Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Infect Immun ; 86(8)2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866909

RESUMEN

Enterotoxigenic Escherichia coli (ETEC) causes diarrheal illness in infants in the developing world and travelers to countries where the disease is endemic, including military personnel. ETEC infection of the host involves colonization of the small intestinal epithelium and toxin secretion, leading to watery diarrhea. There is currently no vaccine licensed to prevent ETEC infection. CFA/I is one of the most common colonization factor antigens (CFAs). The CFA/I adhesin subunit, CfaE, is required for ETEC adhesion to host intestinal cells. Human antibodies against CfaE have the potential to block colonization of ETEC and serve as an immunoprophylactic against ETEC-related diarrhea. Mice transgenic for human immunoglobulin genes were immunized with CfaE to generate a panel of human monoclonal IgG1 antibodies (HuMAbs). The most potent IgG1 antibodies identified in the in vitro functional assays were selected and isotype switched to secretory IgA (sIgA) and tested in animal colonization assays via oral administration. Over 300 unique anti-CfaE IgG1 HuMAbs were identified. The lead IgG1 anti-CfaE HuMAbs completely inhibited hemagglutination and blocked adhesion of ETEC to Caco-2 cells. Epitope mapping studies revealed that HuMAbs recognized epitopes in the N-terminal domain of CfaE near the putative receptor binding site. Oral administration of anti-CfaE antibodies in either IgG or sIgA isotypes inhibited intestinal colonization in mice challenged with ETEC. A 2- to 4-log decrease in CFU was observed in comparison to mice challenged with irrelevant isotype controls. We identified fully human monoclonal antibodies against the CfaE adhesion domain that can be potentially employed as an immunoprophylactic to prevent ETEC-related diarrhea.


Asunto(s)
Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/genética , Escherichia coli Enterotoxigénica/genética , Escherichia coli Enterotoxigénica/inmunología , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/prevención & control , Vacunas contra Escherichia coli/inmunología , Animales , Humanos , Ratones
2.
Vet Pathol ; 55(2): 341-354, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29191134

RESUMEN

The pharmacology, pharmacokinetics, and safety of modified mRNA formulated in lipid nanoparticles (LNPs) were evaluated after repeat intravenous infusion to rats and monkeys. In both species, modified mRNA encoding the protein for human erythropoietin (hEPO) had predictable and consistent pharmacologic and toxicologic effects. Pharmacokinetic analysis conducted following the first dose showed that measured hEPO levels were maximal at 6 hours after the end of intravenous infusion and in excess of 100-fold the anticipated efficacious exposure (17.6 ng/ml) at the highest dose tested.24 hEPO was pharmacologically active in both the rat and the monkey, as indicated by a significant increase in red blood cell mass parameters. The primary safety-related findings were caused by the exaggerated pharmacology of hEPO and included increased hematopoiesis in the liver, spleen, and bone marrow (rats) and minimal hemorrhage in the heart (monkeys). Additional primary safety-related findings in the rat included mildly increased white blood cell counts, changes in the coagulation parameters at all doses, as well as liver injury and release of interferon γ-inducible protein 10 in high-dose groups only. In the monkey, as seen with the parenteral administration of cationic LNPs, splenic necrosis and lymphocyte depletion were observed, accompanied with mild and reversible complement activation. These findings defined a well-tolerated dose level above the anticipated efficacious dose. Overall, these combined studies indicate that LNP-formulated modified mRNA can be administered by intravenous infusion in 2 toxicologically relevant test species and generate supratherapeutic levels of protein (hEPO) in vivo.


Asunto(s)
Lípidos/efectos adversos , Nanopartículas/efectos adversos , ARN Mensajero/administración & dosificación , Animales , Coagulación Sanguínea/efectos de los fármacos , Eritropoyetina/genética , Femenino , Hematopoyesis/efectos de los fármacos , Infusiones Intravenosas/veterinaria , Recuento de Leucocitos/veterinaria , Macaca fascicularis , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
4.
Clin Cancer Res ; 26(23): 6284-6298, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32817076

RESUMEN

PURPOSE: While immune checkpoint inhibitors such as anti-PD-L1 are rapidly becoming the standard of care in the treatment of many cancers, only a subset of treated patients have long-term responses. IL12 promotes antitumor immunity in mouse models; however, systemic recombinant IL12 had significant toxicity and limited efficacy in early clinical trials. EXPERIMENTAL DESIGN: We therefore designed a novel intratumoral IL12 mRNA therapy to promote local IL12 tumor production while mitigating systemic effects. RESULTS: A single intratumoral dose of mouse (m)IL12 mRNA induced IFNγ and CD8+ T-cell-dependent tumor regression in multiple syngeneic mouse models, and animals with a complete response demonstrated immunity to rechallenge. Antitumor activity of mIL12 mRNA did not require NK and NKT cells. mIL12 mRNA antitumor activity correlated with TH1 tumor microenvironment (TME) transformation. In a PD-L1 blockade monotherapy-resistant model, antitumor immunity induced by mIL12 mRNA was enhanced by anti-PD-L1. mIL12 mRNA also drove regression of uninjected distal lesions, and anti-PD-L1 potentiated this response. Importantly, intratumoral delivery of mRNA encoding membrane-tethered mIL12 also drove rejection of uninjected lesions with very limited circulating IL12p70, supporting the hypothesis that local IL12 could induce a systemic antitumor immune response against distal lesions. Furthermore, in ex vivo patient tumor slice cultures, human IL12 mRNA (MEDI1191) induced dose-dependent IL12 production, downstream IFNγ expression and TH1 gene expression. CONCLUSIONS: These data demonstrate the potential for intratumorally delivered IL12 mRNA to promote TH1 TME transformation and robust antitumor immunity.See related commentary by Cirella et al., p. 6080.


Asunto(s)
Neoplasias Colorrectales/prevención & control , Interleucina-12/administración & dosificación , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/prevención & control , ARN Mensajero/administración & dosificación , Células TH1/inmunología , Microambiente Tumoral/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Apoptosis , Antígeno B7-H1/antagonistas & inhibidores , Linfocitos T CD8-positivos , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Resistencia a Antineoplásicos , Femenino , Humanos , Interleucina-12/genética , Melanoma/genética , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones SCID , ARN Mensajero/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Bacteriol ; 190(17): 5972-80, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18621903

RESUMEN

Gram-negative bacteria display either a flat or an irregular outer membrane. The periodontal pathogen Aggregatibacter (Actinobacillus) actinomycetemcomitans has an irregular outer membrane. We have identified a gene that is associated with the biogenesis of this morphology. The gene is part of a three-gene operon and codes for a 141-kDa protein designated morphogenesis protein C (MorC), which is conserved in several gram-negative bacteria including Haemophilus influenzae and Pasteurella multocida. Insertional inactivation of this gene resulted in the conversion of an irregularly shaped membrane to a flat membrane. Associated with this morphological change were the autoaggregation of the bacteria during planktonic growth and a concomitant increase in the surface hydrophobicity of the bacterium. The absence of MorC also resulted in the loss of the secretion of leukotoxin but not the ltxA transcription. Our findings suggest that MorC is critical for membrane morphology and leukotoxin secretion in A. actinomycetemcomitans.


Asunto(s)
Aggregatibacter actinomycetemcomitans/metabolismo , Proteínas Bacterianas/metabolismo , Exotoxinas/metabolismo , Proteínas de la Membrana/metabolismo , Aggregatibacter actinomycetemcomitans/genética , Aggregatibacter actinomycetemcomitans/ultraestructura , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Cromatografía Liquida , Electroforesis en Gel de Poliacrilamida , Interacciones Hidrofóbicas e Hidrofílicas , Immunoblotting , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Microscopía Electrónica de Transmisión , Datos de Secuencia Molecular , Mutagénesis Insercional , Operón/genética , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Espectrometría de Masa por Ionización de Electrospray
6.
Stem Cell Reports ; 9(3): 943-955, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28781076

RESUMEN

Organogenesis and tissue development occur through sequential stepwise processes leading to increased lineage restriction and loss of pluripotency. An exception to this appears in the adult human breast, where rare variant epithelial cells exhibit pluripotency and multilineage differentiation potential when removed from the signals of their native microenvironment. This phenomenon provides a unique opportunity to study mechanisms that lead to cellular reprogramming and lineage plasticity in real time. Here, we show that primary human mammary epithelial cells (HMECs) lose expression of differentiated mammary epithelial markers in a manner dependent on paracrine factors and epigenetic regulation. Furthermore, we demonstrate that HMEC reprogramming is dependent on gene silencing by the DNA methyltransferase DNMT3A and loss of histone transcriptional marks following downregulation of the methyltransferase DOT1L. These results demonstrate that lineage commitment in adult tissues is context dependent and highlight the plasticity of somatic cells when removed from their native tissue microenvironment.


Asunto(s)
Linaje de la Célula/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Epigénesis Genética , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/citología , Metiltransferasas/metabolismo , Diferenciación Celular/genética , Microambiente Celular , Reprogramación Celular/genética , ADN Metiltransferasa 3A , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Humanos , Metilación , Células del Estroma/citología
7.
Cell Cycle ; 15(5): 621-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26822887

RESUMEN

Over the past 20 years tremendous progress has been made in understanding the function of BRCA1 gene products. Yet one question still remains: why is mutation of BRCA1 typically associated with preferential development of breast and ovarian cancers and not tumors in other tissues? Here we discuss recent evidence documenting the effect of BRCA1-haploinsufficiency in different cells and tissues and synthesize a model for how mutations in a single BRCA1 allele in human cells might preferentially confer increased cancer risk in breast epithelial cells.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Animales , Neoplasias de la Mama/patología , Femenino , Genes BRCA1 , Haploinsuficiencia , Humanos , Glándulas Mamarias Humanas , Mutación , Especificidad de Órganos , Neoplasias Ováricas/genética
8.
PLoS One ; 10(3): e0121281, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793264

RESUMEN

Regulators of chromatin structure and gene expression contribute to tumor formation and progression. The co-repressor CoREST1 regulates the localization and activity of associated histone modifying enzymes including lysine specific demethylase 1 (LSD1) and histone deacetylase 1 (HDAC1). Although several CoREST1 associated proteins have been reported to enhance breast cancer progression, the role of CoREST1 in breast cancer is currently unclear. Here we report that knockdown of CoREST1 in the basal-type breast cancer cell line, MDA-MB-231, led to significantly reduced incidence and diminished size of tumors compared to controls in mouse xenograft studies. Notably, CoREST1-depleted cells gave rise to tumors with a marked decrease in angiogenesis. CoREST1 knockdown led to a decrease in secreted angiogenic and inflammatory factors, and mRNA analysis suggests that CoREST1 promotes expression of genes related to angiogenesis and inflammation including VEGF-A and CCL2. CoREST1 knockdown decreased the ability of MDA-MB-231 conditioned media to promote endothelial cell tube formation and migration. Further, tumors derived from CoREST1-depleted cells had reduced macrophage infiltration and the secretome of CoREST1 knockdown cells was deficient in promoting macrophage migration and macrophage-mediated angiogenesis. Taken together, these findings reveal that the epigenetic regulator CoREST1 promotes tumorigenesis in a breast cancer model at least in part through regulation of gene expression patterns in tumor cells that have profound non-cell autonomous effects on endothelial and inflammatory cells in the tumor microenvironment.


Asunto(s)
Carcinogénesis/patología , Comunicación Celular , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Represoras/metabolismo , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proteínas Co-Represoras , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Activación de Macrófagos , Macrófagos/metabolismo , Neoplasias Mamarias Animales/irrigación sanguínea , Neoplasias Mamarias Animales/genética , Ratones , Neovascularización Patológica/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Represoras/genética , Células del Estroma/metabolismo , Células del Estroma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Commun ; 6: 7505, 2015 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-26106036

RESUMEN

Although BRCA1 function is essential for maintaining genomic integrity in all cell types, it is unclear why increased risk of cancer in individuals harbouring deleterious mutations in BRCA1 is restricted to only a select few tissues. Here we show that human mammary epithelial cells (HMECs) from BRCA1-mutation carriers (BRCA1(mut/+)) exhibit increased genomic instability and rapid telomere erosion in the absence of tumour-suppressor loss. Furthermore, we uncover a novel form of haploinsufficiency-induced senescence (HIS) specific to epithelial cells, which is triggered by pRb pathway activation rather than p53 induction. HIS and telomere erosion in HMECs correlate with misregulation of SIRT1 leading to increased levels of acetylated pRb as well as acetylated H4K16 both globally and at telomeric regions. These results identify a novel form of cellular senescence and provide a potential molecular basis for the rapid cell- and tissue- specific predisposition of breast cancer development associated with BRCA1 haploinsufficiency.


Asunto(s)
Senescencia Celular/genética , Células Epiteliales/metabolismo , Genes BRCA1 , Inestabilidad Genómica/genética , Haploinsuficiencia , Glándulas Mamarias Humanas/metabolismo , Acortamiento del Telómero/genética , Daño del ADN , Células Epiteliales/citología , Heterocigoto , Humanos , Glándulas Mamarias Humanas/citología , Mutación , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
MAbs ; 6(6): 1533-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25484044

RESUMEN

A common challenge encountered during development of high concentration monoclonal antibody formulations is preventing self-association. Depending on the antibody and its formulation, self-association can be seen as aggregation, precipitation, opalescence or phase separation. Here we report on an unusual manifestation of self-association, formation of a semi-solid gel or "gelation." Therapeutic monoclonal antibody C4 was isolated from human B cells based on its strong potency in neutralizing bacterial toxin in animal models. The purified antibody possessed the unusual property of forming a firm, opaque white gel when it was formulated at concentrations >30 mg/mL and the temperature was <6°C. Gel formation was reversible with temperature. Gelation was affected by salt concentration or pH, suggesting an electrostatic interaction between IgG monomers. A comparison of the C4 amino acid sequences to consensus germline sequences revealed differences in framework regions. A C4 variant in which the framework sequence was restored to the consensus germline sequence did not gel at 100 mg/mL at temperatures as low as 1°C. Additional genetic analysis was used to predict the key residue(s) involved in the gelation. Strikingly, a single substitution in the native antibody, replacing heavy chain glutamate 23 with lysine (E23K), was sufficient to prevent gelation. These results indicate that the framework region is involved in intermolecular interactions. The temperature dependence of gelation may be related to conformational changes near glutamate 23 or the regions it interacts with. Molecular engineering of the framework can be an effective approach to resolve the solubility issues of therapeutic antibodies.


Asunto(s)
Sustitución de Aminoácidos/genética , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/genética , Ingeniería de Proteínas/métodos , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/genética , Anticuerpos Neutralizantes/uso terapéutico , Toxina Diftérica/antagonistas & inhibidores , Geles/química , Ácido Glutámico/genética , Humanos , Concentración de Iones de Hidrógeno , Cadenas Pesadas de Inmunoglobulina/química , Cadenas Pesadas de Inmunoglobulina/genética , Lisina/genética , Modelos Moleculares , Unión Proteica/genética , Estructura Terciaria de Proteína , Electricidad Estática , Temperatura
11.
Stem Cell Reports ; 2(5): 633-47, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24936451

RESUMEN

Perturbations in stem cell activity and differentiation can lead to developmental defects and cancer. We use an approach involving a quantitative model of cell-state transitions in vitro to gain insights into how SLUG/SNAI2, a key developmental transcription factor, modulates mammary epithelial stem cell activity and differentiation in vivo. In the absence of SLUG, stem cells fail to transition into basal progenitor cells, while existing basal progenitor cells undergo luminal differentiation; together, these changes result in abnormal mammary architecture and defects in tissue function. Furthermore, we show that in the absence of SLUG, mammary stem cell activity necessary for tissue regeneration and cancer initiation is lost. Mechanistically, SLUG regulates differentiation and cellular plasticity by recruiting the chromatin modifier lysine-specific demethylase 1 (LSD1) to promoters of lineage-specific genes to repress transcription. Together, these results demonstrate that SLUG plays a dual role in repressing luminal epithelial differentiation while unlocking stem cell transitions necessary for tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Línea Celular , Linaje de la Célula , Supervivencia sin Enfermedad , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neoplasias/metabolismo , Neoplasias/mortalidad , Neoplasias/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Regeneración , Factores de Transcripción de la Familia Snail , Trasplante de Células Madre , Células Madre/citología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Trasplante Heterólogo
12.
Cell Stem Cell ; 8(2): 149-63, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295272

RESUMEN

Women with inherited mutations in the BRCA1 gene have increased risk of developing breast cancer but also exhibit a predisposition for the development of aggressive basal-like breast tumors. We report here that breast epithelial cells derived from patients harboring deleterious mutations in BRCA1 (BRCA1(mut /+) give rise to tumors with increased basal differentiation relative to cells from BRCA1+/+ patients. Molecular analysis of disease-free breast tissues from BRCA1(mut /+) patients revealed defects in progenitor cell lineage commitment even before cancer incidence. Moreover, we discovered that the transcriptional repressor Slug is an important functional suppressor of human breast progenitor cell lineage commitment and differentiation and that it is aberrantly expressed in BRCA1(mut /+) tissues. Slug expression is necessary for increased basal-like phenotypes prior to and after neoplastic transformation. These findings demonstrate that the genetic background of patient populations, in addition to affecting incidence rates, significantly impacts progenitor cell fate commitment and, therefore, tumor phenotype.


Asunto(s)
Neoplasias de la Mama/metabolismo , Predisposición Genética a la Enfermedad/genética , Células Madre/citología , Adulto , Animales , Neoplasias de la Mama/genética , Femenino , Citometría de Flujo , Genes BRCA1 , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Persona de Mediana Edad , Datos de Secuencia Molecular , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA