Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Biochemistry (Mosc) ; 87(4): 301-318, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35527372

ABSTRACT

Many factors underlie the development of inflammatory bowel disease (IBD) in humans. In particular, imbalance of microbiota and thinning of the mucosal layer in the large intestine play a huge role. Pathogenic microorganisms also exacerbate the course of diseases. In this research the role of mucin 2 deficiency in the formation of intestinal microflora in the experimental model using the Muc2 gene knockout mice in the presence of Helicobacter spp. was investigated. Also, restorative and anti-inflammatory effect of the dietary L-fucose in the Muc2-/- mice on microflora and immunity was evaluated. For this purpose, bacterial diversity in feces was studied in the animals before and after antibiotic therapy and role of the dietary L-fucose in their recovery was assessed. To determine the effect of bacterial imbalance and fucose on the immune system, mRNA levels of the genes encoding pro-inflammatory cytokines (Tnf, Il1a, Il1b, Il6) and transcription factors of T cells (Foxp3 - Treg, Rorc - Th17, Tbx21 - Th1) were determined in the colon tissue of the Muc2-/- mice. Significant elimination of bacteria due to antibiotic therapy caused decrease of the fucose levels in the intestine and facilitated reduction of the regulatory T cell transcription factor (Foxp3). When the dietary L-fucose was added to antibiotics, the level of bacterial DNA of Bacteroides spp. in the feces of the Muc2-/- mice was partially restored. T regulatory cells are involved in the regulation of inflammation in the Muc2-/- mice. Antibiotics reduced the number of regulatory T cell but did not decrease the inflammatory response to infection. Fucose, as a component of mucin 2, helped to maintain the level of Bacteroides spp. during antibiotic therapy of the Muc2-/- mice and restored biochemical parameters, but did not affect the inflammatory response.


Subject(s)
Fucose , Inflammatory Bowel Diseases , Microbiota , Mucin-2 , Animals , Anti-Bacterial Agents/pharmacology , Bacteria , Forkhead Transcription Factors , Fucose/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa , Mice , Mice, Knockout , Models, Theoretical , Mucin-2/genetics
2.
Mol Cancer Ther ; 19(5): 1102-1109, 2020 05.
Article in English | MEDLINE | ID: mdl-32165557

ABSTRACT

2-fluorofucose (2FF) inhibits protein and cellular fucosylation. Afucosylation of IgG antibodies enhances antibody-dependent cell-mediated cytotoxicity by modulating antibody affinity for FcγRIIIa, which can impact secondary T-cell activation. Immune responses toward most common solid tumors are dominated by a humoral immune response rather than the presence of tumor-infiltrating cytotoxic T cells. IgG antibodies directed against numerous tumor-associated proteins are found in the sera of both patients with breast cancer and transgenic mice bearing mammary cancer. We questioned whether 2FF would have antitumor activity in two genetically distinct transgenic models; TgMMTV-neu (luminal B) and C3(1)-Tag (basal) mammary cancer. 2FF treatment significantly improved overall survival. The TgMMTV-neu doubled survival time compared with controls [P < 0.0001; HR, 7.04; 95% confidence interval (CI), 3.31-15.0], and survival was significantly improved in C3(1)-Tag (P = 0.0013; HR, 3.36; 95% CI, 1.58-7.14). 2FF treated mice, not controls, developed delayed-type hypersensitivity and T-cell responses specific for syngeneic tumor lysates (P < 0.0001). Serum IgG from 2FF-treated mice enhanced tumor lysis more efficiently than control sera (P = 0.004). Administration of 2FF for prophylaxis, at two different doses, significantly delayed tumor onset in both TgMMTV-neu; 20 mmol/L (P = 0.0004; HR, 3.55; 95% CI, 1.60-7.88) and 50 mmol/L (P = 0.0002; HR: 3.89; 95% CI, 1.71-8.86) and C3(1)-Tag; 20 mmol/L (P = 0.0020; HR, 2.51; 95% CI, 1.22-5.18), and 50 mmol/L (P = 0.0012; HR, 3.36; 95% CI, 1.57-7.18). Mammary cancer was prevented in 33% of TgMMTV-neu and 26% of C3(1)-Tag. 2FF has potent antitumor effects in mammary cancer models. The agent shows preclinical efficacy for both cancer treatment and prevention.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Fucose/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Protein Processing, Post-Translational/drug effects , Animals , Apoptosis , Cell Proliferation , Female , Fucose/administration & dosage , Glycosylation , Humans , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Tumor Cells, Cultured
3.
Int J Biol Macromol ; 107(Pt A): 803-807, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28962845

ABSTRACT

Recent studies on crude and pure compounds from Sargassum horneri have shown promising bioactive properties. However, anti-inflammatory potentials of fucose-rich sulfated polysaccharides from S. horneri have not yet been discovered. In the present study, we evaluated the in vitro and in vivo anti-inflammatory activities of four polysaccharide fractions separated from membrane filters according to their molecular weights (<5kDa (f1), 5-10kDa (f2), 10-30kDa (f3), and >30kDa (f4)). According to the results, F4 fraction inhibited the lipopolysaccharides (LPS)-induced nitric oxide (NO) (IC50=87.12µg/mL) and prostaglandin E2 production as well as pro-inflammatory cytokine production in RAW 264.7 cells through down-regulating nuclear factor-κB signaling cascade. According to the results, f4 has a potential to down-regulate LPS-induced toxicity, cell death and NO production levels in LPS-induced in vivo zebrafish embryo model. These results suggest that f4 fraction has the potential to develop functional materials or drugs to treat inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/chemistry , Fucose/chemistry , Inflammation/drug therapy , Polysaccharides/chemistry , Animals , Anti-Inflammatory Agents/administration & dosage , Dinoprostone/biosynthesis , Fucose/administration & dosage , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Mice , Molecular Weight , Nitric Oxide/biosynthesis , Polysaccharides/administration & dosage , Polysaccharides/isolation & purification , RAW 264.7 Cells , Sargassum/chemistry , Signal Transduction/drug effects , Sulfates/chemistry , Zebrafish/embryology
4.
Gastroenterology ; 152(1): 193-205.e10, 2017 01.
Article in English | MEDLINE | ID: mdl-27639802

ABSTRACT

BACKGROUND & AIMS: De novo synthesis of guanosine diphosphate (GDP)-fucose, a substrate for fucosylglycans, requires sequential reactions mediated by GDP-mannose 4,6-dehydratase (GMDS) and GDP-4-keto-6-deoxymannose 3,5-epimerase-4-reductase (FX or tissue specific transplantation antigen P35B [TSTA3]). GMDS deletions and mutations are found in 6%-13% of colorectal cancers; these mostly affect the ascending and transverse colon. We investigated whether a lack of fucosylation consequent to loss of GDP-fucose synthesis contributes to colon carcinogenesis. METHODS: FX deficiency and GMDS deletion produce the same biochemical phenotype of GDP-fucose deficiency. We studied a mouse model of fucosylation deficiency (Fx-/- mice) and mice with the full-length Fx gene (controls). Mice were placed on standard chow or fucose-containing diet (equivalent to a control fucosylglycan phenotype). Colon tissues were collected and analyzed histologically or by enzyme-linked immunosorbent assays to measure cytokine levels; T cells also were collected and analyzed. Fecal samples were analyzed by 16s ribosomal RNA sequencing. Mucosal barrier function was measured by uptake of fluorescent dextran. We transplanted bone marrow cells from Fx-/- or control mice (Ly5.2) into irradiated 8-week-old Fx-/- or control mice (Ly5.1). We performed immunohistochemical analyses for expression of Notch and the hes family bHLH transcription factor (HES1) in colon tissues from mice and a panel of 60 human colorectal cancer specimens (27 left-sided, 33 right-sided). RESULTS: Fx-/- mice developed colitis and serrated-like lesions. The intestinal pathology of Fx-/- mice was reversed by addition of fucose to the diet, which restored fucosylation via a salvage pathway. In the absence of fucosylation, dysplasia appeared and progressed to adenocarcinoma in up to 40% of mice, affecting mainly the right colon and cecum. Notch was not activated in Fx-/- mice fed standard chow, leading to decreased expression of its target Hes1. Fucosylation deficiency altered the composition of the fecal microbiota, reduced mucosal barrier function, and altered epithelial proliferation marked by Ki67. Fx-/- mice receiving control bone marrow cells had intestinal inflammation and dysplasia, and reduced expression of cytokines produced by cytotoxic T cells. Human sessile serrated adenomas and right-sided colorectal tumors with epigenetic loss of MutL homolog 1 (MLH1) had lost or had lower levels of HES1 than other colorectal tumor types or nontumor tissues. CONCLUSIONS: In mice, fucosylation deficiency leads to colitis and adenocarcinoma, loss of Notch activation, and down-regulation of Hes1. HES1 loss correlates with the development of human right-sided colorectal tumors with epigenetic loss of MLH1. These findings indicate that carcinogenesis in a subset of colon cancer is consequent to a molecular mechanism driven by fucosylation deficiency and/or HES1-loss.


Subject(s)
Adenocarcinoma/etiology , Carbohydrate Epimerases/deficiency , Colitis/etiology , Colitis/metabolism , Colon/metabolism , Colonic Neoplasms/etiology , Intestinal Mucosa/metabolism , Ketone Oxidoreductases/deficiency , Adenocarcinoma/chemistry , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Bone Marrow Transplantation , Carbohydrate Epimerases/genetics , Carcinogenesis , Cecum/pathology , Cell Proliferation , Colitis/pathology , Colitis/prevention & control , Colon/pathology , Colonic Neoplasms/chemistry , Colonic Neoplasms/pathology , Cytokines/genetics , Cytokines/metabolism , Feces/microbiology , Female , Fucose/administration & dosage , Gastrointestinal Microbiome , Guanosine Diphosphate Fucose/biosynthesis , Guanosine Diphosphate Fucose/deficiency , Humans , Ketone Oxidoreductases/genetics , Male , Mice , Mice, Knockout , Middle Aged , Permeability , RNA, Messenger/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Signal Transduction , Transcription Factor HES-1/analysis , Transcription Factor HES-1/metabolism , Young Adult
5.
PLoS One ; 11(11): e0166070, 2016.
Article in English | MEDLINE | ID: mdl-27851789

ABSTRACT

2´-fucosyllactose (2´-FL) is an abundant human milk oligosaccharide (HMO) in human milk with diverse biological effects. We recently reported ingested 2´-FL stimulates central nervous system (CNS) function, such as hippocampal long term potentiation (LTP) and learning and memory in rats. Conceivably the effect of 2´-FL on CNS function may be via the gut-brain axis (GBA), specifically the vagus nerve, and L-fucose (Fuc) may play a role. This study had two aims: (1) determine if the effect of ingested 2´-FL on the modulation of CNS function is dependent on the integrity of the molecule; and (2) confirm if oral 2´-FL modified hippocampal LTP and associative learning related skills in rats submitted to bilateral subdiaphragmatic vagotomy. Results showed that 2´-FL but not Fuc enhanced LTP, and vagotomy inhibited the effects of oral 2´-FL on LTP and associative learning related paradigms. Taken together, the data show that dietary 2´-FL but not its Fuc moiety affects cognitive domains and improves learning and memory in rats. This effect is dependent on vagus nerve integrity, suggesting GBA plays a role in 2´-FL-mediated cognitive benefits.


Subject(s)
Brain/physiology , Conditioning, Operant/drug effects , Diet , Digestive System/drug effects , Long-Term Potentiation/drug effects , Trisaccharides/pharmacology , Vagus Nerve/physiology , Administration, Oral , Animals , Brain/drug effects , Fucose/administration & dosage , Fucose/pharmacology , Male , Rats, Sprague-Dawley , Trisaccharides/administration & dosage , Vagotomy , Vagus Nerve/drug effects
6.
Nucl Med Biol ; 43(9): 577-580, 2016 09.
Article in English | MEDLINE | ID: mdl-27434609

ABSTRACT

INTRODUCTION: PET with [(18)F]fluoro-2-deoxy-D-galactose ((18)F-FDGal) is a promising imaging modality for detection of hepatocellular carcinoma (HCC). However, it can be difficult to distinguish small intrahepatic HCC lesions from surrounding liver tissue. Ut the competitive inhibition that galactose shows towards hepatic (18)F-FDGal metabolism, we tested the hypothesis that co-administration of galactose, at near-saturating doses, inhibits (18)F-FDGal metabolism to a greater extent in non-malignant hepatocytes than in HCC cells. This would increase the tumor to background ratio in the (18)F-FDGal PET scans with co-administration of galactose. METHODS: Three patients known to have HCC underwent two (18)F-FDGal PET/CT scans on consecutive days, one with and one without simultaneous constant intravenous infusion of galactose. On both days, (18)F-FDGal was injected in the beginning of a 45-min dynamic PET scan of the liver followed by a static PET scan from mid-thigh to the top of the skull starting 60-70min after (18)F-FDGal administration. Parametric images of the hepatic metabolic function expressed in terms of hepatic systemic clearance of (18)F-FDGal were generated from the dynamic PET recordings. RESULTS: Co-administration of galactose did not give significantly better discrimination of the HCC lesions from background. Parametric images of the hepatic metabolic function did not add additional useful information to the detection of HCC lesions compared to the static images of radioactivity concentrations. CONCLUSION: Co-administration of galactose did not improve the interpretation of the (18)F-FDGal PET/CT images and did not improve the detection of intrahepatic HCC lesions, either using static or parametric images.


Subject(s)
Carcinoma, Hepatocellular/diagnostic imaging , Fucose/analogs & derivatives , Galactose/administration & dosage , Liver Neoplasms/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Aged , Fucose/administration & dosage , Humans , Image Processing, Computer-Assisted , Male
7.
Oncotarget ; 7(25): 38586-38597, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27233074

ABSTRACT

Complete remission by induction therapy in acute myelogenous leukemia (AML) can be achieved due to improvements in supportive and optimized therapy. However, more than 20% of patients will still need to undergo salvage therapy, and most will have a poor prognosis. Determining the specificity of drugs to leukemia cells is important since this will maximize the dose of chemotherapeutic agents that can be administered to AML patients. In turn, this would be expected to lead to reduced drug toxicity and its increased efficacy. We targeted Notch-1 positive AML cells utilizing fucose-bound liposomes, since activation of Notch-1 is required for O-fucosylation. Herein, we report that intravenously injected, L-fucose-bound liposomes containing daunorubicin can be successfully delivered to AML cells that express fucosylated antigens. This resulted in efficient tumor growth inhibition in tumor-bearing mice and decreased proliferation of AML patient-derived leukemia cells. Thus, biological targeting by fucose-bound liposomes that takes advantage of the intrinsic characteristics of AML cells could be a promising new strategy for Notch-1 positive-AML treatment.


Subject(s)
Daunorubicin/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Liposomes/administration & dosage , Receptor, Notch1/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Female , Fucose/administration & dosage , Fucose/chemistry , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Liposomes/chemistry , Male , Mice , Middle Aged , Molecular Targeted Therapy , Xenograft Model Antitumor Assays , Young Adult
8.
Regul Toxicol Pharmacol ; 72(1): 39-48, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25728407

ABSTRACT

l-Fucose is a natural monosaccharide present in mammals where it is found predominantly as an O-glycosidically linked component of glycoproteins, glycolipids, and oligosaccharides. It is also present in its free form in human breast milk (human milk monosaccharide). l-Fucose plays important roles in the development of the immune and nervous systems and is involved in cognitive function and memory formation. The human-identical milk monosaccharide l-fucose is therefore proposed for use in infant formulas to better simulate the free saccharides present in human breast milk. As part of the safety evaluation of l-fucose, a subchronic dietary toxicity study preceded by an in utero phase was conducted in Sprague-Dawley rats. l-Fucose was without maternal toxicity or compound-related adverse effects on female reproduction and general growth and development of offspring at a maternal dietary level up to 1%, equivalent to a dose of 1655 mg/kg body weight (bw)/day. During the subchronic phase, no compound-related adverse effects were observed in first generation rats at dietary levels of up to 1% (highest level tested), corresponding to doses of 516 and 665 mg/kg bw/day in males and females, respectively. l-Fucose was non-genotoxic in a series of in vitro genotoxicity/mutagenicity tests. These results support the safe use of l-fucose in infant formula and as a food ingredient at levels equivalent to those present in human breast milk.


Subject(s)
Fucose/administration & dosage , Infant Formula/pharmacology , Milk, Human/metabolism , Monosaccharides/adverse effects , Animals , Female , Humans , Infant , Male , Mutagenicity Tests/methods , Rats , Rats, Sprague-Dawley , Reproduction/drug effects , Safety
9.
Sci Rep ; 5: 8264, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25652335

ABSTRACT

Core fucosylation is an important post-translational modification, which is catalyzed by α1,6-fucosyltransferase (Fut8). Increased expression of Fut8 has been shown in diverse carcinomas including hepatocarcinoma. In this study, we investigated the role of Fut8 expression in liver regeneration by using the 70% partial hepatectomy (PH) model, and found that Fut8 is also critical for the regeneration of liver. Interestingly, we show that the Fut8 activities were significantly increased in the beginning of PH (~4d), but returned to the basal level in the late stage of PH. Lacking Fut8 led to delayed liver recovery in mice. This retardation mainly resulted from suppressed hepatocyte proliferation, as supported not only by a decreased phosphorylation level of epidermal growth factor (EGF) receptor and hepatocyte growth factor (HGF) receptor in the liver of Fut8(-/-) mice in vivo, but by the reduced response to exogenous EGF and HGF of the primary hepatocytes isolated from the Fut8(-/-) mice. Furthermore, an administration of L-fucose, which can increase GDP-fucose synthesis through a salvage pathway, significantly rescued the delayed liver regeneration of Fut8(+/-) mice. Overall, our study provides the first direct evidence for the involvement of Fut8 in liver regeneration.


Subject(s)
Fucosyltransferases/deficiency , Liver Regeneration , Animals , Cell Proliferation/drug effects , Fucose/administration & dosage , Fucose/metabolism , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Gene Expression , Genotype , Hepatectomy , Hepatocytes/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver Regeneration/genetics , Mice , Mice, Knockout , Models, Animal , Receptors, Growth Factor/metabolism , Signal Transduction
10.
Nanomedicine (Lond) ; 10(1): 57-71, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25177920

ABSTRACT

AIM: The aim of this work was to develop fucose-conjugated nanoparticles and control the release of berberine, and demonstrate that these particles come into contact with Helicobacter pylori and enhance the suppressive effect of berberine on H. pylori growth. MATERIALS & METHODS: Fucose-chitosan/heparin nanoparticle-encapsulated berberine was prepared and delivery efficiency was monitored by confocal laser scanning microscopy. Anti-H. pylori activities were investigated by determining the calculated bacterial colonies and immunohistochemistry staining analysis. RESULTS: Analysis of a simulated gastrointestinal medium indicated that the proposed drug carrier effectively controls the release of berberine, which interacts specifically at the site of H. pylori infection, and significantly increases berberine's suppressive effect on H. pylori growth. In an in vivo study, the berberine-loaded fucose-conjugated nanoparticles exhibited an H. pylori clearance effect. CONCLUSION: These findings indicate that berberine-loaded fucose-conjugated nanoparticles exert an H. pylori clearance effect and effectively reduce gastric inflammation in an H. pylori-infected animal study.


Subject(s)
Drug Delivery Systems , Helicobacter Infections/drug therapy , Helicobacter pylori/drug effects , Nanoparticles/administration & dosage , Animals , Berberine/administration & dosage , Berberine/chemistry , Chitosan/administration & dosage , Chitosan/chemistry , Fucose/administration & dosage , Fucose/chemistry , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter pylori/pathogenicity , Heparin/administration & dosage , Heparin/chemistry , Humans , Mice , Microscopy, Confocal , Nanoparticles/chemistry
11.
J Drug Target ; 22(1): 34-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23987115

ABSTRACT

PURPOSE: Oral immunization for mucosal protection against Mycobacterium tuberculosis would be the best option for effective tuberculosis (TB) control. However, this route of vaccine delivery is limited due to the short residence time of the delivery system at the site of absorption. Cytoadhension has made it possible to optimize the targeted delivery of oral vaccine to lymphoid tissues. The purpose of this project was to evaluate the ability of human M-cell specific lectin-labeled microparticles to target the human M-cells of the Peyer's patches. METHOD: Albumin microspheres containing Mycobacterium tuberculosis cell lysate antigens were coupled with Wheat germ agglutinin and Aleuria aurantia lectins and their ability to bind to M cell models as well as their preferential distribution in the Peyer's patches were investigated. RESULTS: The study demonstrated an enhanced delivery of targeted polystyrene and BSA/Lysate microspheres to M cells. It was demonstrated that alpha-l-fucose sugar residue might be the target of these lectins. CONCLUSION: It can be concluded from the study that the lectin-coupled microspheres had better affinity for M-cells and showed preferential binding to the Peyer's patches. This means that the coupling enhanced the targeted delivery of the antigens to the M cells.


Subject(s)
Antigens, Bacterial/administration & dosage , Antigens, Bacterial/chemistry , BCG Vaccine/administration & dosage , BCG Vaccine/chemistry , Lectins/administration & dosage , Lectins/chemistry , Administration, Oral , Albumins/immunology , Alkaline Phosphatase/metabolism , Animals , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Caco-2 Cells , Cell Line, Tumor , Drug Delivery Systems/methods , Fucose/administration & dosage , Fucose/chemistry , Fucose/immunology , Humans , Lectins/immunology , Mice , Microspheres , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Peyer's Patches/immunology , Polystyrenes/administration & dosage , Polystyrenes/chemistry , Polystyrenes/immunology , Tuberculosis/prevention & control
12.
Eur J Pharm Biopharm ; 86(3): 449-58, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24189499

ABSTRACT

The present investigation was aimed at exploring the targeting potential of sulfasalazine (NF-κB inhibitor drug) loaded fucose tethered poly (propylene imine) (PPI) dendritic nanoarchitecture (SSZ-FUCO-PPID) to Kupffer cells for effective management of cytokine-induced liver damage. The SSZ-FUCO-PPID formulation was characterized for entrapment efficiency, in vitro release, stability, toxicological investigations, macrophage uptake, NF-κB inhibition, and in vivo studies. In cell uptake assay the uptake of SSZ-FUCO-PPID was found to be higher and preferentially by J774 macrophage cell line. Cytokine assay suggested that the SSZ-FUCO-PPID potentially inhibited the IL-12 p40 production in LPS activated macrophages. Western blot analysis clearly suggested that SSZ-FUCO-PPID inhibited the activation of NF-κB as indicated by the absence of p-IκB band. Pharmacokinetic study revealed improved bioavailability, half-life and mean residence time of SSZ upon fucosylation of dendrimers. The biodistribution pattern clearly established the higher amount of SSZ-FUCO-PPID in liver. Hematological data suggest that the fucosylated formulations are less immunogenic as compared to unconjugated formulations. The results suggest that the SSZ-FUCO-PPID formulation holds targeting potential to Kupffer cells for the treatment of cytokine-induced liver damage.


Subject(s)
Aziridines/chemistry , Chemical and Drug Induced Liver Injury/drug therapy , Cytokines/toxicity , Dendrimers/chemistry , Fucose/chemistry , Sulfasalazine/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Aziridines/administration & dosage , Chemical and Drug Induced Liver Injury/blood , Dendrimers/administration & dosage , Dose-Response Relationship, Drug , Fucose/administration & dosage , Male , Rats , Rats, Sprague-Dawley , Sulfasalazine/administration & dosage , Treatment Outcome
13.
Am J Physiol Lung Cell Mol Physiol ; 305(5): L352-63, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23792737

ABSTRACT

Treatment of acute and chronic pulmonary infections caused by opportunistic pathogen Pseudomonas aeruginosa is limited by the increasing frequency of multidrug bacterial resistance. Here, we describe a novel adjunctive therapy in which administration of a mix of simple sugars-mannose, fucose, and galactose-inhibits bacterial attachment, limits lung damage, and potentiates conventional antibiotic therapy. The sugar mixture inhibits adhesion of nonmucoid and mucoid P. aeruginosa strains to bronchial epithelial cells in vitro. In a murine model of acute pneumonia, treatment with the sugar mixture alone diminishes lung damage, bacterial dissemination to the subpleural alveoli, and neutrophil- and IL-8-driven inflammatory responses. Remarkably, the sugars act synergistically with anti-Pseudomonas antibiotics, including ß-lactams and quinolones, to further reduce bacterial lung colonization and damage. To probe the mechanism, we examined the effects of sugars in the presence or absence of antibiotics during growth in liquid culture and in an ex vivo infection model utilizing freshly dissected mouse tracheas and lungs. We demonstrate that the sugar mixture induces rapid but reversible formation of bacterial clusters that exhibited enhanced susceptibility to antibiotics compared with individual bacteria. Our findings reveal that sugar inhalation, an inexpensive and safe therapeutic, could be used in combination with conventional antibiotic therapy to more effectively treat P. aeruginosa lung infections.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Carbohydrates/administration & dosage , Cystic Fibrosis/drug therapy , Pneumonia, Bacterial/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Animals , Bacterial Adhesion/drug effects , Bronchi/drug effects , Bronchi/metabolism , Bronchi/microbiology , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis/microbiology , Fucose/administration & dosage , Galactose/administration & dosage , Humans , Interleukin-8/metabolism , Lung Injury/drug therapy , Lung Injury/metabolism , Lung Injury/microbiology , Male , Mannose/administration & dosage , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Polysaccharides/administration & dosage , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/metabolism , Trachea/drug effects , Trachea/metabolism , Trachea/microbiology
14.
Gastroenterology ; 138(3): 1079-90.e1-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19900444

ABSTRACT

BACKGROUND & AIMS: Leukocyte adhesion deficiency II (LAD II) is a rare condition caused by defective protein fucosylation, causing decreased leukocyte rolling, psychomotor retardation, and poor growth. The ligand-binding activity of Notch, a gastrointestinal signaling protein, depends on O-fucosylation. We investigated Notch signaling and intestinal epithelial architecture in a mouse model of LAD II. METHODS: Mice lacking 3,5-epimerase/4-reductase (FX) or FX(-/-) bone marrow chimeras (with either wild-type or FX(-/-) bone marrow) were maintained on a fucose-free diet. Intestinal secretory epithelial cells were quantified by histology and immunohistochemistry. Reverse transcription-polymerase chain reaction and immunoblot analyses were used to detect Notch-regulated genes in isolated crypt epithelium. Intestinal leukocyte-endothelial interaction was quantified by intravital microscopy. The intestinal epithelium of 2-week-old FX(-/-) mice was transfected with an adenoviral vector expressing a constitutively active form of Notch. RESULTS: FX(-/-) mice rapidly exhibited secretory epithelial cell hyperplasia, reduced cell proliferation, and altered epithelial gene expression patterns consistent with reduced Notch signaling. These effects were reversed when mice were given dietary fucose or by adenoviral transfection of the intestinal epithelium with the Notch intracellular domain. CONCLUSIONS: In a mouse model of LAD II, secretory cell hyperplasia occurs in the small intestine and colon; these effects depend on Notch signaling. Defects in Notch signaling might therefore be involved in the pathogenesis of this rare pediatric condition.


Subject(s)
Carbohydrate Epimerases/metabolism , Cell Proliferation , Colon/metabolism , Goblet Cells/metabolism , Hydro-Lyases/metabolism , Ileum/metabolism , Leukocyte Rolling , Leukocyte-Adhesion Deficiency Syndrome/metabolism , Paneth Cells/metabolism , Receptors, Notch/metabolism , Adenoviridae/genetics , Animals , Carbohydrate Epimerases/deficiency , Carbohydrate Epimerases/genetics , Cell Lineage , Colon/pathology , Dietary Carbohydrates/administration & dosage , Disease Models, Animal , Fucose/administration & dosage , Fucose/deficiency , Gene Expression Regulation , Genetic Vectors , Genotype , Goblet Cells/pathology , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Hyperplasia , Ileum/pathology , Immunoblotting , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Video , Paneth Cells/pathology , Phenotype , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection , Weight Gain
15.
Am J Pathol ; 174(5): 1745-55, 2009 May.
Article in English | MEDLINE | ID: mdl-19349363

ABSTRACT

Notch1 is an evolutionarily conserved receptor that regulates cell fate, including such events as differentiation, proliferation, and apoptosis. Myofibroblast differentiation is a key feature of lung fibrosis. Found in inflammatory zone 1 (FIZZ1) has direct fibrogenic properties because of its ability to induce myofibroblast differentiation. However, the downstream signaling pathway that mediates FIZZ1 induction of myofibroblast differentiation remains unknown. The objective of this study was to investigate the involvement of Notch signaling in FIZZ1 induction of lung myofibroblast differentiation and thus explore the potential role of Notch1 in pulmonary fibrosis. The results showed that FIZZ1 increased the expression levels of activated intracellular domain of Notch1 (NIC), its ligand Jagged1, and its target gene Hes1, which were associated with elevated alpha-smooth muscle actin expression levels. Fibroblast alpha-smooth muscle actin expression is induced by the overexpression of NIC but is suppressed by the inhibition of NIC. Moreover, lung fibroblasts that were isolated from mice lacking the GDP-4-keto-6-deoxymannose3,5-epimerase-4-reductase enzyme (FX knockout) exhibited significantly reduced responsiveness to FIZZ1, which was reversed by fucose supplementation. In the absence of exogenous fucose, these FX-deficient cells exhibited defective fucosylation, which is required for Notch signaling. These knockout mice also showed impaired lung fibrosis. These findings suggest that Notch1 signaling in response to FIZZ1 may play a significant role in myofibroblast differentiation during lung fibrosis.


Subject(s)
Cell Differentiation , Fibroblasts/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Lung/cytology , Receptor, Notch1/metabolism , Actins/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bleomycin/toxicity , Blotting, Western , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Fibroblasts/metabolism , Fluorescent Antibody Technique , Fucose/administration & dosage , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hydro-Lyases/physiology , Hydroxyproline/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Lung/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Promoter Regions, Genetic , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Transcription Factor HES-1
16.
Int J Med Sci ; 5(6): 371-6, 2008.
Article in English | MEDLINE | ID: mdl-19043609

ABSTRACT

BACKGROUND: Colonisation of cystic fibrosis (CF) lungs with Pseudomonas aeruginosa is facilitated by two lectins, which bind to the sugar coat of the surface lining epithelia and stop the cilia beating. OBJECTIVES: We hypothesized that P. aeruginosa lung infection should be cleared by inhalation of fucose and galactose, which compete for the sugar binding site of the two lectins and thus inhibit the binding of P. aeruginosa. METHODS: 11 adult CF patients with chronic infection with P. aeruginosa were treated twice daily with inhalation of a fucose/galactose solution for 21 days (4 patients only received inhalation, 7 patients received inhalation and intravenous antibiotics). Microbial counts of P. aeruginosa, lung function measurements, and inflammatory markers were determined before and after treatment. RESULTS: The sugar inhalation was well tolerated and no adverse side effects were observed. Inhalation alone as well as combined therapy (inhalation and antibiotics) significantly decreased P. aeruginosa in sputum (P < 0.05). Both therapies also significantly reduced TNFalpha expression in sputum and peripheral blood cells (P < 0.05). No change in lung function measurements was observed. CONCLUSIONS: Inhalation of simple sugars is a safe and effective measure to reduce the P. aeruginosa counts in CF patients. This may provide an alternative therapeutical approach to treat infection with P. aeruginosa.


Subject(s)
Cystic Fibrosis/complications , Fucose/therapeutic use , Galactose/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Administration, Inhalation , Adult , Aminoglycosides/administration & dosage , Aminoglycosides/therapeutic use , C-Reactive Protein/metabolism , Cell Count , Cephalosporins/administration & dosage , Cephalosporins/therapeutic use , Drug Therapy, Combination , Female , Fucose/administration & dosage , Galactose/administration & dosage , Gene Expression/drug effects , Humans , Immunoglobulin Isotypes/blood , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Liver Function Tests , Male , Neutrophils/cytology , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/isolation & purification , Respiratory Function Tests , Sputum/cytology , Sputum/metabolism , Sputum/microbiology , Treatment Outcome , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics
17.
Vascul Pharmacol ; 44(5): 363-71, 2006 May.
Article in English | MEDLINE | ID: mdl-16540378

ABSTRACT

Although drugs such as sirolimus and paclitaxel are effective in reducing restenosis, their effects on vascular function are often overlooked. In this study, we have examined the effects of local delivery of several anti-restenotic drugs given in vivo after balloon injury on in vitro vascular contraction and relaxation 28 days after injury. Paclitaxel (50 microM), the farnesyl protein transferase inhibitor L744 (25 microM), sirolimus (25 microM) and Van 10/4 (decahydro-1,1,4,7-tetramethyl-1H-cycloprop[e]azulen-4-o-[2-(3-methylpent-2-enoyl)-fucopyranoside]; 25 microM) were delivered to porcine coronary arteries in vivo and the arteries removed 28 days later. Contractions to KCl and 5-hydroxytryptamine (5-HT) and relaxations to calcimycin and 3-morpholinosydnonimine (SIN-1) were measured in control (LCx) and balloon-injured (LAD) rings. In vehicle-infused coronary arteries, contraction to KCl and 5-HT was significantly enhanced 28 days after balloon injury, while the response to calcimycin had recovered fully, indicating endothelial regrowth. The response to SIN-1 was unchanged. None of the four drugs tested had any effect on the enhanced response to KCl 28 days after injury or on recovery of the calcimycin response. The hyper-responsiveness to 5-HT was eliminated by sirolimus, Van 10/4 and L744, but not paclitaxel. This study demonstrates that local drug infusion with structurally different antiproliferative drugs at the time of balloon angioplasty does not affect endothelial recovery and may in some cases prevent hyper-responsiveness to constrictor agents.


Subject(s)
Angioplasty, Balloon, Coronary , Coronary Vessels/drug effects , Growth Inhibitors/pharmacology , Serotonin , Vasoconstriction , Vasoconstrictor Agents , Vasodilation , Animals , Azulenes/administration & dosage , Azulenes/pharmacology , Calcimycin , Coronary Vessels/injuries , Coronary Vessels/physiopathology , Fucose/administration & dosage , Fucose/analogs & derivatives , Fucose/pharmacology , Growth Inhibitors/administration & dosage , Male , Methionine/administration & dosage , Methionine/analogs & derivatives , Methionine/pharmacology , Models, Animal , Sirolimus/administration & dosage , Sirolimus/pharmacology , Swine , Vasodilator Agents
18.
Cells Tissues Organs ; 172(3): 161-73, 2002.
Article in English | MEDLINE | ID: mdl-12476046

ABSTRACT

Leukocyte adhesion deficiency II (LAD II) is a rare congenital disease which is caused by a defect in fucosylation of glycoconjugates. Hypofucosylated structures include ligands for the selectin family of adhesion molecules. This results in a leukocyte adhesion defect causing an immunodeficiency. In addition, LAD II patients show severe mental and growth retardations suggesting a role of fucose in development. Recently, a LAD II patient was treated with oral supplementation of fucose. This simple therapy restored selectin ligands and corrected the immunodeficiency. However, in another patient the treatment protocol had no effect indicating that the biochemical defect in the latter patient is somewhat different. The genetic defect in LAD II has now been located to a gene encoding a GDP-fucose transporter which gates GDP-fucose into the Golgi where fucose is transferred onto glycoconjugates. Point mutations have been detected in this gene in several LAD II patients, which inactivate the transporter function. Thus, LAD II represents the first developmental and immune defect that is based on a malfunctioning nucleotide sugar transporter.


Subject(s)
Fucose/therapeutic use , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/therapy , Monosaccharide Transport Proteins , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , E-Selectin/metabolism , Fucose/administration & dosage , Fucose/metabolism , Golgi Apparatus/metabolism , Humans , Infant , Neutrophils/cytology , Neutrophils/metabolism , P-Selectin/metabolism , Recombinant Fusion Proteins
19.
Klin Padiatr ; 213(5): 285-7, 2001.
Article in English | MEDLINE | ID: mdl-11582527

ABSTRACT

BACKGROUND: Airway infections with Pseudomonas aeruginosa often represent a life-threatening event in immuno-compromised patients or patients with Cystic Fibrosis. The adhesion of this bacterium to surfaces such as the airway epithelium is mediated by two lectins, sugar binding proteins. In addition to their adhesive properties, these lectins have been shown to stop human ciliary beating thus compromising the mucociliary clearance as an important non-specific defence mechanism of the airways. Inhibition of these lectins by their specific sugars galactose and fucose, respectively, could therefore be of benefit in the elimination therapy of P. aeruginosa. CASE REPORT: An infant suffering from P. aeruginosa airway infection after chemotherapy for neuroblastoma, which could not successfully be treated by antibiotics, was subjected to a series of additional galactose/fucose inhalations, which eliminated the germ as evidenced by microbiological testing. This is the first report suggesting the effectiveness of a lectin-based therapeutic principle in P. aeruginosa airway infection. CONCLUSION: The competitive inhibition of P. aeruginosa lectins by the lectin specific sugars galactose and fucose may overcome particular mechanisms of bacterial resistance in patients with P. aeruginosa airway infection. This underlying biochemical mechanism and the outcome of our patient suggest a clinical benefit of this novel therapeutic approach for immunocompromised patients or patients with cystic fibrosis suffering from infection with P. aeruginosa.


Subject(s)
Fucose/therapeutic use , Galactose/therapeutic use , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Respiratory Therapy , Respiratory Tract Infections/drug therapy , Abdominal Neoplasms/surgery , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/metabolism , Drug Resistance, Microbial , Fucose/administration & dosage , Fucose/pharmacology , Galactose/administration & dosage , Galactose/pharmacology , Humans , Infant , Lectins/metabolism , Male , Neuroblastoma/surgery , Postoperative Complications , Pseudomonas aeruginosa/isolation & purification , Pseudomonas aeruginosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...