1
|
Willis LF, Kapur N, Radford SE, Brockwell DJ. Biophysical Analysis of Therapeutic Antibodies in the Early Development Pipeline. Biologics 2024; 18:413-432. [PMID: 39723199 PMCID: PMC11669289 DOI: 10.2147/btt.s486345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
The successful progression of therapeutic antibodies and other biologics from the laboratory to the clinic depends on their possession of "drug-like" biophysical properties. The techniques and the resultant biophysical and biochemical parameters used to characterize their ease of manufacture can be broadly defined as developability. Focusing on antibodies, this review firstly discusses established and emerging biophysical techniques used to probe the early-stage developability of biologics, aimed towards those new to the field. Secondly, we describe the inter-relationships and redundancies amongst developability assays and how in silico methods aid the efficient deployment of developability to bring a new generation of cost-effective therapeutic proteins from bench to bedside more quickly and sustainably.
Collapse
Affiliation(s)
- Leon F Willis
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Sheena E Radford
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - David J Brockwell
- School of Molecular and Cellular Biology, Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
2
|
Liu Y, Hatch HW, Yuan G, Shen VK, Grishaev AV, Panchal J, Blanco M. Extracting Orientation and Distance-Dependent Interaction Potentials between Proteins in Solutions Using Small-Angle X-ray/Neutron Scattering. J Phys Chem Lett 2024; 15:12401-12407. [PMID: 39656919 DOI: 10.1021/acs.jpclett.4c02629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Nonspecific protein-protein interactions (PPIs) are key to understanding the behavior of proteins in solutions. However, experimentally measuring anisotropic PPIs as a function of orientation and distance has been challenging. Here, we propose to measure a new parameter, the generalized second virial coefficient, B22(Q), to address this challenge. B22(Q) can be measured by using small-angle X-ray/neutron scattering (SAXS/SANS) at finite Q values, where Q is the magnitude of the scattering wave vector. We develop the analytical theory here to calculate B22(Q) with any known interprotein potentials including anisotropic interaction potentials. This method overcomes the challenges and limitations of commonly used methods for extracting PPI information, namely, using integral approximations to solve the Ornstein-Zernike equation by fitting SAXS/SANS data. The accuracy of this analytical theory is further evaluated with computer simulations using a model system. Not only can our method greatly extend the capability of SAXS/SANS to investigate PPIs of many proteins, but it is also applicable to a wide variety of colloidal systems where anisotropic interaction potentials are important.
Collapse
Affiliation(s)
- Yun Liu
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Harold W Hatch
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Guangcui Yuan
- Center for Neutron Research, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Vincent K Shen
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Alexander V Grishaev
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Jainik Panchal
- Sterile and Specialty Products, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marco Blanco
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
3
|
Zhang T, Zheng J, Chen M, Li D, Sun Y, Liu R, Sun T. A mini review of polysaccharides from Zanthoxylum bungeanum maxim: Their extraction, purification, structural characteristics, bioactivity and potential applications. Int J Biol Macromol 2024; 282:137007. [PMID: 39486707 DOI: 10.1016/j.ijbiomac.2024.137007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/29/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
Zanthoxylum bungeanum Maxim (Z. bungeanum), commonly known as Sichuan pepper or Chinese prickly ash, is a deciduous shrub in the Rutaceae family, with a lengthy history of use as a food ingredient and traditional medicine in China. Z. bungeanum polysaccharides (ZBPs) represent one of the crucial bioactive components of Z. bungeanum, garnering global attention due to their potential medicinal value, culinary significance, and promising application prospects. The principal methods for extracting ZBPs are hot water extraction, ultrasound-assisted extraction, enzyme-assisted extraction and microbial fermentation extraction. However, the structural characteristics of ZBPs remain ambiguous, necessitating further exploration and elucidation of the structure-activity relationship using the advanced analytical techniques. In addition, ZBPs demonstrate diverse bioactivities, including antioxidant activity, neuroprotective effect, antibacterial activity, and the anti-fatigue effect, positioning them as promising candidates for various therapeutic and health-promoting applications. This review provides a comprehensive overview of the extraction, purification, structural characteristics, bioactivities, and potential applications of ZBPs, emphasizing the significant promise of ZBPs as valuable natural compounds with a range of bioactivities, supporting their further exploitation and application in various fields of industries and therapeutics.
Collapse
Affiliation(s)
- Ting Zhang
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China
| | - Jianfeng Zheng
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China
| | - Mengjie Chen
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China
| | - Dan Li
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China
| | - Yuan Sun
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China.
| | - Rui Liu
- Center of Pharmaceutical Engineering and Technology, Harbin University of Commerce, Harbin 150076, PR China.
| | - Tiedong Sun
- College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, PR China.
| |
Collapse
|
4
|
Fernandes DA. Comprehensive Review on Bubbles: Synthesis, Modification, Characterization and Biomedical Applications. Bioconjug Chem 2024; 35:1639-1686. [PMID: 39377727 DOI: 10.1021/acs.bioconjchem.4c00137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Accurate detection, treatment, and imaging of diseases are important for effective treatment outcomes in patients. In this regard, bubbles have gained much attention, due to their versatility. Bubbles usually 1 nm to 10 μm in size can be produced and loaded with a variety of lipids, polymers, proteins, and therapeutic and imaging agents. This review details the different production and loading methods for bubbles, for imaging and treatment of diseases/conditions such as cancer, tumor angiogenesis, thrombosis, and inflammation. Bubbles can also be used for perfusion measurements, important for diagnostic and therapeutic decision making in cardiac disease. The different factors important in the stability of bubbles and the different techniques for characterizing their physical and chemical properties are explained, for developing bubbles with advanced therapeutic and imaging features. Hence, the review provides important insights for researchers studying bubbles for biomedical applications.
Collapse
|
5
|
Jun T, Shin SH, Won YY. Engineered polymeric excipients for enhancing the stability of protein biologics: Poly(N-isopropylacrylamide)-poly(ethylene glycol) (PNIPAM-PEG) block copolymers. Int J Pharm 2024; 664:124636. [PMID: 39197798 DOI: 10.1016/j.ijpharm.2024.124636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 07/24/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
Protein therapeutics, particularly antibodies, depend on maintaining their native structures for optimal function. Hydrophobic interfaces, such as the air-water interface, can trigger protein aggregation and denaturation. While completely avoiding such interfacial exposures during manufacturing and storage is impractical, minimizing them is crucial for enhancing protein drug stability and extending shelf life. In the biologics industry, surfactants like polysorbates are commonly used as additives (excipients) to mitigate these undesirable interfacial exposures. However, polysorbates, the most prevalent choice, have recognized limitations in terms of polydispersity, purity, and stability, prompting the exploration of alternative excipients. The present study identifies poly(N-isopropylacrylamide)-poly(ethylene glycol) (PNIPAM-PEG) block copolymers as a promising alternative to polysorbates. Due to its stronger affinity for the air-water interface, PNIPAM-PEG significantly outperforms polysorbates in enhancing protein stability. This claim is supported by results from multiple tests. Accelerated dynamic light scattering (DLS) experiments demonstrate PNIPAM-PEG's exceptional efficacy in preserving IgG stability against surface-induced aggregation, surpassing conventional polysorbate excipients (Tween 80 and Tween 20) under high-temperature conditions. Additionally, circular dichroism (CD) spectroscopy results reveal conformational alterations associated with aggregation, with PNIPAM-PEG consistently demonstrates a greater protective effect by mitigating negative shifts at λ ≅ 220 nm, indicative of changes in secondary structure. Overall, this study positions PNIPAM-PEG as a promising excipient for antibody therapeutics, facilitating the development of more stable and effective biopharmaceuticals.
Collapse
Affiliation(s)
- Taesuk Jun
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Sung-Ho Shin
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN 47907, USA; Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
6
|
Liu X, Jean-Gilles R, Baginski J, Cai C, Yan R, Zhang L, Lance K, van der Loo JC, Davidson BL. Evaluation of a rapid multi-attribute combinatorial high-throughput UV-Vis/DLS/SLS analytical platform for rAAV quantification and characterization. Mol Ther Methods Clin Dev 2024; 32:101298. [PMID: 39170800 PMCID: PMC11338085 DOI: 10.1016/j.omtm.2024.101298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
Recombinant adeno-associated virus (rAAV)-based gene therapies are expanding in their application. Despite progress in manufacturing, current analytical methods for product quantification and characterization remain largely unchanged. Although critical for product and process development, in-process testing, and batch release, current analytical methods are labor-intensive, costly, and hampered by extended turnaround times and low throughput. The field requires more efficient, cost-effective analytical techniques capable of handling large sample quantities to accelerate product and process development. Here, we evaluated Stunner from Unchained Labs for quantifying and characterizing rAAVs and compared it with established analytical methods. Stunner is a combinatorial analytic technology platform that interpolates ultraviolet-visible (UV-Vis) absorption with static and dynamic light scattering (SLS/DLS) analysis to determine capsid and genomic titer, empty and full capsid ratio, and assess vector size and polydispersity. The platform offers empirical measurements with minimal sample requirements. Upon testing hundreds of rAAV vectors, comprising various serotypes and transgenes, the data show a strong correlation with established analytical methods and exhibit high reproducibility and repeatability. Some analyses can be applied to in-process samples from different purification stages and processes, fulfilling the demand for rapid, high-throughput analysis during development. In sum, the pipeline presented streamlines small- and large-batch analytics.
Collapse
Affiliation(s)
- Xueyuan Liu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | | | - Julia Baginski
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christina Cai
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ruilan Yan
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lili Zhang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Johannes C.M. van der Loo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly L. Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Sripada SA, Hosseini M, Ramesh S, Wang J, Ritola K, Menegatti S, Daniele MA. Advances and opportunities in process analytical technologies for viral vector manufacturing. Biotechnol Adv 2024; 74:108391. [PMID: 38848795 DOI: 10.1016/j.biotechadv.2024.108391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/14/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Viral vectors are an emerging, exciting class of biologics whose application in vaccines, oncology, and gene therapy has grown exponentially in recent years. Following first regulatory approval, this class of therapeutics has been vigorously pursued to treat monogenic disorders including orphan diseases, entering hundreds of new products into pipelines. Viral vector manufacturing supporting clinical efforts has spurred the introduction of a broad swath of analytical techniques dedicated to assessing the diverse and evolving panel of Critical Quality Attributes (CQAs) of these products. Herein, we provide an overview of the current state of analytics enabling measurement of CQAs such as capsid and vector identities, product titer, transduction efficiency, impurity clearance etc. We highlight orthogonal methods and discuss the advantages and limitations of these techniques while evaluating their adaptation as process analytical technologies. Finally, we identify gaps and propose opportunities in enabling existing technologies for real-time monitoring from hardware, software, and data analysis viewpoints for technology development within viral vector biomanufacturing.
Collapse
Affiliation(s)
- Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Mahshid Hosseini
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Srivatsan Ramesh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA
| | - Junhyeong Wang
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA
| | - Kimberly Ritola
- North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Neuroscience Center, Brain Initiative Neurotools Vector Core, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC, 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Biomanufacturing Training and Education Center, North Carolina State University, 890 Main Campus Dr, Raleigh, NC 27695, USA.
| | - Michael A Daniele
- Joint Department of Biomedical Engineering, North Carolina State University, and University of North Carolina, Chapel Hill, 911 Oval Dr., Raleigh, NC 27695, USA; North Carolina Viral Vector Initiative in Research and Learning (NC-VVIRAL), North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA; Department of Electrical and Computer Engineering, North Carolina State University, 890 Oval Dr, Raleigh, NC 27695, USA.
| |
Collapse
|
8
|
Dürauer A, Jungbauer A, Scharl T. Sensors and chemometrics in downstream processing. Biotechnol Bioeng 2024; 121:2347-2364. [PMID: 37470278 DOI: 10.1002/bit.28499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/14/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
The biopharmaceutical industry is still running in batch mode, mostly because it is highly regulated. In the past, sensors were not readily available and in-process control was mainly executed offline. The most important product parameters are quantity, purity, and potency, in addition to adventitious agents and bioburden. New concepts using disposable single-use technologies and integrated bioprocessing for manufacturing will dominate the future of bioprocessing. To ensure the quality of pharmaceuticals, initiatives such as Process Analytical Technologies, Quality by Design, and Continuous Integrated Manufacturing have been established. The aim is that these initiatives, together with technology development, will pave the way for process automation and autonomous bioprocessing without any human intervention. Then, real-time release would be realized, leading to a highly predictive and robust biomanufacturing system. The steps toward such automated and autonomous bioprocessing are reviewed in the context of monitoring and control. It is possible to integrate real-time monitoring gradually, and it should be considered from a soft sensor perspective. This concept has already been successfully implemented in other industries and requires relatively simple model training and the use of established statistical tools, such as multivariate statistics or neural networks. This review describes a scenario for integrating soft sensors and predictive chemometrics into modern process control. This is exemplified by selective downstream processing steps, such as chromatography and membrane filtration, the most common unit operations for separation of biopharmaceuticals.
Collapse
Affiliation(s)
- Astrid Dürauer
- Institute of Bioprocessing Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Alois Jungbauer
- Institute of Bioprocessing Science and Engineering, University of Natural Resources and Life Sciences, Vienna, Austria
- Austrian Centre of Industrial Biotechnology, Vienna, Austria
| | - Theresa Scharl
- Institute of Statistics, University of Natural Resources and Life Sciences, Vienna, Austria
| |
Collapse
|
9
|
Pispas I, Spiliopoulos N, Papagiannopoulos A. Biocompatible Preparation of Beta-Lactoglobulin/Chondroitin Sulfate Carrier Nanoparticles and Modification of Their Colloidal and Hydropathic Properties by Tween 80. Polymers (Basel) 2024; 16:1995. [PMID: 39065312 PMCID: PMC11280915 DOI: 10.3390/polym16141995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The electrostatic complexation of the protein beta-lactoglobulin (β-LG) with the anionic polysaccharide chondroitin sulfate (CS) and the subsequent stabilization by thermal treatment were studied to achieve the well-defined nanoparticles (NPs). The formation of the well-defined NPs was obtained at pH 4 with a hydrodynamic radius from 60 to 80 nm. NP aggregation was observed at pH 1.5 because of the loss of the anionic charge of chondroitin sulfate on the surface of the NPs. After thermal treatment, the NPs exhibited stability against a pH increase to pH 7 while a stronger aggregation at pH 1.5 was observed. Core-shell structures were found at pH 7 after thermal treatment, indicating a possible mechanism of partial disintegration. The addition of Tween 80 (T80) before thermal treatment led to the formation of T80 self-assemblies inside the NPs. This caused an increase in the hydrophobicity of the inner and outer surfaces of the NPs as it was observed by fluorescence spectroscopy. The ζ-potential of the complexes and NPs was about -20 mV while the presence of T80 did not affect it. FTIR spectra verified changes of the secondary structure of β-LG in its complexes with CS and T80. The thermally treated NPs exhibited high surface and overall hydrophobicity and stability in high salinity and biocompatible solutions. The thermally treated NPs showed colloidal and physicochemical stability for 1 month, which were enhanced by the addition of T80. Due to the nature of the precursors and their colloidal properties, the NPs are highly promising for applications as biocompatible drug delivery nanocarriers while T80 acts as an agent to modify their properties.
Collapse
Affiliation(s)
- Ioannis Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| | | | - Aristeidis Papagiannopoulos
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece;
| |
Collapse
|
10
|
Zacco E, Broglia L, Kurihara M, Monti M, Gustincich S, Pastore A, Plath K, Nagakawa S, Cerase A, Sanchez de Groot N, Tartaglia GG. RNA: The Unsuspected Conductor in the Orchestra of Macromolecular Crowding. Chem Rev 2024; 124:4734-4777. [PMID: 38579177 PMCID: PMC11046439 DOI: 10.1021/acs.chemrev.3c00575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 04/07/2024]
Abstract
This comprehensive Review delves into the chemical principles governing RNA-mediated crowding events, commonly referred to as granules or biological condensates. We explore the pivotal role played by RNA sequence, structure, and chemical modifications in these processes, uncovering their correlation with crowding phenomena under physiological conditions. Additionally, we investigate instances where crowding deviates from its intended function, leading to pathological consequences. By deepening our understanding of the delicate balance that governs molecular crowding driven by RNA and its implications for cellular homeostasis, we aim to shed light on this intriguing area of research. Our exploration extends to the methodologies employed to decipher the composition and structural intricacies of RNA granules, offering a comprehensive overview of the techniques used to characterize them, including relevant computational approaches. Through two detailed examples highlighting the significance of noncoding RNAs, NEAT1 and XIST, in the formation of phase-separated assemblies and their influence on the cellular landscape, we emphasize their crucial role in cellular organization and function. By elucidating the chemical underpinnings of RNA-mediated molecular crowding, investigating the role of modifications, structures, and composition of RNA granules, and exploring both physiological and aberrant phase separation phenomena, this Review provides a multifaceted understanding of the intriguing world of RNA-mediated biological condensates.
Collapse
Affiliation(s)
- Elsa Zacco
- RNA
Systems Biology Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, Via Enrico Melen, 83, 16152 Genova, Italy
| | - Laura Broglia
- RNA
Systems Biology Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, Via Enrico Melen, 83, 16152 Genova, Italy
| | - Misuzu Kurihara
- RNA
Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Michele Monti
- RNA
Systems Biology Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, Via Enrico Melen, 83, 16152 Genova, Italy
| | - Stefano Gustincich
- Central
RNA Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, Via Enrico Melen, 83, 16152 Genova, Italy
| | - Annalisa Pastore
- UK
Dementia Research Institute at the Maurice Wohl Institute of King’s
College London, London SE5 9RT, U.K.
| | - Kathrin Plath
- Department
of Biological Chemistry, David Geffen School
of Medicine at the University of California Los Angeles, Los Angeles, California 90095, United States
| | - Shinichi Nagakawa
- RNA
Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Andrea Cerase
- Blizard
Institute,
Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 4NS, U.K.
- Unit
of Cell and developmental Biology, Department of Biology, Università di Pisa, 56123 Pisa, Italy
| | - Natalia Sanchez de Groot
- Unitat
de Bioquímica, Departament de Bioquímica i Biologia
Molecular, Universitat Autònoma de
Barcelona, 08193 Barcelona, Spain
| | - Gian Gaetano Tartaglia
- RNA
Systems Biology Lab, Center for Human Technologies, Istituto Italiano di Tecnologia, Via Enrico Melen, 83, 16152 Genova, Italy
- Catalan
Institution for Research and Advanced Studies, ICREA, Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
11
|
Farzam F, Dabirmanesh B. Experimental techniques for detecting and evaluating the amyloid fibrils. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 206:183-227. [PMID: 38811081 DOI: 10.1016/bs.pmbts.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Amyloid fibrils are insoluble proteins with intricate β-sheet structures associated with various human diseases, including Parkinson's, Alzheimer's, and prion diseases. Proteins can form aggregates when their structure is misfolded, resulting in highly organized amyloid fibrils or amorphous aggregates. The formation of protein aggregates is a promising research field for mitigating diseases and the pharmaceutical and food industries. It is important to monitor and minimize the appearance of aggregates in these protein products. Several methods exist to assess protein aggregation, that includes from basic investigations to advanced biophysical techniques. Physicochemical parameters such as molecular weight, conformation, structure, and dimension are examined to study aggregation. There is an urgent need to develop methods for the detection of protein aggregation and amyloid fibril formation both in vitro and in vivo. This chapter focuses on a comprehensive discussion of the methods used to characterize and evaluate aggregates and amyloid fibrils.
Collapse
Affiliation(s)
- Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bahareh Dabirmanesh
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
12
|
Renodon-Corniere A, Mikawa T, Kuwabara N, Ito K, Levitsky D, Iwasaki H, Takahashi M. Human Rad51 Protein Requires Higher Concentrations of Calcium Ions for D-Loop Formation than for Oligonucleotide Strand Exchange. Int J Mol Sci 2024; 25:3633. [PMID: 38612444 PMCID: PMC11011376 DOI: 10.3390/ijms25073633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Human Rad51 protein (HsRad51)-promoted DNA strand exchange, a crucial step in homologous recombination, is regulated by proteins and calcium ions. Both the activator protein Swi5/Sfr1 and Ca2+ ions stimulate different reaction steps and induce perpendicular DNA base alignment in the presynaptic complex. To investigate the role of base orientation in the strand exchange reaction, we examined the Ca2+ concentration dependence of strand exchange activities and structural changes in the presynaptic complex. Our results show that optimal D-loop formation (strand exchange with closed circular DNA) required Ca2+ concentrations greater than 5 mM, whereas 1 mM Ca2+ was sufficient for strand exchange between two oligonucleotides. Structural changes indicated by increased fluorescence intensity of poly(dεA) (a poly(dA) analog) reached a plateau at 1 mM Ca2+. Ca2+ > 2 mM was required for saturation of linear dichroism signal intensity at 260 nm, associated with rigid perpendicular DNA base orientation, suggesting a correlation with the stimulation of D-loop formation. Therefore, Ca2+ exerts two different effects. Thermal stability measurements suggest that HsRad51 binds two Ca2+ ions with KD values of 0.2 and 2.5 mM, implying that one step is stimulated by one Ca2+ bond and the other by two Ca2+ bonds. Our results indicate parallels between the Mg2+ activation of RecA and the Ca2+ activation of HsRad51.
Collapse
Affiliation(s)
| | - Tsutomu Mikawa
- RIKEN Center for Biosystems Dynamics Research, Yokohama 230-0045, Japan;
| | - Naoyuki Kuwabara
- Structural Biology Research Center, Institute of Materials Structure Science, High Energy Accelerator Research Organization (KEK), Tsukuba 305-0801, Japan;
| | - Kentaro Ito
- Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan;
| | - Dmitri Levitsky
- Nantes Université, CNRS, US2B, UMR 6286, F-44000 Nantes, France; (A.R.-C.); (D.L.)
| | - Hiroshi Iwasaki
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan;
- Innovative Science Institute, Tokyo Institute of Technology, Yokohama 226-8503, Japan
| | - Masayuki Takahashi
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo 152-8550, Japan;
| |
Collapse
|
13
|
Forder JK, Palakollu V, Adhikari S, Blanco MA, Derebe MG, Ferguson HM, Luthra SA, Munsell EV, Roberts CJ. Electrostatically Mediated Attractive Self-Interactions and Reversible Self-Association of Fc-Fusion Proteins. Mol Pharm 2024; 21:1321-1333. [PMID: 38334418 DOI: 10.1021/acs.molpharmaceut.3c01009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Attractive self-interactions and reversible self-association are implicated in many problematic solution behaviors for therapeutic proteins, such as irreversible aggregation, elevated viscosity, phase separation, and opalescence. Protein self-interactions and reversible oligomerization of two Fc-fusion proteins (monovalent and bivalent) and the corresponding fusion partner protein were characterized experimentally with static and dynamic light scattering as a function of pH (5 and 6.5) and ionic strength (10 mM to at least 300 mM). The fusion partner protein and monovalent Fc-fusion each displayed net attractive electrostatic self-interactions at pH 6.5 and net repulsive electrostatic self-interactions at pH 5. Solutions of the bivalent Fc-fusion contained higher molecular weight species that prevented quantification of typical interaction parameters (B22 and kD). All three of the proteins displayed reversible self-association at pH 6.5, where oligomers dissociated with increased ionic strength. Coarse-grained molecular simulations were used to model the self-interactions measured experimentally, assess net self-interactions for the bivalent Fc-fusion, and probe the specific electrostatic interactions between charged amino acids that were involved in attractive electrostatic self-interactions. Mayer-weighted pairwise electrostatic energies from the simulations suggested that attractive electrostatic self-interactions at pH 6.5 for the two Fc-fusion proteins were due to cross-domain interactions between the fusion partner domain(s) and the Fc domain.
Collapse
Affiliation(s)
- James K Forder
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - Veerabhadraiah Palakollu
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19713, United States
| | - Sudeep Adhikari
- Analytical R&D, Digital & NMR Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marco A Blanco
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Mehabaw Getahun Derebe
- Discovery Biologics, Protein Sciences, Merck & Co., Inc., South San Francisco, California 94080, United States
| | - Heidi M Ferguson
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Suman A Luthra
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Erik V Munsell
- Discovery Pharmaceutical Sciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19713, United States
| |
Collapse
|
14
|
Alfonso C, Sobrinos-Sanguino M, Luque-Ortega JR, Zorrilla S, Monterroso B, Nuero OM, Rivas G. Studying Macromolecular Interactions of Cellular Machines by the Combined Use of Analytical Ultracentrifugation, Light Scattering, and Fluorescence Spectroscopy Methods. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 3234:89-107. [PMID: 38507202 DOI: 10.1007/978-3-031-52193-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Cellular machines formed by the interaction and assembly of macromolecules are essential in many processes of the living cell. These assemblies involve homo- and hetero-associations, including protein-protein, protein-DNA, protein-RNA, and protein-polysaccharide associations, most of which are reversible. This chapter describes the use of analytical ultracentrifugation, light scattering, and fluorescence-based methods, well-established biophysical techniques, to characterize interactions leading to the formation of macromolecular complexes and their modulation in response to specific or unspecific factors. We also illustrate, with several examples taken from studies on bacterial processes, the advantages of the combined use of subsets of these techniques as orthogonal analytical methods to analyze protein oligomerization and polymerization, interactions with ligands, hetero-associations involving membrane proteins, and protein-nucleic acid complexes.
Collapse
Affiliation(s)
- Carlos Alfonso
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain.
| | - Marta Sobrinos-Sanguino
- Molecular Interactions Facility, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Juan Román Luque-Ortega
- Molecular Interactions Facility, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Silvia Zorrilla
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Begoña Monterroso
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Oscar M Nuero
- Molecular Interactions Facility, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Germán Rivas
- Structural and Chemical Biology Department, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
15
|
Schmid SY, Lachowski K, Chiang HT, Pozzo L, De Yoreo J, Zhang S. Mechanisms of Biomolecular Self-Assembly Investigated Through In Situ Observations of Structures and Dynamics. Angew Chem Int Ed Engl 2023; 62:e202309725. [PMID: 37702227 DOI: 10.1002/anie.202309725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Indexed: 09/14/2023]
Abstract
Biomolecular self-assembly of hierarchical materials is a precise and adaptable bottom-up approach to synthesizing across scales with considerable energy, health, environment, sustainability, and information technology applications. To achieve desired functions in biomaterials, it is essential to directly observe assembly dynamics and structural evolutions that reflect the underlying energy landscape and the assembly mechanism. This review will summarize the current understanding of biomolecular assembly mechanisms based on in situ characterization and discuss the broader significance and achievements of newly gained insights. In addition, we will also introduce how emerging deep learning/machine learning-based approaches, multiparametric characterization, and high-throughput methods can boost the development of biomolecular self-assembly. The objective of this review is to accelerate the development of in situ characterization approaches for biomolecular self-assembly and to inspire the next generation of biomimetic materials.
Collapse
Affiliation(s)
- Sakshi Yadav Schmid
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Kacper Lachowski
- Chemical Engineering, University of Washington, Seattle, WA 98105, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98105, USA
| | - Huat Thart Chiang
- Chemical Engineering, University of Washington, Seattle, WA 98105, USA
| | - Lilo Pozzo
- Chemical Engineering, University of Washington, Seattle, WA 98105, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98105, USA
- Materials Science and Engineering, University of Washington, Seattle, WA 98105, USA
| | - Jim De Yoreo
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
- Materials Science and Engineering, University of Washington, Seattle, WA 98105, USA
| | - Shuai Zhang
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA 98105, USA
- Materials Science and Engineering, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
16
|
Willis LF, Toprani V, Wijetunge S, Sievers A, Lin L, Williams J, Crowley TJ, Radford SE, Kapur N, Brockwell DJ. Exploring a role for flow-induced aggregation assays in platform formulation optimisation for antibody-based proteins. J Pharm Sci 2023; 113:S0022-3549(23)00441-0. [PMID: 39492475 DOI: 10.1016/j.xphs.2023.10.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
The development time of therapeutic monoclonal antibodies (mAbs) has been shortened by formulation platforms and the assessment of 'protein stability' using 'developability' assays. A range of assays are used to measure stability to a variety of stresses, including forces induced by hydrodynamic flow. We have previously developed a low-volume Extensional Flow Device (EFD) which subjects proteins to defined fluid flow fields in the presence of glass interfaces and used it to identify robust candidate sequences. Here, we study the aggregation of mAbs and Fc-fusion proteins using the EFD and orbital shaking under different formulations, investigating the relationship between these assays and evaluating their potential in formulation optimisation. EFD experiments identified the least aggregation-prone molecule using a fraction of the material and time involved in traditional screening. We also show that the EFD can differentiate between different formulations and that protective formulations containing polysorbate 80 stabilised poorly developable Fc-fusion proteins against EFD-induced aggregation up to two-fold. Our work highlights common platform formulation additives that affect the extent of aggregation under EFD-stress, as well as identifying factors that modulate the underlying aggregation mechanism. Together, our data could aid the choice of platform formulations early in development for next-generation therapeutics including fusion proteins.
Collapse
Affiliation(s)
- Leon F Willis
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Vishal Toprani
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810.
| | - Sashini Wijetunge
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810
| | - Annette Sievers
- BioMedicine Design, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA 02139
| | - Laura Lin
- BioMedicine Design, Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, MA 02139
| | - Jeanine Williams
- School of Chemistry, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Tom J Crowley
- Pharmaceutical Research and Development, Pfizer Inc. 1 Burtt Road, Andover, Massachusetts, USA, 01810
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT
| | - Nikil Kapur
- School of Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Leeds, Leeds UK LS2 9JT
| | - David J Brockwell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds UK LS2 9JT.
| |
Collapse
|
17
|
Campuzano IDG. A Research Journey: Over a Decade of Denaturing and Native-MS Analyses of Hydrophobic and Membrane Proteins in Amgen Therapeutic Discovery. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2023; 34:2413-2431. [PMID: 37643331 DOI: 10.1021/jasms.3c00175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Membrane proteins and associated complexes currently comprise the majority of therapeutic targets and remain among the most challenging classes of proteins for analytical characterization. Through long-term strategic collaborations forged between industrial and academic research groups, there has been tremendous progress in advancing membrane protein mass spectrometry (MS) analytical methods and their concomitant application to Amgen therapeutic project progression. Herein, I will describe a detailed and personal account of how electrospray ionization (ESI) native mass spectrometry (nMS), ion mobility-MS (IM-MS), reversed phase liquid chromatographic mass spectrometry (RPLC-MS), high-throughput solid phase extraction mass spectrometry, and matrix-assisted laser desorption ionization mass spectrometry methods were developed, optimized, and validated within Amgen Research, and importantly, how these analytical methods were applied for membrane and hydrophobic protein analyses and ultimately therapeutic project support and progression. Additionally, I will discuss all the highly important and productive collaborative efforts, both internal Amgen and external academic, which were key in generating the samples, methods, and associated data described herein. I will also describe some early and previously unpublished nano-ESI (nESI) native-MS data from Amgen Research and the highly productive University of California Los Angeles (UCLA) collaboration. I will also present previously unpublished examples of real-life Amgen biotherapeutic membrane protein projects that were supported by all the MS (and IM) analytical techniques described herein. I will start by describing the initial nESI nMS experiments performed at Amgen in 2011 on empty nanodisc molecules, using a quadrupole time-of-flight MS, and how these experiments progressed on to the 15 Tesla Fourier transform ion cyclotron resonance MS at UCLA. Then described are monomeric and multimeric membrane protein data acquired in both nESI nMS and tandem-MS modes, using multiple methods of ion activation, resulting in dramatic spectral simplification. Also described is how we investigated the far less established and less published subject, that is denaturing RPLC-MS analysis of membrane proteins, and how we developed a highly robust and reproducible RPLC-MS method capable of effective separation of membrane proteins differing in only the presence or absence of an N-terminal post translational modification. Also described is the evolution of the aforementioned RPLC-MS method into a high-throughput solid phase extraction MS method. Finally, I will give my opinion on key developments and how the area of nMS of membrane proteins needs to evolve to a state where it can be applied within the biopharmaceutical research environment for routine therapeutic project support.
Collapse
Affiliation(s)
- Iain D G Campuzano
- Amgen Research, Center for Research Acceleration by Digital Innovation, Molecular Analytics, Thousand Oaks, California 91320, United States
| |
Collapse
|
18
|
Nawalage S, Wathudura P, Wang A, Wamsley M, Zou S, Zhang D. Effects of Cascading Optical Processes: Part I: Impacts on Quantification of Sample Scattering Extinction, Intensity, and Depolarization. Anal Chem 2023; 95:1899-1907. [PMID: 36598877 DOI: 10.1021/acs.analchem.2c03917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Light scattering is a universal matter property that is especially prominent in nanoscale or larger materials. However, the effects of scattering-based cascading optical processes on experimental quantification of sample absorption, scattering, and emission intensities, as well as scattering and emission depolarization, have not been adequately addressed. Using a series of polystyrene nanoparticles (PSNPs) of different sizes as model analytes, we present a computational and experimental study on the effects of cascading light scattering on experimental quantification of NP scattering activities (scattering cross-section or molar coefficient), intensity, and depolarization. Part II and Part III of this series of companion articles explore the effects of cascading optical processes on sample absorption and fluorescence measurements, respectively. A general theoretical model is developed on how forward scattered light complicates the general applicability of Beer's law to the experimental UV-vis spectrum of scattering samples. The correlation between the scattering intensity and PSNP concentration is highly complicated with no robust linearity even when the scatterers' concentration is very low. Such complexity arises from the combination of concentration-dependence of light scattering depolarization and the scattering inner filter effects (IFEs). Scattering depolarization increases with the PSNP scattering extinction (thereby, its concentration) but can never reach unity (isotropic) due to the polarization dependence of the scattering IFE. The insights from this study are important for understanding the strengths and limitations of various scattering-based techniques for material characterization including nanoparticle quantification. They are also foundational for quantitative mechanistic understanding on the effects of light scattering on sample absorption and fluorescence measurements.
Collapse
Affiliation(s)
- Samadhi Nawalage
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Pathum Wathudura
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Ankai Wang
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States
| | - Max Wamsley
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Shengli Zou
- Department of Chemistry, University of Central Florida, Orlando, Florida 32816, United States
| | - Dongmao Zhang
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| |
Collapse
|
19
|
Mock M, Jacobitz AW, Langmead CJ, Sudom A, Yoo D, Humphreys SC, Alday M, Alekseychyk L, Angell N, Bi V, Catterall H, Chen CC, Chou HT, Conner KP, Cook KD, Correia AR, Dykstra A, Ghimire-Rijal S, Graham K, Grandsard P, Huh J, Hui JO, Jain M, Jann V, Jia L, Johnstone S, Khanal N, Kolvenbach C, Narhi L, Padaki R, Pelegri-O'Day EM, Qi W, Razinkov V, Rice AJ, Smith R, Spahr C, Stevens J, Sun Y, Thomas VA, van Driesche S, Vernon R, Wagner V, Walker KW, Wei Y, Winters D, Yang M, Campuzano IDG. Development of in silico models to predict viscosity and mouse clearance using a comprehensive analytical data set collected on 83 scaffold-consistent monoclonal antibodies. MAbs 2023; 15:2256745. [PMID: 37698932 PMCID: PMC10498806 DOI: 10.1080/19420862.2023.2256745] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/16/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
Biologic drug discovery pipelines are designed to deliver protein therapeutics that have exquisite functional potency and selectivity while also manifesting biophysical characteristics suitable for manufacturing, storage, and convenient administration to patients. The ability to use computational methods to predict biophysical properties from protein sequence, potentially in combination with high throughput assays, could decrease timelines and increase the success rates for therapeutic developability engineering by eliminating lengthy and expensive cycles of recombinant protein production and testing. To support development of high-quality predictive models for antibody developability, we designed a sequence-diverse panel of 83 effector functionless IgG1 antibodies displaying a range of biophysical properties, produced and formulated each protein under standard platform conditions, and collected a comprehensive package of analytical data, including in vitro assays and in vivo mouse pharmacokinetics. We used this robust training data set to build machine learning classifier models that can predict complex protein behavior from these data and features derived from predicted and/or experimental structures. Our models predict with 87% accuracy whether viscosity at 150 mg/mL is above or below a threshold of 15 centipoise (cP) and with 75% accuracy whether the area under the plasma drug concentration-time curve (AUC0-672 h) in normal mouse is above or below a threshold of 3.9 × 106 h x ng/mL.
Collapse
Affiliation(s)
- Marissa Mock
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Alex W Jacobitz
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | | | - Athena Sudom
- Structural Biology, Amgen Research, South San Francisco, CA, USA
| | - Daniel Yoo
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Sara C Humphreys
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Mai Alday
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | | | - Nicolas Angell
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | - Vivian Bi
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Hannah Catterall
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Chen-Chun Chen
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Hui-Ting Chou
- Structural Biology, Amgen Research, South San Francisco, CA, USA
| | - Kip P Conner
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Kevin D Cook
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Ana R Correia
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Andrew Dykstra
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | | | - Kevin Graham
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Peter Grandsard
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Joon Huh
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | - John O Hui
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Mani Jain
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Victoria Jann
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Lei Jia
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Sheree Johnstone
- Structural Biology, Amgen Research, South San Francisco, CA, USA
| | - Neelam Khanal
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | - Carl Kolvenbach
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Linda Narhi
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | - Rupa Padaki
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | | | - Wei Qi
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | | | - Austin J Rice
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Richard Smith
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | - Christopher Spahr
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | | | - Yax Sun
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Veena A Thomas
- Pharmacokinetics & Drug Metabolism, Amgen Research, South San Francisco, CA, USA
| | | | - Robert Vernon
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Victoria Wagner
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Kenneth W Walker
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Yangjie Wei
- Process Development, Amgen Operations, Thousand Oaks, CA, USA
| | - Dwight Winters
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | - Melissa Yang
- Biologic Therapeutic Discovery, Amgen Research, Thousand Oaks, CA, USA
| | | |
Collapse
|
20
|
Conner CG, McAndrew J, Menegatti S, Velev OD. An accelerated antibody aggregation test based on time sequenced dynamic light scattering. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
21
|
Luo H, Du Q, Qian C, Mlynarczyk M, Pabst TM, Damschroder M, Hunter AK, Wang WK. Formation of Transient Highly-Charged mAb Clusters Strengthens Interactions with Host Cell Proteins and Results in Poor Clearance of Host Cell Proteins by Protein A Chromatography. J Chromatogr A 2022; 1679:463385. [DOI: 10.1016/j.chroma.2022.463385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
|
22
|
Gao H, Wang ST, Hu F, Shen BB, Sun MF, Wang H, Li L, Fang WJ. Investigation of an Uncommon Artifact during Reducing Capillary Electrophoresis-Sodium Dodecyl Sulfate Analysis of a Monoclonal Antibody with Dynamic Light Scattering and Reversed Phase High-Performance Liquid Chromatography. Pharm Res 2022; 39:1959-1968. [PMID: 35701679 DOI: 10.1007/s11095-022-03303-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
PURPOSES In reducing capillary electrophoresis sodium dodecyl sulfate (CE-SDS) analysis of a monoclonal antibody (mAb-1), the peak area ratio of heavy chain (HC) to light chain (LC) was out of balance, while multiple artifact peaks were observed following the migration of HC. The main purposes of this study were to describe the techniques utilized to eliminate this artifact and clarify the root cause for this interesting phenomenon. METHODS We optimized the CE-SDS analysis of mAb-1 by a vairety of techniques including changing the concentration of protein or replacing SDS with a more hydrophobic surfactant (i.e., sodium hexadecyl sulfate (SHS) or sodium tetradecyl sulfate (STS) instead of SDS) in sample and/or the sieving gel buffer. Dynamic light scattering (DLS) and reversed phase high-performance liquid chromatography (RP-HPLC) were used to study the protein-surfactant complex. RESULTS The artifact could be partially mitigated by reducing the protein concentration and replacing SDS with SHS or STS in the sample and/or the sieving gel buffer solutions. Due to replacing a more hydrophobic surfactant, the HC-surfactant complex formed was more resistant to dissociation, preventing additional hydrophobic HC-HC interaction and aggregation, thus eliminating the artifact problem. CONCLUSIONS DLS and RP-HPLC are powerful supplementary techniques in characterizing the protein-surfactant complex, and hydrophobic surfactants such as SHS and STS could afford more normal electropherograms during the analysis of mAbs.
Collapse
Affiliation(s)
- Han Gao
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Si-Tao Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng Hu
- Zhejiang Bioray Biopharmaceutical Co., Taizhou, 317000, China
| | - Bin-Bin Shen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Min-Fei Sun
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Haibin Wang
- Zhejiang Bioray Biopharmaceutical Co., Taizhou, 317000, China
| | - Lei Li
- Zhejiang Bioray Biopharmaceutical Co., Taizhou, 317000, China
| | - Wei-Jie Fang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China. .,Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou, 310016, China.
| |
Collapse
|
23
|
Effects of Monovalent Salt on Protein-Protein Interactions of Dilute and Concentrated Monoclonal Antibody Formulations. Antibodies (Basel) 2022; 11:antib11020024. [PMID: 35466277 PMCID: PMC9036246 DOI: 10.3390/antib11020024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
In this study, we used sodium chloride (NaCl) to extensively modulate non-specific protein-protein interactions (PPI) of a humanized anti-streptavidin monoclonal antibody class 2 molecule (ASA-IgG2). The changes in PPI with varying NaCl (CNaCl) and monoclonal antibody (mAb) concentration (CmAb) were assessed using the diffusion interaction parameter kD and second virial coefficient B22 measured from solutions with low to moderate CmAb. The effective structure factor S(q)eff measured from concentrated mAb solutions using small-angle X-ray and neutron scattering (SAXS/SANS) was also used to characterize the PPI. Our results found that the nature of net PPI changed not only with CNaCl, but also with increasing CmAb. As a result, parameters measured from dilute and concentrated mAb samples could lead to different predictions on the stability of mAb formulations. We also compared experimentally determined viscosity results with those predicted from interaction parameters, including kD and S(q)eff. The lack of a clear correlation between interaction parameters and measured viscosity values indicates that the relationship between viscosity and PPI is concentration-dependent. Collectively, the behavior of flexible mAb molecules in concentrated solutions may not be correctly predicted using models where proteins are considered to be uniform colloid particles defined by parameters derived from low CmAb.
Collapse
|
24
|
Kittel Y, Kuehne AJC, De Laporte L. Translating Therapeutic Microgels into Clinical Applications. Adv Healthc Mater 2022; 11:e2101989. [PMID: 34826201 DOI: 10.1002/adhm.202101989] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/17/2021] [Indexed: 12/14/2022]
Abstract
Microgels are crosslinked, water-swollen networks with a 10 nm to 100 µm diameter and can be modified chemically or biologically to render them biocompatible for advanced clinical applications. Depending on their intended use, microgels require different mechanical and structural properties, which can be engineered on demand by altering the biochemical composition, crosslink density of the polymer network, and the fabrication method. Here, the fundamental aspects of microgel research and development, as well as their specific applications for theranostics and therapy in the clinic, are discussed. A detailed overview of microgel fabrication techniques with regards to their intended clinical application is presented, while focusing on how microgels can be employed as local drug delivery materials, scavengers, and contrast agents. Moreover, microgels can act as scaffolds for tissue engineering and regeneration application. Finally, an overview of microgels is given, which already made it into pre-clinical and clinical trials, while future challenges and chances are discussed. This review presents an instructive guideline for chemists, material scientists, and researchers in the biomedical field to introduce them to the fundamental physicochemical properties of microgels and guide them from fabrication methods via characterization techniques and functionalization of microgels toward specific applications in the clinic.
Collapse
Affiliation(s)
- Yonca Kittel
- DWI – Leibniz Institute for Interactive Materials Forckenbeckstrasse 50 52074 Aachen Germany
| | - Alexander J. C. Kuehne
- DWI – Leibniz Institute for Interactive Materials Forckenbeckstrasse 50 52074 Aachen Germany
- Institute of Organic and Macromolecular Chemistry Ulm University Albert‐Einstein‐Allee 11 89081 Ulm Germany
- Institute of Technical and Macromolecular Chemistry (ITMC) Polymeric Biomaterials RWTH University Aachen Worringerweg 2 52074 Aachen Germany
| | - Laura De Laporte
- DWI – Leibniz Institute for Interactive Materials Forckenbeckstrasse 50 52074 Aachen Germany
- Max Planck School‐Matter to Life (MtL) Jahnstraße 29 69120 Heidelberg Germany
- Advanced Materials for Biomedicine (AMB) Institute of Applied Medical Engineering (AME) Center for Biohybrid Medical Systems (CBMS) University Hospital RWTH 52074 Aachen Germany
| |
Collapse
|
25
|
Konoplev G, Agafonova D, Bakhchova L, Mukhin N, Kurachkina M, Schmidt MP, Verlov N, Sidorov A, Oseev A, Stepanova O, Kozyrev A, Dmitriev A, Hirsch S. Label-Free Physical Techniques and Methodologies for Proteins Detection in Microfluidic Biosensor Structures. Biomedicines 2022; 10:207. [PMID: 35203416 PMCID: PMC8868674 DOI: 10.3390/biomedicines10020207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/01/2022] [Accepted: 01/11/2022] [Indexed: 12/25/2022] Open
Abstract
Proteins in biological fluids (blood, urine, cerebrospinal fluid) are important biomarkers of various pathological conditions. Protein biomarkers detection and quantification have been proven to be an indispensable diagnostic tool in clinical practice. There is a growing tendency towards using portable diagnostic biosensor devices for point-of-care (POC) analysis based on microfluidic technology as an alternative to conventional laboratory protein assays. In contrast to universally accepted analytical methods involving protein labeling, label-free approaches often allow the development of biosensors with minimal requirements for sample preparation by omitting expensive labelling reagents. The aim of the present work is to review the variety of physical label-free techniques of protein detection and characterization which are suitable for application in micro-fluidic structures and analyze the technological and material aspects of label-free biosensors that implement these methods. The most widely used optical and impedance spectroscopy techniques: absorption, fluorescence, surface plasmon resonance, Raman scattering, and interferometry, as well as new trends in photonics are reviewed. The challenges of materials selection, surfaces tailoring in microfluidic structures, and enhancement of the sensitivity and miniaturization of biosensor systems are discussed. The review provides an overview for current advances and future trends in microfluidics integrated technologies for label-free protein biomarkers detection and discusses existing challenges and a way towards novel solutions.
Collapse
Affiliation(s)
- Georgii Konoplev
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
| | - Darina Agafonova
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
| | - Liubov Bakhchova
- Institute for Automation Technology, Otto-von-Guericke-University Magdeburg, 39106 Magdeburg, Germany;
| | - Nikolay Mukhin
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
- Department of Engineering, University of Applied Sciences Brandenburg, 14770 Brandenburg an der Havel, Germany; (M.K.); (S.H.)
| | - Marharyta Kurachkina
- Department of Engineering, University of Applied Sciences Brandenburg, 14770 Brandenburg an der Havel, Germany; (M.K.); (S.H.)
| | - Marc-Peter Schmidt
- Faculty of Electrical Engineering, University of Applied Sciences Dresden, 01069 Dresden, Germany;
| | - Nikolay Verlov
- Molecular and Radiation Biophysics Division, Petersburg Nuclear Physics Institute Named by B.P. Konstantinov, National Research Centre Kurchatov Institute, 188300 Gatchina, Russia;
| | - Alexander Sidorov
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
- Fuculty of Photonics, ITMO University, 197101 Saint Petersburg, Russia
| | - Aleksandr Oseev
- FEMTO-ST Institute, CNRS UMR-6174, University Bourgogne Franche-Comté, 25000 Besançon, France;
| | - Oksana Stepanova
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
| | - Andrey Kozyrev
- Faculty of Electronics, Saint Petersburg Electrotechnical University “LETI”, 197376 Saint Petersburg, Russia; (D.A.); (A.S.); (O.S.); (A.K.)
| | - Alexander Dmitriev
- Department of Ecological Physiology, Federal State Budgetary Scientific Institution “Institute of Experimental Medicine” (FSBSI “IEM”), 197376 Saint Petersburg, Russia;
| | - Soeren Hirsch
- Department of Engineering, University of Applied Sciences Brandenburg, 14770 Brandenburg an der Havel, Germany; (M.K.); (S.H.)
| |
Collapse
|
26
|
Shmool TA, Constantinou A, Jirkas A, Zhao C, Georgiou TK, Hallett J. Next Generation Strategy for Tuning the Thermoresponsive Properties of Micellar and Hydrogel Drug Delivery Vehicles Using Ionic Liquids. Polym Chem 2022. [DOI: 10.1039/d2py00053a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Amongst the greatest challenges in developing injectable controlled thermoresponsive micellar and hydrogel drug delivery vehicles include tuning the cloud point (CP) and reducing the gelation temperature (Tgel), below 37 °C,...
Collapse
|
27
|
Lai SH, Tamara S, Heck AJ. Single-particle mass analysis of intact ribosomes by mass photometry and Orbitrap-based charge detection mass spectrometry. iScience 2021; 24:103211. [PMID: 34712917 PMCID: PMC8529500 DOI: 10.1016/j.isci.2021.103211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/04/2021] [Accepted: 09/29/2021] [Indexed: 12/28/2022] Open
Abstract
Standard methods for mass analysis measure ensembles of thousand to millions of molecules. This approach enables analysis of monodisperse recombinant proteins, whereas some heterogeneous protein assemblies pose a significant challenge, whereby co-occurring stoichiometries, sub-complexes, and modifications hamper analysis using native mass spectrometry. To tackle the challenges posed by mass heterogeneity, single-particle methods may come to the rescue. Recently, two such approaches have been introduced, namely, mass photometry (MP) and Orbitrap-based charge detection mass spectrometry (CDMS). Both methods assess masses of individual molecules, albeit adhering to distinct physical principles. To evaluate these methods side by side, we analyzed a set of ribosomal particles, representing polydisperse ribonucleoprotein assemblies in the MDa range. MP and CDMS provide accurate masses for intact ribosomes and enable quantitative analysis of concomitant distinct particles within each ribosome sample. Here, we discuss pros and cons of these single-molecule techniques, also in the context of other techniques used for mass analysis.
Collapse
Affiliation(s)
- Szu-Hsueh Lai
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Sem Tamara
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Albert J.R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, The Netherlands
- Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
28
|
Khosla N, Thayil SM, Kaur R, Kesavan AK. MSMEG_3955 from Mycobacterium smegmatis is a FMN bounded homotrimeric NAD(P)H:Flavin mononucleotide (FMN) oxidoreductase. BMC Microbiol 2021; 21:319. [PMID: 34798816 PMCID: PMC8605562 DOI: 10.1186/s12866-021-02330-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 09/27/2021] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) remains an important public health problem since it is the major cause of elevated morbidity and mortality globally. Previous works have shown that Mycobacterium tuberculosis (Mtb); the prime causative agent of the deadly disease has dormancy survival regulator (DosR) regulon, a two-component regulatory system which controls the transcription of more than 50 genes. However, the structure and detailed functions of these DosR regulated genes are largely undetermined. Out of many DosR regulon genes, Rv3131 gets up regulated in hypoxic conditions and was believed to encode for a nitroreductase flavoprotein. The utilization of mycobacteria-specific model systems has greatly added to our understanding of the molecular mechanisms involved in the life cycle and pathogenesis of Mtb. RESULTS In this study the non-pathogenic mycobacterial model organism Mycobacterium smegmatis (Msmeg) was used to reveal the structure and function of MSMEG_3955; which is a homologue of Rv3131 from Mtb. Using chromatography and spectroscopy techniques it was revealed that cofactor flavin mononucleotide (FMN) was bound to flavoprotein MSMEG_3955. Consistent with the homology modelling predictions, Circular Dichroism (CD) analysis indicated that the MSMEG_3955 is composed of 39.3% α-helix and 24.9% β-pleated sheets. In contrast to the current notions, the enzymatic assays performed in the present study revealed that MSMEG_3955 was not capable of reducing nitro substrates but showed NADPH dependent FMN oxidoreductase activity. Also, gel permeation chromatography, dynamic light scattering and native acidic gels showed that MSMEG_3955 exists as a homotrimer. Furthermore, the presence of NADPH dependent FMN oxidoreductase and homotrimeric existence could be an alternative function of the protein to help the bacteria survive in dormant state or may be involved in other biochemical pathways. CONCLUSION MSMEG_3955 is a FMN bound flavoprotein, which exits as a trimer under in vitro conditions. There is no disulphide linkages in between the three protomers of the homotrimer MSMEG_3955. It has a NADPH dependent FMN oxidoreductase activity.
Collapse
Affiliation(s)
- Neha Khosla
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Seema Madhumal Thayil
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Rajinder Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, Punjab, 143005, India
| | - Anup Kumar Kesavan
- Department of Molecular Biology and Biochemistry, Guru Nanak Dev University, Amritsar, Punjab, 143005, India.
- Current Address: Department of Biotechnology & Microbiology, Kannur University, Dr. E.K. Janaki Ammal Campus, Palayad, Thalassery, Kannur, Kerala, India.
| |
Collapse
|
29
|
Clauss ZS, Wardzala CL, Schlirf AE, Wright NS, Saini SS, Onoa B, Bustamante C, Kramer JR. Tunable, biodegradable grafting-from glycopolypeptide bottlebrush polymers. Nat Commun 2021; 12:6472. [PMID: 34753949 PMCID: PMC8578664 DOI: 10.1038/s41467-021-26808-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/01/2021] [Indexed: 11/09/2022] Open
Abstract
The cellular glycocalyx and extracellular matrix are rich in glycoproteins and proteoglycans that play essential physical and biochemical roles in all life. Synthetic mimics of these natural bottlebrush polymers have wide applications in biomedicine, yet preparation has been challenged by their high grafting and glycosylation densities. Using one-pot dual-catalysis polymerization of glycan-bearing α-amino acid N-carboxyanhydrides, we report grafting-from glycopolypeptide brushes. The materials are chemically and conformationally tunable where backbone and sidechain lengths were precisely altered, grafting density modulated up to 100%, and glycan density and identity tuned by monomer feed ratios. The glycobrushes are composed entirely of sugars and amino acids, are non-toxic to cells, and are degradable by natural proteases. Inspired by native lipid-anchored proteoglycans, cholesterol-modified glycobrushes were displayed on the surface of live human cells. Our materials overcome long-standing challenges in glycobrush polymer synthesis and offer new opportunities to examine glycan presentation and multivalency from chemically defined scaffolds. Synthetic mimics of glycoproteins and proteoglycans have wide applications in biomedicine, yet preparation has been challenged by their high grafting and glycosylation densities. Here the authors show one-pot dual-catalysis polymerization of glycan-bearing α-amino acid N-carboxyanhydrides to form glycopolypeptide brushes.
Collapse
Affiliation(s)
- Zachary S Clauss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA
| | - Casia L Wardzala
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA
| | - Austin E Schlirf
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA
| | - Nathaniel S Wright
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA
| | - Simranpreet S Saini
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA
| | - Bibiana Onoa
- Howard Hughes Medical Institute University of California Berkeley, Berkeley, CA, 94720, USA
| | - Carlos Bustamante
- Howard Hughes Medical Institute University of California Berkeley, Berkeley, CA, 94720, USA.,Department of Chemistry, University of California Berkeley, Berkeley, CA, 94720, USA.,Institute for Quantitative Biosciences, University of California, Berkeley, CA, 94720, USA.,Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.,Department of Physics, University of California Berkeley, Berkeley, CA, 94720, USA.,Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, 84102, USA. .,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah, 84102, USA.
| |
Collapse
|
30
|
Britt HM, Cragnolini T, Thalassinos K. Integration of Mass Spectrometry Data for Structural Biology. Chem Rev 2021; 122:7952-7986. [PMID: 34506113 DOI: 10.1021/acs.chemrev.1c00356] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mass spectrometry (MS) is increasingly being used to probe the structure and dynamics of proteins and the complexes they form with other macromolecules. There are now several specialized MS methods, each with unique sample preparation, data acquisition, and data processing protocols. Collectively, these methods are referred to as structural MS and include cross-linking, hydrogen-deuterium exchange, hydroxyl radical footprinting, native, ion mobility, and top-down MS. Each of these provides a unique type of structural information, ranging from composition and stoichiometry through to residue level proximity and solvent accessibility. Structural MS has proved particularly beneficial in studying protein classes for which analysis by classic structural biology techniques proves challenging such as glycosylated or intrinsically disordered proteins. To capture the structural details for a particular system, especially larger multiprotein complexes, more than one structural MS method with other structural and biophysical techniques is often required. Key to integrating these diverse data are computational strategies and software solutions to facilitate this process. We provide a background to the structural MS methods and briefly summarize other structural methods and how these are combined with MS. We then describe current state of the art approaches for the integration of structural MS data for structural biology. We quantify how often these methods are used together and provide examples where such combinations have been fruitful. To illustrate the power of integrative approaches, we discuss progress in solving the structures of the proteasome and the nuclear pore complex. We also discuss how information from structural MS, particularly pertaining to protein dynamics, is not currently utilized in integrative workflows and how such information can provide a more accurate picture of the systems studied. We conclude by discussing new developments in the MS and computational fields that will further enable in-cell structural studies.
Collapse
Affiliation(s)
- Hannah M Britt
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Tristan Cragnolini
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom.,Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, United Kingdom
| | - Konstantinos Thalassinos
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom.,Institute of Structural and Molecular Biology, Birkbeck College, University of London, London WC1E 7HX, United Kingdom
| |
Collapse
|
31
|
Soltermann F, Struwe WB, Kukura P. Label-free methods for optical in vitro characterization of protein-protein interactions. Phys Chem Chem Phys 2021; 23:16488-16500. [PMID: 34342317 PMCID: PMC8359934 DOI: 10.1039/d1cp01072g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions are involved in the regulation and function of the majority of cellular processes. As a result, much effort has been aimed at the development of methodologies capable of quantifying protein-protein interactions, with label-free methods being of particular interest due to the associated simplified workflows and minimisation of label-induced perturbations. Here, we review recent advances in optical technologies providing label-free in vitro measurements of affinities and kinetics. We provide an overview and comparison of existing techniques and their principles, discussing advantages, limitations, and recent applications.
Collapse
Affiliation(s)
- Fabian Soltermann
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Weston B. Struwe
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Philipp Kukura
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| |
Collapse
|
32
|
Detwiler RE, Schlirf AE, Kramer JR. Rethinking Transition Metal Catalyzed N-Carboxyanhydride Polymerization: Polymerization of Pro and AcOPro N-Carboxyanhydrides. J Am Chem Soc 2021; 143:11482-11489. [PMID: 34283588 DOI: 10.1021/jacs.1c03338] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Polyproline (PP) based polypeptides have broad applications as protein mimics, ordered materials, hydrogels, and surface coatings. However, a lack of rapid and efficient preparatory methods has challenged synthesis of well-defined high molecular weight materials. Here, we report facile and high-yielding methods for preparation and polymerization of Pro and trans-4-acetoxy-Pro N-carboxyanhdrides (NCAs). For decades, transition metal initiators of NCA polymerization were assumed to be nonstarters with Pro due to the lack of an amide NH proton. We carefully considered the known steps in the initiation mechanism and applied a Ni initiator that intercepts an intermediate and does not require an NH group. This initiator efficiently catalyzes controlled, living polymerization of Pro NCAs, revealing that routes alternate to the previously proposed mechanism must be at play. We also found Co species can catalyze Pro NCA polymerization, and we improved the synthetic methods to prepare the NCA monomers. Our methods are high-yielding and rapid and give tunable, end-functional PP-based homo, statistical, and block polypeptides. We characterized the conformation of PP and trans-4-hydroxy-PP by CD and confirmed the time scale for quantitative conversion from PPI to PPII helices. Overall, our data shed light on the general propagation mechanism of transition metal catalyzed NCA polymerization and have opened the door for efficient preparation of a desirable class of biomaterials.
Collapse
Affiliation(s)
- Rachel E Detwiler
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Austin E Schlirf
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| | - Jessica R Kramer
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
33
|
Campuzano IDG, Sandoval W. Denaturing and Native Mass Spectrometric Analytics for Biotherapeutic Drug Discovery Research: Historical, Current, and Future Personal Perspectives. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1861-1885. [PMID: 33886297 DOI: 10.1021/jasms.1c00036] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Mass spectrometry (MS) plays a key role throughout all stages of drug development and is now as ubiquitous as other analytical techniques such as surface plasmon resonance, nuclear magnetic resonance, and supercritical fluid chromatography, among others. Herein, we aim to discuss the history of MS, both electrospray and matrix-assisted laser desorption ionization, specifically for the analysis of antibodies, evolving through to denaturing and native-MS analysis of newer biologic moieties such as antibody-drug conjugates, multispecific antibodies, and interfering nucleic acid-based therapies. We discuss challenging therapeutic target characterization such as membrane protein receptors. Importantly, we compare and contrast the MS and hyphenated analytical chromatographic methods used to characterize these therapeutic modalities and targets within biopharmaceutical research and highlight the importance of appropriate MS deconvolution software and its essential contribution to project progression. Finally, we describe emerging applications and MS technologies that are still predominantly within either a development or academic stage of use but are poised to have significant impact on future drug development within the biopharmaceutic industry once matured. The views reflected herein are personal and are not meant to be an exhaustive list of all relevant MS performed within biopharmaceutical research but are what we feel have been historically, are currently, and will be in the future the most impactful for the drug development process.
Collapse
MESH Headings
- Antibodies, Monoclonal/analysis
- Automation, Laboratory
- Biopharmaceutics/methods
- Chromatography, Liquid
- Drug Discovery/methods
- Drug Industry/history
- History, 20th Century
- History, 21st Century
- Humans
- Immunoconjugates/analysis
- Immunoconjugates/chemistry
- Protein Denaturation
- Protein Processing, Post-Translational
- Proteins/analysis
- Spectrometry, Mass, Electrospray Ionization/history
- Spectrometry, Mass, Electrospray Ionization/instrumentation
- Spectrometry, Mass, Electrospray Ionization/methods
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/history
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
Collapse
Affiliation(s)
- Iain D G Campuzano
- Discovery Attribute Sciences, Amgen Research, 1 Amgen Center Drive, Thousand Oaks, California 92130, United States
| | - Wendy Sandoval
- Department of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
34
|
Lakha R, Montero AM, Jabeen T, Costeas CC, Ma J, Vizcarra CL. Variable Autoinhibition among Deafness-Associated Variants of Diaphanous 1 (DIAPH1). Biochemistry 2021; 60:2320-2329. [PMID: 34279089 DOI: 10.1021/acs.biochem.1c00170] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
One of the earliest mapped human deafness genes, DIAPH1, encodes the formin DIAPH1. To date, at least three distinct mutations in the C-terminal domains and two additional mutations in the N-terminal region are associated with autosomal dominant hearing loss. The underlying molecular mechanisms are not known, and the role of formins in the inner ear is not well understood. In this study, we use biochemical assays to test the hypotheses that autoinhibition and/or actin assembly activities are disrupted by DFNA1 mutations. Our results indicate that C-terminal mutant forms of DIAPH1 are functional in vitro and promote actin filament assembly. Similarly, N-terminal mutants are well-folded and have quaternary structures and thermal stabilities similar to those of the wild-type (WT) protein. The strength of the autoinhibitory interactions varies widely among mutants, with the ttaa, A265S, and I530S mutations having an affinity similar to that of WT and the 1213x and Δag mutations completely abolishing autoinhibition. These data indicate that, in some cases, hearing loss may be linked to weakened inhibition of actin assembly.
Collapse
Affiliation(s)
- Rabina Lakha
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Angela M Montero
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Tayyaba Jabeen
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Christina C Costeas
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| | - Jia Ma
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| | - Christina L Vizcarra
- Department of Chemistry, Barnard College, New York, New York 10027, United States
| |
Collapse
|
35
|
Martinez SE, Singh A, De Wijngaert B, Sultana S, Dharia C, Vanbuel H, Shen J, Vasilchuk D, Patel SS, Das K. Assembly and Cryo-EM structure determination of yeast mitochondrial RNA polymerase initiation complex intermediates. STAR Protoc 2021; 2:100431. [PMID: 33870232 PMCID: PMC8044712 DOI: 10.1016/j.xpro.2021.100431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In yeast mitochondria, transcription initiation requires assembly of mitochondrial RNA polymerase and transcription initiation factor MTF1 at the DNA promoter initiation site. This protocol describes the purification of the component proteins and assembly of partially melted and fully melted initiation complex states. Both states co-exist in equilibrium in the same sample as seen by cryoelectron microscopy (cryo-EM) and allow elucidation of MTF1's structural roles in controlling the transition into elongation. We further outline how analysis of the complex by light scattering, thermal shift assay, and ultrafiltration assay exhibits reproducible results. For complete details on the use and execution of this protocol, please refer to De Wijngaert et al. (2021).
Collapse
Affiliation(s)
- Sergio E. Martinez
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Anupam Singh
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Brent De Wijngaert
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Shemaila Sultana
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Chhaya Dharia
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Hans Vanbuel
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Jiayu Shen
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Daniel Vasilchuk
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Smita S. Patel
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Kalyan Das
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
36
|
Lanzaro A, Roche A, Sibanda N, Corbett D, Davis P, Shah M, Pathak JA, Uddin S, van der Walle CF, Yuan XF, Pluen A, Curtis R. Cluster Percolation Causes Shear Thinning Behavior in Concentrated Solutions of Monoclonal Antibodies. Mol Pharm 2021; 18:2669-2682. [PMID: 34121411 DOI: 10.1021/acs.molpharmaceut.1c00198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
High-concentration (>100 g/L) solutions of monoclonal antibodies (mAbs) are typically characterized by anomalously large solution viscosity and shear thinning behavior for strain rates ≥103 s-1. Here, the link between protein-protein interactions (PPIs) and the rheology of concentrated solutions of COE-03 and COE-19 mAbs is studied by means of static and dynamic light scattering and microfluidic rheometry. By comparing the experimental data with predictions based on the Baxter sticky hard-sphere model, we surprisingly find a connection between the observed shear thinning and the predicted percolation threshold. The longest shear relaxation time of mAbs was much larger than that of model sticky hard spheres within the same region of the phase diagram, which is attributed to the anisotropy of the mAb PPIs. Our results suggest that not only the strength but also the patchiness of short-range attractive PPIs should be explicitly accounted for by theoretical approaches aimed at predicting the shear rate-dependent viscosity of dense mAb solutions.
Collapse
Affiliation(s)
- Alfredo Lanzaro
- Institute for Systems Rheology, Guangzhou University, No. 230 West Outer Ring Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, China
| | - Aisling Roche
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Nicole Sibanda
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Daniel Corbett
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| | - Peter Davis
- Department of Molecular Biology and Biotechnology, University of Sheffield UK, Sheffield S10 2TN, United Kingdom
| | - Maryam Shah
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Jai A Pathak
- Dosage Form Design and Development, Biopharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, United Kingdom
| | - Shahid Uddin
- Dosage Form Design and Development, Biopharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, United Kingdom
| | - Christopher F van der Walle
- Dosage Form Design and Development, Biopharmaceuticals Development, R&D, AstraZeneca, Cambridge CB21 6GH, United Kingdom
| | - Xue-Feng Yuan
- Institute for Systems Rheology, Guangzhou University, No. 230 West Outer Ring Road, Guangzhou Higher Education Mega Center, Guangzhou 510006, China
| | - Alain Pluen
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, United Kingdom
| | - Robin Curtis
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
37
|
Pei Y, Hinchliffe BA, Minelli C. Measurement of the Size Distribution of Multimodal Colloidal Systems by Laser Diffraction. ACS OMEGA 2021; 6:14049-14058. [PMID: 34124428 PMCID: PMC8190786 DOI: 10.1021/acsomega.1c00411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/09/2021] [Indexed: 06/01/2023]
Abstract
Laser diffraction (LD) is a well-established tool for the measurement of particle size distribution. Recently, its demand and use for the measurement of complex biological systems have increased. Among the challenges that these types of samples present, there is the presence of multiple particle populations whose modal size may span across several orders of magnitude. In this study, we assessed the accuracy of LD for the measurement of the modal diameter of both single and mixed populations of polystyrene particles with diameters ranging from 60 nm to 40 μm. We discuss the application of different available algorithms to the analysis of the data and their impact on the measurement results. Independent methods were applied to guide the selection of the algorithms and validate the measured size distributions. We found that the modal diameters of the particle size distribution measured by LD for the mixed suspension was accurate within 2 % for particles larger than 1 μm and generally within 25 % for the particles tested. Method repeatability was found to be robust, with deviations below 1%. The method was also found to be useful for estimating the relative concentration of the particle populations in the mixed samples. This study provides confidence in the use of LD for the measurement of complex multimodal colloidal samples.
Collapse
|
38
|
Trimethylamine N-oxide alters structure-function integrity of β-casein: Structural disorder co-regulates the aggregation propensity and chaperone activity. Int J Biol Macromol 2021; 182:921-930. [PMID: 33872615 DOI: 10.1016/j.ijbiomac.2021.04.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 11/23/2022]
Abstract
Intrinsically disordered proteins (IDPs), involved in the regulation and function of various cellular processes like transcription, translation, cell cycle etc., exist as ensembles of rapidly interconverting structures with functional plasticity. Among numerous cellular regulatory mechanisms involved in structural and functional regulation of IDPs, osmolytes are emerging as promising regulatory agents due to their ability to affect the structure-function integrity of IDPs. The present study investigated the effect of methylamine osmolytes on β-casein, an IDP essential for maintaining the overall stability of casein complex in milk. It was observed that trimethylamine N-oxide induces a compact structural state in β-casein with slightly decreased chaperone activity and insignificant aggregation propensity. However, the other two osmolytes from this group, i.e., sarcosine and betaine, had no significant effect on the overall structure and chaperone activity of the IDP. The present study hints towards the possible evolutionary selection of higher structural disorder in β-casein, compared to α-casein, for stability of the casein complex and prevention of amyloidosis in the mammary gland.
Collapse
|
39
|
Zielińska A, Szalata M, Gorczyński A, Karczewski J, Eder P, Severino P, Cabeda JM, Souto EB, Słomski R. Cancer Nanopharmaceuticals: Physicochemical Characterization and In Vitro/In Vivo Applications. Cancers (Basel) 2021; 13:1896. [PMID: 33920840 PMCID: PMC8071188 DOI: 10.3390/cancers13081896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/07/2021] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
Physicochemical, pharmacokinetic, and biopharmaceutical characterization tools play a key role in the assessment of nanopharmaceuticals' potential imaging analysis and for site-specific delivery of anti-cancers to neoplastic cells/tissues. If diagnostic tools and therapeutic approaches are combined in one single nanoparticle, a new platform called nanotheragnostics is generated. Several analytical technologies allow us to characterize nanopharmaceuticals and nanoparticles and their properties so that they can be properly used in cancer therapy. This paper describes the role of multifunctional nanoparticles in cancer diagnosis and treatment, describing how nanotheragnostics can be useful in modern chemotherapy, and finally, the challenges associated with the commercialization of nanoparticles for cancer therapy.
Collapse
Affiliation(s)
- Aleksandra Zielińska
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
- Department of Pharmaceutical Echnology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Marlena Szalata
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Dojazd 11, 60-632 Poznań, Poland
| | - Adam Gorczyński
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland;
| | - Jacek Karczewski
- Department of Environmental Medicine, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland;
| | - Piotr Eder
- Department of Gastroenterology, Dietetics and Internal Diseases, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznań, Poland;
| | - Patrícia Severino
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women & Hospital, Harvard Medical School, 65 Landsdowne Street, Cambridge, MA 02139, USA;
- Biotechnological Postgraduate Program, Institute of Technology and Research (ITP), Nanomedicine and Nanotechnology Laboratory (LNMed), University of Tiradentes (Unit), Av. Murilo Dantas 300, Aracaju 49010-390, Brazil
- Tiradentes Institute, 150 Mt Vernon St, Dorchester, MA 02125, USA
| | - José M. Cabeda
- ESS-FP, Escola Superior de Saúde Fernando Pessoa, Rua Delfim Maia 334, 4200-253 Porto, Portugal;
- FP-ENAS-Fernando Pessoa Energy, Environment and Health Research Unit, Universidade Fernando Pessoa, Praça 9 de Abril, 349, 4249-004 Porto, Portugal
| | - Eliana B. Souto
- Department of Pharmaceutical Echnology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- CEB–Centre of Biological Engineering, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Ryszard Słomski
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479 Poznań, Poland; (M.S.); (R.S.)
| |
Collapse
|
40
|
Shahfar H, Forder JK, Roberts CJ. Toward a Suite of Coarse-Grained Models for Molecular Simulation of Monoclonal Antibodies and Therapeutic Proteins. J Phys Chem B 2021; 125:3574-3588. [PMID: 33821645 DOI: 10.1021/acs.jpcb.1c01903] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of coarse-grained models for molecular simulation of proteins are considered, with emphasis on the application of predicting protein-protein self-interactions for monoclonal antibodies (MAbs). As an illustrative example and for quantitative comparison, the models are used to predict osmotic virial coefficients over a broad range of attractive and repulsive self-interactions and solution conditions for a series of MAbs where the second osmotic virial coefficient has been experimentally determined in prior work. The models are compared based on how well they can predict experimental behavior, their computational burdens, and scalability. An intermediate-resolution model is also introduced that can capture specific electrostatic interactions with improved efficiency and similar or improved accuracy when compared to the previously published models. Guidance is included for the selection of coarse-grained models more generally for capturing a balance of electrostatic, steric, and short-ranged nonelectrostatic interactions for proteins from low to high concentrations.
Collapse
Affiliation(s)
- Hassan Shahfar
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States.,Department of Physics and Astronomy, University of Delaware, Newark, Delaware 19716, United States
| | - James K Forder
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Christopher J Roberts
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
41
|
Al-Sawaftah NM, Abusamra RH, Husseini GA. Carbohydrate-functionalized Liposomes in Cancer Therapy. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716999200626144921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Existing cancer treatments are often accompanied by adverse side effects that can greatly
reduce the quality of life of cancer patients; this sets the platform for the development and application
of nanocarrier-based platforms for the delivery of anticancer drugs. Among these nanocarriers,
liposomes have demonstrated excellent potential in drug delivery applications. Furthermore,
the overexpression of certain receptors on cancer cells has led to the development of active targeting
approaches where liposome surfaces are decorated with ligands against these receptors. Given
the central role that sugars play in cancer biology, more and more researchers are integrating “glycoscience”
into their anticancer therapeutic designs. Carbohydrate functionalized liposomes present
an attractive drug delivery system due to their biocompatibility, biodegradability, low toxicity,
and specific cell targeting ability. This review presents an overview of the preparation methods,
characterization, evaluation, and applications of carbohydrate functionalized liposomes in cancer
therapy.
Collapse
Affiliation(s)
- Nour M. Al-Sawaftah
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Rand H. Abusamra
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
42
|
Comprehensive characterization and quantification of adeno associated vectors by size exclusion chromatography and multi angle light scattering. Sci Rep 2021; 11:3012. [PMID: 33542328 PMCID: PMC7862616 DOI: 10.1038/s41598-021-82599-1] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 01/22/2021] [Indexed: 01/10/2023] Open
Abstract
Adeno associated virus (AAV) capsids are a leading modality for in vivo gene delivery. Complete and precise characterization of capsid particles, including capsid and vector genome concentration, is necessary to safely and efficaciously dose patients. Size exclusion chromatography (SEC) coupled to multiangle light scattering (MALS) offers a straightforward approach to comprehensively characterize AAV capsids. The current study demonstrates that this method provides detailed AAV characterization information, including but not limited to aggregation profile, size-distribution, capsid content, capsid molar mass, encapsidated DNA molar mass, and total capsid and vector genome titer. Currently, multiple techniques are required to generate this information, with varying accuracy and precision. In the current study, a new series of equations for SEC-MALS are used in tandem with intrinsic properties of the capsids and encapsidated DNA to quantify multiple physical AAV attributes in one 20-min run with minimal sample manipulation, high accuracy, and high precision. These novel applications designate this well-established method as a powerful tool for product development and process analytics in future gene therapy programs.
Collapse
|
43
|
Folta-Stogniew E. Characterization of Protein-Nucleic Acid Complexes by Size-Exclusion Chromatography Coupled with Light Scattering, Absorbance, and Refractive Index Detectors. Methods Mol Biol 2021; 2263:381-395. [PMID: 33877609 DOI: 10.1007/978-1-0716-1197-5_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Size-exclusion chromatography (SEC) coupled with multiangle light scattering detection (SEC/MALS) enables determination of the molecular weight, oligomeric state, and stoichiometry of protein-nucleic acid complexes in solution. Often such complexes show anomalous behavior on SEC, thus presenting a challenge in determination of molecular weight and stoichiometry based solely on the elution position from SEC. In contrast to analytical ultracentrifugation, the SEC/MALS analysis is not affected by the shape of the complex. Here we describe the use of SEC/MALS for characterization of the stoichiometry of the complex between the reverse transcriptase (RT) domain from group II intron-maturase from Eubacterium rectale and intron RNA, and for monitoring protein dimerization that is driven by interaction between single-stranded DNA upstream of the P1 promoter, known as FUSE and FUSE binding protein-interacting repressor (FIR).
Collapse
Affiliation(s)
- Ewa Folta-Stogniew
- W.M. Keck Biotechnology Resource Laboratory, Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
44
|
De Wijngaert B, Sultana S, Singh A, Dharia C, Vanbuel H, Shen J, Vasilchuk D, Martinez SE, Kandiah E, Patel SS, Das K. Cryo-EM Structures Reveal Transcription Initiation Steps by Yeast Mitochondrial RNA Polymerase. Mol Cell 2020; 81:268-280.e5. [PMID: 33278362 DOI: 10.1016/j.molcel.2020.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 01/18/2023]
Abstract
Mitochondrial RNA polymerase (mtRNAP) is crucial in cellular energy production, yet understanding of mitochondrial DNA transcription initiation lags that of bacterial and nuclear DNA transcription. We report structures of two transcription initiation intermediate states of yeast mtRNAP that explain promoter melting, template alignment, DNA scrunching, abortive synthesis, and transition into elongation. In the partially melted initiation complex (PmIC), transcription factor MTF1 makes base-specific interactions with flipped non-template (NT) nucleotides "AAGT" at -4 to -1 positions of the DNA promoter. In the initiation complex (IC), the template in the expanded 7-mer bubble positions the RNA and NTP analog UTPαS, while NT scrunches into an NT loop. The scrunched NT loop is stabilized by the centrally positioned MTF1 C-tail. The IC and PmIC states coexist in solution, revealing a dynamic equilibrium between two functional states. Frequent scrunching/unscruching transitions and the imminent steric clashes of the inflating NT loop and growing RNA:DNA with the C-tail explain abortive synthesis and transition into elongation.
Collapse
Affiliation(s)
- Brent De Wijngaert
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Shemaila Sultana
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Anupam Singh
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Chhaya Dharia
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Hans Vanbuel
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Jiayu Shen
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Daniel Vasilchuk
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA
| | - Sergio E Martinez
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Eaazhisai Kandiah
- European Synchrotron Radiation Facility, 71 Avenue des Martyrs, 38043 Grenoble, France
| | - Smita S Patel
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854, USA.
| | - Kalyan Das
- Rega Institute for Medical Research, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
45
|
Faust C, Ochs C, Korn M, Werner U, Jung J, Dittrich W, Schiebler W, Schauder R, Rao E, Langer T. Production of a novel heterodimeric two-chain insulin-Fc fusion protein. Protein Eng Des Sel 2020; 33:5959880. [PMID: 33159202 DOI: 10.1093/protein/gzaa026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 01/12/2023] Open
Abstract
Insulin is a peptide hormone produced by the pancreas. The physiological role of insulin is the regulation of glucose metabolism. Under certain pathological conditions the insulin levels can be reduced leading to the metabolic disorder diabetes mellitus (DM). For type 1 DM and, dependent on the disease progression for type 2 DM, insulin substitution becomes indispensable. To relieve insulin substitution therapy for patients, novel insulin analogs with pharmacokinetic and pharmacodynamic profiles aiming for long-lasting or fast-acting insulins have been developed. The next step in the evolution of novel insulins should be insulin analogs with a time action profile beyond 1-2 days, preferable up to 1 week. Nowadays, insulin is produced in a recombinant manner. This approach facilitates the design and production of further insulin-analogs or insulin-fusion proteins. The usage of the Fc-domain from immunoglobulin as a fusion partner for therapeutic proteins and peptides is widely used to extend their plasma half-life. Insulin consists of two chains, the A- and B-chain, which are connected by two disulfide-bridges. To produce a novel kind of Fc-fusion protein we have fused the A-chain as well as the B-chain to Fc-fragments containing either 'knob' or 'hole' mutations. The 'knob-into-hole' technique is frequently used to force heterodimerization of the Fc-domain. Using this approach, we were able to produce different variants of two-chain-insulin-Fc-protein (tcI-Fc-protein) variants. The tcI-Fc-fusion variants retained activity as shown in in vitro assays. Finally, prolonged blood glucose lowering activity was demonstrated in normoglycemic rats. Overall, we describe here the production of novel insulin-Fc-fusion proteins with prolonged times of action.
Collapse
Affiliation(s)
- Christine Faust
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Christian Ochs
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany.,Provadis School of International Management and Technology AG, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Marcus Korn
- Sanofi-Aventis Deutschland GmbH, R&D TA Diabetes, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Ulrich Werner
- Sanofi-Aventis Deutschland GmbH, R&D TA Diabetes, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Jennifer Jung
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Werner Dittrich
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Werner Schiebler
- Provadis School of International Management and Technology AG, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Rolf Schauder
- Provadis School of International Management and Technology AG, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Ercole Rao
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| | - Thomas Langer
- Sanofi-Aventis Deutschland GmbH, R&D Biologics Research, Industriepark Höchst, 65926 Frankfurt am Main, Germany
| |
Collapse
|
46
|
Optical Characterization of Homogeneous and Heterogeneous Intralipid-Based Samples. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10186234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Different scattering processes take place when photons propagate inside turbid media. Many powerful experimental techniques exploiting these processes have been developed and applied over the years in a large variety of situations from fundamental and applied research to industrial applications. In the present paper, we intend to take advantage of Static Light Scattering (SLS), Dynamic Light Scattering (DLS), and Time-Resolved Transmittance (TRT) for investigating all the different scattering regimes by using scattering suspensions in a very large range of scatterer concentrations. The suspensions were prepared using Intralipid 20%, a material largely employed in studies of the optical properties of turbid media, with concentrations from 10−5% to 50%. By the analysis of the angular and temporal dependence of the scattered light, a more reliable description of the scattering process occurring in these samples can be obtained. TRT measurements allowed us to obtain information on the reduced scattering coefficient, an important parameter largely used in the description of the optical properties of turbid media. TRT was also employed for the detection of inclusions embedded in Intralipid suspensions, by using a properly designed data analysis. The present study allowed us to better elucidate the dependence of scattering properties of Intralipid suspensions in a very large concentration range and the occurrence of the different scattering processes involved in the propagation of light in turbid media for the first time to our knowledge. In so doing, the complementary contribution of SLS, DLS, and TRT in the characterization of turbid media from an optical and structural point of view is strongly evidenced.
Collapse
|
47
|
Sanaeifar N, Mäder K, Hinderberger D. Nanoscopic Characterization of Stearic Acid Release from Bovine Serum Albumin Hydrogels. Macromol Biosci 2020; 20:e2000126. [PMID: 32567224 DOI: 10.1002/mabi.202000126] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/11/2020] [Indexed: 12/22/2022]
Abstract
The release behavior of 16-doxyl stearic acid (16-DSA) from hydrogels made from bovine serum albumin (BSA) is characterized. 16-DSA serves as a model tracer molecule for amphiphilic drugs. Various hydrogel preparation procedures are tested and the fatty acid release from the different gels is compared in detail. These comparisons reach from the macroscopic level, the viscoelastic behavior via rheological characterization to changes on the nanoscopic level concerning the secondary structure of the protein during gelation through infrared (ATR-IR) spectroscopy. 16-DSA-BSA interaction via continuous wave electron paramagnetic resonance (CW EPR) spectroscopy in addition gives a nanoscopic view of small molecule-hydrogel interaction. The combined effects of fatty acid concentration, hydrogel incubation time, and gelation procedures on release behavior are studied via CW EPR spectroscopy and dynamic light scattering (DLS) measurements, which provide deep insight on the interaction of 16-DSA with BSA hydrogels and the nature and size of the released components, respectively. It is found that the release rate of the fatty acid from BSA hydrogels depends on and can thus be tuned through its loading percentage, duration of hydrogel formation and the type of gelation methods. All of the results confirm the potential of these gels as delivery hosts in pharmaceutical applications allowing the sustained release of drug.
Collapse
Affiliation(s)
- Niuosha Sanaeifar
- Institute of Chemistry, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 4, Halle (Saale), 06120, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str.4, Halle (Saale), 06120, Germany
| | - Dariush Hinderberger
- Institute of Chemistry, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 4, Halle (Saale), 06120, Germany
| |
Collapse
|
48
|
Gentiluomo L, Svilenov HL, Augustijn D, El Bialy I, Greco ML, Kulakova A, Indrakumar S, Mahapatra S, Morales MM, Pohl C, Roche A, Tosstorff A, Curtis R, Derrick JP, Nørgaard A, Khan TA, Peters GHJ, Pluen A, Rinnan Å, Streicher WW, van der Walle CF, Uddin S, Winter G, Roessner D, Harris P, Frieß W. Advancing Therapeutic Protein Discovery and Development through Comprehensive Computational and Biophysical Characterization. Mol Pharm 2020; 17:426-440. [PMID: 31790599 DOI: 10.1021/acs.molpharmaceut.9b00852] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Therapeutic protein candidates should exhibit favorable properties that render them suitable to become drugs. Nevertheless, there are no well-established guidelines for the efficient selection of proteinaceous molecules with desired features during early stage development. Such guidelines can emerge only from a large body of published research that employs orthogonal techniques to characterize therapeutic proteins in different formulations. In this work, we share a study on a diverse group of proteins, including their primary sequences, purity data, and computational and biophysical characterization at different pH and ionic strength. We report weak linear correlations between many of the biophysical parameters. We suggest that a stability comparison of diverse therapeutic protein candidates should be based on a computational and biophysical characterization in multiple formulation conditions, as the latter can largely determine whether a protein is above or below a certain stability threshold. We use the presented data set to calculate several stability risk scores obtained with an increasing level of analytical effort and show how they correlate with protein aggregation during storage. Our work highlights the importance of developing combined risk scores that can be used for early stage developability assessment. We suggest that such scores can have high prediction accuracy only when they are based on protein stability characterization in different solution conditions.
Collapse
Affiliation(s)
- Lorenzo Gentiluomo
- Wyatt Technology Europe GmbH , Hochstrasse 18 , 56307 Dernbach , Germany.,Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| | - Hristo L Svilenov
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| | - Dillen Augustijn
- Department of Food Science, Faculty of Science , Copenhagen University , Rolighedsvej 26 , 1958 Frederiksberg , Denmark
| | - Inas El Bialy
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| | - Maria Laura Greco
- Dosage Form Design and Development , AstraZeneca , Sir Aaron Klug Building, Granta Park , Cambridge CB21 6GH , U.K
| | - Alina Kulakova
- Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , 2800 Kongens Lyngby , Denmark
| | - Sowmya Indrakumar
- Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , 2800 Kongens Lyngby , Denmark
| | | | - Marcello Martinez Morales
- Dosage Form Design and Development , AstraZeneca , Sir Aaron Klug Building, Granta Park , Cambridge CB21 6GH , U.K
| | - Christin Pohl
- Novozymes A/S , Krogshoejvej 36 , 2880 Bagsvaerd , Denmark
| | - Aisling Roche
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology , The University of Manchester , 131 Princess Street , Manchester M1 7DN , U.K
| | - Andreas Tosstorff
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| | - Robin Curtis
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology , The University of Manchester , 131 Princess Street , Manchester M1 7DN , U.K
| | - Jeremy P Derrick
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre , The University of Manchester , Oxford Road , Manchester M13 9PT , U.K
| | - Allan Nørgaard
- Novozymes A/S , Krogshoejvej 36 , 2880 Bagsvaerd , Denmark
| | - Tarik A Khan
- Pharmaceutical Development & Supplies, Pharma Technical Development Biologics Europe , F. Hoffmann-La Roche Ltd. , Grenzacherstrasse 124 , 4070 Basel , Switzerland
| | - Günther H J Peters
- Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , 2800 Kongens Lyngby , Denmark
| | - Alain Pluen
- School of Chemical Engineering and Analytical Science, Manchester Institute of Biotechnology , The University of Manchester , 131 Princess Street , Manchester M1 7DN , U.K
| | - Åsmund Rinnan
- Department of Food Science, Faculty of Science , Copenhagen University , Rolighedsvej 26 , 1958 Frederiksberg , Denmark
| | | | - Christopher F van der Walle
- Dosage Form Design and Development , AstraZeneca , Sir Aaron Klug Building, Granta Park , Cambridge CB21 6GH , U.K
| | - Shahid Uddin
- Dosage Form Design and Development , AstraZeneca , Sir Aaron Klug Building, Granta Park , Cambridge CB21 6GH , U.K
| | - Gerhard Winter
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| | - Dierk Roessner
- Wyatt Technology Europe GmbH , Hochstrasse 18 , 56307 Dernbach , Germany
| | - Pernille Harris
- Department of Chemistry , Technical University of Denmark , Kemitorvet 207 , 2800 Kongens Lyngby , Denmark
| | - Wolfgang Frieß
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics , Ludwig-Maximilians-Universitaet Muenchen , Butenandtstrasse 5 , 81377 Munich , Germany
| |
Collapse
|
49
|
Schleinitz M, Nolte L, Brandenbusch C. Predicting protein-protein interactions using the ePC-SAFT equation-of-state. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2019.112011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
50
|
Gentiluomo L, Roessner D, Streicher W, Mahapatra S, Harris P, Frieß W. Characterization of Native Reversible Self-Association of a Monoclonal Antibody Mediated by Fab-Fab Interaction. J Pharm Sci 2020; 109:443-451. [DOI: 10.1016/j.xphs.2019.09.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/22/2019] [Accepted: 09/16/2019] [Indexed: 11/28/2022]
|