Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 19(1): 76-84, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29180808

RESUMEN

Exciting progress in the field of cancer immunotherapy has renewed the urgency of the need for basic studies of immunoregulation in both adaptive cell lineages and innate cell lineages. Here we found a central role for major histocompatibility complex (MHC) class I in controlling the phagocytic function of macrophages. Our results demonstrated that expression of the common MHC class I component ß2-microglobulin (ß2M) by cancer cells directly protected them from phagocytosis. We further showed that this protection was mediated by the inhibitory receptor LILRB1, whose expression was upregulated on the surface of macrophages, including tumor-associated macrophages. Disruption of either MHC class I or LILRB1 potentiated phagocytosis of tumor cells both in vitro and in vivo, which defines the MHC class I-LILRB1 signaling axis as an important regulator of the effector function of innate immune cells, a potential biomarker for therapeutic response to agents directed against the signal-regulatory protein CD47 and a potential target of anti-cancer immunotherapy.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/inmunología , Receptor Leucocitario Tipo Inmunoglobulina B1/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Fagocitosis/inmunología , Animales , Línea Celular Tumoral , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inmunoterapia/métodos , Receptor Leucocitario Tipo Inmunoglobulina B1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/terapia , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia
2.
Nat Commun ; 8: 14802, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28378740

RESUMEN

CD47 is a cell surface molecule that inhibits phagocytosis of cells that express it by binding to its receptor, SIRPα, on macrophages and other immune cells. CD47 is expressed at different levels by neoplastic and normal cells. Here, to reveal mechanisms by which different neoplastic cells generate this dominant 'don't eat me' signal, we analyse the CD47 regulatory genomic landscape. We identify two distinct super-enhancers (SEs) associated with CD47 in certain cancer cell types. We show that a set of active constituent enhancers, located within the two CD47 SEs, regulate CD47 expression in different cancer cell types and that disruption of CD47 SEs reduces CD47 gene expression. Finally we report that the TNF-NFKB1 signalling pathway directly regulates CD47 by interacting with a constituent enhancer located within a CD47-associated SE specific to breast cancer. These results suggest that cancers can evolve SE to drive CD47 overexpression to escape immune surveillance.


Asunto(s)
Neoplasias de la Mama/metabolismo , Antígeno CD47/fisiología , Elementos de Facilitación Genéticos , Mediadores de Inflamación/metabolismo , Inflamación/metabolismo , Transducción de Señal , Regulación hacia Arriba , Animales , Neoplasias de la Mama/patología , Antígeno CD47/genética , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Subunidad p50 de NF-kappa B/metabolismo , Fagocitosis , Unión Proteica , Factor de Necrosis Tumoral alfa/metabolismo
3.
PLoS Negl Trop Dis ; 7(6): e2263, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23818995

RESUMEN

Ascaris suum and Ascaris lumbricoides are two closely related geo-helminth parasites that ubiquitously infect pigs and humans, respectively. Ascaris suum infection in pigs is considered a good model for A. lumbricoides infection in humans because of a similar biology and tissue migration to the intestines. Ascaris lumbricoides infections in children are associated with malnutrition, growth and cognitive stunting, immune defects, and, in extreme cases, life-threatening blockage of the digestive tract and aberrant migration into the bile duct and peritoneum. Similar effects can be seen with A. suum infections in pigs related to poor feed efficiency and performance. New strategies to control Ascaris infections are needed largely due to reduced treatment efficacies of current anthelmintics in the field, the threat of resistance development, and the general lack of new drug development for intestinal soil-transmitted helminths for humans and animals. Here we demonstrate for the first time that A. suum expresses the receptors for Bacillus thuringiensis crystal protein and novel anthelmintic Cry5B, which has been previously shown to intoxicate hookworms and which belongs to a class of proteins considered non-toxic to vertebrates. Cry5B is able to intoxicate A. suum larvae and adults and triggers the activation of the p38 mitogen-activated protein kinase pathway similar to that observed with other nematodes. Most importantly, two moderate doses of 20 mg/kg body weight (143 nM/kg) of Cry5B resulted in a near complete cure of intestinal A. suum infections in pigs. Taken together, these results demonstrate the excellent potential of Cry5B to treat Ascaris infections in pigs and in humans and for Cry5B to work effectively in the human gastrointestinal tract.


Asunto(s)
Antihelmínticos/uso terapéutico , Ascariasis/veterinaria , Ascaris suum/efectos de los fármacos , Ascaris suum/fisiología , Bacillus thuringiensis/metabolismo , Proteínas Bacterianas/uso terapéutico , Endotoxinas/uso terapéutico , Proteínas Hemolisinas/uso terapéutico , Enfermedades de los Porcinos/tratamiento farmacológico , Animales , Antihelmínticos/aislamiento & purificación , Antihelmínticos/farmacología , Ascariasis/tratamiento farmacológico , Ascariasis/parasitología , Toxinas de Bacillus thuringiensis , Proteínas Bacterianas/aislamiento & purificación , Proteínas Bacterianas/farmacología , Endotoxinas/aislamiento & purificación , Endotoxinas/farmacología , Femenino , Proteínas Hemolisinas/aislamiento & purificación , Proteínas Hemolisinas/farmacología , Larva/efectos de los fármacos , Larva/fisiología , Porcinos , Enfermedades de los Porcinos/parasitología , Resultado del Tratamiento
4.
PLoS One ; 8(7): e70702, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23869246

RESUMEN

Soil-transmitted helminths are parasitic nematodes that inhabit the human intestine. These parasites, which include two hookworm species, Ancylostomaduodenale and Necator americanus, the whipworm Trichuristrichiura, and the large roundworm Ascarislumbricoides, infect upwards of two billion people and are a major cause of disease burden in children and pregnant women. The challenge with treating these diseases is that poverty, safety, and inefficient public health policy have marginalized drug development and distribution to control infection in humans. Anthelmintics (anti-worm drugs) have historically been developed and tested for treatment of non-human parasitic nematodes that infect livestock and companion animals. Here we systematically compare the in vitro efficacy of all major anthelmintic classes currently used in human therapy (benzimidazoles, nicotinic acetylcholine receptor agonists, macrocyclic lactones, nitazoxanide) against species closely related to human parasitic nematodes-Ancylostoma ceylanicum, Trichurismuris, and Ascarissuum--- as well as a rodent parasitic nematode used in veterinary drug discovery, Heligmosomoidesbakeri, and the free-living nematode Caenorhabditis elegans. Extensive in vitro data is complemented with single-dose in vivo data in three rodent models of parasitic diseases. We find that the effects of the drugs in vitro and in vivo can vary greatly among these nematode species, e.g., the efficacy of albendazole is strong on A. ceylanicum but weak on H. bakeri. Nonetheless, certain commonalities of the in vitro effects of the drugs can be seen, e.g., nitazoxanide consistently shows an all-or-nothing response. Our in vitro data suggest that further optimization of the clinical efficacy of some of these anthelmintics could be achieved by altering the treatment routine and/or dosing. Most importantly, our in vitro and in vivo data indicate that the hookworm A. ceylanicum is a particularly sensitive and useful model for anthelmintic studies and should be incorporated early on in drug screens for broad-spectrum human soil-transmitted helminth therapies.


Asunto(s)
Antihelmínticos/farmacología , Nematodos/efectos de los fármacos , Albendazol/farmacología , Albendazol/uso terapéutico , Animales , Antihelmínticos/uso terapéutico , Cricetinae , Resistencia a Medicamentos , Femenino , Ivermectina/farmacología , Ivermectina/uso terapéutico , Masculino , Ratones , Infecciones por Nematodos/tratamiento farmacológico , Nitrocompuestos , Pruebas de Sensibilidad Parasitaria , Pirantel/farmacología , Pirantel/uso terapéutico , Especificidad de la Especie , Tiazoles/farmacología , Tiazoles/uso terapéutico
5.
PLoS Negl Trop Dis ; 6(11): e1900, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23145203

RESUMEN

BACKGROUND: Hookworm infections are one of the most important parasitic infections of humans worldwide, considered by some second only to malaria in associated disease burden. Single-dose mass drug administration for soil-transmitted helminths, including hookworms, relies primarily on albendazole, which has variable efficacy. New and better hookworm therapies are urgently needed. Bacillus thuringiensis crystal protein Cry5B has potential as a novel anthelmintic and has been extensively studied in the roundworm Caenorhabditis elegans. Here, we ask whether single-dose Cry5B can provide therapy against a hookworm infection and whether C. elegans mechanism-of-action studies are relevant to hookworms. METHODOLOGY/PRINCIPAL FINDINGS: To test whether the C. elegans invertebrate-specific glycolipid receptor for Cry5B is relevant in hookworms, we fed Ancylostoma ceylanicum hookworm adults Cry5B with and without galactose, an inhibitor of Cry5B-C. elegans glycolipid interactions. As with C. elegans, galactose inhibits Cry5B toxicity in A. ceylanicum. Furthermore, p38 mitogen-activated protein kinase (MAPK), which controls one of the most important Cry5B signal transduction responses in C. elegans, is functionally operational in hookworms. A. ceylanicum hookworms treated with Cry5B up-regulate p38 MAPK and knock down of p38 MAPK activity in hookworms results in hypersensitivity of A. ceylanicum adults to Cry5B attack. Single-dose Cry5B is able to reduce by >90% A. ceylanicum hookworm burdens from infected hamsters, in the process eliminating hookworm egg shedding in feces and protecting infected hamsters from blood loss. Anthelmintic activity is increased about 3-fold, eliminating >97% of the parasites with a single 3 mg dose (∼30 mg/kg), by incorporating a simple formulation to help prevent digestion in the acidic stomach of the host mammal. CONCLUSIONS/SIGNIFICANCE: These studies advance the development of Cry5B protein as a potent, safe single-dose anthelmintic for hookworm therapy and make available the information of how Cry5B functions in C. elegans in order to study and improve Cry5B function against hookworms.


Asunto(s)
Ancylostoma/efectos de los fármacos , Anquilostomiasis/tratamiento farmacológico , Antihelmínticos/administración & dosificación , Proteínas Bacterianas/administración & dosificación , Endotoxinas/administración & dosificación , Proteínas Hemolisinas/administración & dosificación , Animales , Toxinas de Bacillus thuringiensis , Cricetinae , Modelos Animales de Enfermedad , Heces/parasitología , Femenino , Masculino , Mesocricetus , Recuento de Huevos de Parásitos , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...