CONFOCHECK. Innovation with Integrity. Infrared Protein Analysis FT-IR

Similar documents
Analysis of Polymers and Plastics. Innovation with Integrity. Quality Control & Failure Analysis FT-IR

Application Note # AN112 Failure analysis of packaging materials

Analysis of Polymers and Plastics. Innovation with Integrity. Quality Control & Failure Analysis FTIR

EM 27/SUN Series. Innovation with Integrity. For Atmospheric Measurements FT-IR

Chirascan 6-Cell Peltier Cell Holder: Rapid Optimisation of Buffer Conditions for Stabilising Protein Therapeutics

Everyday NMR. Innovation with Integrity. Why infer when you can be sure? NMR

Use of DLS/Raman to study the thermal unfolding process of lysozyme

Agilent Cary 630 FTIR Spectrometer Supporting Organic Synthesis in Academic Teaching Labs

ALPHA. Innovation with Integrity. The very compact and smart FTIR spectrometer FTIR

Physical principles of IR and Raman. Infrared Spectroscopy

Meet Stunner: The one-shot protein concentration and sizing combo

Agilent Cary 630 FTIR sample interfaces combining high performance and versatility

Meet Our Uncle: 12 Stability Applications on One Platform

Supporting Information

Detection of trace contamination on metal surfaces using the handheld Agilent 4100 ExoScan FTIR

Understanding the conformational stability of protein therapeutics using Raman spectroscopy

Ligand Controlled Assembly of Hexamers, Dihexamers, and Linear Multihexamer Structures by the Engineered, Acylated, Insulin Degludec

Introduction to Fourier Transform Infrared Spectroscopy

Quantification Strategies Using the High Sensitivity Protein 250 Assay for the Agilent 2100 Bioanalyzer. Technical Note. Abstract

Highly automated protein formulation development: a case study

Acidic electrolyzed water (ph: 2.7; ORP:1150 mv; free chlorine: 60 ppm) was generated at 9-12

Isothermal experiments characterize time-dependent aggregation and unfolding

S2 PICOFOX. Innovation with Integrity. Spectrometry Solutions TXRF

Introduction to Fourier Transform Infrared Spectroscopy

Understanding the colloidal stability of protein therapeutics using dynamic light scattering

The DialPath Solution an Easier Way to Analyze Liquids By FTIR. Yanqia Wang, Ph.D. Application Engineer - FTIR Agilent Technologies May 7, 2015

Bruker Daltonics. EASY-nLC. Tailored HPLC for nano-lc-ms Proteomics. Nano-HPLC. think forward

Natural Products. Innovation with Integrity. High Performance NMR Solutions for Analysis NMR

Far UV Performance of the LAMBDA 850/950 UV/Vis and UV/Vis/NIR Research Spectrophotometers

Application Note LCMS-112 A Fully Automated Two-Step Procedure for Quality Control of Synthetic Peptides

Supplementary Figures

MM 2. Innovation with Integrity. The new Generation of Mobile Mass Spectrometers. Defence CBRNE

Protein separation and characterization

INSTRUCTION MANUAL. Lowry Assay Kit. Kit for Protein Quantification. (Cat. No )

BIOLIGHT STUDIO IN ROUTINE UV/VIS SPECTROSCOPY

The Plus for your lab!

Intelligent NMR productivity tools

Process Analytical Technology Diagnosis, Optimization and Monitoring of Chemical Processes

S8 TIGER Series 2 for ASTM D 6443

Precision and accuracy of protein size determination using the ActiPix TDA200 Nano-Sizing System

High-throughput Quantification of DNA for NGS Library Prep with the Zephyr G3 Workstation and the VICTOR Nivo Plate Reader

Protein Structure. W. M. Grogan, Ph.D. OBJECTIVES

Gyrolab Protein A Kit Quantification of residual Protein A ligands in the presence of excess amounts of IgG

ESPRIT Feature. Innovation with Integrity. Particle detection and chemical classification EDS

IR Biotyper. Innovation with Integrity. Microbial typing for real-time epidemiology FT-IR

Principles of Physical Biochemistry

IFS 125HR. Innovation with Integrity. FT-IR Spectrometer for Highest Spectral Resolution FT-IR

IRDye 800CW Protein Labeling Kit Low MW

MALVERN ADVANCED GPC/SEC DETECTORS SETTING THE STANDARD MOLECULAR WEIGHT MOLECULAR SIZE MOLECULAR STRUCTURE

4.3A: Electronic transitions

HYPHENATED TECHNOLOGY GUIDE

SUPPLEMENTARY INFORMATION

Key Terms (1 point each). Fill in the blank with the proper term. Each term may be used only once.

Data sheet. UV-Tracer TM Biotin-Maleimide. For Labeling of Thiol-groups with UV-detectable Biotin CLK-B Description

Dendritic Star Polymer of Polyacrylamide Based on β-cyclodextrin Trimer: A. Flocculant and Drug Vehicle

Denaturation and renaturation of proteins

ProPac WCX-10 Columns

Chemical Analysis. Low Level Oxygenates Analyzer. Trace Analysis of Oxygenates in Hydrocarbon Matrices. Gas Chromatography.

ion mobility spectrometry IR spectroscopy

Accurate Mass Measurement for Intact Proteins using ESI-oa-TOF. Application Note. Donghui Yi and Christine Miller Agilent Technologies

Methods for the study of the conformation of folding intermediates

Biomolecules: lecture 10

HYPHENATED TECHNOLOGY GUIDE

Mid-IR Sampling Techniques for Biological Molecules

Supporting Information

Advanced Pharmaceutical Analysis

Preview Slides for Introduction to Protein Formulation and Delivery

Design and Development of a Smartphone Based Visible Spectrophotometer for Analytical Applications

for XPS surface analysis

Analysis of a hybrid TATA box binding protein originating from mesophilic and thermophilic donor organisms

Thermo Scientific LTQ Orbitrap Velos Hybrid FT Mass Spectrometer

Dental Biochemistry EXAM I

Non-Interfering Protein Assay Kit Cat. No

1 Introduction FOURIER TRANSFORM INFRARED STUDIES IN SOLID EGG WHITE LYSOZYME. 1C/94/403 INTERNAL REPORT (Limited Distribution)

Alkaline Phosphatase Labeling Kit-NH2

Fragment-based drug discovery

11,12-EET/DHET ELISA kit

Sepax SRT -C, Zenix -C SEC Phases

1 st European Union Science Olympiad in Dublin, Ireland. task B

Supporting information

Compact Knowledge: Absorbance Spectrophotometry. Flexible. Reliable. Personal.

Finnigan LCQ Advantage MAX

penta-hilic UHPLC COLUMNS

Strategies to compare far-uv Circular Dichroism spectra of similar proteins using Chirascan -plus ACD

Exploration of Protein Folding

Infrared Spectroscopy of Proteins

Supporting Information. Labeled Ligand Displacement: Extending NMR-based Screening of Protein Targets

Bis sulfone Reagents. Figure 1.

Thermo Finnigan LTQ. Specifications

WALKUP LC/MS FOR PHARMACEUTICAL R&D

Application Note 12: Fully Automated Compound Screening and Verification Using Spinsolve and MestReNova

10 REASONS THAT THE ZETASIZER NANO IS CHOSEN FOR SCIENTIFIC SUCCESS

International Journal of Chemical Sciences

Diffusion of urea through membranes

Human rheumatoid factor (RF) antibody (IgM) ELISA Kit

Investigation of physiochemical interactions in

Chemical Analysis. PIONA+ Analyzer. Characterization of Engine Fuels by Hydrocarbon Group Type. Gas Chromatography. think forward

HPLC Columns. HILICpak VT-50 2D MANUAL. Shodex HPLC Columns Europe, Middle East, Africa, Russia

Optical density measurements automatically corrected to a 1-cm pathlength with PathCheck Technology

Contents. xiii. Preface v

Transcription:

CONFOCHECK Infrared Protein Analysis Innovation with Integrity FT-IR

CONFOCHECK: FT-IR System for Protein Analytics FT-IR Protein Analysis Infrared spectroscopy measures molecular vibrations due to the specific absorption of infrared (IR) radiation by chemical bonds. For over 35 years, it has been known that the shape and frequency of the amide I band, which is assigned to the C=O stretching vibration within peptide bonds, is characteristic for the structure of the studied protein. From this single band the presence and relative percentage of secondary structure elements (α-helix, β-strand) in a protein can be determined and structural changes detected with high sensitivity. The CONFOCHECK is a dedicated Fourier transform infrared (FT-IR) spectrometer system for protein analysis in aqueous solution. The main applications are the detection of conformational changes, concentration analysis and determination of the secondary structure. Its specific design facilitates a fast data acquisition with a high sample throughput and includes calibrations for protein concentration and secondary structure analysis.

FT-IR Protein Analysis Information Content of IR Protein Spectra The information content of the IR protein spectrum is not limited to the amide I band. Since IR spectroscopic data follows Lambert-Beer s law, the protein spectrum can be utilized for concentration determination. Also, side chains of various amino acids can be monitored and directly compared under varying environmental conditions (ph, salts, ligands), or in mutant proteins with altered primary sequence. Moreover, all other chemical components used in protein samples like certain ph buffers or proteinstabilizing agents are also represented in IR spectra by characteristic bands. Therefore, this method is also applied to reveal any changes of the chemical environment within the aqueous protein solution. In general the concentration of any molecular analyte dissolved in water can be determined by the CONFOCHECK, often down to the ppm level. Fig. 1A: Secondary Structure information Fig. 1B: Lysozyme in water, low concentrations Fig. 1C: Lysozyme in water, high concentrations Figure 1A: The amide I band of the IR spectrum of a protein reflects its secondary structure. Here, IR spectra of Hemoglobin and Tendamistat are shown. The Hemoglobin amide I band has a position (~1652 cm -1 ) and shape clearly indicative of a protein with a high α-helical content. The Tendamistat amide I band is characteristic (broad peak at ~1635 cm -1 ) of a protein with a high β-sheet content. Figures 1B and 1C: IR protein spectra follow Lambert-Beer s law and are informative of concentration. A dilution series of Lysozyme in phosphate buffer saline (PBS) was generated and the IR spectrum at each concentration was acquired. Figure 1B contains Lysozyme IR spectra from a low concentration (0.15 μg/μl - 2.2 μg/μl), 100 sec acquisition time. Figure 1C shows Lysozyme IR spectra from high concentrations (2.2 μg/μl - 45 μg/μl), 25 sec acquisition time. The spectra clearly demonstrate the correlation between IR signal intensity and protein amount over a large dynamic range.

FT-IR Protein Determination and Structure Analysis The demanding research lab and biopharmaceutical environments necessitate that an IR system provides accurate and reproducible protein measurements, rapid analysis (< 1 minute), and ease of use. The CONFO- CHECK system satisfies these demands. The system is unique in that it combines the necessary hardware with software for precise spectral measurement and interpretation using internal calibration models. Stability Study Stability studies of an antibody using the AquaSpec transmission accessory. Fig. 2A: Native and stressed antibody The stability of the system permits high sensitivity (dynamic range: 0.03 to >200 µg/µl) and direct analysis of proteins in any biological buffer with a spectrum acquisition time of under 1 minute. Due to its large dynamic range dilution of protein samples is not required for infrared protein analysis which makes it superior to CD spectroscopy and biochemical staining techniques. IR spectroscopy can also be applied to quantify, in parallel, other components of aqueous samples like detergents, sugars, buffer components, and contaminants (e.g. urea, acetonitrile, etc). The robustness of the system enables the resolution of small differences (<1 %) in protein structure, critical to assessing protein stability to differing stresses, formulations, and conjugations. These structural changes include conversion form α-helices to β-sheets; from intra- to intermolecular β-sheets; and unfolding of α-helix and β-sheet. Also, the degree of protein aggregation can be monitored by quantifying intermolecular beta sheet formation. The inclusion of internal concentration and secondary structure calibrations minimizes user setup time and simplifies spectra interpretation. Figure 2A: Examination of antibody stability by difference spectroscopy. The difference spectrum (green) of the stressed antibody (red IR spectrum) minus the unstressed antibody (blue IR spectrum) reveals a loss of intramolecular β-sheets (negative band at 1640 cm -1 ) and formation of intermolecular β-sheet (positive band at 1622 cm -1 ). These secondary structure changes are quantifiable. Fig. 2B: Antibody stability in different formulations Even structural effects on a protein upon binding to drugs or interacting with substrates can be measured with FT-IR-spectroscopy. The CONFOCHECK enables also to study the formation of multimeric structures like aggregates or fibrills, often in real time. Typical applications in Pharma: Pre-formulation and formulation studies: determine the effects of differing formulations on protein stability, quantify protein aggregation, analyze protein conjugates Forced degradation studies: examine protein stability and aggregation, perform accelerated stability tests (ph, temperature, mutations, etc ) Quality control: test protein concentration, structure, stability, and purity in the production environment Binding studies: analyze the effects of protein ligand binding on protein structure Figure 2B: Identification of the optimum antibody stabilizing formulation. The blue spectrum is the initial protein spectrum of the unstressed reference sample against which the difference spectra were calculated. The difference spectra originate from six similarly stressed samples containing the same antibody but being formulated differently. More stabilizing formulations will generate difference spectra with lower intensity. Therefore, the most stable protein formulation is formulation number 5. The ratio of each difference spectrum with the amide I band of the native protein correlates to the amount of structurally changed protein. Therefore, conformational changes of proteins are not only detected but also quantified by IR spectroscopy.

Temperature Ramp Fig. 3A: Antibody spectra in temperature ramp Thermal stability studies of a therapeutic antibody using the BioATR II accessory. The CONFOCHECK software permits the user designated configuration of temperature ramps. When the temperature range (maximum from 0-95 C), increment and equilibration time are set, the system starts to acquire automatically a spectrum at each predefined temperature. The IR spectra of an antibody at different temperatures (25-95 C) are shown (Figure 3A). As indicated by the unchanged amide I band shape the protein maintains its native β-sheet structure up to ~60 C. Further heating causes the amide I band maximum at 1636 cm -1 to decrease in intensity indicative of intramolecular-β-sheets unfolding. Unfolding of the native secondary structure leads to intermolecularβ-sheet formation reflected by an increasing IR signal at 1623 cm -1. Fig. 3B: Temperature induced structure changes lgg The percentages of the structural changes (Figure 3B) at each temperature were determined by calculating the relative signal change in the amide I band (difference spectroscopy as shown in Fig. 2B). Using the derivative of the curves of Figure 3B, two melting points of different structural domains of the antibody can be distinguished; one at ~66 C and one at ~79 C (Figure 3C). Fig. 3C: Denaturation temperature lgg Protein Dynamics software

CONFOCHECK: A Dedicated FT-IR System for Protein Analysis The CONFOCHECK is a compact IR spectrometer system dedicated to the analysis of proteins in aqueous solutions. Its robust design permits fast data acquisition while maintaining high sensitivity thus making high sample throughput possible. The system is controlled by a user friendly software interface that guides the user through the measurement procedure for spectral acquisition and temperature ramps. The system includes internal calibrations for protein concentration determination and secondary structure prediction, greatly simplifying experimental setup and spectral interpretation. For analysis, a protein sample is injected into the AquaSpec IR transmission cell in a flow-through manner. Typically, 20 μl of protein sample are sufficient to load the cell and to obtain a high quality protein spectrum within a minute. From the spectrum, the α-helix and β-sheet content within the protein and the protein s concentration are determined within seconds. To follow temperature induced structural changes, 20 μl of a protein solution are pipetted onto the measurement crystal of the BioATR II accessory. The sampling area is sealed before the user defined ramping program is started. The sample chamber temperature is controlled by the user software via a thermostat: The measurements are performed automatically in the programmed temperature range (max. 0 C - 95 C) with defined increments and equilibration times. The resulting set of protein spectra allows the assignment of the type of structural changes exhibited over increasing temperatures and the calculation of the melting point of the protein secondary structures. In addition to temperature ramping experiments, the BioATR II is also very suitable for analyzing insoluble (membrane bound) or precipitated proteins as well as monitoring protein aggregation or fibrillation processes. Protein lyophilizates can also be analyzed with the CONFOCHECK system, requiring low amounts of dried proteins (1-10 mg) for a typical measurement.

CONFOCHECK utilizes three FT-IR sampling accessory cells to cover all protein applications. AquaSpec cell This flow through transmission cell is used to study proteins in aqueous solution. The analysis of conformational stability, concentration and secondary structure composition are typical applications. For biocompatibility, all tubes are made from PTFE. In-line filters are integrated to prevent contamination of the cell volume by aggregated and precipitated protein. BioATR II This ATR-accessory is dedicated to perform temperature ramp measurements for the investigation of thermally induced structural changes in proteins. Its optical path is optimized for the sensitive measurement of aqueous samples. Furthermore the BioATR II is suitable for analyzing aggregation and precipitation processes of water-soluble proteins and for the investigation of membrane proteins. Applications for CONFOCHECK Protein quantification (down to 0.03 mg/ml in aqueous solution) Detection of conformational changes in proteins Protein dynamics: Unfolding, aggregation (temperature induced conformational changes) Determination of secondary structure Quantification of any molecular analyte in aqueous samples BioATR II with dialysis functionality With this ATR-accessory dialysis experiments performed in the sampling compartment can be followed IR-spectroscopically. Low molecular weight compounds (ligands, salts, protons) can be applied to the protein of interest by dialysis in order to reveal structural alterations caused by the protein-ligand-interaction.

Application Support Bruker is staffed by expert life scientists and engineers that have in depth knowledge of protein analytics and instrumentation. Our product specialists are prepared to assist you with method development, selection and usage of sampling accessories, choices of optical components and software operation. We offer customized instruction and support packages to fit your needs. Bruker systems are designed to provide many years of trouble-free operation, but should a problem occur a network of Bruker Optics offices and representatives throughout the world are ready to promptly respond to your needs. Professional installations and a high standard of post-delivery service are commitments Bruker makes to each of its customers. www.bruker.com/optics Bruker Optics Inc. Bruker Optik GmbH Bruker Hong Kong Ltd. Billerica, MA USA Phone +1 (978) 439-9899 Fax +1 (978) 663-9177 info@brukeroptics.com Ettlingen Germany Phone +49 (7243) 504-2000 Fax +49 (7243) 504-2050 info@brukeroptics.de Hong Kong Phone +852 2796-6100 Fax +852 2796-6109 hk@brukeroptics.com.hk Bruker Optics is continually improving its products and reserves the right to change specifications without notice. 2014 Bruker Optics BOPT-4000058-02