Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Front Bioeng Biotechnol ; 12: 1397465, 2024.
Article in English | MEDLINE | ID: mdl-38812919

ABSTRACT

Protein crystallization as opposed to well-established chromatography processes has the benefits to reduce production costs while reaching a comparable high purity. However, monitoring crystallization processes remains a challenge as the produced crystals may interfere with analytical measurements. Especially for capturing proteins from complex feedstock containing various impurities, establishing reliable process analytical technology (PAT) to monitor protein crystallization processes can be complicated. In heterogeneous mixtures, important product characteristics can be found by multivariate analysis and chemometrics, thus contributing to the development of a thorough process understanding. In this project, an analytical set-up is established combining offline analytics, on-line ultraviolet visible light (UV/Vis) spectroscopy, and in-line Raman spectroscopy to monitor a stirred-batch crystallization process with multiple phases and species being present. As an example process, the enzyme Lactobacillus kefir alcohol dehydrogenase (LkADH) was crystallized from clarified Escherichia coli (E. coli) lysate on a 300 mL scale in five distinct experiments, with the experimental conditions changing in terms of the initial lysate solution preparation method and precipitant concentration. Since UV/Vis spectroscopy is sensitive to particles, a cross-flow filtration (cross-flow filtration)-based bypass enabled the on-line analysis of the liquid phase providing information on the lysate composition regarding the nucleic acid to protein ratio. A principal component analysis (PCA) of in situ Raman spectra supported the identification of spectra and wavenumber ranges associated with productspecific information and revealed that the experiments followed a comparable, spectral trend when crystals were present. Based on preprocessed Raman spectra, a partial least squares (PLS) regression model was optimized to monitor the target molecule concentration in real-time. The off-line sample analysis provided information on the crystal number and crystal geometry by automated image analysis as well as the concentration of LkADH and host cell proteins (HCPs) In spite of a complex lysate suspension containing scattering crystals and various impurities, it was possible to monitor the target molecule concentration in a heterogeneous, multi-phase process using spectroscopic methods. With the presented analytical set-up of off-line, particle-sensitive on-line, and in-line analyzers, a crystallization capture process can be characterized better in terms of the geometry, yield, and purity of the crystals.

2.
Anal Bioanal Chem ; 414(21): 6379-6391, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35661232

ABSTRACT

Since preparative chromatography is a sustainability challenge due to large amounts of consumables used in downstream processing of biomolecules, protein crystallization offers a promising alternative as a purification method. While the limited crystallizability of proteins often restricts a broad application of crystallization as a purification method, advances in molecular biology, as well as computational methods are pushing the applicability towards integration in biotechnological downstream processes. However, in industrial and academic settings, monitoring protein crystallization processes non-invasively by microscopic photography and automated image evaluation remains a challenging problem. Recently, the identification of single crystal objects using deep learning has been the subject of increased attention for various model systems. However, the advancement of crystal detection using deep learning for biotechnological applications is limited: robust models obtained through supervised machine learning tasks require large-scale and high-quality data sets usually obtained in large projects through extensive manual labeling, an approach that is highly error-prone for dense systems of transparent crystals. For the first time, recent trends involving the use of synthetic data sets for supervised learning are transferred, thus generating photorealistic images of virtual protein crystals in suspension (PCS) through the use of ray tracing algorithms, accompanied by specialized data augmentations modelling experimental noise. Further, it is demonstrated that state-of-the-art models trained with the large-scale synthetic PCS data set outperform similar fine-tuned models based on the average precision metric on a validation data set, followed by experimental validation using high-resolution photomicrographs from stirred tank protein crystallization processes.


Subject(s)
Machine Learning , Neural Networks, Computer , Algorithms , Crystallization , Image Processing, Computer-Assisted/methods , Proteins
3.
Biotechnol J ; 15(11): e2000010, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32302461

ABSTRACT

Technical crystallization is an attractive method to purify recombinant proteins. However, it is rarely applied due to the limited crystallizability of many proteins. To overcome this limitation, single amino acid exchanges are rationally introduced to enhance intermolecular interactions at the crystal contacts of the industrially relevant biocatalyst Lactobacillus brevis alcohol dehydrogenase (LbADH). The wildtype (WT) and the best crystallizing and enzymatically active LbADH mutants K32A, D54F, Q126H, and T102E are produced with Escherichia coli and subsequently crystallized from cell lysate in stirred mL-crystallizers. Notwithstanding the high host cell protein (HCP) concentrations in the lysate, all mutants crystallize significantly faster than the WT. Combinations of mutations result in double mutants with faster crystallization kinetics than the respective single mutants, demonstrating a synergetic effect. The almost entire depletion of the soluble LbADH fraction at crystallization equilibrium is observed, proving high yields. The HCP concentration is reduced to below 0.5% after crystal dissolution and recrystallization, and thus a 100-fold HCP reduction is achieved after two successive crystallization steps. The combination of fast kinetics, high yields, and high target protein purity highlights the potential of crystal contact engineering to transform technical crystallization into an efficient protein capture and purification step in biotechnological downstream processes.


Subject(s)
Biotechnology , Oxidoreductases , Alcohol Dehydrogenase/genetics , Crystallization , Crystallography, X-Ray , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...