ABSTRACT
Inflammation is the body's biological reaction to endogenous and exogenous stimuli. Recent studies have demonstrated several anti-inflammatory properties of Ferula species. In this paper, we decided to study the anti-inflammatory effect of ethanolic extract of Ferula assafoetida oleo-gum-resin (asafoetida) against TNF-α-stimulated human umbilical vein endothelial cells (HUVECs). HUVECs were cultured in a flat-bottom plate and then treated with ethanolic extract of asafoetida (EEA, 0-500 µg/ml) and TNF-α (0-100 ng/ml) for 24 h. We used the MTT test to assess cell survival. In addition, the LC-MS analysis was performed to determine the active substances. HUVECs were pretreated with EEA and then induced by TNF-α. Intracellular reactive oxygen species (ROS) and adhesion of peripheral blood mononuclear cells (PBMCs) to HUVECs were evaluated with DCFH-DA and CFSE fluorescent probes, respectively. Gene expression of intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and E-selectin and surface expression of ICAM-1 protein were measured using real-time PCR and flow cytometry methods, respectively. While TNF-α significantly increased intracellular ROS formation and PBMC adhesion to TNF-α-induced HUVECs, the pretreatment of HUVECs with EEA (125 and 250 µg/ml) significantly reduced the parameters. In addition, EEA pretreatment decreased TNF-α-induced mRNA expression of VCAM-1 and surface protein expression of ICAM-1 in the target cells. Taken together, the results indicated that EEA prevented ROS generation, triggered by TNF-α, and inhibited the expression of VCAM-1 and ICAM-1, leading to reduced PBMC adhesion. These findings suggest that EEA can probably have anti-inflammatory properties.
Subject(s)
Anti-Inflammatory Agents , Cell Adhesion Molecules , Ferula , Human Umbilical Vein Endothelial Cells , Plant Extracts , Anti-Inflammatory Agents/pharmacology , Cell Adhesion , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , E-Selectin/biosynthesis , E-Selectin/genetics , E-Selectin/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Leukocytes, Mononuclear/immunology , Plant Extracts/pharmacology , Reactive Oxygen Species/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunologyABSTRACT
Aryl-hydrocarbon receptor (Ahr) plays an important role in the regulation of intestinal homeostasis. Diabetes is characterized by vascular complications and intestinal dysfunction. We aimed at understanding the relationship between intestinal defense impairment and inflammation in diabetes and effects of Ahr ligands on diabetes-induced insulin resistance, endovascular inflammation, and intercellular adhesion molecule (ICAM) and flavin mono-oxygenase (FMO3) expression. Effects of Ahr ligands, such as tryptophan (Trp) and indole-3-carbinol (I3C) on intestinal barrier and inflammation of Ins2Akita mice were examined. Myeloid differentiation primary response 88 (MYD88) is the adaptor for inflammatory signaling pathways. Ins2Akita-MyD88-/- mice were used to study the role of MyD88. Ins2Akita mice demonstrated decreased Ahr and regenerating islet-derived 3-ß (Reg3ß) expression, and increased Klebsiella pneumoniae translocation. Ins2Akita mice demonstrated increased inducible nitric oxide synthase (iNOS) expression of intestine; ICAM, iNOS, interleukin 1 beta (IL-1ß), and FMO3 expression of liver; and ICAM, iNOS, and FMO3 expression in aorta. Trp and I3C decreased diabetes-induced translocation and increased Ahr and Reg3ß expression of intestine. Ahr ligands reduced diabetes-induced ICAM and FMO3 expression in liver and aorta; IL-6, tumor necrosis factor alpha (TNF-α), and iNOS expression in Kupffer cells; plasma IL-6 and TNF-α levels; dipeptidyl peptidase (DPP4) activity; and insulin insensitivity. Ins2Akita-MyD88-/- mice demonstrated decreased expression of p-NF-κB of liver and ICAM of aorta compared with Ins2Akita mice. Altogether, our data suggest that diabetes induces ICAM and FMO3 expression through the decrease in intestinal defense and MyD88. Ahr ligands reverse diabetes-induced intestinal defense impairment, insulin insensitivity, FMO3/ICAM expression, and systemic inflammation.
Subject(s)
Diabetes Mellitus/drug therapy , Indoles/administration & dosage , Insulin Resistance , Tryptophan/administration & dosage , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Diabetes Mellitus/genetics , Diabetes Mellitus/immunology , Dietary Supplements/analysis , Disease Models, Animal , Humans , Kupffer Cells/drug effects , Kupffer Cells/immunology , Ligands , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Oxygenases/genetics , Oxygenases/immunology , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/immunologyABSTRACT
INTRODUCTION: Mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expression in gut-associated lymphoid tissue is upregulated in patients with inflammatory bowel disease (IBD). Blocking adhesion molecules and thereby inhibiting migration of lymphocytes into sites of inflammation in the gut is an attractive new treatment target in drug development for IBD. AREAS COVERED: This review discusses the preclinical and clinical experience on SHP647 (previously called PF-00547659 and PF-00547,659), a fully human IgG2K monoclonal antibody that binds to MAdCAM-1 to selectively reduce lymphocyte homing to the intestinal tract. EXPERT OPINION: Blocking endothelial adhesion molecule MAdCAM-1 could represent an attractive target for the treatment of IBD. In the next years, the results from the phase III studies as well as data to support therapeutic drug monitoring based on drug levels to guide and optimize individual therapy will become available. Furthermore, much effort is put in the development of clinical prediction models to predict which drug is optimal for an individual patient.
Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Cell Adhesion Molecules/immunology , Inflammatory Bowel Diseases/drug therapy , Mucoproteins/immunology , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Clinical Trials as Topic , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Crohn Disease/drug therapy , Crohn Disease/pathology , Drug Evaluation, Preclinical , Humans , Inflammatory Bowel Diseases/pathologyABSTRACT
BACKGROUND: To investigate the potential roles of periostin (POSTN), an extracellular matrix preferentially expressed in Th2-skewed conditions in the pathophysiology of allergic conjunctivitis. METHODS: The roles of POSTN in ragweed-induced experimental allergic conjunctivitis (RW-EAC) were evaluated using both POSTN-knockout (KO) and congenic BALB/c wild-type mice. Histological analysis was carried out to enumerate eosinophils/basophils in the conjunctival tissue. Th2 cytokine expression was evaluated by quantitative polymerase chain reaction (Q-PCR), and microarray analysis was performed to elucidate genes differentially expressed in POSTN-KO and wild-type mice in the RW-EAC model. RESULTS: Upregulation of POSTN expression and eosinophil infiltration was observed in subconjunctival tissue of RW-EAC in the wild-type mice. The number of infiltrating eosinophils in the conjunctivae of RW-EAC was diminished in POSTN-KO mice compared to wild-type mice. Q-PCR analysis of conjunctival tissue showed induction of Th2 cytokine (Ccl5, Il4, Il5, Il13) expression in the RW-EAC and attenuated Ccl5, Il4, Il13 mRNA expression in the conjunctivae of the RW-EAC using POSTN-KO mice. Microarray analysis and immunohistochemical analysis showed diminished basophil marker (Mcpt8) expression and reduced numbers of infiltrating basophils in the conjunctivae of RW-EAC in POSTN-KO mice. CONCLUSIONS: POSTN expression in conjunctival tissue plays an indispensable role in the late-phase reaction of the RW-EAC model by facilitating eosinophil/basophil infiltration and augmenting Th2 cytokine expression.
Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Conjunctivitis, Allergic/immunology , Allergens , Ambrosia , Animals , Antigens, Plant , Basophils/immunology , Conjunctiva/immunology , Cytokines/immunology , Disease Models, Animal , Eosinophils/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , PollenABSTRACT
C4.4A (LYPD3) has been identified as a cancer- and metastasis-associated internalizing cell surface protein that is expressed in non-small cell lung cancer (NSCLC), with particularly high prevalence in the squamous cell carcinoma (SCC) subtype. With the exception of skin keratinocytes and esophageal endothelial cells, C4.4A expression is scarce in normal tissues, presenting an opportunity to selectively treat cancers with a C4.4A-directed antibody-drug conjugate (ADC). We have generated BAY 1129980 (C4.4A-ADC), an ADC consisting of a fully human C4.4A-targeting mAb conjugated to a novel, highly potent derivative of the microtubule-disrupting cytotoxic drug auristatin via a noncleavable alkyl hydrazide linker. In vitro, C4.4A-ADC demonstrated potent antiproliferative efficacy in cell lines endogenously expressing C4.4A and inhibited proliferation of C4.4A-transfected A549 lung cancer cells showing selectivity compared with a nontargeted control ADC. In vivo, C4.4A-ADC was efficacious in human NSCLC cell line (NCI-H292 and NCI-H322) and patient-derived xenograft (PDX) models (Lu7064, Lu7126, Lu7433, and Lu7466). C4.4A expression level correlated with in vivo efficacy, the most responsive being the models with C4.4A expression in over 50% of the cells. In the NCI-H292 NSCLC model, C4.4A-ADC demonstrated equal or superior efficacy compared to cisplatin, paclitaxel, and vinorelbine. Furthermore, an additive antitumor efficacy in combination with cisplatin was observed. Finally, a repeated dosing with C4.4A-ADC was well tolerated without changing the sensitivity to the treatment. Taken together, C4.4A-ADC is a promising therapeutic candidate for the treatment of NSCLC and other cancers expressing C4.4A. A phase I study (NCT02134197) with the C4.4A-ADC BAY 1129980 is currently ongoing. Mol Cancer Ther; 16(5); 893-904. ©2017 AACR.
Subject(s)
Antibodies, Monoclonal/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Adhesion Molecules/immunology , Immunoconjugates/administration & dosage , Aminobenzoates/chemistry , Aminobenzoates/immunology , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Cell Adhesion Molecules/antagonists & inhibitors , Cell Line, Tumor , Cisplatin/administration & dosage , Cisplatin/immunology , GPI-Linked Proteins/antagonists & inhibitors , GPI-Linked Proteins/immunology , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Mice , Oligopeptides/chemistry , Oligopeptides/immunology , Paclitaxel/administration & dosage , Paclitaxel/immunology , Vinblastine/administration & dosage , Vinblastine/analogs & derivatives , Vinblastine/immunology , Vinorelbine , Xenograft Model Antitumor AssaysABSTRACT
INTRODUCTION: As many inflammatory bowel disease (IBD) patients do not benefit from long-term anti-tumour necrosis factor treatment, new anti-inflammatories are urgently needed. After the discovery of the interleukin (IL) 23/17 axis being pivotal in IBD pathogenesis, many different compounds were developed, targeting different components within this pathway. Areas covered: A literature search to March 2016 was performed to identify the most relevant reports on the role of the IL-23/IL-17 axis in IBD and on the different molecules targeting this pathway. First, the authors briefly summarize the immunology of the IL-23/IL-17 pathway to elucidate the mode of action of all different agents. Second, they describe all different molecules targeting this pathway. Besides discussing efficacy and safety data, they also explore immunogenicity, exposure during pregnancy and pharmacokinetics. Expert opinion: A new era in IBD treatment has recently been initiated: besides immunomodulators and TNF-antagonists, anti-adhesion molecules and monoclonal antibodies targeting the IL-23/IL-17 pathway have been developed. Biomarkers for personalized medicine are urgently needed. This therapeutic (r)evolution will further improve disease-related and patient-reported outcome, though a lot of questions should still be addressed in future years.
Subject(s)
Biological Therapy/methods , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/metabolism , Interleukin-17/metabolism , Interleukin-23/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biological Therapy/trends , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Clinical Trials as Topic/methods , Humans , Inflammatory Bowel Diseases/immunology , Interleukin-17/antagonists & inhibitors , Interleukin-17/immunology , Interleukin-23/antagonists & inhibitors , Interleukin-23/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolismABSTRACT
In this issue of Blood, complementary studies by Amin et al and Linley et al demonstrate that sugar moieties linked to surface immunoglobulin (sIg) of follicular lymphoma (FL) cells directly interact with endogenous lectins within the lymphoma niche and lead to activation of downstream B-cell receptor (BCR) signaling pathways. In addition to providing further insight into the role of the microenvironment in lymphomagenesis, these findings expose a unique molecular interaction that may represent a viable target for therapeutic intervention.
Subject(s)
B-Lymphocytes/metabolism , Calcium Signaling , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Gene Expression Regulation/immunology , Immunoglobulin M/immunology , Immunoglobulin Variable Region/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphoma, Follicular/immunology , Lymphoma, Follicular/metabolism , MAP Kinase Signaling System , Macrophages/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signal Transduction/immunology , Female , Humans , MaleABSTRACT
Bispecific antibodies are on the cusp of coming of age as therapeutics more than half a century after they were first described. Two bispecific antibodies, catumaxomab (Removab(®), anti-EpCAM×anti-CD3) and blinatumomab (Blincyto(®), anti-CD19×anti-CD3) are approved for therapy, and >30 additional bispecific antibodies are currently in clinical development. Many of these investigational bispecific antibody drugs are designed to retarget T cells to kill tumor cells, whereas most others are intended to interact with two different disease mediators such as cell surface receptors, soluble ligands and other proteins. The modular architecture of antibodies has been exploited to create more than 60 different bispecific antibody formats. These formats vary in many ways including their molecular weight, number of antigen-binding sites, spatial relationship between different binding sites, valency for each antigen, ability to support secondary immune functions and pharmacokinetic half-life. These diverse formats provide great opportunity to tailor the design of bispecific antibodies to match the proposed mechanisms of action and the intended clinical application.
Subject(s)
Antibodies, Bispecific/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Antigens, CD19/immunology , Antigens, CD19/metabolism , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , CD3 Complex/immunology , CD3 Complex/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Epithelial Cell Adhesion Molecule , Humans , Models, Immunological , Neoplasms/immunology , Neoplasms/metabolismABSTRACT
Photothermal ablation (PTA) is a promising avenue in the area of cancer therapeutics that destroys tumor cells through conversion of near-infrared (NIR) laser light to heat. Hollow gold nanospheres (HGNs) are one of the few materials that are capable of converting light to heat and have been previously used for photothermal ablation studies. Selective delivery of functional nanoparticles to the tumor site is considered as an effective therapeutic approach. In this paper, we demonstrated the anti-cancer potential of HGNs. HGNs were conjugated with monoclonal antibody (anti-TROP2) in order to target cervical cancer cells (HeLa) that contain abundant trophoblast cell surface antigen 2 (TROP2) on the cell surface. The efficient uptake and intracellular location of these functionalized HGNs were studied through application of inductively coupled plasma atomic emission spectroscopy (ICP-AES) and transmission electron microscopy (TEM). Cytotoxicity induced by PTA was measured using CCK-8 assay. HeLa cells incubated with naked HGNs (0.3-3 nmol L(-1)) within 48 h did not show obvious cytotoxicity. Under laser irradiation at suitable power, anti-TROP2 conjugated HGNs achieved significant tumor cell growth inhibition in comparison to the effects of non-specific PEGylated HGNs (P < 0.05). γH2AX assay results revealed higher occurrences of DNA-DSBs with anti-TROP2 conjugated HGNs plus laser radiation as compared to treatment with laser alone. Flow cytometry analysis showed that the amount of cell apoptosis was increased after laser irradiation with anti-TROP2 conjugated HGNs (P < 0.05). Anti-TROP2 conjugated HGNs resulted in down-regulation of Bcl-2 expression and up-regulation of Bax expression. Our study results confirmed that anti-TROP2 conjugated HGNs can selectively destroy cervical cancer cells through inducing its apoptosis and DNA damages. We propose that HGNs have the potentials to mediate targeted cancer treatment.
Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Gold/therapeutic use , Lasers , Nanospheres/therapeutic use , Phototherapy , Uterine Cervical Neoplasms/therapy , Down-Regulation , Female , Hot Temperature , Humans , Nanospheres/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation , bcl-2-Associated X Protein/geneticsABSTRACT
The purpose of this study is to optimize ELISA conditions to quantify the colorectal cancer antigen GA733 linked to the Fc antibody fragment fused to KDEL, an ER retention motif (GA733-FcK) expressed in transgenic plant. Variable conditions of capture antibody, blocking buffer, and detection antibody for ELISA were optimized with application of leaf extracts from transgenic plant expressing GA733-FcK. In detection antibody, anti-EpCAM/CD362 IgG recognizing the GA733 did not detect any GA733-FcK whereas anti-human Fc IgG recognizing the human Fc existed in plant leaf extracts. For blocking buffer conditions, 3% BSA buffer clearly blocked the plate, compared to the 5% skim-milk buffer. For capture antibody, monoclonal antibody (MAb) CO17-1A was applied to coat the plate with different amounts (1, 0.5, and 0.25 µg/well). Among the amounts of the capture antibody, 1 and 0.5 µg/well (capture antibody) showed similar absorbance, whereas 0.25 µg/well of the capture antibody showed significantly less absorbance. Taken together, the optimized conditions to quantify plant-derived GA733-FcK were 0.5 µg/well of MAb CO17-1A per well for the capture antibody, 3% BSA for blocking buffer, and anti-human Fc conjugated HRP. To confirm the optimized ELISA conditions, correlation analysis was conducted between the quantified amount of GA733-FcK in ELISA and its protein density values of different leaf samples in Western blot. The co-efficient value R(2) between the ELISA quantified value and protein density was 0.85 (p<0.01), which indicates that the optimized ELISA conditions feasibly provides quantitative information of GA733-FcK expression in transgenic plant.
Subject(s)
Antigen-Antibody Complex/genetics , Antigens, Neoplasm/genetics , Cell Adhesion Molecules/genetics , Colorectal Neoplasms/immunology , Immunoglobulin Fc Fragments/genetics , Plants, Genetically Modified/genetics , Antibodies, Monoclonal/immunology , Antigen-Antibody Complex/analysis , Antigens, Neoplasm/immunology , Cancer Vaccines , Cell Adhesion Molecules/immunology , Colorectal Neoplasms/genetics , Enzyme-Linked Immunosorbent Assay/methods , Epithelial Cell Adhesion Molecule , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Plant Extracts/immunology , Plant Leaves/metabolism , Receptors, Peptide/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/geneticsABSTRACT
Atherosclerosis is the most common pathological process underlying cardiovascular diseases. Current therapies are largely focused on alleviating hyperlipidaemia and preventing thrombotic complications, but do not completely eliminate risk of suffering recurrent acute ischaemic events. Specifically targeting the inflammatory processes may help to reduce this residual risk of major adverse cardiovascular events in atherosclerotic patients. The involvement of neutrophils in the pathophysiology of atherosclerosis is an emerging field, where evidence for their causal contribution during various stages of atherosclerosis is accumulating. Therefore, the identification of neutrophils as a potential therapeutic target may offer new therapeutic perspective to reduce the current atherosclerotic burden. This narrative review highlights the expanding role of neutrophils in atherogenesis and discusses on the potential treatment targeting neutrophil-related inflammation and associated atherosclerotic plaque vulnerability.
Subject(s)
Antibodies, Monoclonal/therapeutic use , Atherosclerosis/therapy , Inflammation/therapy , Neutrophils/drug effects , Thrombosis/therapy , Animals , Atherosclerosis/immunology , Cell Adhesion Molecules/immunology , Chemokines/immunology , Clinical Trials as Topic , Drug Evaluation, Preclinical , Humans , Inflammation/immunology , Molecular Targeted Therapy , Neutrophil Infiltration/drug effects , Neutrophils/immunology , Receptors, Chemokine/immunology , Thrombosis/immunologyABSTRACT
Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6) is over-expressed in pancreatic cancer cells, and it is associated with the progression of pancreatic cancer. We tested a single domain antibody (sdAb) targeting CEACAM6, 2A3, which was isolated previously from a llama immune library, and an Fc conjugated version of this sdAb, to determine how they affect the pancreatic cancer cell line BxPC3. We also compared the effects of the antibodies to gemcitabine. Gemcitabine and 2A3 slowed down cancer cell proliferation. However, only 2A3 retarded cancer cell invasion, angiogenesis within the cancer mass and BxPC3 cell MMP-9 activity, three features important for tumour growth and metastasis. The IC50s for 2A3, 2A3-Fc and gemcitabine were determined as 6.5µM, 8µM and 12nM, respectively. While the 2A3 antibody inhibited MMP-9 activity by 33% compared to non-treated control cells, gemcitabine failed to inhibit MMP-9 activity. Moreover, 2A3 and 2A3-Fc inhibited invasion of BxPC3 by 73% compared to non-treated cells. When conditioned media that were produced using 2A3- or 2A3-Fc-treated BxPC3 cells were used in a capillary formation assay, the capillary length was reduced by 21% and 49%, respectively. Therefore 2A3 is an ideal candidate for treating tumours that over-express CEACAM6.
Subject(s)
Antigens, CD/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion Molecules/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/pathology , Single-Domain Antibodies/pharmacology , Animals , Camelids, New World , Carcinoma, Pancreatic Ductal/blood supply , Cells, Cultured , Drug Evaluation, Preclinical , GPI-Linked Proteins/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Neoplasm Invasiveness , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Pancreatic Neoplasms/blood supply , Pancreatic NeoplasmsABSTRACT
The blood-brain barrier (BBB) is an integral part of the neurovascular unit (NVU). The NVU is comprised of endothelial cells that are interconnected by tight junctions resting on a parenchymal basement membrane ensheathed by pericytes, smooth muscle cells and a layer of astrocyte end feet. Circulating blood cells, such as leukocytes, complete the NVU. BBB disruption is common in several neurological diseases, but the molecular mechanisms involved remain largely unknown. We analyzed the role of TWIK-related potassium channel-1 (TREK1, encoded by KCNK2) in human and mouse endothelial cells and the BBB. TREK1 was downregulated in endothelial cells by treatment with interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). Blocking TREK1 increased leukocyte transmigration, whereas TREK1 activation had the opposite effect. We identified altered mitogen-activated protein (MAP) kinase signaling, actin remodeling and upregulation of cellular adhesion molecules as potential mechanisms of increased migration in TREK1-deficient (Kcnk2(-/-)) cells. In Kcnk2(-/-) mice, brain endothelial cells showed an upregulation of the cellular adhesion molecules ICAM1, VCAM1 and PECAM1 and facilitated leukocyte trafficking into the CNS. Following the induction of experimental autoimmune encephalomyelitis (EAE) by immunization with a myelin oligodendrocyte protein (MOG)35-55 peptide, Kcnk2(-/-) mice showed higher EAE severity scores that were accompanied by increased cellular infiltrates in the central nervous system (CNS). The severity of EAE was attenuated in mice given the amyotrophic lateral sclerosis drug riluzole or fed a diet enriched with linseed oil (which contains the TREK-1 activating omega-3 fatty acid α-linolenic acid). These beneficial effects were reduced in Kcnk2(-/-) mice, suggesting TREK-1 activating compounds may be used therapeutically to treat diseases related to BBB dysfunction.
Subject(s)
Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Actins/metabolism , Animals , Anticonvulsants/pharmacology , Blood-Brain Barrier/immunology , Brain/immunology , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/immunology , Cell Movement , Cells, Cultured , Coculture Techniques , Dendritic Cells , Down-Regulation , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , HEK293 Cells , Humans , Intercellular Adhesion Molecule-1/immunology , Intercellular Adhesion Molecule-1/metabolism , Interferon-alpha/pharmacology , Leukocytes/metabolism , Linseed Oil/administration & dosage , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments , Potassium Channels, Tandem Pore Domain/genetics , Riluzole/pharmacology , Transendothelial and Transepithelial MigrationABSTRACT
ETHNOPHARMACOLOGICAL RELEVANCE: Illicium verum is a traditional herbal medicine with anti-inflammatory properties used in Asia. However, its usefulness in the treatment of allergic diseases remains unclear. This study evaluated the anti-inflammatory and antiallergic effects of I. verum extract (IVE) in a mouse model of atopic dermatitis. MATERIALS AND METHODS: We investigated the effects of IVE on compound 48/80-induced histamine release, and phorbol 12-myristate13-acetate and calcium ionophore A23187-stimulated cytokines secretion in MC/9 mast cells. Atopic dermatitis was induced in NC/Nga mice by exposure to extract of house dust mite (Dermatophagoides farinae). After a topical application of IVE on ear and skin lesions, we evaluated the severity of skin symptoms, ear thickness, inflammatory cell infiltration, and serum levels of immunoglobulin E (IgE), histamine, interleukin (IL)-6, and intercellular adhesion molecule (ICAM)-1. In addition, we determined the expression of IL-4, IL-6, tumor necrosis factor (TNF)-α, interferon (IFN)-γ thymus- and activation-regulated chemokine (TARC), regulated on activation, normal T cell expressed and secreted (RANTES), ICAM-1, and vascular cell adhesion molecule (VCAM)-1 in ear tissues. RESULTS: IVE inhibited secretion of histamine, IL-4, IL-6, and TNF-α from mast cells in a dose-dependent manner. Topical application of IVE significantly reduced dermatitis scores, ear thickness, and serum levels of IgE, histamine, IL-6, and ICAM-1. Histopathological analysis demonstrated decreased epidermal thickening and dermal infiltration by inflammatory cells. In the ear lesions, IVE treatment reduced expression of IL-4, IL-6, TNF-α, TARC, RANTES, ICAM-1, and VCAM-1, but not IFN-γ. CONCLUSIONS: These results indicate that IVE inhibits atopic dermatitis-like skin lesions by suppressing the expression of cytokines, chemokines, and adhesion molecules. These results suggest that IVE may be a potential therapeutic candidate for atopic dermatitis.
Subject(s)
Anti-Allergic Agents/therapeutic use , Dermatitis, Atopic/drug therapy , Illicium , Phytotherapy , Plant Extracts/therapeutic use , Administration, Topical , Allergens/immunology , Animals , Anti-Allergic Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cell Adhesion Molecules/immunology , Cytokines/blood , Cytokines/immunology , Dermatitis, Atopic/pathology , Dermatophagoides farinae/immunology , Ethanol/chemistry , Fruit , Histamine/blood , Immunoglobulin E/blood , Male , Mice , Plant Extracts/pharmacology , Solvents/chemistry , Vascular Cell Adhesion Molecule-1/immunologyABSTRACT
VB6-845 is a recombinant immunotoxin comprised of deBouganin (a de-immunized plant toxin) genetically linked to an epithelial cell adhesion molecule (EpCAM)-targeting humanized Fab fragment (4D5MOCB). EpCAM is highly expressed on a wide range of epithelial tumors but has limited expression on most normal epithelia and therefore represents an excellent target for immunotherapy. A comprehensive preclinical evaluation was performed to determine the safety and suitability of VB6-845 as a systemically administered drug for the treatment of solid tumors. Efficacy studies in mice demonstrated that VB6-845 specifically and potently targeted EpCAM-positive tumors. In a dose-ranging study in Sprague-Dawley rats, single doses of VB6-845 were well-tolerated resulting in a no-observable adverse effect level (NOAEL) of 100 mg/kg whereas repeated doses of VB6-845 resulted in vascular leak-associated symptoms particularly at higher dose levels. However, much higher doses in Cynomolgus monkeys were well-tolerated when given as a 3-hour infusion mimicking the intended route of administration in the clinic. In addition, VB6-845 proved to be minimally immunogenic in monkeys. The toxicological data obtained in Cynomolgus monkeys indicated an excellent safety profile with a NOAEL value of 30 mg/kg (equivalent to a 10 mg/kg dose in humans). These results are supportive of an exploratory Phase I trial.
Subject(s)
Immunotoxins/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epithelial Cell Adhesion Molecule , Female , Humans , Immunotherapy/methods , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor AssaysABSTRACT
In this report, a label-free reflectometric interference spectroscopy (RIfS) based microchip biosensor for the detection of circulating tumour cells (CTCs) is demonstrated. Highly ordered nanoporous anodic aluminium oxide (AAO) fabricated by electrochemical anodization of aluminium foil was used as the RIfS sensing platform. Biotinylated anti-EpCAM antibody that specifically binds to human cancer cells of epithelial origin such as pancreatic cancer cells (PANC-1) was covalently attached to the AAO surface through multiple surface functionalization steps. Whole blood or phosphate buffer saline spiked with low numbers of pancreatic cancer cells were successfully detected by specially designed microfluidic device incorporating an AAO RIfS sensor, without labour intensive fluorescence labelling and/or pre-enhancement process. Our results show that the developed device is capable of selectively detecting of cancer cells, within a concentrations range of 1000-100,000 cells/mL, with a detection limit of <1000 cells/mL, a response time of <5 min and sample volume of 50 µL of. The presented RIfS method shows considerable promise for translation to a rapid and cost-effective point-of-care diagnostic device for the detection of CTCs in patients with metastatic cancer.
Subject(s)
Biosensing Techniques/instrumentation , Neoplastic Cells, Circulating/pathology , Aluminum Oxide , Antibodies, Immobilized , Antibodies, Neoplasm , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Equipment Design , Humans , Lab-On-A-Chip Devices , Limit of Detection , Metal Nanoparticles/ultrastructure , Microfluidic Analytical Techniques/instrumentation , Microscopy, Electron, Scanning , Neoplastic Cells, Circulating/immunology , Porosity , Spectrum Analysis/instrumentation , Surface PropertiesABSTRACT
Carcinoembryonic antigen related cell adhesion molecule (CEACAM) 6 is over-expressed in different types of cancer cells. In addition, it has also been implicated in some infectious diseases. Targeting this molecule by an antibody might have applications in diverse tumor models. Single domain antibody (sdAb) is becoming very useful format in antibody engineering as potential tools for treating acute and chronic disease conditions such as cancer for both diagnostic as well as therapeutic application. Generally, sdAbs with good affinity are isolated from an immune library. Discovery of a new target antigen would require a new immunization with purified antigen which is not always easy. In this study, we have isolated, by phage display, an sdAb against CEACAM6 with an affinity of 5 nM from a llama immunized with cancer cells. The antibody has good biophysical properties, and it binds to the cells expressing the target antigen. Furthermore, it reduces cancer cells proliferation in vitro and shows an excellent tumor targeting in vivo. This sdAb could be useful in diagnosis as well as therapy of CEACAM6 expressing tumors. Finally, we envisage it would be feasible to isolate good sdAbs against other interesting tumor associated antigens from this library. Therefore, this immunization method could be a general strategy for isolating sdAbs against any surface antigen without immunizing the animal with the antigen of interest each time.
Subject(s)
Antibodies, Neoplasm/isolation & purification , Single-Chain Antibodies/therapeutic use , Amino Acid Sequence , Animals , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/immunology , Antibodies, Neoplasm/therapeutic use , Antibody Affinity , Antigens, CD/administration & dosage , Antigens, CD/immunology , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Base Sequence , Camelids, New World , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/immunology , Cell Line, Tumor , DNA, Complementary/genetics , Epitope Mapping , GPI-Linked Proteins/administration & dosage , GPI-Linked Proteins/immunology , Humans , Immunization , Molecular Sequence Data , Neoplasms/immunology , Neoplasms/therapy , Peptide Library , Protein Engineering/methods , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/isolation & purificationABSTRACT
Garlic ( Allium sativum ) possesses anti-inflammatory effects. This study investigated the effects of garlic oil on endotoxin-induced neutrophil infiltration in the small intestine. Wistar rats received by gavage 10, 50, or 100 mg/kg body wt garlic oil (GO) or the vehicle (corn oil; 2 mL/kg body wt) every other day for 2 weeks before being injected with endotoxin (ip, 5 mg/kg body wt). Control rats were administered corn oil and injected with sterile saline. Blood samples for the measurement of soluble adhesion molecules were collected at various time points after injection, and all other samples were collected 18 h after injection. The 10 and 50 mg/kg doses suppressed endotoxin-induced neutrophilia, serum levels of sL-selectin and sICAM-1, cellular CD11b on neutrophils, intestinal ICAM-1 content, and neutrophil infiltration (P < 0.05). The 100 mg/kg dose significantly lowered local ICAM-1 and cellular CD11b on neutrophils (P < 0.05) but did not have a beneficial effect on neutrophil infiltration. In addition, 100 mg/kg of GO worsened the elevation of the local TNF-α level and neutrophilia. Appropriate doses of garlic oil have a preventive effect on endotoxin-induced neutrophil infiltration and damage to the small intestine.
Subject(s)
Cell Adhesion Molecules/immunology , Endotoxemia/immunology , Endotoxins/toxicity , Garlic/chemistry , Intestine, Small/immunology , Neutrophil Infiltration/drug effects , Neutrophils/drug effects , Plant Oils/pharmacology , Animals , CD18 Antigens/genetics , CD18 Antigens/immunology , Cell Adhesion Molecules/blood , Disease Models, Animal , Endotoxemia/drug therapy , Endotoxemia/genetics , Endotoxins/administration & dosage , Endotoxins/immunology , Humans , Intestine, Small/drug effects , Male , Neutrophils/immunology , Plant Oils/administration & dosage , Rats , Rats, WistarABSTRACT
Trichosanthin (TCS), extracted from the Chinese medicinal herb Trichosanthes kirilowi, has shown promise for the inhibition of tumor growth. However, its immunomodulatory effect on tumor-host interaction remains unknown. In this study, we focused on the effect of TCS on murine anti-tumor immune response in the 3LL Lewis lung carcinoma tumor model and explored the possible molecular pathways involved. In addition to inhibiting cell proliferation and inducing apoptosis in the 3LL tumor, TCS retarded tumor growth and prolonged mouse survival more significantly in C57BL/6 immunocompetent mice than in nude mice. This reflected the fact that the host immune system was involved in tumor eradication. Using FACS analysis, we found that TCS increased the percentage of effector T cells, particularly Interferon-gamma (IFN-γ) producing CD4(+) and CD8(+) T cells from tumor-bearing mice. TCS also promoted the vigorous proliferation of antigen-specific effector T cells, markedly increased Th1 cytokine secretion and elicited more memory T cells in tumor-bearing mice, consequently enhancing the anti-tumor response and inducing immune protection. Furthermore, we found that TCS upregulated the expression of tumor suppressor in lung cancer 1 (TSLC1) in 3LL tumor cells and the expression of its ligand, class I-restricted T cell-associated molecule (CRTAM), in effector T cells. Blocking TSLC1 expression with small interfering RNA (siRNA) significantly eliminated the effects of TCS on the proliferation and cytokine secretion of effector T cells, suggesting that TCS enhances anti-tumor immune response at least partially by boosting the interaction between TSLC1 and CRTAM. Collectively, our data demonstrate that TCS not only affects tumor cells directly, but also enhances anti-tumor immunity via the interaction between TSLC1 and CRTAM. These findings may lead to the development of a novel approach for tumor regression.
Subject(s)
Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/immunology , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Immunoglobulins/immunology , Immunoglobulins/metabolism , Trichosanthin/pharmacology , Animals , Apoptosis/drug effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Lewis Lung/metabolism , Cell Adhesion Molecule-1 , Cell Adhesion Molecules/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytokines/metabolism , Female , Immunoglobulins/genetics , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mice, Nude , RNA, Small Interfering/pharmacology , Th1 Cells/drug effects , Th1 Cells/metabolism , Trichosanthin/immunologyABSTRACT
TSG-6 (TNF-α-stimulated gene/protein 6), a hyaluronan (HA)-binding protein, has been implicated in the negative regulation of inflammatory tissue destruction. However, little is known about the tissue/cell-specific expression of TSG-6 in inflammatory processes, due to the lack of appropriate reagents for the detection of this protein in vivo. Here, we report on the development of a highly sensitive detection system and its use in cartilage proteoglycan (aggrecan)-induced arthritis, an autoimmune murine model of rheumatoid arthritis. We found significant correlation between serum concentrations of TSG-6 and arthritis severity throughout the disease process, making TSG-6 a better biomarker of inflammation than any of the other arthritis-related cytokines measured in this study. TSG-6 was present in arthritic joint tissue extracts together with the heavy chains of inter-α-inhibitor (IαI). Whereas TSG-6 was broadly detectable in arthritic synovial tissue, the highest level of TSG-6 was co-localized with tryptases in the heparin-containing secretory granules of mast cells. In vitro, TSG-6 formed complexes with the tryptases murine mast cell protease-6 and -7 via either heparin or HA. In vivo TSG-6-tryptase association could also be detected in arthritic joint extracts by co-immunoprecipitation. TSG-6 has been reported to suppress inflammatory tissue destruction by enhancing the serine protease-inhibitory activity of IαI against plasmin. TSG-6 achieves this by transferring heavy chains from IαI to HA, thus liberating the active bikunin subunit of IαI. Because bikunin is also present in mast cell granules, we propose that TSG-6 can promote inhibition of tryptase activity via a mechanism similar to inhibition of plasmin.