Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biotechnol Bioeng ; 117(11): 3277-3285, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32648943

RESUMEN

Vaccines provide effective protection against many infectious diseases as well as therapeutics for select pathologies, such as cancer. Many viral vaccines require amplification of virus in cell cultures during manufacture. Traditionally, cell cultures, such as VERO, have been used for virus production in bovine serum-containing culture media. However, due to concerns of potential adventitious agents present in fetal bovine serum (FBS), regulatory agencies suggest avoiding the use of bovine serum in vaccine production. Current serum-free media suitable for VERO-based virus production contains high concentrations of undefined plant hydrolysates. Although these media have been extensively used, the lack of chemical definition has the potential to adversely affect cell growth kinetics and subsequent virus production. As plant hydrolysates are made from plant raw materials, performance variations could be significant among different lots of production. We developed a chemically defined, serum-free medium, OptiVERO, which was optimized specifically for VERO cells. VERO cell growth kinetics were demonstrated to be equivalent to EMEM-10% FBS in this chemically defined medium while the plant hydrolysate-containing medium demonstrated a slower doubling time in both two-dimensional (2D) and 3D cultures. Virus production comparisons demonstrated that the chemically defined OptiVERO medium performed at least as good as the EMEM-10%FBS and better than the plant hydrolysate-containing media. We report the success in using recombinant proteins to replace undefined plant hydrolysates to formulate a chemically defined medium that can efficiently support VERO cell expansion and virus production.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medio de Cultivo Libre de Suero , Células Vero , Cultivo de Virus/métodos , Animales , Chlorocebus aethiops , Medio de Cultivo Libre de Suero/química , Medio de Cultivo Libre de Suero/metabolismo , Preparaciones de Plantas , Proteínas Recombinantes , Células Vero/citología , Células Vero/metabolismo , Ensayo de Placa Viral
2.
J Crohns Colitis ; 11(9): 1101-1112, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28472424

RESUMEN

BACKGROUND AND AIMS: Inflammatory bowel disease [IBD] is characterised by a disruption of immune homeostasis, which is tightly regulated to protect against harmful pathogens yet not react to commensal antigens. Animal studies indicate that regulatory T cells [Treg] modulate the immune response to prevent IBD development. Lactoferrin [LF] is an endogenous anti-inflammatory pleiotropic protein secreted at high concentrations in colostrum and at mucosal sites. However, the effect of LF on specific T lymphocyte populations has not been studied. Here, we identify a novel mechanism by which a recombinant human LF, VEN-120, regulates T cell populations in health and disease. METHODS: Two murine models of intestinal inflammation, the dextran sodium sulphate colitis model and the TNFΔARE/+ model of ileitis, were used to study the anti-inflammatory and T cell modulating ability of VEN-120. Flow cytometry was used to evaluate T cell populations within the lamina propria and mesenteric lymph nodes, and to evaluate the effect of VEN-120 on CD4+ T cells in vitro. RESULTS: VEN-120 reduced inflammation in both models of IBD, accompanied by increased Tregs in the intestinal lamina propria. Treatment of CD4+ T cells in vitro resulted in an upregulation of Treg genes and skewing towards a Treg population. This in vitro T cell skewing translated to an increase of Treg homing to the intestinal lamina propria and associated lymph tissue in healthy mice. CONCLUSIONS: These data provide a novel immunological mechanism by which VEN-120 modulates T cells to restrict inflammatory T cell-driven disease.


Asunto(s)
Colitis/inmunología , Ileítis/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Lactoferrina/inmunología , Fenotipo , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/inmunología
3.
Bioengineered ; 5(3): 180-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24776984

RESUMEN

Stem cell-based therapy has the potential to treat an array of human diseases. However, to study the therapeutic potential and safety of these cells, a scalable cell culture medium is needed that is free of human or bovine-derived serum proteins. Thus, cost-effective recombinant serum proteins and cytokines are needed to produce such mediums. One such cytokine, leukemia inhibitory factor (LIF), has been shown to be a critical paracrine factor that maintains stem cell pluripotency in murine embryonic stem cells and human naïve stem cells while simultaneously inhibiting differentiation. We recently produced recombinant human LIF (rhLIF) in a rice-based protein expression system known as ExpressTec. (12) We described expression of rice-derived rhLIF and demonstrated its biological equivalency to E. coli-derived rhLIF in traditional and embryonic mouse stem cell systems. Here we describe the expression yield of rice-derived rhLIF and the scale up production capacity. We provide further evidence of the efficacy of rice-derived rhLIF in additional stem cell systems including human neural stem cells and mouse induced pluripotent stem (iPS) cells. The expression level, biological activity, and potential for production at commercial scale of rice-derived rhLIF provides a proof-of-principal for ExpressTec-derived proteins to produce regulatory-friendly, high performance, and dependable stem cell media.


Asunto(s)
Células Madre Pluripotentes Inducidas/efectos de los fármacos , Factor Inhibidor de Leucemia/farmacología , Células-Madre Neurales/efectos de los fármacos , Oryza/genética , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de los fármacos , Codón , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Glicosilación , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Factor Inhibidor de Leucemia/biosíntesis , Factor Inhibidor de Leucemia/genética , Ratones , Proteína Homeótica Nanog , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oryza/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología
4.
J Biotechnol ; 172: 67-72, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24380819

RESUMEN

Embryonic and induced pluripotent stem cells have the ability to differentiate into any somatic cell type, and thus have potential to treat a number of diseases that are currently incurable. Application of these cells for clinical or industrial uses would require an increase in production to yield adequate numbers of viable cells. However, the relatively high costs of cytokines and growth factors required for maintenance of stem cells in the undifferentiated state have the potential to limit translational research. Leukemia inhibitory factor (LIF), a member of the IL-6 cytokine family, is a key regulator in the maintenance of naïve states for both human and mouse stem cells. In this study, we describe a new recombinant human LIF (rhLIF) using a plant-based (rice) expression system. We found that rice-derived rhLIF possessed the same specific activity as commercial Escherichia coli-derived LIF and was capable of supporting mouse embryonic stem cell proliferation in the undifferentiated state as evidenced from pluripotency marker level analysis. Retention of the pluripotent state was found to be indistinguishable between rice-derived rhLIF and other recombinant LIF proteins currently on the market.


Asunto(s)
Células Madre Embrionarias/inmunología , Factor Inhibidor de Leucemia/metabolismo , Oryza/genética , Proteínas Recombinantes/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Embrión de Mamíferos , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Ratones , Ratones Endogámicos C57BL , Oryza/metabolismo , Plantas Modificadas Genéticamente , Investigación Biomédica Traslacional
5.
Mol Cancer Ther ; 9(1): 190-201, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20053778

RESUMEN

Patients with anaplastic thyroid carcinoma (ATC) typically succumb to their disease months after diagnosis despite aggressive therapy. A large percentage of ATCs have been shown to harbor the V600E B-Raf point mutation, leading to the constitutive activation of the mitogen-activated protein kinase pathway. ATC invasion, metastasis, and angiogenesis are in part dependent on the gelatinase class of matrix metalloproteinases (MMP). The explicit targeting of these two tumor markers may provide a novel therapeutic strategy for the treatment of ATC. The MMP-activated anthrax lethal toxin (LeTx), a novel recombinant protein toxin combination, shows potent mitogen-activated protein kinase pathway inhibition in gelatinase-expressing V600E B-Raf tumor cells in vitro. However, preliminary in vivo studies showed that the MMP-activated LeTx also exhibited dramatic antitumor activity against xenografts that did not show significant antiproliferative responses to the LeTx in vitro. Here, we show that the MMP-activated LeTx inhibits orthotopic ATC xenograft progression in both toxin-sensitive and toxin-resistant ATC cells via reduced endothelial cell recruitment and subsequent tumor vascularization. This in turn translates to an improved long-term survival that is comparable with that produced by the multikinase inhibitor sorafenib. Our results also indicate that therapy with the MMP-activated LeTx is extremely effective against advanced tumors with well-established vascular networks. Taken together, these results suggest that the MMP-activated LeTx-mediated endothelial cell targeting is the primary in vivo antitumor mechanism of this novel toxin. Therefore, the MMP-activated LeTx could be used not only in the clinical management of V600E B-Raf ATC but potentially in any solid tumor.


Asunto(s)
Antígenos Bacterianos/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Carcinoma/irrigación sanguínea , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Tiroides/irrigación sanguínea , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Bencenosulfonatos/farmacología , Carcinoma/tratamiento farmacológico , Carcinoma/enzimología , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piridinas/farmacología , Sorafenib , Análisis de Supervivencia , Neoplasias de la Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/patología , Factores de Tiempo
6.
Bioconjug Chem ; 20(10): 1975-82, 2009 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-19785403

RESUMEN

Immunotoxin potency is dependent on cell surface binding specificity as well as internalization efficiency. Current approaches for immunotoxin development are dependent on existing antibodies that were selected for high affinity and/or high production yield. However, these antibodies may demonstrate low internalization efficiency upon cell surface binding and thus are not necessarily the best candidates for immunotoxin design. Here, we have developed an assay with a novel protein, DTG3, to compare and evaluate the internalization efficiency of monoclonal antibodies in order to circumvent the possibility of low internalization. DTG3 is a fusion protein containing the N-terminus of diphtheria toxin (DT) and three copies of streptococci Protein G immunoglobulin binding domains. We show that antibody-DTG3 complexes formed in the test tube are able to bind their antigen on the target cell surface, resulting in cell internalization, DT-mediated protein synthesis inhibition, and host cell apoptosis. We tested this system with two well-studied antibodies, antihuman CD3ε, and anti-PSMA antibodies and were able to show efficiency of this assay. We further examined commercially available anti-CD123 antibodies for potential leukemia-targeting immunotoxin development. Finally, we applied this system in the early-stage screening of newly generated anti-CD123 hybridomas. Our data showed that this internalization assay system is sensitive, time efficient, and reproducible, and has provided a tool to compare monoclonal antibodies for the clinical development of effective immunotoxins for the treatment of a variety of neoplasms.


Asunto(s)
Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/inmunología , Antígenos de Superficie/inmunología , Antígenos de Superficie/metabolismo , Toxina Diftérica/inmunología , Inmunoensayo , Inmunoglobulina G/genética , Inmunotoxinas/química , Inmunotoxinas/inmunología , Terapia Molecular Dirigida/métodos , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Reacciones Antígeno-Anticuerpo , Cricetinae , Toxina Diftérica/metabolismo , Toxina Diftérica/uso terapéutico , Humanos , Hibridomas , Inmunoglobulina G/metabolismo , Inmunotoxinas/metabolismo , Inmunotoxinas/uso terapéutico , Leucemia/inmunología , Leucemia/terapia , Ratones , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico
7.
Mol Cancer Res ; 7(4): 452-61, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19372576

RESUMEN

Solid tumor growth is dependent on angiogenesis, the formation of neovasculature from existing vessels. Endothelial activation of the extracellular signal-regulated kinase 1/2, c-jun NH(2)-terminal kinase, and p38 mitogen-activated protein kinase pathways is central to this process, and thus presents an attractive target for the development of angiogenesis inhibitors. Anthrax lethal toxin (LeTx) has potent catalytic mitogen-activated protein kinase inhibition activity. Preclinical studies showed that LeTx induced potent tumor growth inhibition via the inhibition of xenograft vascularization. However, LeTx receptors and the essential furin-like activating proteases are expressed in many normal tissues, potentially limiting the specificity of LeTx as an antitumor agent. To circumvent nonspecific LeTx activation and simultaneously enhance tumor vascular targeting, a substrate preferably cleaved by the gelatinases class of matrix metalloproteinases (MMP) was substituted for the furin LeTx activation site. In vivo efficacy studies showed that this MMP-activated LeTx inhibited tumor xenografts growth via the reduced migration of endothelial cells into the tumor parenchyma. Here we have expanded on these initial findings by showing that this MMP-activated LeTx reduces endothelial proangiogenic MMP expression, thus causing a diminished proteolytic capacity for extracellular matrix remodeling and endothelial differentiation into capillary networks. Additionally, our data suggest that inhibition of the c-jun NH(2)-terminal kinase and p38, but not extracellular signal-regulated kinase-1/2, pathways is significant in the antiangiogenic activity of the MMP-activated LeTx. Collectively, these results support the clinical development of the MMP-activated LeTx for the treatment of solid tumors.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Endotelio Vascular/efectos de los fármacos , Metaloproteinasas de la Matriz/metabolismo , Neovascularización Patológica/prevención & control , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/antagonistas & inhibidores , MAP Quinasa Quinasa 2/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Mol Cancer Ther ; 7(5): 1218-26, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18483309

RESUMEN

Anthrax lethal toxin (LeTx) shows potent mitogen-activated protein kinase pathway inhibition and apoptosis in melanoma cells that harbor the activating V600E B-RAF mutation. LeTx is composed of two proteins, protective antigen and lethal factor. Uptake of the toxin into cells is dependent on proteolytic activation of protective antigen by the ubiquitously expressed furin or furin-like proteases. To circumvent nonspecific LeTx activation, a substrate preferably cleaved by gelatinases was substituted for the furin LeTx activation site. Here, we have shown that the toxicity of this matrix metalloproteinase (MMP)-activated LeTx is dependent on host cell surface MMP-2 and MMP-9 activity as well as the presence of the activating V600E B-RAF mutation, making this toxin dual specific. This additional layer of tumor cell specificity would potentially decrease systemic toxicity from the reduction of nonspecific toxin activation while retaining antitumor efficacy in patients with V600E B-RAF melanomas. Moreover, our results indicate that cell surface-associated gelatinase expression can be used to predict sensitivity among V600E B-RAF melanomas. This finding will aid in the better selection of patients that will potentially respond to MMP-activated LeTx therapy.


Asunto(s)
Antígenos Bacterianos/toxicidad , Antineoplásicos/toxicidad , Toxinas Bacterianas/toxicidad , Gelatinasas/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Melanoma/enzimología , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/genética , Línea Celular Tumoral , Humanos , Melanoma/metabolismo , Mutación , Proteínas Proto-Oncogénicas B-raf/metabolismo , Células Tumorales Cultivadas
9.
Cell Cycle ; 7(6): 745-9, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18245947

RESUMEN

Angiogenesis is a critical step in solid tumor progression. The mitogen-activated protein kinase (MAPK) signaling pathways are central to this process, and thus present attractive targets for angiogenesis inhibition. Anthrax Lethal Toxin (LeTx), secreted from the gram positive Bacillus anthracis, demonstrates potent MAPK pathway inhibition. In vivo efficacy studies revealed that LeTx has broad anti-tumor efficacy via the targeting of angiogenesis. However, specificity in animal models was limited due to the presence of receptors on many normal tissues and the ubiquitous expression of furin in tissues. Further, half-life of LeTx was short due to circulating furin-like proteases. Gelatinases are expressed on tumor angiogenic sprouts and only to a limited extent in normal tissues or blood. In order to circumvent nonspecific LeTx activation, enhance tumor vascular targeting, and improve plasma half-life, a substrate preferably cleaved by gelatinases was substituted for the furin LeTx activation site. The MMP-activated LeTx showed potent angiogenic inhibition in vivo in the absence of systemic toxicity. Based on these studies, this attenuated toxin has clinical potential as a broad anti-tumor agent.


Asunto(s)
Antígenos Bacterianos/farmacología , Toxinas Bacterianas/farmacología , Metaloproteinasas de la Matriz/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Humanos
10.
J Biol Chem ; 283(1): 529-540, 2008 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-17974567

RESUMEN

Anthrax lethal toxin (LT), a virulence factor secreted by Bacillus anthracis, is selectively toxic to human melanomas with the BRAF V600E activating mutation because of its proteolytic activities toward the mitogen-activated protein kinase kinases (MEKs). To develop LT variants with lower in vivo toxicity and high tumor specificity, and therefore greater potential for clinical use, we generated a mutated LT that requires activation by matrix metalloproteinases (MMPs). This engineered toxin was less toxic than wild-type LT to mice because of the limited expression of MMPs by normal cells. Moreover, the systemically administered toxin produced greater anti-tumor effects than wild-type LT toward human xenografted tumors. This was shown to result from its greater bioavailability, a consequence of the limited uptake and clearance of the modified toxin by normal cells. Furthermore, the MMP-activated LT had very potent anti-tumor activity not only to human melanomas containing the BRAF mutation but also to other tumor types, including lung and colon carcinomas regardless of their BRAF status. Tumor histology and in vivo angiogenesis assays showed that this anti-tumor activity is due largely to the indirect targeting of tumor vasculature and angiogenic processes. Thus, even tumors genetically deficient in anthrax toxin receptors were still susceptible to the toxin therapy in vivo. Moreover, the modified toxin also displayed lower immunogenicity compared with the wild-type toxin. All these properties suggest that this MMP-activated anti-tumor toxin has potential for use in cancer therapy.


Asunto(s)
Antígenos Bacterianos/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Metaloproteinasas de la Matriz/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Animales , Antígenos Bacterianos/genética , Antígenos Bacterianos/metabolismo , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática/efectos de los fármacos , Femenino , Células HT29 , Humanos , Inmunohistoquímica , Interleucina-8/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Neoplasias Experimentales/irrigación sanguínea , Neoplasias Experimentales/patología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA