Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Nat Cardiovasc Res ; 3(2): 145-165, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-39196193

RESUMEN

Preclinical data have confirmed that human pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can remuscularize the injured or diseased heart, with several clinical trials now in planning or recruitment stages. However, because ventricular arrhythmias represent a complication following engraftment of intramyocardially injected PSC-CMs, it is necessary to provide treatment strategies to control or prevent engraftment arrhythmias (EAs). Here, we show in a porcine model of myocardial infarction and PSC-CM transplantation that EAs are mechanistically linked to cellular heterogeneity in the input PSC-CM and resultant graft. Specifically, we identify atrial and pacemaker-like cardiomyocytes as culprit arrhythmogenic subpopulations. Two unique surface marker signatures, signal regulatory protein α (SIRPA)+CD90-CD200+ and SIRPA+CD90-CD200-, identify arrhythmogenic and non-arrhythmogenic cardiomyocytes, respectively. Our data suggest that modifications to current PSC-CM-production and/or PSC-CM-selection protocols could potentially prevent EAs. We further show that pharmacologic and interventional anti-arrhythmic strategies can control and potentially abolish these arrhythmias.


Asunto(s)
Arritmias Cardíacas , Miocitos Cardíacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/trasplante , Animales , Arritmias Cardíacas/terapia , Humanos , Modelos Animales de Enfermedad , Infarto del Miocardio/terapia , Porcinos , Células Cultivadas , Diferenciación Celular , Células Madre Pluripotentes Inducidas/trasplante , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de los fármacos , Fenotipo , Biomarcadores/metabolismo , Células Madre Pluripotentes/trasplante , Trasplante de Células Madre/métodos , Antiarrítmicos/uso terapéutico , Antiarrítmicos/farmacología , Frecuencia Cardíaca/fisiología
2.
MAbs ; 14(1): 2083465, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35737825

RESUMEN

The reliable and cost-efficient manufacturing of monoclonal antibodies (mAbs) is essential to fulfil their ever-growing demand. Cell death in bioreactors reduces productivity and product quality, and is largely attributed to apoptosis. In perfusion bioreactors, this leads to the necessity of a bleed stream, which negatively affects the overall process economy. To combat this limitation, death-resistant Chinese hamster ovary cell lines were developed by simultaneously knocking out the apoptosis effector proteins Bak1, Bax, and Bok with CRISPR technology. These cell lines were cultured in fed-batch and perfusion bioreactors and compared to an unmodified control cell line. In fed-batch, the death-resistant cell lines showed higher cell densities and longer culture durations, lasting nearly a month under standard culture conditions. In perfusion, the death-resistant cell lines showed slower drops in viability and displayed an arrest in cell division after which cell size increased instead. Pertinently, the death-resistant cell lines demonstrated the ability to be cultured for several weeks without bleed, and achieved similar volumetric productivities at lower cell densities than that of the control cell line. Perfusion culture reduced fragmentation of the mAb produced, and the death-resistant cell lines showed increased glycosylation in the light chain in both bioreactor modes. These data demonstrate that rationally engineered death-resistant cell lines are ideal for mAb production in perfusion culture, negating the need to bleed the bioreactor whilst maintaining product quantity and quality.


Asunto(s)
Anticuerpos Monoclonales , Reactores Biológicos , Animales , Anticuerpos Monoclonales/farmacología , Técnicas de Cultivo Celular por Lotes , Células CHO , Cricetinae , Cricetulus , Perfusión
3.
Biotechnol Bioeng ; 119(6): 1380-1391, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35180317

RESUMEN

Chinese hamster ovary (CHO) cells are the primary platform for the production of biopharmaceuticals. To increase yields, many CHO cell lines have been genetically engineered to resist cell death. However, the kinetics that governs cell fate in bioreactors are confounded by many variables associated with batch processes. Here, we used CRISPR-Cas9 to create combinatorial knockouts of the three known BCL-2 family effector proteins: Bak1, Bax, and Bok. To assess the response to apoptotic stimuli, cell lines were cultured in the presence of four cytotoxic compounds with different mechanisms of action. A population-based model was developed to describe the behavior of the resulting viable cell dynamics as a function of genotype and treatment. Our results validated the synergistic antiapoptotic nature of Bak1 and Bax, while the deletion of Bok had no significant impact. Importantly, the uniform application of apoptotic stresses permitted direct observation and quantification of a delay in the onset of cell death through Bayesian inference of meaningful model parameters. In addition to the classical death rate, a delay function was found to be essential in the accurate modeling of the cell death response. These findings represent an important bridge between cell line engineering strategies and biological modeling in a bioprocess context.


Asunto(s)
Apoptosis , Proteínas Proto-Oncogénicas c-bcl-2 , Animales , Apoptosis/genética , Teorema de Bayes , Células CHO , Cricetinae , Cricetulus , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
4.
Biotechnol Bioeng ; 118(1): 481-490, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32865815

RESUMEN

Chinese hamster ovary (CHO) cells are widely used in biopharmaceutical production. Improvements to cell lines and bioprocesses are constantly being explored. One of the major limitations of CHO cell culture is that the cells undergo apoptosis, leading to rapid cell death, which impedes reaching high recombinant protein titres. While several genetic engineering strategies have been successfully employed to reduce apoptosis, there is still room to further enhance CHO cell lines performance. 'Omics analysis is a powerful tool to better understand different phenotypes and for the identification of gene targets for engineering. Here, we present a comprehensive review of previous CHO 'omics studies that revealed changes in the expression of apoptosis-related genes. We highlight targets for genetic engineering that have reduced, or have the potential to reduce, apoptosis or to increase cell proliferation in CHO cells, with the final aim of increasing productivity.


Asunto(s)
Apoptosis , Proliferación Celular , Proteómica , Animales , Células CHO , Cricetulus , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
5.
Lancet Infect Dis ; 20(4): 445-454, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32027842

RESUMEN

BACKGROUND: The monoclonal antibody m102.4 is a potent, fully human antibody that neutralises Hendra and Nipah viruses in vitro and in vivo. We aimed to investigate the safety, tolerability, pharmacokinetics, and immunogenicity of m102.4 in healthy adults. METHODS: In this double-blind, placebo-controlled, single-centre, dose-escalation, phase 1 trial of m102.4, we randomly assigned healthy adults aged 18-50 years with a body-mass index of 18·0-35·0 kg/m2 to one of five cohorts. A sentinel pair for each cohort was randomly assigned to either m102.4 or placebo. The remaining participants in each cohort were randomly assigned (5:1) to receive m102.4 or placebo. Cohorts 1-4 received a single intravenous infusion of m102.4 at doses of 1 mg/kg (cohort 1), 3 mg/kg (cohort 2), 10 mg/kg (cohort 3), and 20 mg/kg (cohort 4), and were monitored for 113 days. Cohort 5 received two infusions of 20 mg/kg 72 h apart and were monitored for 123 days. The primary outcomes were safety and tolerability. Secondary outcomes were pharmacokinetics and immunogenicity. Analyses were completed according to protocol. The study was registered on the Australian New Zealand Clinical Trials Registry, ACTRN12615000395538. FINDINGS: Between March 27, 2015, and June 16, 2016, 40 (52%) of 77 healthy screened adults were enrolled in the study. Eight participants were assigned to each cohort (six received m102.4 and two received placebo). 86 treatment-emergent adverse events were reported, with similar rates between placebo and treatment groups. The most common treatment-related event was headache (12 [40%] of 30 participants in the combined m102.4 group, and three [30%] of ten participants in the pooled placebo group). No deaths or severe adverse events leading to study discontinuation occurred. Pharmacokinetics based on those receiving m102.4 (n=30) were linear, with a median half-life of 663·3 h (range 474·3-735·1) for cohort 1, 466·3 h (382·8-522·3) for cohort 2, 397·0 h (333·9-491·8) for cohort 3, and 466·7 h (351·0-889·6) for cohort 4. The elimination kinetics of those receiving repeated dosing (cohort 5) were similar to those of single-dose recipients (median elimination half-time 472·0 [385·6-592·0]). Anti-m102.4 antibodies were not detected at any time-point during the study. INTERPRETATION: Single and repeated dosing of m102.4 were well tolerated and safe, displayed linear pharmacokinetics, and showed no evidence of an immunogenic response. This study will inform future dosing regimens for m102.4 to achieve prolonged exposure for systemic efficacy to prevent and treat henipavirus infections. FUNDING: Queensland Department of Health, the National Health and Medical Research Council, and the National Hendra Virus Research Program.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacocinética , Glicoproteínas/inmunología , Voluntarios Sanos , Henipavirus/inmunología , Inmunogenicidad Vacunal , Seguridad , Adulto , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/inmunología , Australia , Método Doble Ciego , Femenino , Cefalea/etiología , Humanos , Infusiones Intravenosas , Masculino
6.
Biotechnol Bioeng ; 117(4): 1187-1203, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31930480

RESUMEN

Chinese hamster ovary (CHO) cells are the predominant host cell line for the production of biopharmaceuticals, a growing industry currently worth more than $188 billion USD in global sales. CHO cells undergo programmed cell death (apoptosis) following different stresses encountered in cell culture, such as substrate limitation, accumulation of toxic by-products, and mechanical shear, hindering production. Genetic engineering strategies to reduce apoptosis in CHO cells have been investigated with mixed results. In this review, a contemporary understanding of the real complexity of apoptotic mechanisms and signaling pathways is described; followed by an overview of antiapoptotic cell line engineering strategies tested so far in CHO cells.


Asunto(s)
Apoptosis , Productos Biológicos/metabolismo , Células CHO , Ingeniería Celular , Animales , Técnicas de Cultivo de Célula , Cricetinae , Cricetulus
7.
Biotechnol J ; 13(3): e1700231, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29316330

RESUMEN

The development of next-generation sequencing technologies has opened new opportunities to better characterize complex eukaryotic cells. Chinese hamster ovary (CHO) cells play a primary role in therapeutic protein production, with currently five of the top ten blockbuster drugs produced in CHO. However, engineering superior CHO cells with improved production features has had limited success to date and cell lines are still developed through the generation and screening of large strain pools. Here, we applied RNA sequencing to contrast a high and a low monoclonal antibody producing cell line. Rigorous experimental design achieved high reproducibility between biological replicates, remarkably reducing variation to less than 10%. More than 14 000 gene-transcripts are identified and surprisingly 58% are classified as differentially expressed, including 2900 with a fold change higher than 1.5. The largest differences are found for gene-transcripts belonging to regulation of apoptosis, cell death, and protein intracellular transport GO term classifications, which are found to be significantly enriched in the high producing cell line. RNA sequencing is also performed on subclones, where down-regulation of genes encoding secreted glycoproteins is found to be the most significant change. The large number of significant differences even between subclones challenges the notion of identifying and manipulating a few key genes to generate high production CHO cell lines.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Células CHO , Evolución Clonal/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Linaje de la Célula/genética , Cricetulus
8.
Tissue Eng Part C Methods ; 24(3): 146-157, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29239281

RESUMEN

Human pluripotent stem cells (hPSCs) are viewed as promising candidates for applications in regenerative medicine and therapy due to their proliferative and pluripotent properties. However, obtaining clinically significant numbers of hPSCs remains a limiting factor and impedes their use in therapeutic applications. Conventionally, hPSCs are cultured on two-dimensional surfaces coated with a suitable substrate, such as Matrigel™. This method, however, requires a large surface area to generate sufficient cell numbers to meet clinical needs and is therefore impractical as a manufacturing platform for cell expansion. In addition, the use of enzymes for cell detachment and small molecule inhibitors to increase plating efficiency may impact future cell behavior when used for routine subculturing. In this study, we describe a protocol to generate and maintain hPSC aggregates in a three-dimensional suspension culture by utilizing thermoresponsive nanobridges. The property of the polymer used in the nanobridges enables passaging and expansion through a temperature change in combination with mechanically applied shear to dissociate aggregates; thus, we eliminate the need of enzymes or small molecules for cell dissociation and viability, respectively. Utilizing this platform, maintenance of human embryonic stem cells for three continuous passages demonstrated high expression levels in key pluripotent markers.


Asunto(s)
Técnicas de Cultivo de Célula/instrumentación , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Células Madre Embrionarias Humanas/citología , Nanotecnología/métodos , Polímeros/química , Temperatura , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias Humanas/fisiología , Humanos
9.
Biotechnol Bioeng ; 114(8): 1825-1836, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28436007

RESUMEN

For decades, Chinese hamster ovary (CHO) cells have been the preferred host for therapeutic monoclonal antibody (mAb) production; however, increasing mAb titer by rational engineering remains a challenge. Our previous proteomic analysis in CHO cells suggested that a higher content of glutathione (GSH) might be related to higher productivity. GSH is an important antioxidant, cell detoxifier, and is required to ensure the formation of native disulfide bonds in proteins. To investigate the involvement of GSH in mAb production, we generated stable CHO cell lines overexpressing genes involved in the first step of GSH synthesis; namely the glutamate-cysteine ligase catalytic subunit (Gclc) and the glutamate-cysteine ligase modifier subunit (Gclm). The two genes were reconstructed from our RNA-Seq de novo assembly and then were functionally annotated. Once the sequences of the genes were confirmed using proteogenomics, a transiently expressed mAb was introduced into cell lines overexpressing either Gclc or Gclm. The new cell lines were compared for mAb production to the parental cell line and changes at the proteome level were measured using SWATH. As per our previous proteomics observations, overexpressing Gclm improved productivity, titer, and the frequency of high producer clones by 70%. In contrast, overexpressing Gclc, which produced a higher amount of GSH, did not increase mAb production. We show that GSH cannot be linked to higher productivity and that Gclm may be controlling other cellular processes involved in mAb production yet to be elucidated. Biotechnol. Bioeng. 2017;114: 1825-1836. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Células CHO/fisiología , Mejoramiento Genético/métodos , Glutamato-Cisteína Ligasa/metabolismo , Ingeniería de Proteínas/métodos , Regulación hacia Arriba/fisiología , Animales , Anticuerpos Monoclonales/genética , Células CHO/citología , Catálisis , Cricetulus , Glutamato-Cisteína Ligasa/genética , Subunidades de Proteína
10.
J Mater Chem B ; 3(43): 8477-8485, 2015 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-32262687

RESUMEN

There is an increasing demand of efficient nano-carriers for intracellular delivery of therapeutic proteins. This study reports on a novel "neck-enhancing" approach to synthesize stable rough silica nanoparticles (RSNs) with controllable surface roughness. By increasing the shell particle size from 13 to 98 nm while fixing the core size at 211 nm, the interspace size between neighboring shell particles of RSNs is enlarged from 7 to 38 nm. Cytochrome c, IgG fragment and IgG antibody are preferably adsorbed onto one of the RSNs with the interspace size of 14, 21 and 38 nm, respectively. The binding activity of the IgG fragment loaded onto RSNs is maintained as confirmed by surface plasmon resonance. The hydrophobically modified RSN with the interspace size of 38 nm effectively deliver the therapeutic anti-pAkt antibody into breast cancer cells, causing significant cell inhibition by blocking pAkt and the downstream anti-apoptotic protein Bcl-2.

11.
J Proteome Res ; 14(2): 609-18, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25495469

RESUMEN

Chinese hamster ovary (CHO) cells are the preferred production host for therapeutic monoclonal antibodies (mAb) due to their ability to perform post-translational modifications and their successful approval history. The completion of the genome sequence for CHO cells has reignited interest in using quantitative proteomics to identify markers of good production lines. Here we applied two different proteomic techniques, iTRAQ and SWATH, for the identification of expression differences between a high- and low-antibody-producing CHO cell lines derived from the same transfection. More than 2000 proteins were quantified with 70 of them classified as differentially expressed in both techniques. Two biological processes were identified as differentially regulated by both methods: up-regulation of glutathione biosynthesis and down-regulation of DNA replication. Metabolomic analysis confirmed that the high producing cell line displayed higher intracellular levels of glutathione. SWATH further identified up-regulation of actin filament processes and intracellular transport and down regulation of several growth-related processes. These processes may be important for conferring high mAb production and as such are promising candidates for targeted engineering of high-expression cell lines.


Asunto(s)
Anticuerpos Monoclonales/biosíntesis , Glutatión/biosíntesis , Ovario/inmunología , Regulación hacia Arriba , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Transporte de Proteínas
12.
Biomacromolecules ; 15(10): 3569-76, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25156109

RESUMEN

Understanding the pathways for nuclear entry could see vast improvements in polymer design for the delivery of genetic materials to cells. Here, we use a novel diblock copolymer complexed with plasmid DNA (pDNA) to determine both its cellular entry and nuclear pathways. The diblock copolymer (A-C3) is specifically designed to bind and protect pDNA, release it at a specific time, but more importantly, rapidly escape the endosome. The copolymer was taken up by HEK293 cells preferentially via the clathrin-mediated endocytosis (CME) pathway, and the pDNA entered the nucleus to produce high gene expression levels in all cells after 48 h, a similar observation to the commercially available polymer transfection agent, PEI Max. This demonstrates that the polymers must first escape the endosome and then mediate transport of pDNA to the nucleus for occurrence of gene expression. The amount of pDNA within the nucleus was found to be higher for our A-C3 polymer than PEI Max, with our polymer delivering 7 times more pDNA than PEI Max after 24 h. We further found that entry into the nucleus was primarily through the small nuclear pores and did not occur during mitosis when the nuclear envelope becomes compromised. The observation that the polymers are also found in the nucleus supports the hypothesis that the large pDNA/polymer complex (size ~200 nm) must dissociate prior to nucleus entry and that cationic and hydrophobic monomer units on the polymer may facilitate active transport of the pDNA through the nuclear pore.


Asunto(s)
ADN/metabolismo , Endosomas/metabolismo , Plásmidos/metabolismo , Polímeros/metabolismo , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular/fisiología , Cationes/metabolismo , Núcleo Celular/metabolismo , Endocitosis/fisiología , Células HEK293 , Humanos , Transfección/métodos
13.
N Biotechnol ; 31(3): 214-20, 2014 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-24518824

RESUMEN

Therapeutic recombinant monoclonal antibodies (mAbs) are commonly produced by high-expressing, clonal, mammalian cells. Creation of these clones for manufacturing remains heavily reliant on stringent selection and gene amplification, which in turn can lead to genetic instability, variable expression, product heterogeneity and prolonged development timelines. Inclusion of cis-acting ubiquitous chromatin opening elements (UCOE™) in mammalian expression vectors has been shown to improve productivity and facilitate high-level gene expression irrespective of the chromosomal integration site without lengthy gene amplification protocols. In this study we have used high-throughput robotic clone selection in combination with UCOE™ containing expression vectors to develop a rapid, streamlined approach for early-stage cell line development and isolation of high-expressing clones for mAb production using Chinese hamster ovary (CHO) cells. Our results demonstrate that it is possible to go from transfection to stable clones in only 4 weeks, while achieving specific productivities exceeding 20 pg/cell/day. Furthermore, we have used this approach to quickly screen several process-crucial parameters including IgG subtype, enhancer-promoter combination and UCOE™ length. The use of UCOE™-containing vectors in combination with automated robotic selection provides a rapid method for the selection of stable, high-expressing clones.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Cromatina/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Animales , Secuencia de Bases , Técnicas de Cultivo Celular por Lotes , Células CHO , Células Clonales , Cricetinae , Cricetulus , Vectores Genéticos/metabolismo , Cobayas , Humanos , Inmunoglobulina G/metabolismo , Regiones Promotoras Genéticas/genética , Transfección
14.
Biomacromolecules ; 15(3): 844-55, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24571238

RESUMEN

The development of robust suspension cultures of human embryonic stem cells (hESCs) without the use of cell membrane disrupting enzymes or inhibitors is critical for future clinical applications in regenerative medicine. We have achieved this by using long, flexible, and thermoresponsive polymer worms decorated with a recombinant vitronectin subdomain that bridge hESCs, aiding in hESC's natural ability to form embryoid bodies (EBs) and satisfying their inherent requirement for cell-cell and cell-extracellular matrix contact. When the EBs reached an optimal upper size where cytokine and nutrient penetration becomes limiting, these long and flexible polymer worms facilitated EB breakdown via a temperature shift from 37 to 25 °C. The thermoresponsive nature of the worms enabled a cyclical dissociation and propagation of the cells. Repeating the process for three cycles (over eighteen days) provided a >30-fold expansion in cell number while maintaining pluripotency, thereby providing a simple, nondestructive process for the 3D expansion of hESC.


Asunto(s)
Técnicas de Cultivo de Célula , Cuerpos Embrioides/química , Células Madre Embrionarias/citología , Matriz Extracelular/química , Diferenciación Celular/genética , Proliferación Celular , Cuerpos Embrioides/citología , Humanos , Polímeros/química , Medicina Regenerativa , Temperatura
15.
J Biol Chem ; 288(21): 15269-79, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23482564

RESUMEN

Human group IIA secreted phospholipase A2 (hGIIA) promotes tumor growth and inflammation and can act independently of its well described catalytic lipase activity via an alternative poorly understood signaling pathway. With six chemically diverse inhibitors we show that it is possible to selectively inhibit hGIIA signaling over catalysis, and x-ray crystal structures illustrate that signaling involves a pharmacologically distinct surface to the catalytic site. We demonstrate in rheumatoid fibroblast-like synoviocytes that non-catalytic signaling is associated with rapid internalization of the enzyme and colocalization with vimentin. Trafficking of exogenous hGIIA was monitored with immunofluorescence studies, which revealed that vimentin localization is disrupted by inhibitors of signaling that belong to a rare class of small molecule inhibitors that modulate protein-protein interactions. This study provides structural and pharmacological evidence for an association between vimentin, hGIIA, and arachidonic acid metabolism in synovial inflammation, avenues for selective interrogation of hGIIA signaling, and new strategies for therapeutic hGIIA inhibitor design.


Asunto(s)
Ácido Araquidónico/metabolismo , Artritis Reumatoide/metabolismo , Inhibidores Enzimáticos/farmacología , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Membrana Sinovial/metabolismo , Vimentina/metabolismo , Animales , Ácido Araquidónico/genética , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Células CHO , Cricetinae , Cricetulus , Diseño de Fármacos , Inhibidores Enzimáticos/uso terapéutico , Femenino , Fosfolipasas A2 Grupo II/genética , Fosfolipasas A2 Grupo II/metabolismo , Humanos , Masculino , Transducción de Señal/genética , Membrana Sinovial/patología , Vimentina/genética
16.
Biotechnol Prog ; 28(3): 887-91, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22505017

RESUMEN

Biologic drugs, such as monoclonal antibodies, are commonly made using mammalian cells in culture. The cell lines used for manufacturing should ideally be clonal, meaning derived from a single cell, which represents a technically challenging process. Fetal bovine serum is often used to support low cell density cultures, however, from a regulatory perspective, it is preferable to avoid animal-derived components to increase process consistency and reduce the risk of contamination from adventitious agents. Chinese hamster ovary (CHO) cells are the most widely used cell line in industry and a large number of serum-free, protein-free, and fully chemically defined growth media are commercially available, although these media alone do not readily support efficient single cell cloning. In this work, we have developed a simple, fully defined, single-cell cloning media, specifically for CHO cells, using commercially available reagents. Our results show that a 1:1 mixture of CD-CHO™ and DMEM/F12 supplemented with 1.5 g/L of recombinant albumin (Albucult®) supports single cell cloning. This formulation can support recovery of single cells in 43% of cultures compared to 62% in the presence of serum.


Asunto(s)
Albúminas/metabolismo , Clonación de Organismos , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Proteínas Recombinantes/metabolismo
17.
PLoS One ; 7(12): e52214, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284940

RESUMEN

Human embryonic stem cell (hESC) derivatives show promise as viable cell therapy options for multiple disorders in different tissues. Recent advances in stem cell biology have lead to the reliable production and detailed molecular characterisation of a range of cell-types. However, the role of mitochondria during differentiation has yet to be fully elucidated. Mitochondria mediate a cells response to altered energy requirements (e.g. cardiomyocyte contraction) and, as such, the mitochondrial phenotype is likely to change during the dynamic process of hESC differentiation. We demonstrate that manipulating mitochondrial biogenesis alters mesendoderm commitment. To investigate mitochondrial localisation during early lineage specification of hESCs we developed a mitochondrial reporter line, KMEL2, in which sequences encoding the green fluorescent protein (GFP) are targeted to the mitochondria. Differentiation of KMEL2 lines into the three germ layers showed that the mitochondria in these differentiated progeny are GFP positive. Therefore, KMEL2 hESCs facilitate the study of mitochondria in a range of cell types and, importantly, permit real-time analysis of mitochondria via the GFP tag.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Mitocondrias/metabolismo , Línea Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Cariotipo , Fosforilación Oxidativa
18.
MAbs ; 3(5): 440-52, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21822050

RESUMEN

Therapeutic monoclonal antibodies (mAbs) currently dominate the biologics marketplace. Development of a new therapeutic mAb candidate is a complex, multistep process and early stages of development typically begin in an academic research environment. Recently, a number of facilities and initiatives have been launched to aid researchers along this difficult path and facilitate progression of the next mAb blockbuster. Complementing this, there has been a renewed interest from the pharmaceutical industry to reconnect with academia in order to boost dwindling pipelines and encourage innovation. In this review, we examine the steps required to take a therapeutic mAb from discovery through early stage preclinical development and toward becoming a feasible clinical candidate. Discussion of the technologies used for mAb discovery, production in mammalian cells and innovations in single-use bioprocessing is included. We also examine regulatory requirements for product quality and characterization that should be considered at the earliest stages of mAb development. We provide details on the facilities available to help researchers and small-biotech build value into early stage product development, and include examples from within our own facility of how technologies are utilized and an analysis of our client base.


Asunto(s)
Centros Médicos Académicos/organización & administración , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/uso terapéutico , Industria Farmacéutica/organización & administración , Tecnología Farmacéutica/métodos , Animales , Células CHO , Cricetinae , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Ratones , Proyectos de Investigación
19.
Mol Biotechnol ; 48(2): 109-15, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21104043

RESUMEN

Transient gene expression systems in mammalian cells continue to grow in popularity due to their capacity to produce significant amounts of recombinant protein in a rapid and scalable manner, without the lengthy time periods and resources required for stable cell line development. Traditionally, production of recombinant monoclonal antibodies for pre-clinical assessment by transient expression in CHO cells has been hampered by low titers. In this report, we demonstrate transient monoclonal antibody titers of 140 mg/l with CHO cells using the episomal-based transient expression system, Epi-CHO. Such titers were achieved by implementing an optimized transfection protocol incorporating mild-hypothermia and through screening of a variety of chemically defined and serum-free media for their ability to support elevated and prolonged viable cell densities post-transfection, and in turn, improve recombinant protein yields. Further evidence supporting Epi-CHO's capacity to enhance transgene expression is provided, where we demonstrate higher transgene mRNA and protein levels of two monoclonal antibodies and a destabilized enhanced green fluorescent protein with Epi-CHO compared to cell lines deficient in plasmid DNA replication and/or retention post-transfection. The results demonstrate the Epi-CHO system's capacity for the rapid production of CHO cell-derived recombinant monoclonal antibodies in serum-free conditions.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Anticuerpos Monoclonales/genética , Western Blotting , Células CHO , Cricetinae , Cricetulus , Electroforesis en Gel de Poliacrilamida , Liposomas , Proteínas Recombinantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
20.
Stem Cell Res ; 6(1): 1-12, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21075697

RESUMEN

Use of stem cells, whether adult or embryonic for clinical applications to treat diseases such as Parkinson's, macular degeneration or Type I diabetes will require a homogenous population of mature, terminally differentiated cells. A current area of intense interest is the development of defined surfaces for stem cell derivation, maintenance, proliferation and subsequent differentiation, which are capable of replicating the complex cellular environment existing in vivo. During development many cellular cues result from integrin signalling induced by the local extracellular matrix. There are 24 known integrin heterodimers comprised of one of 18 α subunits and one of 8 ß subunits and these have a diverse range of functions mediating cell-cell adhesion, growth factor receptor responses and intracellular signalling cascades for cell migration, differentiation, survival and proliferation. We discuss here a brief summary of defined conditions for human embryonic stem cell culture together with a description of integrin function and signalling pathways. The importance of integrin expression during development is highlighted as critical for lineage specific cell function and how consideration of the integrin expression profile should be made while differentiating stem cells for use in therapy. In addition this review summarises the known integrin expression profiles for human embryonic stem cells and 3 common adult stem cell types: mesenchymal, haematopoietic and neural. We then outline some of the possible technologies available for investigating cell-extracellular matrix interactions and subsequent integrin mediated cell responses.


Asunto(s)
Técnicas de Cultivo de Célula , Tratamiento Basado en Trasplante de Células y Tejidos , Integrinas/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular , Humanos , Integrinas/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA