Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Chin Med J (Engl) ; 135(24): 2956-2967, 2022 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-36728287

RESUMEN

BACKGROUND: Cholangiocarcinoma (CCA) is a malignant biliary tract tumor with an extremely poor prognosis. There is an urgent demand to explore novel therapeutic strategies. L-fucose has been confirmed to participate in anti-inflammation and antitumor activities. However, the effect of L-fucose on the progression of CCA has not been well investigated. This study aimed to determine whether L-fucose induced the inhibition of CCA and its possible mechanism. METHODS: The anti-growth activity was determined using Cell Counting Kit-8 assay, colony formation assays, Annexin V-fluorescein isothiocyanate/propidium iodide (FITC/PI) assay, and cell cycle analysis. The anti-metastasis activity was determined by wound healing, transwell, and invasion assays. The anti-angiogenesis activity was determined by tube formation and transwell assays. MicroRNAs that may be involved in the L-fucose-induced CCA inhibition was analyzed using bioinformatics methods. The preclinical therapeutic efficacy was mainly estimated by ultrasound in xenograft nude mouse models. Differences were analyzed via Student's t test or one-way analysis of variance. RESULTS: L-Fucose induced apoptosis and G0/G1 cell cycle arrest, inhibited cell epithelial-mesenchymal transition of CCA cells, and additionally inhibited tube formation of human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner, leading to a decrease in cell proliferation, metastasis, and angiogenesis. Mechanistically, L-fucose induced microRNA-200b (miR-200b) upregulation, and mitogen-activated protein kinase 7 (MAPK7) downregulation was found to be targeted by miR-200b, with decreased cell proliferation and metastasis. Additionally, phosphorylated signal transducer and activator of transcription 3 was found to be downregulated after L-fucose treatment. Finally, in vivo experiments in CCA xenograft models also confirmed the antitumor properties of L-fucose. CONCLUSION: L-Fucose inhibited the progression of CCA via the miR-200b/MAPK7 and signal transducer and activator of transcription 3 signaling pathways.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , MicroARNs , Animales , Ratones , Humanos , Factor de Transcripción STAT3/metabolismo , Fucosa/uso terapéutico , Fucosa/farmacología , Células Endoteliales/metabolismo , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/metabolismo , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Proteína Quinasa 7 Activada por Mitógenos/metabolismo
2.
Int J Biol Macromol ; 190: 739-753, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34509519

RESUMEN

Naturally occurring polysaccharide-structured nanoparticles have developed as promising materials for treatment of bone health disorders. Silver nanoparticle (ST-AgNP) structured from sulfated polygalacto-fucopyranose comprising of recurring structural entities of 2-SO3-α-(1 â†’ 3)-fucopyranose and 6-O-acetyl-ß-(1 â†’ 4)-galactopyranose isolated from marine macroalga Sargassum tenerrimum demonstrated potential activities associated with osteogenesis. Subsequent treatment with ST-AgNP, activity of alkaline phosphatase (63 mU/mg) was raised in osteoblast stem cells (human mesenchymal, hMSC) than that in control (30 mU/mg). Intense growth of mineralized nodule on the surface of hMSC was apparent following treatment with ST-AgNP. Increased population of bone morphogenic protein-2 (23%) and osteocalcin+ cells (50%) on M2 macrophages were apparent following treatment with ST-AgNP (0.25 mg/mL). Glucocorticoid-induced in vivo animal model studies of ST-AgNP exhibited significant recovery of serum biochemical parameters along with serum estradiol and parathyroid hormone compared to disease control. Disease-induced groups treated with ST-AgNP showed the disappearance of osteoporotic cavities in the trabecular bone. Following treatment with ST-AgNP, serum calcium and phosphorus contents were significantly recovered.


Asunto(s)
Fucosa/uso terapéutico , Galactanos/uso terapéutico , Glucocorticoides/efectos adversos , Nanopartículas/química , Osteoporosis/inducido químicamente , Osteoporosis/tratamiento farmacológico , Fosfatasa Alcalina/metabolismo , Animales , Antioxidantes/farmacología , Peso Corporal/efectos de los fármacos , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Espectroscopía de Resonancia Magnética con Carbono-13 , Línea Celular , Supervivencia Celular/efectos de los fármacos , Digestión/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Femenino , Fémur/efectos de los fármacos , Fémur/patología , Fucosa/aislamiento & purificación , Fucosa/farmacología , Galactanos/aislamiento & purificación , Galactanos/farmacología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteocalcina/metabolismo , Espectroscopía de Protones por Resonancia Magnética , Ratas Wistar , Espectrometría por Rayos X , Espectrofotometría Ultravioleta
3.
J Inherit Metab Dis ; 44(6): 1441-1452, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34389986

RESUMEN

Fucosylation is essential for intercellular and intracellular recognition, cell-cell interaction, fertilization, and inflammatory processes. Only five types of congenital disorders of glycosylation (CDG) related to an impaired fucosylation have been described to date: FUT8-CDG, FCSK-CDG, POFUT1-CDG SLC35C1-CDG, and the only recently described GFUS-CDG. This review summarizes the clinical findings of all hitherto known 25 patients affected with those defects with regard to their pathophysiology and genotype. In addition, we describe five new patients with novel variants in the SLC35C1 gene. Furthermore, we discuss the efficacy of fucose therapy approaches within the different defects.


Asunto(s)
Trastornos Congénitos de Glicosilación/tratamiento farmacológico , Trastornos Congénitos de Glicosilación/genética , Fucosa/uso terapéutico , Proteínas de Transporte de Monosacáridos/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Glicoproteínas , Glicosilación , Humanos , Lactante , Masculino , Resultado del Tratamiento , Adulto Joven
4.
Clin Immunol ; 221: 108599, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32992000

RESUMEN

Individuals with the Bombay phenotype (Oh) in the ABO blood group system do not express the H, A, and B antigens but have no clinical symptoms. Bombay phenotype with clinical symptoms has been described in leukocyte adhesion deficiency type II (LAD II), a fucosylation disorder caused by mutations in SLC35C1. Only few LAD II patients have been described so far. Here we describe an additional patient, a 22-year old male, born to unrelated parents, presenting with inflammatory skin disease, periodontitis, growth, and mental retardation, admitted to the department of dentistry for treatment under general anesthesia. Pre-operative routine investigations revealed the presence of the Bombay phenotype (Oh). Genomic sequencing identified two novel triplet deletions of the SLC35C1 gene. Functional investigations confirmed the diagnosis of LAD II. Therapy with oral fucose led to the disappearance of the chronic skin infections and improvements in behavior and attention span.


Asunto(s)
Síndrome de Deficiencia de Adhesión del Leucocito/diagnóstico , Sistema del Grupo Sanguíneo ABO , Adulto , Tipificación y Pruebas Cruzadas Sanguíneas , Eritrocitos , Fucosa/uso terapéutico , Humanos , Síndrome de Deficiencia de Adhesión del Leucocito/sangre , Síndrome de Deficiencia de Adhesión del Leucocito/tratamiento farmacológico , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Leucocitos , Masculino , Proteínas de Transporte de Monosacáridos/genética , Adulto Joven
5.
Nutrients ; 12(2)2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053891

RESUMEN

Growing evidence suggests that intestinal mucosa homeostasis impacts immunity, metabolism, the Central Nervous System (CNS), and behavior. Here, we investigated the effect of the monosaccharide fucose on inflammation, metabolism, intestinal microbiota, and social behavior in the Dextran Sulfate Sodium (DSS)-induced chronic colitis mouse model. Our data show that chronic colitis is accompanied by the decrease of the serum tryptophan level and the depletion of the intestinal microbiota, specifically tryptophan-producing E. coli and Bifidobacterium. These changes are associated with defects in the male mouse social behavior such as a lack of preference towards female bedding in an odor preference test. The addition of fucose to the test animals' diet altered the bacterial community, increased the abundance of tryptophan-producing E. coli, normalized blood tryptophan levels, and ameliorated social behavior deficits. At the same time, we observed no ameliorating effect of fucose on colon morphology and colitis. Our results suggest a possible mechanism by which intestinal inflammation affects social behavior in male mice. We propose fucose as a promising prebiotic, since it creates a favorable environment for the beneficial bacteria that promote normalization of serum tryptophan level and amelioration of the behavioral abnormalities in the odor preference test.


Asunto(s)
Conducta Animal/efectos de los fármacos , Colitis/psicología , Fucosa/farmacología , Triptófano/metabolismo , Animales , Bifidobacterium/metabolismo , Enfermedad Crónica , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis/microbiología , Colon/metabolismo , Colon/microbiología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Escherichia coli/metabolismo , Fucosa/uso terapéutico , Microbioma Gastrointestinal/efectos de los fármacos , Inflamación , Mucosa Intestinal , Masculino , Ratones Endogámicos C57BL , Conducta Social
6.
Sci Rep ; 9(1): 14904, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31624296

RESUMEN

A recently described bangle lectin (PHL) from the bacterium Photorhabdus asymbiotica was identified as a mainly fucose-binding protein that could play an important role in the host-pathogen interaction and in the modulation of host immune response. Structural studies showed that PHL is a homo-dimer that contains up to seven L-fucose-specific binding sites per monomer. For these reasons, potential ligands of the PHL lectin: α-L-fucopyranosyl-containing mono-, di-, tetra-, hexa- and dodecavalent ligands were tested. Two types of polyvalent structures were investigated - calix[4]arenes and dendrimers. The shared feature of all these structures was a C-glycosidic bond instead of the more common but physiologically unstable O-glycosidic bond. The inhibition potential of the tested structures was assessed using different techniques - hemagglutination, surface plasmon resonance, isothermal titration calorimetry, and cell cross-linking. All the ligands proved to be better than free L-fucose. The most active hexavalent dendrimer exhibited affinity three orders of magnitude higher than that of standard L-fucose. To determine the binding mode of some ligands, crystal complex PHL/fucosides 2 - 4 were prepared and studied using X-ray crystallography. The electron density in complexes proved the presence of the compounds in 6 out of 7 fucose-binding sites.


Asunto(s)
Antibacterianos/farmacología , Infecciones Bacterianas/tratamiento farmacológico , Proteínas Bacterianas/antagonistas & inhibidores , Lectinas/antagonistas & inhibidores , Photorhabdus/metabolismo , Antibacterianos/química , Antibacterianos/uso terapéutico , Infecciones Bacterianas/microbiología , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Dendrímeros/química , Dendrímeros/farmacología , Dendrímeros/uso terapéutico , Eritrocitos , Fucosa/análogos & derivados , Fucosa/farmacología , Fucosa/uso terapéutico , Hemaglutinación/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Lectinas/química , Lectinas/aislamiento & purificación , Lectinas/metabolismo , Ligandos , Modelos Moleculares , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie
7.
Int Immunopharmacol ; 73: 379-388, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31132733

RESUMEN

Previous studies reported that L-fucose had anti-inflammatory effects in respiratory and cutaneous system. However, the effect of L-fucose on colitis and the underlying mechanism is poorly understood. We studied the anti-inflammatory effects of L-fucose on Dextran sulfate sodium (DSS)-induced acute colitis in vivo and on LPS/ATP-induced bone marrow derived macrophages (BMDMs) damage in vitro. Our results show that L-fucose significantly alleviated weight loss and disease activity index (DAI) scores in colitis and reduced the infiltration of macrophages and neutrophils. In addition, L-fucose can inhibit macrophage M1 polarization, inactivate the NLRP3 inflammasome and reduce the release of TNFα, IL1ß, IL6 pro-inflammatory cytokines. In vitro studies showed that L-fucose ameliorated cell damage resulting from the administration of LPS with ATP in BMDMs, inhibited NLRP3 inflammasome activation and reduced the release of corresponding pro-inflammatory cytokines. Finally, L-fucose can inhibit the expression of p-NF-kB in vivo and in vitro. Overall, our results show that L-fucose can attenuate colitis by inhibiting macrophage M1 polarization, inhibiting NLRP3 inflammasome and NF-kB activation, and down-regulation of pro-inflammatory cytokines.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Fucosa/uso terapéutico , Enfermedad Aguda , Animales , Antiinflamatorios/farmacología , Colitis/inducido químicamente , Colitis/inmunología , Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Citocinas/genética , Citocinas/inmunología , Sulfato de Dextran , Fucosa/farmacología , Inflamasomas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología
8.
J Transl Med ; 16(1): 344, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30526624

RESUMEN

BACKGROUND: L-Fucose (Fuc), a six-deoxy hexose monosaccharide, is present endogenously in humans and animals and has a wide range of biological functions. In the present study, we aimed to examine the effect of Fuc on obesity and hepatic steatosis in mice fed a high-fat diet (HFD). METHODS: C57BL/6 mice were fed a normal chow (NC) or HFD for 18 weeks to induce obesity and fatty liver. Fuc was administered intragastrically from the 8th week to the end of the experiment (18 weeks). RESULTS: Metagenomic analysis showed that HFD altered the genomic profile of gut microbiota in the mice; specifically, expression of alpha-L-fucosidase, the gene responsible for Fuc generation, was markedly reduced in the HFD group compared with that in the NC group. Fuc treatment decreased body weight gain, fat accumulation, and hepatic triglyceride elevation in HFD-fed mice. In addition, Fuc decreased the levels of endotoxin-producing bacteria of the Desulfovibrionaceae family and restored HFD-induced enteric dysbiosis at both compositional and functional levels. CONCLUSION: Our findings suggest that Fuc might be a novel strategy to treat HFD-induced obesity and fatty liver.


Asunto(s)
Dieta Alta en Grasa , Fucosa/uso terapéutico , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Adiposidad/efectos de los fármacos , Animales , Ciego/efectos de los fármacos , Ciego/microbiología , Disbiosis/complicaciones , Disbiosis/microbiología , Disbiosis/patología , Conducta Alimentaria , Fucosa/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Insulina , Metagenómica , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/microbiología , Obesidad/microbiología , Aumento de Peso/efectos de los fármacos
9.
Int J Mol Sci ; 19(5)2018 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-29702557

RESUMEN

Congenital disorders of glycosylation (CDG) are a group of genetic disorders that affect protein and lipid glycosylation and glycosylphosphatidylinositol synthesis. More than 100 different disorders have been reported and the number is rapidly increasing. Since glycosylation is an essential post-translational process, patients present a large range of symptoms and variable phenotypes, from very mild to extremely severe. Only for few CDG, potentially curative therapies are being used, including dietary supplementation (e.g., galactose for PGM1-CDG, fucose for SLC35C1-CDG, Mn2+ for TMEM165-CDG or mannose for MPI-CDG) and organ transplantation (e.g., liver for MPI-CDG and heart for DOLK-CDG). However, for the majority of patients, only symptomatic and preventive treatments are in use. This constitutes a burden for patients, care-givers and ultimately the healthcare system. Innovative diagnostic approaches, in vitro and in vivo models and novel biomarkers have been developed that can lead to novel therapeutic avenues aiming to ameliorate the patients’ symptoms and lives. This review summarizes the advances in therapeutic approaches for CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/terapia , Suplementos Dietéticos , Terapia Genética , Trasplante de Órganos , Animales , Biomarcadores , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Fucosa/uso terapéutico , Galactosa/uso terapéutico , Glicosilación , Humanos , Manosa/uso terapéutico
10.
Int J Med Sci ; 5(6): 371-6, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19043609

RESUMEN

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.


Asunto(s)
Fibrosis Quística/complicaciones , Fucosa/uso terapéutico , Galactosa/uso terapéutico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Administración por Inhalación , Adulto , Aminoglicósidos/administración & dosificación , Aminoglicósidos/uso terapéutico , Proteína C-Reactiva/metabolismo , Recuento de Células , Cefalosporinas/administración & dosificación , Cefalosporinas/uso terapéutico , Quimioterapia Combinada , Femenino , Fucosa/administración & dosificación , Galactosa/administración & dosificación , Expresión Génica/efectos de los fármacos , Humanos , Isotipos de Inmunoglobulinas/sangre , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/metabolismo , Pruebas de Función Hepática , Masculino , Neutrófilos/citología , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/aislamiento & purificación , Pruebas de Función Respiratoria , Esputo/citología , Esputo/metabolismo , Esputo/microbiología , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/genética
11.
Expert Opin Biol Ther ; 7(9): 1401-13, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17727329

RESUMEN

Recombinant monoclonal antibody (rMAb) therapy may be instituted to achieve one of two broad outcomes: i) killing of cells or organisms (e.g., cancer cells, bacteria); and ii) neutralisation of soluble molecules (e.g., cytokines in chronic disease or toxins in infection). The choice of rMAb isotype is a critical decision in the development of a therapeutic antibody as it will determine the biological activities triggered in vivo. It is not possible, however, to accurately predict the in vivo activity because multiple parameters impact on the functional outcome, for example, IgG subclass, IgG-Fc glycoform, epitope density, cellular Fc receptors polymorphisms and so on. The present understanding of the molecular interactions between IgG-Fc and effector ligands in vitro has allowed the generation of new antibody structures with altered/improved effector function profiles that may prove optimal for given disease indications. Thus, when maximal antibody-dependent cell-mediated cytotoxicity activity is indicated a non-fucosylated IgG1 format may be optimal; when minimal activity is indicated an aglycosylated IgG2 may be the form of choice.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos , Fucosa/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/inmunología , Isotipos de Inmunoglobulinas/inmunología , Receptores de IgG/fisiología , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Fucosa/metabolismo , Glicosilación , Humanos , Neoplasias/inmunología , Neoplasias/terapia
12.
Expert Opin Biol Ther ; 6(11): 1161-73, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17049014

RESUMEN

Most of the existing therapeutic antibodies that have been licensed and developed as medical agents are of the human IgG1 isotype, the molecular weight of which is approximately 150 kDa. Human IgG1 is a glycoprotein bearing two N-linked biantennary complex-type oligosaccharides bound to the antibody constant region (Fc), in which the majority of the oligosaccharides are core fucosylated, and it exercises the effector functions of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity through the interaction of the Fc with either leukocyte receptors (FcgammaRs) or complement. Recently, therapeutic antibodies have been shown to improve overall survival as well as time to disease progression in a variety of human malignancies, such as breast, colon and haematological cancers, and genetic analysis of FcgammaR polymorphisms of cancer patients has demonstrated that ADCC is a major antineoplasm mechanism responsible for clinical efficacy. However, the ADCC of existing licensed therapeutic antibodies has been found to be strongly inhibited by serum due to nonnpecific IgG competing for binding of the therapeutics to FcgammaRIIIa on natural killer cells, which leads to the requirement of a significant amount of drug and very high costs associated with such therapies. Moreover, enhanced ADCC of non-fucosylated forms of therapeutic antibodies through improved FcgammaRIIIa binding is shown to be inhibited by the fucosylated counterparts. In fact, non-fucosylated therapeutic antibodies, not including the fucosylated forms, exhibit the strongest and most saturable in vitro and ex vivo ADCC among such antibody variants with improved FcgammaRIIIa binding as those bearing naturally occurring oligosaccharide heterogeneities and artificial amino acid mutations, even in the presence of plasma IgG. Robust stable production of completely non-fucosylated therapeutic antibodies in a fixed quality has been achieved by the generation of a unique host cell line, in which the endogenous alpha-1,6-fucosyltransferase (FUT8) gene is knocked out. Thus, the application of non-fucosylated antibodies is expected to be a promising approach as next-generation therapeutic antibodies with improved efficacy, even when administrated at low doses in humans in vivo. Clinical trials using non-fucosylated antibody therapeutics are underway at present.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Citotoxicidad Celular Dependiente de Anticuerpos , Fucosa/uso terapéutico , Animales , Anticuerpos/genética , Anticuerpos/metabolismo , Anticuerpos/uso terapéutico , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Citotoxicidad Celular Dependiente de Anticuerpos/genética , Fucosa/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/terapia
13.
Ophthalmologica ; 219(6): 324-33, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16286790

RESUMEN

Corneal wound healing often leads to the development of scar tissue with loss of transparency. Reconstitution of transparent corneal stroma depends on the regulation of the biosynthetic activities of postlesional keratocytes and also to a large extent on the limitation of matrix degradation, attributed essentially to the upregulation of matrix metalloproteases and especially MMP-9. Using a standardized method for the production of reproducible corneal lesions by burning with iodine vapors, we could show that the local application of 0.5 mg/ml L-fucose reduced significantly MMP-9 upregulation and accelerated the recovery of the epithelial layer of the cornea. The iodine vapor used in the experiments produces a rapid loss of epithelium with no or slight effect below the basement membrane. A relatively rapid regrowth of epithelium was observed. The speed of this reepithelialization was stimulated by the local application of fucose. At 48 h after burn, there was a difference between fucose-treated and control corneas (epithelial thickness was about 50 mum for fucose-treated corneas and 37 microm for control corneas). Culture media of in vivo fucose-treated corneas showed an important decrease of MMP-9 activity (-51%, n = 6, p < 0.01). It appears that the in vivo fucose treatment reduced the MMP-9 activity released in the media. This effect is significant 24 h after iodine vapor burn. In order to study the effect of fucose on normal corneas, it was added to rabbit as well as human cornea explant cultures, and the production and release of MMP-9 was determined by zymography. Fucose at a concentration of 0.5 mg/ml produced a 70% decrease of MMP-9 activity released in the medium by corneal explant cultures. Other mono- and oligosaccharides were also tested. Besides lactose, fucose-rich oligosaccharides also produced significant inhibition. Galactose, melibiose, mannose and glucose were inactive. These results justify the use of fucose for the local treatment of corneal wounds.


Asunto(s)
Quemaduras Químicas/tratamiento farmacológico , Epitelio Corneal/efectos de los fármacos , Quemaduras Oculares/inducido químicamente , Fucosa/uso terapéutico , Metaloproteinasa 9 de la Matriz/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Animales , Quemaduras Químicas/enzimología , Quemaduras Químicas/patología , Regulación hacia Abajo , Epitelio Corneal/ultraestructura , Quemaduras Oculares/tratamiento farmacológico , Quemaduras Oculares/enzimología , Yodo/toxicidad , Técnicas de Cultivo de Órganos , Conejos
15.
Blood ; 101(5): 1705-12, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12406889

RESUMEN

Leukocyte adhesion deficiency type 2 (LADII) is characterized by defective selectin ligand formation, recurrent infection, and mental retardation. This rare syndrome has only been described in 2 kindreds of Middle Eastern descent who have differentially responded to exogenous fucose treatment. The molecular defect was recently ascribed to single and distinct missense mutations in a putative Golgi guanosine diphosphate (GDP)-fucose transporter. Here, we describe a patient of Brazilian origin with features of LADII. Sequencing of the GDP-fucose transporter revealed a novel single nucleotide deletion producing a shift in the open-reading frame and severe truncation of the polypeptide. Overexpression of the mutant protein in the patient's fibroblasts did not rescue fucosylation, suggesting that the deletion ablated the activity of the transporter. Administration of oral L-fucose to the patient produced molecular and clinical responses, as measured by the appearance of selectin ligands, normalization of neutrophil counts, and prevention of infectious recurrence. The lower neutrophil counts paralleled improved neutrophil interactions with activated endothelium in cremasteric venules of nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. However, fucose supplementation induced autoimmune neutropenia and the appearance of H antigen on erythrocytes, albeit without evidence of intravascular hemolysis. The robust response to fucose despite a severely truncated transporter suggests alternative means to transport GDP-fucose into the Golgi complex.


Asunto(s)
Proteínas Portadoras/genética , Fucosa/uso terapéutico , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Proteínas de Transporte de Monosacáridos , Polimorfismo de Nucleótido Simple , Anomalías Múltiples/genética , Animales , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/inducido químicamente , Proteínas Portadoras/fisiología , Células Cultivadas , Discapacidades del Desarrollo/genética , Selectina E/metabolismo , Eritrocitos/inmunología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fucosa/efectos adversos , Fucosa/farmacología , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Lactante , Recuento de Leucocitos , Rodamiento de Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/tratamiento farmacológico , Síndrome de Deficiencia de Adhesión del Leucocito/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neutropenia/inducido químicamente , Neutrófilos/inmunología , Selectina-P/metabolismo , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína
16.
Cells Tissues Organs ; 172(3): 161-73, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12476046

RESUMEN

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.


Asunto(s)
Fucosa/uso terapéutico , Síndrome de Deficiencia de Adhesión del Leucocito/genética , Síndrome de Deficiencia de Adhesión del Leucocito/terapia , Proteínas de Transporte de Monosacáridos , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Selectina E/metabolismo , Fucosa/administración & dosificación , Fucosa/metabolismo , Aparato de Golgi/metabolismo , Humanos , Lactante , Neutrófilos/citología , Neutrófilos/metabolismo , Selectina-P/metabolismo , Proteínas Recombinantes de Fusión
17.
Arterioscler Thromb Vasc Biol ; 22(10): 1604-9, 2002 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12377737

RESUMEN

OBJECTIVE: Smooth muscle cell (SMC) proliferation within the intima is regulated by heparan sulfates. We studied a low molecular weight (LMW) fucoidan (sulfated polysaccharide from brown seaweed) on SMC proliferation in vitro and intimal hyperplasia in vivo. METHODS AND RESULTS: In vitro study revealed that LMW fucoidan reduces rabbit SMC proliferation and is internalized in SMC perinuclear vesicles. On rabbit iliac arteries perfused in vivo with fluorolabeled LMW fucoidan after angioplasty, the labeling was mainly located on sites of injury. Pharmacokinetic studies showed that LMW fucoidan exhibited in rats an elimination half-life of 56+/-25 minutes (n=8) after intravenous administration and a constant plasma rate for > or =6 hours after intramuscular administration. After stent implantation in their iliac arteries, rabbits were also treated with LMW fucoidan (5 mg/kg IM twice a day). Histomorphometric analysis at day 14 indicated that LMW fucoidan reduced intimal hyperplasia by 59% (1.79+/-0.4 versus 0.73+/-0.2 mm2, P<0.0001) and luminal cross-sectional area narrowing by 58% (0.38+/-0.08 versus 0.16+/-0.04, P<0.0001). Blood samples showed no anticoagulant activity due to LMW fucoidan. CONCLUSIONS: This natural polysaccharide with high affinity for SMCs and sustained plasma concentration markedly reduced intimal hyperplasia, suggesting its use for the prevention of human in-stent restenosis.


Asunto(s)
Constricción Patológica/complicaciones , Hiperplasia/prevención & control , Arteria Ilíaca/patología , Polisacáridos/uso terapéutico , Stents/efectos adversos , Túnica Íntima/patología , Angioplastia de Balón/efectos adversos , Animales , División Celular , Células Cultivadas , Constricción Patológica/prevención & control , Modelos Animales de Enfermedad , Fucosa/farmacocinética , Fucosa/uso terapéutico , Arteria Ilíaca/lesiones , Arteria Ilíaca/metabolismo , Masculino , Peso Molecular , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/crecimiento & desarrollo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Polisacáridos/farmacocinética , Conejos , Recurrencia
19.
Ann Plast Surg ; 47(5): 540-6, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11716267

RESUMEN

The objective of this study was to examine whether a decrease in neutrophil-mediated tissue injury using Fucoidin, a nontoxic neutrophil rolling inhibitor, would improve flap survival in an island flap model after ischemia-reperfusion. Myeloperoxidase activity (an indirect index of tissue neutrophil count) and malondialdehyde (an indicator of lipid peroxidation), the degree of neutrophil infiltration by direct counting, and macroscopic flap survival were assessed in the flap after arterial ischemia-reperfusion. Epigastric island skin flaps were elevated in 56 rats. The first group of 21 rats was subjected to 6 hours of arterial ischemia. The second group of 21 rats was subjected to 10 hours of arterial ischemia, and the rest of the rats were used as nonischemic controls (sham flaps). For inhibiting neutrophil rolling, a nontoxic polysaccharide agent-Fucoidin-was used. Each ischemic group was divided further into three subgroups: Subgroup I (control rats) received saline, subgroup II received 10 mg per kilogram Fucoidin, and subgroup III received 25 mg per kilogram Fucoidin before reperfusion. The results were evaluated as tissue neutrophil counts, tissue malondialdehyde content, tissue myeloperoxidase activity, and flap survival. Neutrophil counts and tissue myeloperoxidase activity were decreased significantly (p <0.001) in subgroup III, but lipid peroxidation by means of tissue malondialdehyde content was not affected by Fucoidin administration. The authors conclude that administration of Fucoidin before reperfusion can limit tissue injury apparently by inhibiting neutrophil rolling in a dose-dependent manner.


Asunto(s)
Fucosa/uso terapéutico , Infiltración Neutrófila/efectos de los fármacos , Polisacáridos/uso terapéutico , Daño por Reperfusión/prevención & control , Colgajos Quirúrgicos/irrigación sanguínea , Animales , Supervivencia de Injerto , Malondialdehído/metabolismo , Neutrófilos/patología , Peroxidasa/metabolismo , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Colgajos Quirúrgicos/patología
20.
Klin Padiatr ; 213(5): 285-7, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11582527

RESUMEN

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.


Asunto(s)
Fucosa/uso terapéutico , Galactosa/uso terapéutico , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Terapia Respiratoria , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Neoplasias Abdominales/cirugía , Antibacterianos/uso terapéutico , Proteínas Bacterianas/metabolismo , Farmacorresistencia Microbiana , Fucosa/administración & dosificación , Fucosa/farmacología , Galactosa/administración & dosificación , Galactosa/farmacología , Humanos , Lactante , Lectinas/metabolismo , Masculino , Neuroblastoma/cirugía , Complicaciones Posoperatorias , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA