Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 34(22): 2775-88, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26459514

RESUMEN

The intracellular chaperone heat-shock protein 70 (Hsp70) can be secreted from cells, but its extracellular role is unclear, as the protein has been reported to both activate and suppress the innate immune response. Potential immunomodulatory receptors on myelomonocytic lineage cells that bind extracellular Hsp70 are not well defined. Siglecs are Ig-superfamily lectins on mammalian leukocytes that recognize sialic acid-bearing glycans and thereby modulate immune responses. Siglec-5 and Siglec-14, expressed on monocytes and neutrophils, share identical ligand-binding domains but have opposing signaling functions. Based on phylogenetic analyses of these receptors, we predicted that endogenous sialic acid-independent ligands should exist. An unbiased screen revealed Hsp70 as a ligand for Siglec-5 and Siglec-14. Hsp70 stimulation through Siglec-5 delivers an anti-inflammatory signal, while stimulation through Siglec-14 is pro-inflammatory. The functional consequences of this interaction are also addressed in relation to a SIGLEC14 polymorphism found in humans. Our results demonstrate that an endogenous non-sialic acid-bearing molecule can be either a danger-associated or self-associated signal through paired Siglecs, and may explain seemingly contradictory prior reports on extracellular Hsp70 action.


Asunto(s)
Antígenos CD/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Proteínas HSP70 de Choque Térmico/inmunología , Lectinas/inmunología , Monocitos/inmunología , Receptores de Superficie Celular/inmunología , Transducción de Señal/inmunología , Antígenos CD/genética , Antígenos de Diferenciación Mielomonocítica/genética , Línea Celular Tumoral , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lectinas/genética , Monocitos/patología , Receptores de Superficie Celular/genética , Transducción de Señal/genética
2.
Proc Natl Acad Sci U S A ; 113(1): 74-9, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26621708

RESUMEN

The individuals of most vertebrate species die when they can no longer reproduce. Humans are a rare exception, having evolved a prolonged postreproductive lifespan. Elders contribute to cooperative offspring care, assist in foraging, and communicate important ecological and cultural knowledge, increasing the survival of younger individuals. Age-related deterioration of cognitive capacity in humans compromises these benefits and also burdens the group with socially costly members. We investigated the contribution of the immunoregulatory receptor CD33 to a uniquely human postreproductive disease, Alzheimer's dementia. Surprisingly, even though selection at advanced age is expected to be weak, a CD33 allele protective against Alzheimer's disease is derived and unique to humans and favors a functional molecular state of CD33 resembling that of the chimpanzee. Thus, derived alleles may be compensatory and restore interactions altered as a consequence of human-specific brain evolution. We found several other examples of derived alleles at other human loci that protect against age-related cognitive deterioration arising from neurodegenerative disease or cerebrovascular insufficiency. Selection by inclusive fitness may be strong enough to favor alleles protecting specifically against cognitive decline in postreproductive humans. Such selection would operate by maximizing the contributions of postreproductive individuals to the fitness of younger kin.


Asunto(s)
Enfermedad de Alzheimer/genética , Encéfalo/fisiopatología , Trastornos del Conocimiento/genética , Aptitud Genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/fisiología , Alelos , Empalme Alternativo , Animales , Apolipoproteínas E/genética , Evolución Biológica , Trastornos Cerebrovasculares/genética , Fertilidad/genética , Sitios Genéticos , Humanos , Pan troglodytes , Selección Genética , Lectina 3 Similar a Ig de Unión al Ácido Siálico/genética
3.
J Biol Chem ; 292(7): 2557-2570, 2017 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-28049733

RESUMEN

All vertebrate cell surfaces display a dense glycan layer often terminated with sialic acids, which have multiple functions due to their location and diverse modifications. The major sialic acids in most mammalian tissues are N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc), the latter being derived from Neu5Ac via addition of one oxygen atom at the sugar nucleotide level by CMP-Neu5Ac hydroxylase (Cmah). Contrasting with other organs that express various ratios of Neu5Ac and Neu5Gc depending on the variable expression of Cmah, Neu5Gc expression in the brain is extremely low in all vertebrates studied to date, suggesting that neural expression is detrimental to animals. However, physiological exploration of the reasons for this long term evolutionary selection has been lacking. To explore the consequences of forced expression of Neu5Gc in the brain, we have established brain-specific Cmah transgenic mice. Such Neu5Gc overexpression in the brain resulted in abnormal locomotor activity, impaired object recognition memory, and abnormal axon myelination. Brain-specific Cmah transgenic mice were also lethally sensitive to a Neu5Gc-preferring bacterial toxin, even though Neu5Gc was overexpressed only in the brain and other organs maintained endogenous Neu5Gc expression, as in wild-type mice. Therefore, the unusually strict evolutionary suppression of Neu5Gc expression in the vertebrate brain may be explained by evasion of negative effects on neural functions and by selection against pathogens.


Asunto(s)
Evolución Biológica , Encéfalo/metabolismo , Ácidos Neuramínicos/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Endotelio Vascular/metabolismo , Locomoción , Espectrometría de Masas , Trastornos de la Memoria/metabolismo , Ratones , Ratones Transgénicos
4.
Proc Natl Acad Sci U S A ; 112(2): 542-7, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548184

RESUMEN

A well known, epidemiologically reproducible risk factor for human carcinomas is the long-term consumption of "red meat" of mammalian origin. Although multiple theories have attempted to explain this human-specific association, none have been conclusively proven. We used an improved method to survey common foods for free and glycosidically bound forms of the nonhuman sialic acid N-glycolylneuraminic acid (Neu5Gc), showing that it is highly and selectively enriched in red meat. The bound form of Neu5Gc is bioavailable, undergoing metabolic incorporation into human tissues, despite being a foreign antigen. Interactions of this antigen with circulating anti-Neu5Gc antibodies could potentially incite inflammation. Indeed, when human-like Neu5Gc-deficient mice were fed bioavailable Neu5Gc and challenged with anti-Neu5Gc antibodies, they developed evidence of systemic inflammation. Such mice are already prone to develop occasional tumors of the liver, an organ that can incorporate dietary Neu5Gc. Neu5Gc-deficient mice immunized against Neu5Gc and fed bioavailable Neu5Gc developed a much higher incidence of hepatocellular carcinomas, with evidence of Neu5Gc accumulation. Taken together, our data provide an unusual mechanistic explanation for the epidemiological association between red meat consumption and carcinoma risk. This mechanism might also contribute to other chronic inflammatory processes epidemiologically associated with red meat consumption.


Asunto(s)
Inflamación/etiología , Neoplasias Hepáticas Experimentales/etiología , Carne/efectos adversos , Carne/análisis , Ácidos Neuramínicos/efectos adversos , Animales , Anticuerpos Bloqueadores/metabolismo , Progresión de la Enfermedad , Análisis de los Alimentos , Humanos , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Ácido N-Acetilneuramínico/análisis , Ácidos Neuramínicos/análisis , Ácidos Neuramínicos/inmunología , Factores de Riesgo
5.
Proc Natl Acad Sci U S A ; 111(16): 5998-6003, 2014 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-24711415

RESUMEN

Compelling evidence for naturally occurring immunosurveillance against malignancies informs and justifies some current approaches toward cancer immunotherapy. However, some types of immune reactions have also been shown to facilitate tumor progression. For example, our previous studies showed that although experimental tumor growth is enhanced by low levels of circulating antibodies directed against the nonhuman sialic acid N-glycolyl-neuraminic acid (Neu5Gc), which accumulates in human tumors, growth could be inhibited by anti-Neu5Gc antibodies from a different source, in a different model. However, it remains generally unclear whether the immune responses that mediate cancer immunosurveillance vs. those responsible for inflammatory facilitation are qualitatively and/or quantitatively distinct. Here, we address this question using multiple murine tumor growth models in which polyclonal antibodies against tumor antigens, such as Neu5Gc, can alter tumor progression. We found that although growth was stimulated at low antibody doses, it was inhibited by high doses, over a linear and remarkably narrow range, defining an immune response curve (IRC; i.e., inverse hormesis). Moreover, modulation of immune responses against the tumor by altering antibody avidity or by enhancing innate immunity shifted the IRC in the appropriate direction. Thus, the dualistic role of immunosurveillance vs. inflammation in modulating tumor progression can be quantitatively distinguished in multiple model systems, and can occur over a remarkably narrow range. Similar findings were made in a human tumor xenograft model using a narrow range of doses of a monoclonal antibody currently in clinical use. These findings may have implications for the etiology, prevention, and treatment of cancer.


Asunto(s)
Anticuerpos Antineoplásicos/inmunología , Hormesis/inmunología , Neoplasias/inmunología , Neoplasias/patología , Inmunidad Adaptativa/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Afinidad de Anticuerpos/inmunología , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunoglobulina G/inmunología , Inflamación/patología , Células Asesinas Naturales/inmunología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ácidos Neuramínicos/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(39): 14211-6, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25225409

RESUMEN

Certain pathogenic bacteria are known to modulate the innate immune response by decorating themselves with sialic acids, which can engage the myelomonocytic lineage inhibitory receptor Siglec-9, thereby evading immunosurveillance. We hypothesized that the well-known up-regulation of sialoglycoconjugates by tumors might similarly modulate interactions with innate immune cells. Supporting this hypothesis, Siglec-9-expressing myelomonocytic cells found in human tumor samples were accompanied by a strong up-regulation of Siglec-9 ligands. Blockade of Siglec-9 enhanced neutrophil activity against tumor cells in vitro. To investigate the function of inhibitory myelomonocytic Siglecs in vivo we studied mouse Siglec-E, the murine functional equivalent of Siglec-9. Siglec-E-deficient mice showed increased in vivo killing of tumor cells, and this effect was reversed by transgenic Siglec-9 expression in myelomonocytic cells. Siglec-E-deficient mice also showed enhanced immunosurveillance of autologous tumors. However, once tumors were established, they grew faster in Siglec-E-deficient mice. In keeping with this, Siglec-E-deficient macrophages showed a propensity toward a tumor-promoting M2 polarization, indicating a secondary role of CD33-related Siglecs in limiting cancer-promoting inflammation and tumor growth. Thus, we define a previously unidentified impact of inhibitory myelomonocytic Siglecs in cancer biology, with distinct roles that reflect the dual function of myelomonocytic cells in cancer progression. In keeping with this, a human polymorphism that reduced Siglec-9 binding to carcinomas was associated with improved early survival in non-small-cell lung cancer patients, which suggests that Siglec-9 might be therapeutically targeted within the right time frame and stage of disease.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos B/metabolismo , Inmunidad Innata , Neoplasias/inmunología , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/metabolismo , Animales , Antígenos CD/genética , Antígenos de Diferenciación de Linfocitos B/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Línea Celular Tumoral , Femenino , Humanos , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Monocitos/inmunología , Activación Neutrófila , Polimorfismo de Nucleótido Simple , Lectinas Similares a la Inmunoglobulina de Unión a Ácido Siálico/genética , Microambiente Tumoral/inmunología
7.
Am J Physiol Gastrointest Liver Physiol ; 310(5): G310-22, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26702135

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) and obesity are characterized by altered gut microbiota, inflammation, and gut barrier dysfunction. Here, we investigated the role of mucin-2 (Muc2) as the major component of the intestinal mucus layer in the development of fatty liver disease and obesity. We studied experimental fatty liver disease and obesity induced by feeding wild-type and Muc2-knockout mice a high-fat diet (HFD) for 16 wk. Muc2 deficiency protected mice from HFD-induced fatty liver disease and obesity. Compared with wild-type mice, after a 16-wk HFD, Muc2-knockout mice exhibited better glucose homeostasis, reduced inflammation, and upregulated expression of genes involved in lipolysis and fatty acid ß-oxidation in white adipose tissue. Compared with wild-type mice that were fed the HFD as well, Muc2-knockout mice also displayed higher intestinal and plasma levels of IL-22 and higher intestinal levels of the IL-22 target genes Reg3b and Reg3g. Our findings indicate that absence of the intestinal mucus layer activates the mucosal immune system. Higher IL-22 levels protect mice from diet-induced features of the metabolic syndrome.


Asunto(s)
Endotoxinas/inmunología , Microbioma Gastrointestinal , Inflamación , Interleucinas/metabolismo , Mucosa Intestinal , Mucina 2 , Enfermedad del Hígado Graso no Alcohólico , Obesidad , Tejido Adiposo/metabolismo , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Noqueados , Mucina 2/deficiencia , Mucina 2/metabolismo , Mucina 2/farmacología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/metabolismo , Obesidad/patología , Obesidad/prevención & control , Proteínas Asociadas a Pancreatitis , Sustancias Protectoras/metabolismo , Sustancias Protectoras/farmacología , Proteínas/metabolismo , Regeneración/inmunología , Interleucina-22
8.
Proc Natl Acad Sci U S A ; 110(50): 20218-23, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24284176

RESUMEN

The endocytic Ashwell-Morell receptor (AMR) of hepatocytes detects pathogen remodeling of host glycoproteins by neuraminidase in the bloodstream and mitigates the lethal coagulopathy of sepsis. We have investigated the mechanism of host protection by the AMR during the onset of sepsis and in response to the desialylation of blood glycoproteins by the NanA neuraminidase of Streptococcus pneumoniae. We find that the AMR selects among potential glycoprotein ligands unmasked by microbial neuraminidase activity in pneumococcal sepsis to eliminate from blood circulation host factors that contribute to coagulation and thrombosis. This protection is attributable in large part to the rapid induction of a moderate thrombocytopenia by the AMR. We further show that neuraminidase activity in the blood can be manipulated to induce the clearance of AMR ligands including platelets, thereby preactivating a protective response in pneumococcal sepsis that moderates the severity of disseminated intravascular coagulation and enables host survival.


Asunto(s)
Receptor de Asialoglicoproteína/inmunología , Hepatocitos/inmunología , Sepsis/prevención & control , Streptococcus pneumoniae/inmunología , Análisis de Varianza , Animales , Receptor de Asialoglicoproteína/metabolismo , Tiempo de Sangría , Plaquetas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neuraminidasa/administración & dosificación , Neuraminidasa/metabolismo , Sepsis/inmunología , Sepsis/microbiología
9.
Nature ; 456(7222): 648-52, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18971931

RESUMEN

AB(5) toxins comprise an A subunit that corrupts essential eukaryotic cell functions, and pentameric B subunits that direct target-cell uptake after binding surface glycans. Subtilase cytotoxin (SubAB) is an AB(5) toxin secreted by Shiga toxigenic Escherichia coli (STEC), which causes serious gastrointestinal disease in humans. SubAB causes haemolytic uraemic syndrome-like pathology in mice through SubA-mediated cleavage of BiP/GRP78, an essential endoplasmic reticulum chaperone. Here we show that SubB has a strong preference for glycans terminating in the sialic acid N-glycolylneuraminic acid (Neu5Gc), a monosaccharide not synthesized in humans. Structures of SubB-Neu5Gc complexes revealed the basis for this specificity, and mutagenesis of key SubB residues abrogated in vitro glycan recognition, cell binding and cytotoxicity. SubAB specificity for Neu5Gc was confirmed using mouse tissues with a human-like deficiency of Neu5Gc and human cell lines fed with Neu5Gc. Despite lack of Neu5Gc biosynthesis in humans, assimilation of dietary Neu5Gc creates high-affinity receptors on human gut epithelia and kidney vasculature. This, and the lack of Neu5Gc-containing body fluid competitors in humans, confers susceptibility to the gastrointestinal and systemic toxicities of SubAB. Ironically, foods rich in Neu5Gc are the most common source of STEC contamination. Thus a bacterial toxin's receptor is generated by metabolic incorporation of an exogenous factor derived from food.


Asunto(s)
Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/toxicidad , Ácidos Neuramínicos/metabolismo , Polisacáridos/química , Polisacáridos/metabolismo , Subtilisinas/química , Subtilisinas/toxicidad , Animales , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Muerte Celular/efectos de los fármacos , Línea Celular , Cristalografía por Rayos X , Chaperón BiP del Retículo Endoplásmico , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Modelos Moleculares , Ácidos Neuramínicos/administración & dosificación , Ácidos Neuramínicos/farmacología , Unión Proteica , Subunidades de Proteína , Escherichia coli Shiga-Toxigénica/química , Escherichia coli Shiga-Toxigénica/patogenicidad , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Especificidad de la Especie , Especificidad por Sustrato , Subtilisinas/genética , Subtilisinas/metabolismo , Análisis de Supervivencia
11.
J Biol Chem ; 287(34): 28852-64, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22692204

RESUMEN

Although N-acetyl groups are common in nature, N-glycolyl groups are rare. Mammals express two major sialic acids, N-acetylneuraminic acid and N-glycolylneuraminic acid (Neu5Gc). Although humans cannot produce Neu5Gc, it is detected in the epithelial lining of hollow organs, endothelial lining of the vasculature, fetal tissues, and carcinomas. This unexpected expression is hypothesized to result via metabolic incorporation of Neu5Gc from mammalian foods. This accumulation has relevance for diseases associated with such nutrients, via interaction with Neu5Gc-specific antibodies. Little is known about how ingested sialic acids in general and Neu5Gc in particular are metabolized in the gastrointestinal tract. We studied the gastrointestinal and systemic fate of Neu5Gc-containing glycoproteins (Neu5Gc-glycoproteins) or free Neu5Gc in the Neu5Gc-free Cmah(-/-) mouse model. Ingested free Neu5Gc showed rapid absorption into the circulation and urinary excretion. In contrast, ingestion of Neu5Gc-glycoproteins led to Neu5Gc incorporation into the small intestinal wall, appearance in circulation at a steady-state level for several hours, and metabolic incorporation into multiple peripheral tissue glycoproteins and glycolipids, thus conclusively proving that Neu5Gc can be metabolically incorporated from food. Feeding Neu5Gc-glycoproteins but not free Neu5Gc mimics the human condition, causing tissue incorporation into human-like sites in Cmah(-/-) fetal and adult tissues, as well as developing tumors. Thus, glycoproteins containing glycosidically linked Neu5Gc are the likely dietary source for human tissue accumulation, and not the free monosaccharide. This human-like model can be used to elucidate specific mechanisms of Neu5Gc delivery from the gut to tissues, as well as general mechanisms of metabolism of ingested sialic acids.


Asunto(s)
Antígenos Heterófilos/metabolismo , Autoantígenos/metabolismo , Tracto Gastrointestinal/metabolismo , Glicoproteínas/metabolismo , Productos de la Carne , Ácidos Neuramínicos/metabolismo , Animales , Glicoproteínas/genética , Humanos , Ratones , Ratones Noqueados , Especificidad de la Especie
12.
Virol J ; 10: 321, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24261589

RESUMEN

BACKGROUND: Influenza A virus (IAV) neuraminidase (NA) cleaves sialic acids (Sias) from glycans. Inhibiting NA with oseltamivir suppresses both viral infection, and viral release from cultured human airway epithelial cells. The role of NA in viral exit is well established: it releases budding virions by cleaving Sias from glycoconjugates on infected cells and progeny virions. The role of NA in viral entry remains unclear. Host respiratory epithelia secrete a mucus layer rich in heavily sialylated glycoproteins; these could inhibit viral entry by mimicking sialylated receptors on the cell surface. It has been suggested that NA allows influenza to penetrate the mucus by cleaving these sialylated decoys, but the exact mechanism is not yet established. METHODS: We tested IAV interaction with secreted mucus using frozen human trachea/bronchus tissue sections, and bead-bound purified human salivary mucins (HSM) and purified porcine submaxillary mucins (PSM). The protective effect of mucus was analyzed using MDCK cells coated with purified HSM and PSM with known Sia content. Oseltamivir was used to inhibit NA activity, and the fluorescent reporter substrate, 4MU-Neu5Ac, was used to quantify NA activity. RESULTS: IAV binds to the secreted mucus layer of frozen human trachea/bronchus tissues in a Sia dependent manner. HSM inhibition of IAV infection is Sia dose-dependent, but PSM cannot inhibit infection of underlying cells. HSM competitively inhibits NA cleavage of 4MU-Neu5Ac, reporter substrate. Human IAV effectively cleaves Sias from HSM but not from PSM, and binds to HSM but not to PSM. CONCLUSION: IAV interacts with human mucus on frozen tissue sections and mucus-coated beads. Inhibition of IAV infection by sialylated human mucus is dose-dependent, and enhanced when NA is inhibited with oseltamivir. Thus NA cleaves sialylated decoys during initial stages of infection. Understanding IAV interactions with host mucins is a promising new avenue for drug development.


Asunto(s)
Virus de la Influenza A/enzimología , Virus de la Influenza A/fisiología , Moco/metabolismo , Neuraminidasa/metabolismo , Ácidos Siálicos/metabolismo , Proteínas Virales/metabolismo , Internalización del Virus , Animales , Bronquios/química , Línea Celular , Perros , Humanos , Saliva/química , Tráquea/química
14.
Blood ; 114(25): 5225-35, 2009 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-19828701

RESUMEN

Humans are genetically unable to synthesize the common mammalian sialic acid N-glycolylneuraminic acid (Neu5Gc). However, Neu5Gc can be metabolically incorporated and covalently expressed on cultured human cell surfaces. Meanwhile, humans express varying and sometimes high titers of polyclonal anti-Neu5Gc antibodies. Here, a survey of human tissues by immunohistochemistry with both a monospecific chicken anti-Neu5Gc antibody and with affinity-purified human anti-Neu5Gc antibodies demonstrates endothelial expression of Neu5Gc, likely originating from Neu5Gc-rich foods like red meats. We hypothesized that the combination of Neu5Gc incorporation and anti-Neu5Gc antibodies can induce endothelial activation. Indeed, the incubation of high-titer human sera with Neu5Gc-fed endothelial cells led to Neu5Gc-dependent antibody binding, complement deposition, endothelial activation, selectin expression, increased cytokine secretion, and monocyte binding. The proinflammatory cytokine tumor necrosis factor-alpha also selectively enhanced human anti-Neu5Gc antibody reactivity. Anti-Neu5Gc antibodies affinity-purified from human serum also directed Neu5Gc-dependent complement deposition onto cultured endothelial cells. These data indicate a novel human-specific mechanism in which Neu5Gc-rich foods deliver immunogenic Neu5Gc to the endothelium, giving anti-Neu5Gc antibody- and complement-dependent activation, and potentially contributing to human vascular pathologies. In the case of atherosclerosis, Neu5Gc is present both in endothelium overlying plaques and in subendothelial regions, providing multiple pathways for accelerating inflammation in this disease.


Asunto(s)
Anticuerpos/inmunología , Endotelio Vascular/inmunología , Ácidos Siálicos/inmunología , Animales , Anticuerpos/sangre , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Unión Competitiva/efectos de los fármacos , Vasos Sanguíneos/metabolismo , Línea Celular , Células Cultivadas , Pollos , Colon/irrigación sanguínea , Endotelio Vascular/metabolismo , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Masculino , Placenta/irrigación sanguínea , Embarazo , Ácidos Siálicos/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
15.
Proc Natl Acad Sci U S A ; 105(48): 18936-41, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19017806

RESUMEN

Patients with cancer have circulating heterophile antibodies that agglutinate animal red cells via recognition of the mammalian cell surface sialic acid N-glycolylneuraminic acid (Neu5Gc), which was long considered an oncofetal antigen in humans. However, humans are genetically deficient in Neu5Gc production and instead metabolically accumulate Neu5Gc from dietary sources, particularly red meats and milk products. Moreover, mice with a human-like defect showed no alternate pathway for Neu5Gc synthesis and even normal humans express anti-Neu5Gc antibodies. We show here that human tumors accumulate Neu5Gc that is covalently attached to multiple classes of glycans. The paradox of human tumor Neu5Gc accumulation in the face of circulating anti-Neu5Gc antibodies was hypothesized to be due to facilitation of tumor progression by the resulting low-grade chronic inflammation. Indeed, murine tumors expressing human-like levels of Neu5Gc show accelerated growth in syngeneic mice with a human-like Neu5Gc deficiency, coincident with the induction of anti-Neu5Gc antibodies and increased infiltration of inflammatory cells. Transfer of polyclonal monospecific syngeneic mouse anti-Neu5Gc serum also enhanced growth of transplanted syngeneic tumors bearing human-like levels of Neu5Gc, with tumors showing evidence for antibody deposition, enhanced angiogenesis and chronic inflammation. These effects were suppressed by a cyclooxygenase-2 inhibitor, a drug type known to reduce human carcinoma risk. Finally, affinity-purified human anti-Neu5Gc antibodies also accelerate growth of Neu5Gc-containing tumors in Neu5Gc-deficient mice. Taken together, the data suggest that the human propensity to develop diet-related carcinomas is contributed to by local chronic inflammation, resulting from interaction of metabolically-accumulated dietary Neu5Gc with circulating anti-Neu5Gc antibodies.


Asunto(s)
Anticuerpos Heterófilos/sangre , Carcinoma , Dieta , Inflamación/inmunología , Ácidos Neuramínicos/inmunología , Animales , Anticuerpos Heterófilos/inmunología , Carcinoma/sangre , Carcinoma/inmunología , Carcinoma/patología , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Humanos , Inflamación/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ácidos Neuramínicos/química , Factores de Riesgo
16.
Kidney Int ; 76(2): 140-4, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19387473

RESUMEN

Hemolytic-uremic syndrome (HUS) is a systemic disease characterized by microvascular endothelial damage, mainly in the gastrointestinal tract and the kidneys. A major cause of HUS is Shiga toxigenic Escherichia coli (STEC) infection. In addition to Shiga toxin, additional STEC virulence factors may contribute to HUS. One is the newly discovered subtilase cytotoxin (SubAB), which is highly toxic to eukaryotic cells, and when injected intraperitoneally into mice causes pathology resembling that associated with human HUS. Recent data show that SubAB exhibits a strong preference for glycans terminating in alpha2-3-linked N-glycolylneuraminic acid (Neu5Gc), a sialic acid that humans are unable to synthesize, because we genetically lack the necessary enzyme. However, Neu5Gc can still be found on human cells due to metabolic incorporation from the diet. Dietary incorporation happens to be highest in human endothelium and to a lesser extent in the intestinal epithelium, the two affected cell types in STEC-induced HUS. Mammalian-derived foods such as red meat and dairy products appear to be the primary source of dietary Neu5Gc. Ironically, these are also common sources of STEC contamination. Taken together, these findings suggest a 'two-hit' process in the pathogenesis of human SubAB-induced disease. First, humans eat Neu5Gc-rich food, leading to incorporation of Neu5Gc on the surfaces of endothelial and intestinal cells. Second, when exposed to a SubAB-producing STEC strain, the toxin produced would be able to bind to the intestinal epithelial cells, perhaps causing acute gastrointestinal symptoms, and eventually damaging endothelial cells in other organs like the kidney, thereby causing HUS.


Asunto(s)
Dieta/efectos adversos , Proteínas de Escherichia coli/metabolismo , Síndrome Hemolítico-Urémico/etiología , Ácidos Neuramínicos/efectos adversos , Subtilisinas/metabolismo , Animales , Proteínas de Escherichia coli/toxicidad , Síndrome Hemolítico-Urémico/inducido químicamente , Síndrome Hemolítico-Urémico/microbiología , Síndrome Hemolítico-Urémico/patología , Humanos , Carne/efectos adversos , Carne/microbiología , Ácidos Neuramínicos/farmacocinética , Escherichia coli Shiga-Toxigénica/patogenicidad , Subtilisinas/toxicidad
17.
Cancer Cell ; 35(3): 504-518.e7, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30827889

RESUMEN

Ionizing radiation (IR) and chemotherapy are standard-of-care treatments for glioblastoma (GBM) patients and both result in DNA damage, however, the clinical efficacy is limited due to therapeutic resistance. We identified a mechanism of such resistance mediated by phosphorylation of PTEN on tyrosine 240 (pY240-PTEN) by FGFR2. pY240-PTEN is rapidly elevated and bound to chromatin through interaction with Ki-67 in response to IR treatment and facilitates the recruitment of RAD51 to promote DNA repair. Blocking Y240 phosphorylation confers radiation sensitivity to tumors and extends survival in GBM preclinical models. Y240F-Pten knockin mice showed radiation sensitivity. These results suggest that FGFR-mediated pY240-PTEN is a key mechanism of radiation resistance and is an actionable target for improving radiotherapy efficacy.


Asunto(s)
Neoplasias Encefálicas/terapia , Núcleo Celular/metabolismo , Glioma/terapia , Fosfohidrolasa PTEN/metabolismo , Pirimidinas/administración & dosificación , Tolerancia a Radiación/efectos de los fármacos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Animales , Neoplasias Encefálicas/metabolismo , Reparación del ADN/efectos de los fármacos , Femenino , Glioma/metabolismo , Humanos , Masculino , Ratones , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Recombinasa Rad51/metabolismo , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 66(3): 1536-42, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452210

RESUMEN

Hematogenous carcinoma metastasis is supported by aggregated platelets and leukocytes, forming tumor cell emboli. Early tumor cell-platelet interactions can be mediated by P-selectin binding to tumor cell surface ligands and this process is blocked by heparin. We previously showed that L-selectin deficiency also attenuates experimental metastasis. However, the mechanisms and timing of L-selectin action remained unknown. Here, we study how L-selectin facilitates establishment of pulmonary metastatic foci in syngeneic mice by using experimental metastasis to time events following entry of tumor cells into the bloodstream. Although L-selectin deficiency did not affect platelet aggregation or initial tumor cell embolization, the association of leukocytes with tumor cells was reduced and tumor cell survival was diminished 24 hours later. Temporal inhibition of L-selectin by a function-blocking antibody reduced metastasis. Moreover, although selectin blockade by heparin 6 to 18 hours after tumor cell injection was synergistic with P-selectin deficiency in reducing metastasis, there was no further effect in L-selectin-deficient animals. Thus, heparin apparently works at these time points primarily by blocking L-selectin. Endogenous L-selectin ligands were concomitantly induced adjacent to established intravascular tumor cell emboli in a similar time window when leukocytes were also present. Metastasis was attenuated in mice missing these induced endogenous L-selectin ligands due to fucosyltransferase-7 deficiency. Thus, L-selectin facilitation of metastasis progression involves leukocyte-endothelial interactions at sites of intravascular arrest supported by local induction of L-selectin ligands via fucosyltransferase-7. These data provide the first explanation for how leukocyte L-selectin facilitates tumor metastasis.


Asunto(s)
Neoplasias del Colon/patología , Fucosiltransferasas/metabolismo , Selectina L/fisiología , Neoplasias Pulmonares/secundario , Animales , Neoplasias del Colon/enzimología , Fucosiltransferasas/deficiencia , Heparina/farmacología , Selectina L/inmunología , Selectina L/metabolismo , Ligandos , Neoplasias Pulmonares/enzimología , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba
20.
Cell Rep ; 21(7): 1870-1882, 2017 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-29141219

RESUMEN

Liver cancer has become the second most deadly malignant disease, with no efficient targeted or immune therapeutic agents available yet. While dissecting the roles of cytoplasmic signaling molecules in hepatocarcinogenesis using an inducible mouse gene targeting system, Mx1-cre, we identified a potent liver tumor-inhibitory effect of synthetic double-stranded RNA (dsRNA), polyinosinic-polycytidylic acid (pIC), an inducer of the Mx1-cre system. Injection of pIC at the pre-cancer stage robustly suppressed liver tumorigenesis either induced by chemical carcinogens or by Pten loss and associated hepatosteatosis. The immunostimulatory dsRNA inhibited liver cancer initiation, apparently by boosting multiple anti-tumor activities of innate immunity, including induction of immunoregulatory cytokines, activation of NK cells and dendritic cells, and reprogramming of macrophage polarization. This study paves the way for the development of preventive and early interfering strategies for liver cancer to reduce the rapidly increasing incidences of liver cancer in an ever-growing population with chronic liver disorders.


Asunto(s)
Carcinogénesis/inmunología , Inmunidad Innata , Inductores de Interferón/farmacología , Neoplasias Hepáticas/inmunología , Poli I-C/farmacología , Animales , Carcinogénesis/efectos de los fármacos , Células Dendríticas/inmunología , Inductores de Interferón/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/prevención & control , Activación de Macrófagos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Poli I-C/uso terapéutico , ARN Bicatenario/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA