and have led to a Food and Drug Administration Fast-Track designation for the latter. A large phase III programme in pouchitis is underway. Phase II studies with the anti-MAdCAM-1 antibody [SHP647] delivered positive results in ulcerative colitis and anti-inflammatory signals in Crohn's disease. Furthermore, it was shown that SHP647 does not affect the number and composition of cells in cerebrospinal fluid, suggesting that the compound is not affecting immune surveillance in the central nervous system. In addition, both alicaforsen and SHP647 are promising compounds based on the clear safety profile observed so far.


Subject(s)
Gastrointestinal Agents/therapeutic use , Immunoglobulins/metabolism , Inflammatory Bowel Diseases/drug therapy , Intercellular Adhesion Molecule-1/metabolism , Mucoproteins/metabolism , Phosphorothioate Oligonucleotides/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Adhesion Molecules , Cell Movement , Humans , Immunoglobulins/immunology , Intercellular Adhesion Molecule-1/immunology , Leukocytes/immunology , Molecular Targeted Therapy , Mucoproteins/antagonists & inhibitors , Mucoproteins/immunology , Pouchitis/drug therapy
8.
J Crohns Colitis ; 12(1): 105-119, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-28961959

ABSTRACT

The therapeutic landscape for inflammatory bowel disease [IBD] is rapidly evolving. Two new biologic drugs, vedolizumab and ustekinumab, have recently entered the marketplace, the first biosimilars have been introduced, and several other agents are at an advanced stage of clinical development. In parallel, therapeutic goals have shifted from symptom control towards mucosal healing and prevention of bowel damage. In the coming years, gastroenterologists will be faced with unprecedented choices when selecting the best treatment for their patients with IBD. In this article, we review existing data on the mechanisms of action, efficacy, and safety of recently approved and late-stage pipeline therapies, and use this information to speculate on the positioning of these drugs, alone or in combination, in therapeutic algorithms for Crohn's disease and ulcerative colitis.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Algorithms , Antibodies, Monoclonal, Humanized/adverse effects , Cell Adhesion Molecules , Drug Therapy, Combination , Humans , Immunoglobulins , Immunosuppressive Agents/therapeutic use , Integrins/antagonists & inhibitors , Interleukin-12/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Janus Kinases/antagonists & inhibitors , Mucoproteins/antagonists & inhibitors , Protein Kinase Inhibitors/adverse effects , Smad7 Protein/antagonists & inhibitors , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Vascular Cell Adhesion Molecule-1/antagonists & inhibitors
9.
Curr Gastroenterol Rep ; 18(9): 51, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27461274

ABSTRACT

Tumor necrosis factor (TNF) antagonists are the cornerstone of therapy for moderately to severely active inflammatory bowel disease (IBD). Although our understanding of pharmacokinetics, pharmacodynamics, and treatment optimization for these agents has evolved considerably over the past decade, a substantial majority of individuals fail to respond or lose response to TNF-antagonists over time. A need therefore remains for efficacious treatment options in these patients. Alternative immunological targets have now been identified, and several novel therapeutic agents are in development for IBD. In this review article, we discuss these novel therapeutic agents, with a particular focus on those demonstrated to be efficacious in phase 2 and 3 clinical trials. We further discuss considerations to be made when integrating these agents into routine practice over the next decade.


Subject(s)
Biological Products/therapeutic use , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Cell Adhesion Molecules , Humans , Immunoglobulins , Indans/therapeutic use , Mucoproteins/antagonists & inhibitors , Oligonucleotides/therapeutic use , Oxadiazoles/therapeutic use , Piperidines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Ustekinumab/therapeutic use
10.
Inflamm Bowel Dis ; 22(7): 1737-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27306074

ABSTRACT

Multiple new biological treatments for Crohn's disease and ulcerative colitis are becoming available. Specifically, vedolizumab and ustekinumab are monoclonal antibodies that target molecular pathways relevant to disease pathogenesis. What can Cochrane reviews tell us about the efficacy, safety, and immunogenicity of these new agents? A Cochrane inflammatory bowel disease group symposium held at the 2015 Digestive Diseases Week annual meeting addressed these questions. This article reviews the data presented at that session.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Colitis, Ulcerative/drug therapy , Crohn Disease/drug therapy , Gastrointestinal Agents/therapeutic use , Ustekinumab/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Biological Products/immunology , Biological Products/therapeutic use , Cell Adhesion Molecules , Congresses as Topic , Evidence-Based Medicine , Gastrointestinal Agents/adverse effects , Humans , Immunoglobulins , Lymphocytes/drug effects , Lymphocytes/physiology , Maintenance Chemotherapy , Mucoproteins/antagonists & inhibitors , Remission Induction , Review Literature as Topic , Ustekinumab/adverse effects
11.
Gastroenterol Clin North Am ; 43(3): 581-601, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25110260

ABSTRACT

Lymphocyte homing antagonists represent promising therapeutic agents for the treatment of idiopathic inflammatory bowel disease (IBD). Several critical molecules involved in the recruitment of inflammatory cells in the intestine, including integrins and chemokine receptors, have been successfully targeted for the treatment of IBD. These agents have shown great promise for the induction and maintenance of remission for both Crohn disease and ulcerative colitis. This article discusses currently approved prototypic agents for the treatment of IBD (natalizumab, anti-α4 integrin; vedolizumab, anti-α4ß7 integrin), and several other agents in the same class currently under development.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Integrins/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Cell Adhesion Molecules , Humans , Immunoglobulins , Intercellular Adhesion Molecule-1 , Mucoproteins/antagonists & inhibitors , Natalizumab , Phenylalanine/analogs & derivatives , Phenylalanine/therapeutic use , Phosphorothioate Oligonucleotides/therapeutic use , Quinazolinones/therapeutic use , Receptors, CCR/antagonists & inhibitors , Sulfonamides/therapeutic use
12.
Neurosci Lett ; 576: 40-4, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24905173

ABSTRACT

Calcium-activated chloride channels (CaCCs) are thought to regulate neuronal excitability, and recently chloride (Cl(-)) regulation in DRG neurons has attracted much attention in pain research. Furthermore, the activity of CaCCs is modified by a family of CLCA proteins. In acute antigen-induced arthritis (AIA), a remarkable up-regulation of the murine chloride channel accessory 3 (mClca3) was shown in dorsal root ganglion (DRG) neurons. Therefore we tested the hypothesis that mClca3 is involved in arthritic pain perception. In mClca3 knock-out mice and wild-type control mice, AIA was induced and measures of inflammation and pain were assessed. In the very acute phase of AIA, joint swelling was reduced in mClca3 knock-out mice. This effect disappeared during the course of AIA. We could not show significant differences in mechanical hyperalgesia between both groups of mice, neither at the acute nor at the chronic stage (21 days of AIA). Additional experiments on thermal hyperalgesia in wild-type and mClca3 knock-out mice in the first 3 days of AIA did not show a difference either. In addition, niflumic acid, an antagonist at CaCCs, did not significantly influence hyperalgesia during AIA. Thus, we were not able to provide evidence for a role of CaCCs, and in particular of mClca3, on the expression of arthritis or inflammation-evoked hyperalgesia.


Subject(s)
Arthritis/metabolism , Chloride Channels/metabolism , Mucoproteins/metabolism , Pain/metabolism , Animals , Arthritis/physiopathology , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Freund's Adjuvant , Ganglia, Spinal/metabolism , Hot Temperature , Hyperalgesia/physiopathology , Hyperalgesia/psychology , Mice, Knockout , Mucoproteins/antagonists & inhibitors , Mucoproteins/genetics , Neurons/metabolism , Niflumic Acid/pharmacology , Pain/physiopathology , Pain/psychology , Pain Perception
13.
BMC Plant Biol ; 12: 126, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22853005

ABSTRACT

BACKGROUND: In dicotyledonous plant, the first asymmetric zygotic division and subsequent several cell divisions are crucial for proembryo pattern formation and later embryo development. Arabinogalactan proteins (AGPs) are a family of extensively glycosylated cell surface proteins that are thought to have important roles in various aspects of plant growth and development, including embryogenesis. Previous results from our laboratory show that AGPs are concerned with tobacco egg cell fertilization and zygotic division. However, how AGPs interact with other factors involved in zygotic division and proembryo development remains unknown. RESULTS: In this study, we used the tobacco in vitro zygote culture system and series of meticulous cell biology techniques to investigate the roles of AGPs in zygote and proembryo cell division. For the first time, we examined tobacco proembryo division patterns detailed to every cell division. The bright-field images and statistical results both revealed that with the addition of an exogenous AGPs inhibitor, beta-glucosyl Yariv (beta-GlcY) reagent, the frequency of aberrant division increased remarkably in cultured tobacco zygotes and proembryos, and the cell plate specific locations of AGPs were greatly reduced after beta-GlcY treatment. In addition, the accumulations of new cell wall materials were also significantly affected by treating with beta-GlcY. Detection of cellulose components by Calcofluor white stain showed that strong fluorescence was located in the newly formed wall of daughter cells after the zygotic division of in vivo samples and the control samples from in vitro culture without beta-GlcY treatment; while there was only weak fluorescence in the newly formed cell walls with beta-GlcY treatment. Immunocytochemistry examination with JIM5 and JIM7 respectively against the low- and high-esterified pectins displayed that these two pectins located in opposite positions of zygotes and proembryos in vivo and the polarity was not affected by beta-GlcY. Furthermore, FM4-64 staining revealed that endosomes were distributed in the cell plates of proembryos, and the localization pattern was also affected by beta-GlcY treatment. These results were further confirmed by subsequent observation with transmission electron microscopy. Moreover, the changes to proembryo cell-organelles induced by beta-GlcY reagent were also observed using fluorescent dye staining technique. CONCLUSIONS: These results imply that AGPs may not only relate to cell plate position decision, but also to the location of new cell wall components. Correlated with other factors, AGPs further influence the zygotic division and proembryo pattern establishment in tobacco.


Subject(s)
Cell Division , Glucosides/pharmacology , Mucoproteins/chemistry , Nicotiana/cytology , Phloroglucinol/analogs & derivatives , Seeds/ultrastructure , Benzenesulfonates/chemistry , Cell Cycle Proteins/chemistry , Cell Polarity , Cell Wall/chemistry , Cell Wall/drug effects , Cellulose/chemistry , Endosomes/chemistry , Fluorescence , Fluorescent Antibody Technique , Immunohistochemistry , Microscopy, Confocal , Microscopy, Electron, Transmission , Mucoproteins/antagonists & inhibitors , Pectins/chemistry , Phloroglucinol/pharmacology , Plant Cells/chemistry , Plant Proteins/antagonists & inhibitors , Plant Proteins/chemistry , Pyridinium Compounds/chemistry , Quaternary Ammonium Compounds/chemistry , Seeds/chemistry , Seeds/drug effects , Seeds/embryology , Tissue Culture Techniques , Nicotiana/chemistry , Nicotiana/drug effects , Nicotiana/embryology
14.
Biochem Biophys Res Commun ; 404(1): 261-7, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21111712

ABSTRACT

Th2 cytokines and their downstream Janus kinase (JAK)-signal transducer and activation of transcription (STAT) pathways play a critical role in allergic asthma. We studied the effects of a pan-JAK inhibitor, pyridone 6 (P6), on asthmatic responses in a mouse model and investigated the mechanism for its biological effects. Mice were sensitized and challenged by ovalbumin (OVA). P6 treatment during the challenge phase suppressed eosinophilia in bronchoalveolar lavage (BAL) fluids but did not affect airway hyperresponsiveness (AHR). To improve the efficacy of the JAK inhibitor, P6 was encapsulated in polylactic-coglycolic acid nanoparticles (P6-PLGA). P6-PLGA treatment just before OVA challenge suppressed both airway eosinophilia and AHR. Although the IL-13 levels in BAL fluids and the OVA-specific IgE levels in serum after the challenge phase treatment with P6-PLGA were similar to those after a sham treatment, the eotaxin levels in BAL fluids and lung mCLCA3/Gob-5 expression were decreased in P6-PLGA-treated mice. Interestingly, the local IL-13 levels and serum OVA-specific IgE were decreased, while IL-17-producing T cells were increased by P6-PLGA treatment during the sensitization plus challenge phases. In vitro, P6 strongly suppressed the differentiation of Th2 from naive CD4 T cells, but it partly enhanced Th17 differentiation. P6 potently suppressed IL-13-mediated STAT6 activation and mCLCA3/Gob-5 expression in mouse tracheal epithelial cells. These findings suggest that the JAK inhibitor P6 suppresses asthmatic responses by inhibiting Th2 inflammation and that application of PLGA nanoparticles improves the therapeutic potency of P6.


Subject(s)
Asthma/drug therapy , Benzimidazoles/therapeutic use , Bronchial Hyperreactivity/drug therapy , Janus Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyridones/therapeutic use , Animals , Asthma/immunology , Asthma/physiopathology , Benzimidazoles/administration & dosage , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Capsules , Chloride Channels/antagonists & inhibitors , Chloride Channels/biosynthesis , Eosinophilia/drug therapy , Eosinophilia/immunology , Interleukin-13/immunology , Lactic Acid/chemistry , Lung/immunology , Mice , Mucoproteins/antagonists & inhibitors , Mucoproteins/biosynthesis , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Ovalbumin/immunology , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Pyridones/administration & dosage , STAT6 Transcription Factor/metabolism , Th2 Cells/immunology
15.
Bioorg Med Chem Lett ; 18(7): 2509-12, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18331794

ABSTRACT

A novel series of barbituric acid derivatives were identified as selective inhibitors of alpha4beta7 MAdCAM (mucosal addressin cell adhesion molecule-1) interactions via a high throughput screening exercise. These inhibitors were optimized to submicromolar potencies in whole cell adhesion assays, retaining their selectivity over alpha4beta1 VCAM.


Subject(s)
Barbiturates/pharmacology , Cell Adhesion/drug effects , Integrins/antagonists & inhibitors , Mucoproteins/antagonists & inhibitors , Animals , Barbiturates/chemical synthesis , Binding Sites , Cell Adhesion/physiology , Cell Adhesion Molecules , Immunoglobulins/metabolism , Integrin alpha4beta1/metabolism , Integrins/metabolism , Mice , Models, Chemical , Mucoproteins/metabolism , Structure-Activity Relationship , Vascular Cell Adhesion Molecule-1/metabolism
16.
Cancer Res ; 68(2): 492-7, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18199544

ABSTRACT

The AGR2 gene encodes a secretory protein that is highly expressed in adenocarcinomas of the esophagus, pancreas, breast, and prostate. This study explores the effect of AGR2 expression with well-established in vitro and in vivo assays that screen for cellular transformation and tumor growth. AGR2 expression in SEG-1 esophageal adenocarcinoma cells was reduced with RNA interference. Cellular transformation was examined using NIH3T3 cells that express AGR2 after stable transfection. The cell lines were studied in vitro with assays for density-dependent and anchorage-independent growth, and in vivo as tumor xenografts in nude mice. SEG-1 cells with reduced AGR2 expression showed an 82% decrease in anchorage-independent colony growth and a 60% reduction in tumor xenograft size. In vitro assays of AGR2-expressing NIH3T3 cells displayed enhanced foci formation and anchorage-independent growth. In vivo, AGR2-expressing NIH3T3 cells established tumors in nude mice. Thus, AGR2 expression promotes tumor growth in esophageal adenocarcinoma cells and is able to transform NIH3T3 cells. Immunohistochemistry of the normal mouse intestine detected AGR2 expression in proliferating and differentiated intestinal cells of secretory lineage. AGR2 may be important for the growth and development of the intestine as well as esophageal adenocarcinomas.


Subject(s)
Adenocarcinoma/pathology , Antigens, Neoplasm/physiology , Cell Movement/genetics , Cell Transformation, Neoplastic/genetics , Esophageal Neoplasms/pathology , Mucoproteins/physiology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Proliferation , DNA-Binding Proteins/genetics , Disease Progression , Esophageal Neoplasms/genetics , Gene Expression Regulation , Intestinal Mucosa/metabolism , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Mice , Mucoproteins/antagonists & inhibitors , Mucoproteins/genetics , Mucoproteins/metabolism , NIH 3T3 Cells , Neoplasm Transplantation , Oncogene Proteins , RNA Interference , Rats , Transcription Factors/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
17.
Transpl Immunol ; 17(4): 271-7, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17493530

ABSTRACT

The effect of blocking the expression of the mucosal addressin cell adhesion molecule-1 (MAdCAM-1) in a graft by an antibody, and immunohistochemical changes in the graft were monitored, using a rat small intestinal transplantation model. Dark Agouti (DA) rat small intestines were heterotopically transplanted into Lewis (LEW) rats. The graft was treated with or without an anti-MAdCAM-1 antibody, F(ab')(2), during the operation. The survival of the grafts and histological changes, such as lymphocyte infiltration and destruction of the intestinal architecture in epithelium villus thickness, villus height and submucosal thickness of the graft, were examined. The expression of MAdCAM-1 and beta 7 integrin in the graft was also checked by immunostaining. Furthermore, graft infiltrating lymphocytes, in mesenteric lymph nodes (MLN) and Peyer's patches (PP) were measured by FACS analysis. Survival was prolonged in the DA graft with anti-MAdCAM-1 F(ab')(2) treatment; DA to LEW: 7.0+/-3.3, DA to LEW with the antibody: 24.6+/-8.4 days (p<0.05). Histological findings and scoring of the grafts were consistent with this conclusion. Moreover, MAdCAM-1 expression itself was suppressed in grafts of the antibody-treated group. While a FACS analysis showed no difference in the % of CD4+ T cells and CD8+ T cells in the PP of the graft, CD4+ T cells in the MLN of the antibody-treated graft were significantly low. A strategy directed at blocking the adhesion molecule, MAdCAM-1, in the small intestinal grafts could be useful in the prevention of acute rejection.


Subject(s)
Antibodies, Blocking/pharmacology , Graft Rejection/prevention & control , Intestine, Small/transplantation , Mucoproteins/antagonists & inhibitors , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Graft Survival , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulins/analysis , Immunoglobulins/immunology , Integrin beta Chains/analysis , Intestine, Small/cytology , Intestine, Small/immunology , Male , Models, Animal , Mucoproteins/analysis , Mucoproteins/immunology , Rats , Rats, Inbred Strains
18.
JPEN J Parenter Enteral Nutr ; 29(1): 44-7, 2005.
Article in English | MEDLINE | ID: mdl-15715273

ABSTRACT

BACKGROUND: Parenteral nutrition (PN) decreases gut-associated lymphoid tissue (GALT), the intestinal IgA stimulating cytokines IL-4 and IL-10 in gut homogenates, intestinal IgA levels and the expression of Peyer patch (PP) mucosal cellular adhesion molecule-1 (MAdCAM-1), an adhesion molecule found on the high endothelial venules of PP and other tissues. IL-4 in PP stimulates MAdCAM expression in vitro. MAdCAM-1 blockade with MECA-367 reduces GALT cell populations to PN levels but maintains intestinal IgA levels if the animals are chow fed. This study compares IL-4 levels in PP of chow and PN fed mice and measures the effects of MAdCAM blockade on IL-4 and IL-10 levels in gut homogenates of chow fed mice. We hypothesized that in vivo IL-4 levels drop in PP of PN fed mice and IL-4 and IL-10 levels are maintained after MAdCAM-1 blockade in chow fed mice. METHODS: Exp 1: 18 mice received chow or PN for 5 days to determine PP IL-4 levels. Exp 2: 44 mice were randomized to chow + control monoclonal antibody (mAb), chow + MECA-367 (anti-MAdCAM-1 mAb) or PN for 4 days before measurement of IL-4 and IL-10 levels in gut homogenates. RESULTS: Exp 1: IL-4 levels in vivo were lower in PP of PN-fed mice than chow fed mice (92.0 +/- 15.1 pg/mL vs 251.1 +/- 14.8, p = .0003). Exp 2: IL-4 levels were significantly higher in chow + control mAb (187.1 +/- 44.1 pg/mL) and chow + MECA-367 (110.9 +/- 19.1 pg/mL) groups than PN mice (21.8 +/- 30.6 pg/mL, p < .02 vs chow + control or chow + MECA-367). IL-10 levels were significantly lower with PN (23.1 +/- 40.9 pg/mL) with chow+control (174.0 +/- 22.2 pg/mL p < .01), or chow + MECA-367 (181.7 +/- 23.1 pg/mL, p < .02 vs PN). CONCLUSIONS: PN-feeding reduces in vivo IL-4 levels in PP (consistent with lowered MAdCAM-1 expression) and IL-4 and IL-10 levels in gut homogenates compared with chow. Despite MAdCAM-1 blockade, enteral feeding preserved gut IL-4 levels and increased IL-10 levels consistent with preserved IgA levels.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Enteral Nutrition , Immunity, Mucosal/immunology , Immunoglobulins/immunology , Interleukin-10/biosynthesis , Interleukin-4/biosynthesis , Mucoproteins/immunology , Peyer's Patches/metabolism , Animals , Cell Adhesion Molecules , Immunoglobulin A/metabolism , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred ICR , Mucoproteins/antagonists & inhibitors , Parenteral Nutrition/adverse effects , Random Allocation
19.
J Immunol ; 174(4): 2343-52, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699171

ABSTRACT

CD4+ T cells are essential for development and perpetuation of Crohn's disease, a chronic immune-mediated condition that affects primarily the small intestine. Using novel models of Crohn's disease-like ileitis (i.e., SAMP1/YitFc and CD4+ T cell transfer models), we have begun to understand the adhesive pathways that mediate lymphocyte trafficking to the chronically inflamed small bowel. Expansion of the CD4/beta7+ population and increased mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expression were observed within the intestinal lamina propria with disease progression. However, Ab blockade of the beta7 integrin, the alpha4beta7 heterodimer, MAdCAM-1, or L-selectin did not attenuate inflammation. Blockade of two pathways (L-selectin and MAdCAM-1 or alpha4 integrins) was required to improve ileitis. Further analyses showed that 55 +/- 7% of the mesenteric lymph node alpha4beta7+CD4 expressed L-selectin. These L-selectin+ T cells were the main producers of TNF-alpha and the predominant ileitis-inducing subpopulation. Mechanistically, combined blockade of L-selectin and MAdCAM-1 depleted the intestinal lamina propria of CD4+ T cells that aberrantly coexpressed alpha4beta7 and alpha4beta1 integrins, markedly decreasing local production of TNF-alpha and IFN-gamma. Thus, pathogenic CD4+ T cells not only use the physiologic alpha4beta7/MAdCAM-1 pathway, but alternatively engage alpha4beta1 and L-selectin to recirculate to the chronically inflamed small intestine.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Ileitis/immunology , Ileitis/pathology , Integrin alpha4beta1/physiology , Integrins/physiology , L-Selectin/physiology , Animals , Antibodies, Blocking/administration & dosage , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion Molecules , Chronic Disease , Disease Progression , Female , Ileitis/genetics , Immunoglobulins/biosynthesis , Integrin alpha4beta1/biosynthesis , Integrins/antagonists & inhibitors , Integrins/biosynthesis , L-Selectin/biosynthesis , L-Selectin/immunology , Male , Mice , Mice, Inbred AKR , Mice, Mutant Strains , Mice, SCID , Mucoproteins/antagonists & inhibitors , Mucoproteins/biosynthesis , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/genetics , Up-Regulation/immunology
20.
Brain ; 127(Pt 6): 1313-31, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15130951

ABSTRACT

In multiple sclerosis, the immune system attacks the white matter of the brain and spinal cord, leading to disability and/or paralysis. Myelin, oligodendrocytes and neurons are lost due to the release by immune cells of cytotoxic cytokines, autoantibodies and toxic amounts of the excitatory neurotransmitter glutamate. Experimental autoimmune encephalomyelitis (EAE) is an animal model that exhibits the clinical and pathological features of multiple sclerosis. Current therapies that suppress either the inflammation or glutamate excitotoxicity are partially effective when administered at an early stage of EAE, but cannot block advanced disease. In a multi-faceted approach to combat EAE, we blocked inflammation with an anti-MAdCAM-1 (mucosal addressin cell adhesion molecule-1) monoclonal antibody and simultaneously protected oligodendrocytes and neurons against glutamate-mediated damage with the alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA)/kainate antagonist 2,3-dihydroxy-6-nitro-7- sulfamoylbenzo(f)quinoxaline (NBQX) and the neuroprotector glycine-proline-glutamic acid (GPE; N-terminal tripeptide of insulin-like growth factor). Remarkably, administration at an advanced stage of unremitting EAE of either a combination of NBQX and GPE, or preferably all three latter reagents, resulted in amelioration of disease and repair of the CNS, as assessed by increased oligodendrocyte survival and remyelination, and corresponding decreased paralysis, inflammation, CNS apoptosis and axonal damage. Each treatment reduced the expression of nitric oxide and a large panel of proinflammatory and immunoregulatory cytokines, in particular IL-6 which plays a critical role in mediating EAE. Mice displayed discernible improvements in all physical features examined. Disease was suppressed for 5 weeks, but relapsed when treatment was suspended, suggesting treatment must be maintained to be effective. The above approaches, which allow CNS repair by inhibiting inflammation and/or simultaneously protect neurons and oligodendrocytes from damage, could thus be effective therapies for multiple sclerosis.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Mucoproteins/antagonists & inhibitors , Neuroprotective Agents/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Apoptosis/drug effects , Axons/pathology , Cell Adhesion Molecules , Cytokines/metabolism , Disease Progression , Drug Therapy, Combination , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Excitatory Amino Acid Antagonists/therapeutic use , Immunoglobulins/immunology , Integrins/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mucoproteins/immunology , Multiple Sclerosis/drug therapy , Oligodendroglia/pathology , Quinoxalines , Receptors, Glutamate/metabolism , Treatment Outcome , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL