1
|
Vetter J, Palagi I, Waisman A, Blaeser A. Recent advances in blood-brain barrier-on-a-chip models. Acta Biomater 2025; 197:1-28. [PMID: 40127880 DOI: 10.1016/j.actbio.2025.03.041] [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: 11/20/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
The blood-brain barrier is a physiological barrier between the vascular system and the nervous system. Under healthy conditions, it restricts the passage of most biomolecules into the brain, making drug development exceedingly challenging. Conventional cell-based in vitro models provide valuable insights into certain features of the BBB. Nevertheless, these models often lack the three-dimensional structure and dynamic interactions of the surrounding microenvironment, which greatly influence cell functionality. Consequently, considerable efforts have been made to enhance in vitro models for drug development and disease research. Recently, microfluidic organ-on-a-chip systems have emerged as promising candidates to better mimic the dynamic nature of the BBB. This review provides a comprehensive overview of recent BBB-on-chip devices. The typical building blocks, chip designs, the perfusion infrastructure, and readouts used to characterize and evaluate BBB formation are presented, analyzed, and discussed in detail. STATEMENT OF SIGNIFICANCE: The blood-brain barrier (BBB) is a highly selective barrier that controls what can enter the brain. While it protects the brain from harmful substances, it also hinders the delivery of treatments for neurological diseases such as Alzheimer's and Parkinson's. Due to its complexity, studying the BBB in living organisms remains difficult. However, recent advances in "organ-on-a-chip" technology have allowed scientists to create small, engineered models that replicate the BBB. These models provide a powerful platform to study diseases and test potential drugs with greater accuracy than traditional methods. Organ-on-a-chip devices are designed to mimic the behavior of organs or tissues in the human body, offering a more realistic and controlled environment for research. This review highlights recent breakthroughs in BBB-on-a-chip technology, showing how these models enhance current research and have the potential to transform the way we study brain diseases and develop new drugs. By integrating biology and engineering, BBB-on-a-chip technology has the potential to transform neuroscience research, improve drug development, and enhance our understanding of brain disorders.
Collapse
Affiliation(s)
- Johanna Vetter
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
| | - Ilaria Palagi
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany.
| |
Collapse
|
2
|
Huang B, Abedi M, Ahn G, Coventry B, Sappington I, Tang C, Wang R, Schlichthaerle T, Zhang JZ, Wang Y, Goreshnik I, Chiu CW, Chazin-Gray A, Chan S, Gerben S, Murray A, Wang S, O'Neill J, Yi L, Yeh R, Misquith A, Wolf A, Tomasovic LM, Piraner DI, Duran Gonzalez MJ, Bennett NR, Venkatesh P, Ahlrichs M, Dobbins C, Yang W, Wang X, Sahtoe DD, Vafeados D, Mout R, Shivaei S, Cao L, Carter L, Stewart L, Spangler JB, Roybal KT, Greisen PJ, Li X, Bernardes GJL, Bertozzi CR, Baker D. Designed endocytosis-inducing proteins degrade targets and amplify signals. Nature 2025; 638:796-804. [PMID: 39322662 PMCID: PMC11839401 DOI: 10.1038/s41586-024-07948-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/13/2024] [Indexed: 09/27/2024]
Abstract
Endocytosis and lysosomal trafficking of cell surface receptors can be triggered by endogenous ligands. Therapeutic approaches such as lysosome-targeting chimaeras1,2 (LYTACs) and cytokine receptor-targeting chimeras3 (KineTACs) have used this to target specific proteins for degradation by fusing modified native ligands to target binding proteins. Although powerful, these approaches can be limited by competition with native ligands and requirements for chemical modification that limit genetic encodability and can complicate manufacturing, and, more generally, there may be no native ligands that stimulate endocytosis through a given receptor. Here we describe computational design approaches for endocytosis-triggering binding proteins (EndoTags) that overcome these challenges. We present EndoTags for insulin-like growth factor 2 receptor (IGF2R) and asialoglycoprotein receptor (ASGPR), sortilin and transferrin receptors, and show that fusing these tags to soluble or transmembrane target protein binders leads to lysosomal trafficking and target degradation. As these receptors have different tissue distributions, the different EndoTags could enable targeting of degradation to different tissues. EndoTag fusion to a PD-L1 antibody considerably increases efficacy in a mouse tumour model compared to antibody alone. The modularity and genetic encodability of EndoTags enables AND gate control for higher-specificity targeted degradation, and the localized secretion of degraders from engineered cells. By promoting endocytosis, EndoTag fusion increases signalling through an engineered ligand-receptor system by nearly 100-fold. EndoTags have considerable therapeutic potential as targeted degradation inducers, signalling activators for endocytosis-dependent pathways, and cellular uptake inducers for targeted antibody-drug and antibody-RNA conjugates.
Collapse
Affiliation(s)
- Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Mohamad Abedi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Green Ahn
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Isaac Sappington
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Cong Tang
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Rong Wang
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas Schlichthaerle
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jason Z Zhang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Yujia Wang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Ching Wen Chiu
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Adam Chazin-Gray
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Stacey Gerben
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Analisa Murray
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Shunzhi Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Li Yi
- Novo Nordisk, Måløv, Denmark
| | | | | | | | - Luke M Tomasovic
- Departments of Biomedical Engineering and Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dan I Piraner
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Maria J Duran Gonzalez
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Nathaniel R Bennett
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Preetham Venkatesh
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Maggie Ahlrichs
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Craig Dobbins
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinru Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Dionne Vafeados
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Rubul Mout
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Shirin Shivaei
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Longxing Cao
- School of Life Sciences, Westlake University, Hangzhou, China
| | - Lauren Carter
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lance Stewart
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Kole T Roybal
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA, USA
| | | | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gonçalo J L Bernardes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Chen Z, Ji M, Qian J, Zhang Z, Zhang X, Gao H, Wang H, Wang R, Qi Y. ProBID-Net: a deep learning model for protein-protein binding interface design. Chem Sci 2024; 15:19977-19990. [PMID: 39568891 PMCID: PMC11575592 DOI: 10.1039/d4sc02233e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
Protein-protein interactions are pivotal in numerous biological processes. The computational design of these interactions facilitates the creation of novel binding proteins, crucial for advancing biopharmaceutical products. With the evolution of artificial intelligence (AI), protein design tools have swiftly transitioned from scoring-function-based to AI-based models. However, many AI models for protein design are constrained by assuming complete unfamiliarity with the amino acid sequence of the input protein, a feature most suited for de novo design but posing challenges in designing protein-protein interactions when the receptor sequence is known. To bridge this gap in computational protein design, we introduce ProBID-Net. Trained using natural protein-protein complex structures and protein domain-domain interface structures, ProBID-Net can discern features from known target protein structures to design specific binding proteins based on their binding sites. In independent tests, ProBID-Net achieved interface sequence recovery rates of 52.7%, 43.9%, and 37.6%, surpassing or being on par with ProteinMPNN in binding protein design. Validated using AlphaFold-Multimer, the sequences designed by ProBID-Net demonstrated a close correspondence between the design target and the predicted structure. Moreover, the model's output can predict changes in binding affinity upon mutations in protein complexes, even in scenarios where no data on such mutations were provided during training (zero-shot prediction). In summary, the ProBID-Net model is poised to significantly advance the design of protein-protein interactions.
Collapse
Affiliation(s)
- Zhihang Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Menglin Ji
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Jie Qian
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Zhe Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Xiangying Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Haotian Gao
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Haojie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Renxiao Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| | - Yifei Qi
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University 826 Zhangheng Road Shanghai 201203 People's Republic of China
| |
Collapse
|
4
|
Brylev VA, Ryabukhina EV, Nazarova EV, Samoylenkova NS, Gulyak EL, Sapozhnikova KA, Dzarieva FM, Ustinov AV, Pronin IN, Usachev DY, Kopylov AM, Golovin AV, Pavlova GV, Ryazantsev DY, Korshun VA. Towards Aptamer-Targeted Drug Delivery to Brain Tumors: The Synthesis of Ramified Conjugates of an EGFR-Specific Aptamer with MMAE on a Cathepsin B-Cleavable Linker. Pharmaceutics 2024; 16:1434. [PMID: 39598559 PMCID: PMC11597439 DOI: 10.3390/pharmaceutics16111434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Targeted delivery of chemotherapeutic agents is a well-established approach to cancer therapy. Antibody-drug conjugates (ADCs) typically carry toxic payloads attached to a tumor-associated antigen-targeting IgG antibody via an enzyme-cleavable linker that releases the drug inside the cell. Aptamers are a promising alternative to antibodies in terms of antigen targeting; however, their polynucleotide nature and smaller size result in a completely different PK/PD profile compared to an IgG. This may prove advantageous: owing to their lower molecular weight, aptamer-drug conjugates may achieve better penetration of solid tumors compared to ADCs. Methods: On the way to therapeutic aptamer-drug conjugates, we aimed to develop a versatile and modular approach for the assembly of aptamer-enzymatically cleavable payload conjugates of various drug-aptamer ratios. We chose the epidermal growth factor receptor (EGFR), a transmembrane protein often overexpressed in brain tumors, as the target antigen. We used the 46 mer EGFR-targeting DNA sequence GR-20, monomethylauristatin E (MMAE) on the cathepsin-cleavable ValCit-p-aminobenzylcarbamate linker as the payload, and pentaerythritol-based tetraazide as the branching point for the straightforward synthesis of aptamer-drug conjugates by means of a stepwise Cu-catalyzed azide-alkyne cycloaddition (CuAAC) click reaction. Results: Branched aptamer conjugates of 1:3, 2:2, and 3:1 stoichiometry were synthesized and showed higher cytotoxic activity compared to a 1:1 conjugate, particularly on several glioma cell lines. Conclusions: This approach is convenient and potentially applicable to any aptamer sequence, as well as other payloads and cleavable linkers, thus paving the way for future development of aptamer-drug therapeutics by easily providing a range of branched conjugates for in vitro and in vivo testing.
Collapse
Affiliation(s)
- Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Ekaterina V. Ryabukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | | | - Nadezhda S. Samoylenkova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Fatima M. Dzarieva
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Igor N. Pronin
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Dmitry Y. Usachev
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Alexey M. Kopylov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
| | - Andrey V. Golovin
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
- Department of Microbiology, Virology and Immunology, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Galina V. Pavlova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
- Department of Medical Genetics, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Dmitry Yu. Ryazantsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| |
Collapse
|
5
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
6
|
Jiang H, Jude KM, Wu K, Fallas J, Ueda G, Brunette TJ, Hicks DR, Pyles H, Yang A, Carter L, Lamb M, Li X, Levine PM, Stewart L, Garcia KC, Baker D. De novo design of buttressed loops for sculpting protein functions. Nat Chem Biol 2024; 20:974-980. [PMID: 38816644 PMCID: PMC11288887 DOI: 10.1038/s41589-024-01632-2] [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: 02/18/2023] [Accepted: 04/29/2024] [Indexed: 06/01/2024]
Abstract
In natural proteins, structured loops have central roles in molecular recognition, signal transduction and enzyme catalysis. However, because of the intrinsic flexibility and irregularity of loop regions, organizing multiple structured loops at protein functional sites has been very difficult to achieve by de novo protein design. Here we describe a solution to this problem that designs tandem repeat proteins with structured loops (9-14 residues) buttressed by extensive hydrogen bonding interactions. Experimental characterization shows that the designs are monodisperse, highly soluble, folded and thermally stable. Crystal structures are in close agreement with the design models, with the loops structured and buttressed as designed. We demonstrate the functionality afforded by loop buttressing by designing and characterizing binders for extended peptides in which the loops form one side of an extended binding pocket. The ability to design multiple structured loops should contribute generally to efforts to design new protein functions.
Collapse
Affiliation(s)
- Hanlun Jiang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kevin M Jude
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kejia Wu
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Biological Physics, Structure and Design Graduate Program, University of Washington, Seattle, WA, USA
| | - Jorge Fallas
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - George Ueda
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - T J Brunette
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R Hicks
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Harley Pyles
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Aerin Yang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mila Lamb
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul M Levine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
7
|
Sahtoe DD, Andrzejewska EA, Han HL, Rennella E, Schneider MM, Meisl G, Ahlrichs M, Decarreau J, Nguyen H, Kang A, Levine P, Lamb M, Li X, Bera AK, Kay LE, Knowles TPJ, Baker D. Design of amyloidogenic peptide traps. Nat Chem Biol 2024; 20:981-990. [PMID: 38503834 PMCID: PMC11288891 DOI: 10.1038/s41589-024-01578-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 02/09/2024] [Indexed: 03/21/2024]
Abstract
Segments of proteins with high β-strand propensity can self-associate to form amyloid fibrils implicated in many diseases. We describe a general approach to bind such segments in β-strand and β-hairpin conformations using de novo designed scaffolds that contain deep peptide-binding clefts. The designs bind their cognate peptides in vitro with nanomolar affinities. The crystal structure of a designed protein-peptide complex is close to the design model, and NMR characterization reveals how the peptide-binding cleft is protected in the apo state. We use the approach to design binders to the amyloid-forming proteins transthyretin, tau, serum amyloid A1 and amyloid β1-42 (Aβ42). The Aβ binders block the assembly of Aβ fibrils as effectively as the most potent of the clinically tested antibodies to date and protect cells from toxic Aβ42 species.
Collapse
Affiliation(s)
- Danny D Sahtoe
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- HHMI, University of Washington, Seattle, WA, USA.
- Hubrecht Institute, Utrecht, the Netherlands.
| | - Ewa A Andrzejewska
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Hannah L Han
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Enrico Rennella
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | | | - Georg Meisl
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Maggie Ahlrichs
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Hannah Nguyen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Paul Levine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mila Lamb
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinting Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lewis E Kay
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Tuomas P J Knowles
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- Cavendish Laboratory, University of Cambridge, Cambridge, UK
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- HHMI, University of Washington, Seattle, WA, USA.
| |
Collapse
|
8
|
Liu C, Wu K, Choi H, Han H, Zhang X, Watson JL, Shijo S, Bera AK, Kang A, Brackenbrough E, Coventry B, Hick DR, Hoofnagle AN, Zhu P, Li X, Decarreau J, Gerben SR, Yang W, Wang X, Lamp M, Murray A, Bauer M, Baker D. Diffusing protein binders to intrinsically disordered proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603789. [PMID: 39071267 PMCID: PMC11275890 DOI: 10.1101/2024.07.16.603789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Proteins which bind intrinsically disordered proteins (IDPs) and intrinsically disordered regions (IDRs) with high affinity and specificity could have considerable utility for therapeutic and diagnostic applications. However, a general methodology for targeting IDPs/IDRs has yet to be developed. Here, we show that starting only from the target sequence of the input, and freely sampling both target and binding protein conformation, RFdiffusion can generate binders to IDPs and IDRs in a wide range of conformations. We use this approach to generate binders to the IDPs Amylin, C-peptide and VP48 in a range of conformations with Kds in the 3 -100nM range. The Amylin binder inhibits amyloid fibril formation and dissociates existing fibers, and enables enrichment of amylin for mass spectrometry-based detection. For the IDRs G3bp1, common gamma chain (IL2RG) and prion, we diffused binders to beta strand conformations of the targets, obtaining 10 to 100 nM affinity. The IL2RG binder colocalizes with the receptor in cells, enabling new approaches to modulating IL2 signaling. Our approach should be widely useful for creating binders to flexible IDPs/IDRs spanning a wide range of intrinsic conformational preferences.
Collapse
Affiliation(s)
- Caixuan Liu
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kejia Wu
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Biological Physics, Structure and Design Graduate Program, University of Washington, Seattle, WA, USA
| | - Hojun Choi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Hannah Han
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xulie Zhang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Joseph L Watson
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sara Shijo
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98105, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Evans Brackenbrough
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Brian Coventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R Hick
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Andrew N Hoofnagle
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98105, USA
| | - Ping Zhu
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xingting Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Stacey R Gerben
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Xinru Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Mila Lamp
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Analisa Murray
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Magnus Bauer
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| |
Collapse
|
9
|
Zhou X, Wang Y, Li X, Zhou J, Yang W, Wang X, Jiao S, Zuo W, You Z, Ying W, Wu C, Bao J. O-GlcNAcylation regulates the stability of transferrin receptor (TFRC) to control the ferroptosis in hepatocellular carcinoma cells. Redox Biol 2024; 73:103182. [PMID: 38744192 PMCID: PMC11103954 DOI: 10.1016/j.redox.2024.103182] [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: 04/02/2024] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Ferroptosis is an iron-dependent programmed cell death (PCD) enforced by lipid peroxidation accumulation. Transferrin receptor (TFRC), one of the signature proteins of ferroptosis, is abundantly expressed in hepatocellular carcinoma (HCC). However, post-translational modification (PTM) of TFRC and the underlying mechanisms for ferroptosis regulation remain less understood. In this study, we found that TFRC undergoes O-GlcNAcylation, influencing Erastin-induced ferroptosis sensitivity in hepatocytes. Further mechanistic studies found that Erastin can trigger de-O-GlcNAcylation of TFRC at serine 687 (Ser687), which diminishes the binding of ubiquitin E3 ligase membrane-associated RING-CH8 (MARCH8) and decreases polyubiquitination on lysine 665 (Lys665), thereby enhancing TFRC stability that favors labile iron accumulation. Therefore, our findings report O-GlcNAcylation on an important regulatory protein of ferroptosis and reveal an intriguing mechanism by which HCC ferroptosis is controlled by an iron metabolism pathway.
Collapse
Affiliation(s)
- Xunyu Zhou
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Yida Wang
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Xiaoyu Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jing Zhou
- West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wanyi Yang
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Xin Wang
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Sitong Jiao
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Weibo Zuo
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Ziming You
- School of Life Sciences, Sichuan University, Chengdu, 610041, China
| | - Wantao Ying
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 102206, China.
| | - Chuanfang Wu
- School of Life Sciences, Sichuan University, Chengdu, 610041, China.
| | - Jinku Bao
- School of Life Sciences, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
10
|
Flynn CD, Chang D. Artificial Intelligence in Point-of-Care Biosensing: Challenges and Opportunities. Diagnostics (Basel) 2024; 14:1100. [PMID: 38893627 PMCID: PMC11172335 DOI: 10.3390/diagnostics14111100] [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: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
The integration of artificial intelligence (AI) into point-of-care (POC) biosensing has the potential to revolutionize diagnostic methodologies by offering rapid, accurate, and accessible health assessment directly at the patient level. This review paper explores the transformative impact of AI technologies on POC biosensing, emphasizing recent computational advancements, ongoing challenges, and future prospects in the field. We provide an overview of core biosensing technologies and their use at the POC, highlighting ongoing issues and challenges that may be solved with AI. We follow with an overview of AI methodologies that can be applied to biosensing, including machine learning algorithms, neural networks, and data processing frameworks that facilitate real-time analytical decision-making. We explore the applications of AI at each stage of the biosensor development process, highlighting the diverse opportunities beyond simple data analysis procedures. We include a thorough analysis of outstanding challenges in the field of AI-assisted biosensing, focusing on the technical and ethical challenges regarding the widespread adoption of these technologies, such as data security, algorithmic bias, and regulatory compliance. Through this review, we aim to emphasize the role of AI in advancing POC biosensing and inform researchers, clinicians, and policymakers about the potential of these technologies in reshaping global healthcare landscapes.
Collapse
Affiliation(s)
- Connor D. Flynn
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, IL 60208, USA
| | - Dingran Chang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
11
|
Sjöström DJ, Grill B, Ambrosetti E, Veetil AA, Mohlin C, Teixeira AI, Oberdofer G, Bjelic S. Affinity Maturated Transferrin Receptor Apical Domain Blocks Machupo Virus Glycoprotein Binding. J Mol Biol 2023; 435:168262. [PMID: 37678707 DOI: 10.1016/j.jmb.2023.168262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023]
Abstract
Transferrin receptor 1 (TfR) delivers iron across cellular membranes by shuttling the ion carrier protein transferrin. This ability to deliver large protein ligands inside cells is taken advantage of by pathogens to infiltrate human cells. Notably, the receptor's outermost ectodomain, the apical domain, is used as a point of attachment for several viruses including hemorrhagic arenaviruses. To better understand interactions with the receptor it would be advantageous to probe sequence determinants in the apical domain with viral spike proteins. Here, we carried out affinity maturation of our computationally designed apical domain from human TfR to identify underlying driving forces that lead to better binding. The improved variants were confirmed by in vitro surface plasmon resonance measurements with dissociation constants obtained in the lower nanomolar range. It was found that the strong binding affinities for the optimized variants matched the strength of interactions with the native receptor. The structure of the best variant was determined experimentally indicating that the conformational change in the hairpin binding motif at the protein-protein interface plays a crucial role. The experimental methodology can be straightforwardly applied to other arenavirus or pathogens that use the apical domain. It can further be useful to probe host-virus compatibility or therapeutic strategies based on the transferrin receptor decoys.
Collapse
Affiliation(s)
- Dick J Sjöström
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Birgit Grill
- Department of Biochemistry, Graz University of Technology, Graz, Austria
| | - Elena Ambrosetti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden
| | | | - Camilla Mohlin
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Sweden
| | - Gustav Oberdofer
- Department of Biochemistry, Graz University of Technology, Graz, Austria
| | - Sinisa Bjelic
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden.
| |
Collapse
|
12
|
Yu Y, Rué Casamajo A, Finnigan W, Schnepel C, Barker R, Morrill C, Heath RS, De Maria L, Turner NJ, Scrutton NS. Structure-Based Design of Small Imine Reductase Panels for Target Substrates. ACS Catal 2023; 13:12310-12321. [PMID: 37736118 PMCID: PMC10510103 DOI: 10.1021/acscatal.3c02278] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/20/2023] [Indexed: 09/23/2023]
Abstract
Biocatalysis is important in the discovery, development, and manufacture of pharmaceuticals. However, the identification of enzymes for target transformations of interest requires major screening efforts. Here, we report a structure-based computational workflow to prioritize protein sequences by a score based on predicted activities on substrates, thereby reducing a resource-intensive laboratory-based biocatalyst screening. We selected imine reductases (IREDs) as a class of biocatalysts to illustrate the application of the computational workflow termed IREDFisher. Validation by using published data showed that IREDFisher can retrieve the best enzymes and increase the hit rate by identifying the top 20 ranked sequences. The power of IREDFisher is confirmed by computationally screening 1400 sequences for chosen reductive amination reactions with different levels of complexity. Highly active IREDs were identified by only testing 20 samples in vitro. Our speed test shows that it only takes 90 min to rank 85 sequences from user input and 30 min for the established IREDFisher database containing 591 IRED sequences. IREDFisher is available as a user-friendly web interface (https://enzymeevolver.com/IREDFisher). IREDFisher enables the rapid discovery of IREDs for applications in synthesis and directed evolution studies, with minimal time and resource expenditure. Future use of the workflow with other enzyme families could be implemented following the modification of the workflow scoring function.
Collapse
Affiliation(s)
- Yuqi Yu
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
- Augmented
Biologics Discovery & Design, Department of Biologics Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB21 6GH, U.K.
| | - Arnau Rué Casamajo
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - William Finnigan
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Christian Schnepel
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Rhys Barker
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Charlotte Morrill
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Rachel S. Heath
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Leonardo De Maria
- Medicinal
Chemistry, Research and Early Development, Respiratory and Immunology
(RI), BioPharmaceuticals R&D, AstraZeneca, Gothenburg 43150, Sweden
| | - Nicholas J. Turner
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| | - Nigel S. Scrutton
- Department
of Chemistry, The University of Manchester,
Manchester Institute of Biotechnology, 131 Princess Street, Manchester M1 7DN, U.K.
| |
Collapse
|
13
|
Zhao C, Wang Z, Tang X, Qin J, Jiang Z. Recent advances in sensor-integrated brain-on-a-chip devices for real-time brain monitoring. Colloids Surf B Biointerfaces 2023; 229:113431. [PMID: 37473652 DOI: 10.1016/j.colsurfb.2023.113431] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 06/18/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
Brain science has remained in the global spotlight as an important field of scientific and technological discovery. Numerous in vitro and in vivo animal studies have been performed to understand the pathological processes involved in brain diseases and develop strategies for their diagnosis and treatment. However, owing to species differences between animals and humans, several drugs have shown high rates of treatment failure in clinical settings, hindering the development of diagnostic and treatment modalities for brain diseases. In this scenario, microfluidic brain-on-a-chip (BOC) devices, which allow the direct use of human tissues for experiments, have emerged as novel tools for effectively avoiding species differences and performing screening for new drugs. Although microfluidic BOC technology has achieved significant progress in recent years, monitoring slight changes in neurochemicals, neurotransmitters, and environmental states in the brain has remained challenging owing to the brain's complex environment. Hence, the integration of BOC with new sensors that have high sensitivity and high selectivity is urgently required for the real-time dynamic monitoring of BOC parameters. As sensor-based technologies for BOC have not been summarized, here, we review the principle, fabrication process, and application-based classification of sensor-integrated BOC, and then summarize the opportunities and challenges for their development. Generally, sensor-integrated BOC enables real-time monitoring and dynamic analysis, accurately measuring minute changes in the brain and thus enabling the realization of in vivo brain analysis and drug development.
Collapse
Affiliation(s)
- Chen Zhao
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Zihao Wang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaoying Tang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| | - Jieling Qin
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China; Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Zhenqi Jiang
- School of Medical Technology, School of Life Science, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China.
| |
Collapse
|
14
|
Jiang H, Jude KM, Wu K, Fallas J, Ueda G, Brunette TJ, Hicks D, Pyles H, Yang A, Carter L, Lamb M, Li X, Levine PM, Stewart L, Garcia KC, Baker D. De novo design of buttressed loops for sculpting protein functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.554384. [PMID: 37662224 PMCID: PMC10473674 DOI: 10.1101/2023.08.22.554384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
In natural proteins, structured loops play central roles in molecular recognition, signal transduction and enzyme catalysis. However, because of the intrinsic flexibility and irregularity of loop regions, organizing multiple structured loops at protein functional sites has been very difficult to achieve by de novo protein design. Here we describe a solution to this problem that generates structured loops buttressed by extensive hydrogen bonding interactions with two neighboring loops and with secondary structure elements. We use this approach to design tandem repeat proteins with buttressed loops ranging from 9 to 14 residues in length. Experimental characterization shows the designs are folded and monodisperse, highly soluble, and thermally stable. Crystal structures are in close agreement with the computational design models, with the loops structured and buttressed by their neighbors as designed. We demonstrate the functionality afforded by loop buttressing by designing and characterizing binders for extended peptides in which the loops form one side of an extended binding pocket. The ability to design multiple structured loops should contribute quite generally to efforts to design new protein functions.
Collapse
Affiliation(s)
- Hanlun Jiang
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Kevin M Jude
- Howard Hughes Medical Institute, Stanford University School of Medicine
| | - Kejia Wu
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
- Biological Physics, Structure and Design Graduate Program, University of Washington
| | - Jorge Fallas
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - George Ueda
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - T J Brunette
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Derrick Hicks
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Harley Pyles
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Aerin Yang
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine
| | - Lauren Carter
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Mila Lamb
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Xinting Li
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Paul M Levine
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - Lance Stewart
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine
- Department of Structural Biology, Stanford University School of Medicine
| | - David Baker
- Department of Biochemistry, University of Washington
- Institute for Protein Design, University of Washington
- Howard Hughes Medical Institute, Stanford University School of Medicine
- Howard Hughes Medical Institute, University of Washington
| |
Collapse
|
15
|
Deng S, Li C, Cao J, Cui Z, Du J, Fu Z, Yang H, Chen P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023; 13:4526-4558. [PMID: 37649608 PMCID: PMC10465229 DOI: 10.7150/thno.87266] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 08/02/2023] [Indexed: 09/01/2023] Open
Abstract
Drug evaluation has always been an important area of research in the pharmaceutical industry. However, animal welfare protection and other shortcomings of traditional drug development models pose obstacles and challenges to drug evaluation. Organ-on-a-chip (OoC) technology, which simulates human organs on a chip of the physiological environment and functionality, and with high fidelity reproduction organ-level of physiology or pathophysiology, exhibits great promise for innovating the drug development pipeline. Meanwhile, the advancement in artificial intelligence (AI) provides more improvements for the design and data processing of OoCs. Here, we review the current progress that has been made to generate OoC platforms, and how human single and multi-OoCs have been used in applications, including drug testing, disease modeling, and personalized medicine. Moreover, we discuss issues facing the field, such as large data processing and reproducibility, and point to the integration of OoCs and AI in data analysis and automation, which is of great benefit in future drug evaluation. Finally, we look forward to the opportunities and challenges faced by the coupling of OoCs and AI. In summary, advancements in OoCs development, and future combinations with AI, will eventually break the current state of drug evaluation.
Collapse
Affiliation(s)
- Shiwen Deng
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Caifeng Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Junxian Cao
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhao Cui
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jiang Du
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
| | - Zheng Fu
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Hongjun Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| | - Peng Chen
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China
- Yunnan Biovalley Pharmaceutical Co., Ltd, Kunming 650503, China
- Robot Intelligent Laboratory of Traditional Chinese Medicine, Experimental Research Center, China Academy of Chinese Medical Sciences & MEGAROBO, Beijing 100700, China
| |
Collapse
|
16
|
Mármol I, Abizanda-Campo S, Ayuso JM, Ochoa I, Oliván S. Towards Novel Biomimetic In Vitro Models of the Blood-Brain Barrier for Drug Permeability Evaluation. Bioengineering (Basel) 2023; 10:bioengineering10050572. [PMID: 37237642 DOI: 10.3390/bioengineering10050572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Current available animal and in vitro cell-based models for studying brain-related pathologies and drug evaluation face several limitations since they are unable to reproduce the unique architecture and physiology of the human blood-brain barrier. Because of that, promising preclinical drug candidates often fail in clinical trials due to their inability to penetrate the blood-brain barrier (BBB). Therefore, novel models that allow us to successfully predict drug permeability through the BBB would accelerate the implementation of much-needed therapies for glioblastoma, Alzheimer's disease, and further disorders. In line with this, organ-on-chip models of the BBB are an interesting alternative to traditional models. These microfluidic models provide the necessary support to recreate the architecture of the BBB and mimic the fluidic conditions of the cerebral microvasculature. Herein, the most recent advances in organ-on-chip models for the BBB are reviewed, focusing on their potential to provide robust and reliable data regarding drug candidate ability to reach the brain parenchyma. We point out recent achievements and challenges to overcome in order to advance in more biomimetic in vitro experimental models based on OOO technology. The minimum requirements that should be met to be considered biomimetic (cellular types, fluid flow, and tissular architecture), and consequently, a solid alternative to in vitro traditional models or animals.
Collapse
Affiliation(s)
- Inés Mármol
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Sara Abizanda-Campo
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jose M Ayuso
- Department of Dermatology, Department of Biomedical Engineering, and Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ignacio Ochoa
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
- CIBER Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sara Oliván
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain
| |
Collapse
|
17
|
Abstract
The failure of animal models to predict therapeutic responses in humans is a major problem that also brings into question their use for basic research. Organ-on-a-chip (organ chip) microfluidic devices lined with living cells cultured under fluid flow can recapitulate organ-level physiology and pathophysiology with high fidelity. Here, I review how single and multiple human organ chip systems have been used to model complex diseases and rare genetic disorders, to study host-microbiome interactions, to recapitulate whole-body inter-organ physiology and to reproduce human clinical responses to drugs, radiation, toxins and infectious pathogens. I also address the challenges that must be overcome for organ chips to be accepted by the pharmaceutical industry and regulatory agencies, as well as discuss recent advances in the field. It is evident that the use of human organ chips instead of animal models for drug development and as living avatars for personalized medicine is ever closer to realization.
Collapse
Affiliation(s)
- Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, USA.
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, USA.
| |
Collapse
|
18
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Sahtoe DD, Praetorius F, Courbet A, Hsia Y, Wicky BI, Edman NI, Miller LM, Timmermans BJR, Decarreau J, Morris HM, Kang A, Bera AK, Baker D. Reconfigurable asymmetric protein assemblies through implicit negative design. Science 2022; 375:eabj7662. [PMID: 35050655 PMCID: PMC9881579 DOI: 10.1126/science.abj7662] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Asymmetric multiprotein complexes that undergo subunit exchange play central roles in biology but present a challenge for design because the components must not only contain interfaces that enable reversible association but also be stable and well behaved in isolation. We use implicit negative design to generate β sheet-mediated heterodimers that can be assembled into a wide variety of complexes. The designs are stable, folded, and soluble in isolation and rapidly assemble upon mixing, and crystal structures are close to the computational models. We construct linearly arranged hetero-oligomers with up to six different components, branched hetero-oligomers, closed C4-symmetric two-component rings, and hetero-oligomers assembled on a cyclic homo-oligomeric central hub and demonstrate that such complexes can readily reconfigure through subunit exchange. Our approach provides a general route to designing asymmetric reconfigurable protein systems.
Collapse
Affiliation(s)
- Danny D. Sahtoe
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195,HHMI, University of Washington, Seattle, WA 98195
| | - Florian Praetorius
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Alexis Courbet
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195,HHMI, University of Washington, Seattle, WA 98195
| | - Yang Hsia
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Basile I.M. Wicky
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Natasha I. Edman
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA.,Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Lauren M. Miller
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Bart J. R. Timmermans
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Justin Decarreau
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Hana M. Morris
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Alex Kang
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Asim K. Bera
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195,Institute for Protein Design, University of Washington, Seattle, WA 98195,HHMI, University of Washington, Seattle, WA 98195,Corresponding author.
| |
Collapse
|
20
|
Sjöström DJ, Mohlin C, Ambrosetti E, Garforth SJ, Teixeira AI, Bjelic S. Motif-driven protein binder design towards transferrin receptor helical domain. FEBS J 2021; 289:2935-2947. [PMID: 34862739 DOI: 10.1111/febs.16311] [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: 06/24/2021] [Revised: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 10/19/2022]
Abstract
Human transferrin receptor 1 (TfR) is necessary for the delivery of the iron carrier protein transferrin into cells and can be utilized for targeted delivery across cellular membranes. Binding of transferrin to the receptor is regulated by hereditary hemochromatosis protein (HFE), an iron regulatory protein that partly shares a binding site with transferrin on TfR. Here, we derived essential binding interactions from HFE and computationally grafted these into a library of small protein scaffolds. One of the designed proteins, TB08, was further optimized computationally and experimentally to identify variants with improved binding to TfR. The optimized variant, TB08 S3.1, expressed well in the E. coli expression system and had an affinity to TfR in the low micromolar range, Kd ≈ 1 μm, as determined by surface plasmon resonance. A binding competition assay with transferrin further confirmed the interaction of the evolved variant to TfR at the shared binding surface. Additionally, the GFP-tagged evolved variant of TB08 demonstrated cellular internalization as determined by fluorescent and confocal microscopy in HeLa cells. The designed protein is small, allows for robust cargo tagging, and interacts specifically with TfR, thus making it a valuable tool for the characterization of TfR-mediated cellular transport mechanisms and for the assessment of engineering strategies for cargo delivery across cell membranes.
Collapse
Affiliation(s)
- Dick J Sjöström
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Camilla Mohlin
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| | - Elena Ambrosetti
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Scott J Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana I Teixeira
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Sinisa Bjelic
- Department of Chemistry and Biomedical Sciences, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
21
|
Tajeddin A, Mustafaoglu N. Design and Fabrication of Organ-on-Chips: Promises and Challenges. MICROMACHINES 2021; 12:1443. [PMID: 34945293 PMCID: PMC8707724 DOI: 10.3390/mi12121443] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/14/2021] [Accepted: 11/21/2021] [Indexed: 02/07/2023]
Abstract
The advent of the miniaturization approach has influenced the research trends in almost all disciplines. Bioengineering is one of the fields benefiting from the new possibilities of microfabrication techniques, especially in cell and tissue culture, disease modeling, and drug discovery. The limitations of existing 2D cell culture techniques, the high time and cost requirements, and the considerable failure rates have led to the idea of 3D cell culture environments capable of providing physiologically relevant tissue functions in vitro. Organ-on-chips are microfluidic devices used in this context as a potential alternative to in vivo animal testing to reduce the cost and time required for drug evaluation. This emerging technology contributes significantly to the development of various research areas, including, but not limited to, tissue engineering and drug discovery. However, it also brings many challenges. Further development of the technology requires interdisciplinary studies as some problems are associated with the materials and their manufacturing techniques. Therefore, in this paper, organ-on-chip technologies are presented, focusing on the design and fabrication requirements. Then, state-of-the-art materials and microfabrication techniques are described in detail to show their advantages and also their limitations. A comparison and identification of gaps for current use and further studies are therefore the subject of the final discussion.
Collapse
Affiliation(s)
- Alireza Tajeddin
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34596, Istanbul, Turkey;
| | - Nur Mustafaoglu
- Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34596, Istanbul, Turkey;
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Tuzla 34596, Istanbul, Turkey
| |
Collapse
|
22
|
Antibody-Based Inhibition of Pathogenic New World Hemorrhagic Fever Mammarenaviruses by Steric Occlusion of the Human Transferrin Receptor 1 Apical Domain. J Virol 2021; 95:e0186820. [PMID: 34132574 PMCID: PMC8354235 DOI: 10.1128/jvi.01868-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Pathogenic clade B New World mammarenaviruses (NWM) can cause Argentine, Venezuelan, Brazilian, and Bolivian hemorrhagic fevers. Sequence variability among NWM glycoproteins (GP) poses a challenge to the development of broadly neutralizing therapeutics against the entire clade of viruses. However, blockade of their shared binding site on the apical domain of human transferrin receptor 1 (hTfR1/CD71) presents an opportunity for the development of effective and broadly neutralizing therapeutics. Here, we demonstrate that the murine monoclonal antibody OKT9, which targets the apical domain of hTfR1, can sterically block cellular entry by viral particles presenting clade B NWM glycoproteins (GP1-GP2). OKT9 blockade is also effective against viral particles pseudotyped with glycoproteins of a recently identified pathogenic Sabia-like virus. With nanomolar affinity for hTfR1, the OKT9 antigen binding fragment (OKT9-Fab) sterically blocks clade B NWM-GP1s and reduces infectivity of an attenuated strain of Junin virus. Binding of OKT9 to the hTfR1 ectodomain in its soluble, dimeric state produces stable assemblies that are observable by negative-stain electron microscopy. A model of the OKT9-sTfR1 complex, informed by the known crystallographic structure of sTfR1 and a newly determined structure of the OKT9 antigen binding fragment (Fab), suggests that OKT9 and the Machupo virus GP1 share a binding site on the hTfR1 apical domain. The structural basis for this interaction presents a framework for the design and development of high-affinity, broadly acting agents targeting clade B NWMs. IMPORTANCE Pathogenic clade B NWMs cause grave infectious diseases, the South American hemorrhagic fevers. Their etiological agents are Junin (JUNV), Guanarito (GTOV), Sabiá (SABV), Machupo (MACV), Chapare (CHAV), and a new Sabiá-like (SABV-L) virus recently identified in Brazil. These are priority A pathogens due to their high infectivity and mortality, their potential for person-to-person transmission, and the limited availability of effective therapeutics and vaccines to curb their effects. While low homology between surface glycoproteins of NWMs foils efforts to develop broadly neutralizing therapies targeting NWMs, this work provides structural evidence that OKT9, a monoclonal antibody targeting a single NWM glycoprotein binding site on hTfR1, can efficiently prevent their entry into cells.
Collapse
|
23
|
Sahtoe DD, Coscia A, Mustafaoglu N, Miller LM, Olal D, Vulovic I, Yu TY, Goreshnik I, Lin YR, Clark L, Busch F, Stewart L, Wysocki VH, Ingber DE, Abraham J, Baker D. Transferrin receptor targeting by de novo sheet extension. Proc Natl Acad Sci U S A 2021; 118:e2021569118. [PMID: 33879614 PMCID: PMC8092486 DOI: 10.1073/pnas.2021569118] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The de novo design of polar protein-protein interactions is challenging because of the thermodynamic cost of stripping water away from the polar groups. Here, we describe a general approach for designing proteins which complement exposed polar backbone groups at the edge of beta sheets with geometrically matched beta strands. We used this approach to computationally design small proteins that bind to an exposed beta sheet on the human transferrin receptor (hTfR), which shuttles interacting proteins across the blood-brain barrier (BBB), opening up avenues for drug delivery into the brain. We describe a design which binds hTfR with a 20 nM Kd, is hyperstable, and crosses an in vitro microfluidic organ-on-a-chip model of the human BBB. Our design approach provides a general strategy for creating binders to protein targets with exposed surface beta edge strands.
Collapse
Affiliation(s)
- Danny D Sahtoe
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- HHMI, University of Washington, Seattle, WA 98195
| | - Adrian Coscia
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Nur Mustafaoglu
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115
| | - Lauren M Miller
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Daniel Olal
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Ivan Vulovic
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Ta-Yi Yu
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- Department of Bioengineering, University of Washington, Seattle, WA 98195
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Yu-Ru Lin
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Lars Clark
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Florian Busch
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH 43210
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Institute for Protein Design, University of Washington, Seattle, WA 98195
| | - Vicki H Wysocki
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210
- Resource for Native Mass Spectrometry Guided Structural Biology, The Ohio State University, Columbus, OH 43210
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA 02115
- Department of Surgery and Vascular Biology Program, Harvard Medical School and Boston Children's Hospital, Boston, MA 02115
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02539
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115;
- Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115
- Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA 98195;
- Institute for Protein Design, University of Washington, Seattle, WA 98195
- HHMI, University of Washington, Seattle, WA 98195
| |
Collapse
|