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.
Nat Commun ; 11(1): 1908, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32313013

RESUMEN

Host cell proteins (HCPs) are process-related impurities generated during biotherapeutic protein production. HCPs can be problematic if they pose a significant metabolic demand, degrade product quality, or contaminate the final product. Here, we present an effort to create a "clean" Chinese hamster ovary (CHO) cell by disrupting multiple genes to eliminate HCPs. Using a model of CHO cell protein secretion, we predict that the elimination of unnecessary HCPs could have a non-negligible impact on protein production. We analyze the HCP content of 6-protein, 11-protein, and 14-protein knockout clones. These cell lines exhibit a substantial reduction in total HCP content (40%-70%). We also observe higher productivity and improved growth characteristics in specific clones. The reduced HCP content facilitates purification of a monoclonal antibody. Thus, substantial improvements can be made in protein titer and purity through large-scale HCP deletion, providing an avenue to increased quality and affordability of high-value biopharmaceuticals.


Asunto(s)
Ingeniería Metabólica/métodos , Proteínas Recombinantes/biosíntesis , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Productos Biológicos , Células CHO , Cromatografía , Cricetulus , Técnicas de Inactivación de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Rituximab , Biología Sintética
2.
Nat Commun ; 11(1): 68, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31896772

RESUMEN

In mammalian cells, >25% of synthesized proteins are exported through the secretory pathway. The pathway complexity, however, obfuscates its impact on the secretion of different proteins. Unraveling its impact on diverse proteins is particularly important for biopharmaceutical production. Here we delineate the core secretory pathway functions and integrate them with genome-scale metabolic reconstructions of human, mouse, and Chinese hamster ovary cells. The resulting reconstructions enable the computation of energetic costs and machinery demands of each secreted protein. By integrating additional omics data, we find that highly secretory cells have adapted to reduce expression and secretion of other expensive host cell proteins. Furthermore, we predict metabolic costs and maximum productivities of biotherapeutic proteins and identify protein features that most significantly impact protein secretion. Finally, the model successfully predicts the increase in secretion of a monoclonal antibody after silencing a highly expressed selection marker. This work represents a knowledgebase of the mammalian secretory pathway that serves as a novel tool for systems biotechnology.


Asunto(s)
Genoma , Mamíferos/genética , Mamíferos/metabolismo , Proteínas/metabolismo , Vías Secretoras/genética , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Células CHO , Simulación por Computador , Cricetulus , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reproducibilidad de los Resultados
3.
Metab Eng ; 56: 120-129, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31526854

RESUMEN

Chinese hamster ovary (CHO) cells are the preferred host for producing biopharmaceuticals. Amino acids are biologically important precursors for CHO metabolism; they serve as building blocks for proteogenesis, including synthesis of biomass and recombinant proteins, and are utilized for growth and cellular maintenance. In this work, we studied the physiological impact of disrupting a range of amino acid catabolic pathways in CHO cells. We aimed to reduce secretion of growth inhibiting metabolic by-products derived from amino acid catabolism including lactate and ammonium. To achieve this, we engineered nine genes in seven different amino acid catabolic pathways using the CRISPR-Cas9 genome editing system. For identification of target genes, we used a metabolic network reconstruction of amino acid catabolism to follow transcriptional changes in response to antibody production, which revealed candidate genes for disruption. We found that disruption of single amino acid catabolic genes reduced specific lactate and ammonium secretion while specific growth rate and integral of viable cell density were increased in many cases. Of particular interest were Hpd and Gad2 disruptions, which show unchanged AA uptake rates, while having growth rates increased up to 19%, and integral of viable cell density as much as 50% higher, and up to 26% decrease in specific ammonium production and to a lesser extent (up to 22%) decrease in lactate production. This study demonstrates the broad potential of engineering amino acid catabolism in CHO cells to achieve improved phenotypes for bioprocessing.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Reprogramación Celular , Edición Génica , Redes y Vías Metabólicas/genética , Animales , Células CHO , Cricetulus
4.
J Biotechnol ; 306: 24-31, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31465797

RESUMEN

In recombinant protein expression using Chinese hamster ovary (CHO) cells, chemically defined media contain essential amino acids such as branched chain amino acids (BCAAs) leucine, isoleucine and valine. Availability of amino acids is critical as these are building blocks for protein synthesis. However, breakdown of amino acids can lead to build up of toxic intermediates and metabolites that decrease cell growth, productivity and product quality. BCAA catabolism also hampers the usage of BCAAs for protein synthesis. In this work we studied the effects of disrupting the genes responsible for the first step of BCAA catabolism: branched chain aminotransferase 1 (Bcat1) and branched chain aminotransferase 2 (Bcat2). We evaluated the effect of disrupting the genes individually and in combination, and examined the effects in producer and non-producer host cells. Our experiments show that Bcat1 disruption improves cell growth in producer cells, but not in non-producers. Conversely, Bcat2 has a minor negative effect on growth in producer cells, and none in non-producers. Combined Bcat1 and Bcat2 disruption improves growth in producer cells. By-product metabolism is cell line-, clone- and producer-dependent. Overall, our results show that the effects of targeting Bcat1 and/or Bcat2 are cell line-dependent, and seemingly linked to the burden of recombinant protein expression.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Transaminasas/metabolismo , Animales , Células CHO , Proliferación Celular , Supervivencia Celular , Cricetulus , Medios de Cultivo/metabolismo , Mutación , Biosíntesis de Proteínas , Transaminasas/genética
5.
Methods Mol Biol ; 1674: 147-161, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28921435

RESUMEN

Fed-batch culture is the most commonly used upstream process in industry today for recombinant monoclonal antibody production using Chinese hamster ovary (CHO) cells. Developing and optimizing this process in the lab is crucial for establishing process knowledge, which enables rapid and predictable tech-transfer to manufacturing scale. In this chapter, we describe stepwise how to carry out fed-batch CHO cell culture for lab-scale antibody production.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Formación de Anticuerpos/fisiología , Técnicas de Cultivo Celular por Lotes/métodos , Reactores Biológicos , Células CHO , Técnicas de Cultivo de Célula/métodos , Línea Celular , Cricetulus
6.
Sci Rep ; 7: 40388, 2017 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-28091612

RESUMEN

Recombinant protein production coopts the host cell machinery to provide high protein yields of industrial enzymes or biotherapeutics. However, since protein translation is energetically expensive and tightly controlled, it is unclear if highly expressed recombinant genes are translated as efficiently as host genes. Furthermore, it is unclear how the high expression impacts global translation. Here, we present the first genome-wide view of protein translation in an IgG-producing CHO cell line, measured with ribosome profiling. Through this we found that our recombinant mRNAs were translated as efficiently as the host cell transcriptome, and sequestered up to 15% of the total ribosome occupancy. During cell culture, changes in recombinant mRNA translation were consistent with changes in transcription, demonstrating that transcript levels influence specific productivity. Using this information, we identified the unnecessary resistance marker NeoR to be a highly transcribed and translated gene. Through siRNA knock-down of NeoR, we improved the production- and growth capacity of the host cell. Thus, ribosomal profiling provides valuable insights into translation in CHO cells and can guide efforts to enhance protein production.


Asunto(s)
Proteínas/metabolismo , Ribosomas/metabolismo , Animales , Células CHO , Recuento de Células , Proliferación Celular/genética , Supervivencia Celular/genética , Cricetinae , Cricetulus , Técnicas de Silenciamiento del Gen , Inmunoglobulina G/metabolismo , Nucleótidos/metabolismo , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Transcripción Genética
7.
Cell Syst ; 3(5): 434-443.e8, 2016 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-27883890

RESUMEN

Chinese hamster ovary (CHO) cells dominate biotherapeutic protein production and are widely used in mammalian cell line engineering research. To elucidate metabolic bottlenecks in protein production and to guide cell engineering and bioprocess optimization, we reconstructed the metabolic pathways in CHO and associated them with >1,700 genes in the Cricetulus griseus genome. The genome-scale metabolic model based on this reconstruction, iCHO1766, and cell-line-specific models for CHO-K1, CHO-S, and CHO-DG44 cells provide the biochemical basis of growth and recombinant protein production. The models accurately predict growth phenotypes and known auxotrophies in CHO cells. With the models, we quantify the protein synthesis capacity of CHO cells and demonstrate that common bioprocess treatments, such as histone deacetylase inhibitors, inefficiently increase product yield. However, our simulations show that the metabolic resources in CHO are more than three times more efficiently utilized for growth or recombinant protein synthesis following targeted efforts to engineer the CHO secretory pathway. This model will further accelerate CHO cell engineering and help optimize bioprocesses.


Asunto(s)
Genoma , Animales , Células CHO , Consenso , Cricetinae , Cricetulus , Humanos , Redes y Vías Metabólicas , Proteínas Recombinantes
8.
Biotechnol Bioeng ; 112(11): 2373-87, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25995028

RESUMEN

Chinese hamster ovary (CHO) cells are the preferred production host for many therapeutic proteins. The production of heterologous proteins in CHO cells imposes a burden on the host cell metabolism and impact cellular physiology on a global scale. In this work, a multi-omics approach was applied to study the production of erythropoietin (EPO) in a panel of CHO-K1 cells under growth-limited and unlimited conditions in batch and chemostat cultures. Physiological characterization of the EPO-producing cells included global transcriptome analysis, targeted metabolome analysis, including intracellular pools of glycolytic intermediates, NAD(P)H/NAD(P)(+) , adenine nucleotide phosphates (ANP), and extracellular concentrations of sugars, organic acids, and amino acids. Potential impact of EPO expression on the protein secretory pathway was assessed at multiple stages using quantitative PCR (qPCR), reverse transcription PCR (qRT-PCR), Western blots (WB), and global gene expression analysis to assess EPO gene copy numbers, EPO gene expression, intracellular EPO retention, and differentially expressed genes functionally related to secretory protein processing, respectively. We found no evidence supporting the existence of production bottlenecks in energy metabolism (i.e., glycolytic metabolites, NAD(P)H/NAD(P)(+) and ANPs) in batch culture or in the secretory protein production pathway (i.e., gene dosage, transcription and post-translational processing of EPO) in chemostat culture at specific productivities up to 5 pg/cell/day. Time-course analysis of high- and low-producing clones in chemostat culture revealed rapid adaptation of transcription levels of amino acid catabolic genes in favor of EPO production within nine generations. Interestingly, the adaptation was followed by an increase in specific EPO productivity.


Asunto(s)
Células Epiteliales/fisiología , Eritropoyetina/biosíntesis , Proteínas Recombinantes/biosíntesis , Animales , Células CHO , Cricetulus , Células Epiteliales/metabolismo , Eritropoyetina/genética , Femenino , Perfilación de la Expresión Génica , Metabolómica , Proteoma/análisis , Proteínas Recombinantes/genética
9.
Mol Brain ; 8: 4, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25599691

RESUMEN

BACKGROUND: Homeostatic intrinsic plasticity encompasses the mechanisms by which neurons stabilize their excitability in response to prolonged and destabilizing changes in global activity. However, the milieu of molecular players responsible for these regulatory mechanisms is largely unknown. RESULTS: Using whole-cell patch clamp recording and unbiased gene expression profiling in rat dissociated hippocampal neurons cultured at high density, we demonstrate here that chronic activity blockade induced by the sodium channel blocker tetrodotoxin leads to a homeostatic increase in action potential firing and down-regulation of potassium channel genes. In addition, chronic activity blockade reduces total potassium current, as well as protein expression and current of voltage-gated Kv1 and Kv7 potassium channels, which are critical regulators of action potential firing. Importantly, inhibition of N-Methyl-D-Aspartate receptors alone mimics the effects of tetrodotoxin, including the elevation in firing frequency and reduction of potassium channel gene expression and current driven by activity blockade, whereas inhibition of L-type voltage-gated calcium channels has no effect. CONCLUSIONS: Collectively, our data suggest that homeostatic intrinsic plasticity induced by chronic activity blockade is accomplished in part by decreased calcium influx through N-Methyl-D-Aspartate receptors and subsequent transcriptional down-regulation of potassium channel genes.


Asunto(s)
Regulación hacia Abajo/genética , Homeostasis , Plasticidad Neuronal/genética , Canales de Potasio/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Potenciales de Acción , Animales , Canales de Calcio Tipo L/metabolismo , Células Cultivadas , Ontología de Genes , Redes Reguladoras de Genes , Hipocampo/citología , Modelos Neurológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Canales de Potasio/metabolismo , Células Piramidales/metabolismo , Ratas Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Sinapsis/metabolismo
10.
Appl Environ Microbiol ; 79(19): 5899-906, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23872558

RESUMEN

There continues to be a need for developing efficient and environmentally friendly treatments for Bacillus anthracis, the causative agent of anthrax. One emerging approach for inactivation of vegetative B. anthracis is the use of bacteriophage endolysins or lytic enzymes encoded by bacterial genomes (autolysins) with highly evolved specificity toward bacterium-specific peptidoglycan cell walls. In this work, we performed in silico analysis of the genome of Bacillus anthracis strain Ames, using a consensus binding domain amino acid sequence as a probe, and identified a novel lytic enzyme that we termed AmiBA2446. This enzyme exists as a homodimer, as determined by size exclusion studies. It possesses N-acetylmuramoyl-l-alanine amidase activity, as determined from liquid chromatography-mass spectrometry (LC-MS) analysis of muropeptides released due to the enzymatic digestion of peptidoglycan. Phylogenetic analysis suggested that AmiBA2446 was an autolysin of bacterial origin. We characterized the effects of enzyme concentration and phase of bacterial growth on bactericidal activity and observed close to a 5-log reduction in the viability of cells of Bacillus cereus 4342, a surrogate for B. anthracis. We further tested the bactericidal activity of AmiBA2446 against various Bacillus species and demonstrated significant activity against B. anthracis and B. cereus strains. We also demonstrated activity against B. anthracis spores after pretreatment with germinants. AmiBA2446 enzyme was also stable in solution, retaining its activity after 4 months of storage at room temperature.


Asunto(s)
Antibacterianos/metabolismo , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/enzimología , Bacteriólisis , Viabilidad Microbiana , N-Acetil Muramoil-L-Alanina Amidasa/metabolismo , Bacillus anthracis/genética , Bacillus cereus/efectos de los fármacos , Cromatografía Liquida , Análisis por Conglomerados , Hidrólisis , Espectrometría de Masas , N-Acetil Muramoil-L-Alanina Amidasa/genética , Peptidoglicano/metabolismo , Filogenia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...