1
|
Imai K, Muguruma K, Nakamura A, Kusakari Y, Chang TC, Pradipta AR, Tanaka K. In Vivo Synthetic Anticancer Approach by Resourcing Mouse Blood Albumin as a Biocompatible Artificial Metalloenzyme. Angew Chem Int Ed Engl 2024; 63:e202411225. [PMID: 38989662 DOI: 10.1002/anie.202411225] [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: 06/14/2024] [Accepted: 07/08/2024] [Indexed: 07/12/2024]
Abstract
Methods for producing drugs directly at the cancer site, particularly using bioorthogonal metal catalysts, are being explored to mitigate the side effects of therapy. Albumin-based artificial metalloenzymes (ArMs) catalyze reactions in living mice while protecting the catalyst in the hydrophobic pocket. Here, we describe the in situ preparation and application of biocompatible tumor-targeting ArMs using circulating albumin, which is abundant in the bloodstream. The ArM was formed using blood albumin through the intravenous injection of ruthenium conjugated with an albumin-binding ligand; the tumor-targeting unit was conjugated to the ArM using its catalytic activity, and the ArM was transported to the cancer site. The delivered ArM catalyzed a second tagging reaction of the proapoptotic peptide on the cancer surface, successfully suppressing cancer proliferation. This approach, which efficiently leveraged the persisting reactivity twice in vivo, holds promise for future in vivo metal-catalyzed drug synthesis utilizing endogenous albumin.
Collapse
Affiliation(s)
- Kyosuke Imai
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Kyohei Muguruma
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
- Laboratory for Chemistry and Life Science, Tokyo Institute of Technology, R1-11, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - Akiko Nakamura
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Yuriko Kusakari
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| | - Tsung-Che Chang
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Ambara R Pradipta
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
| | - Katsunori Tanaka
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 Ookayama, Meguro, Tokyo, 152-8552, Japan
- Biofunctional Synthetic Chemistry Laboratory, Cluster for Pioneering Research, RIKEN, 2-1 Hirosawa, Wako, Saitama, 351-0198, Japan
| |
Collapse
|
2
|
Ludwig-Husemann A, Schertl P, Shrivastava A, Geckle U, Hafner J, Schaarschmidt F, Willenbacher N, Freudenberg U, Werner C, Lee-Thedieck C. A Multifunctional Nanostructured Hydrogel as a Platform for Deciphering Niche Interactions of Hematopoietic Stem and Progenitor Cells. Adv Healthc Mater 2024; 13:e2304157. [PMID: 38870600 DOI: 10.1002/adhm.202304157] [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: 11/24/2023] [Revised: 06/10/2024] [Indexed: 06/15/2024]
Abstract
For over half a century, hematopoietic stem cells (HSCs) have been used for transplantation therapy to treat severe hematologic diseases. Successful outcomes depend on collecting sufficient donor HSCs as well as ensuring efficient engraftment. These processes are influenced by dynamic interactions of HSCs with the bone marrow niche, which can be revealed by artificial niche models. Here, a multifunctional nanostructured hydrogel is presented as a 2D platform to investigate how the interdependencies of cytokine binding and nanopatterned adhesive ligands influence the behavior of human hematopoietic stem and progenitor cells (HSPCs). The results indicate that the degree of HSPC polarization and motility, observed when cultured on gels presenting the chemokine SDF-1α and a nanoscale-defined density of a cellular (IDSP) or extracellular matrix (LDV) α4β1 integrin binding motif, are differently influenced on hydrogels functionalized with the different ligand types. Further, SDF-1α promotes cell polarization but not motility. Strikingly, the degree of differentiation correlates negatively with the nanoparticle spacing, which determines ligand density, but only for the cellular-derived IDSP motif. This mechanism potentially offers a means of predictably regulating early HSC fate decisions. Consequently, the innovative multifunctional hydrogel holds promise for deciphering dynamic HSPC-niche interactions and refining transplantation therapy protocols.
Collapse
Affiliation(s)
- Anita Ludwig-Husemann
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
- Institute of Functional Interfaces, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Peter Schertl
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Ananya Shrivastava
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Udo Geckle
- Institute for Applied Materials - Energy Storage Systems, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Johanna Hafner
- Institute for Mechanical Process Engineering and Mechanics, Applied Mechanics Group, Karlsruhe Institute of Technology (KIT), Gotthard-Franz-Str. 3, 76131, Karlsruhe, Germany
| | - Frank Schaarschmidt
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| | - Norbert Willenbacher
- Institute for Mechanical Process Engineering and Mechanics, Applied Mechanics Group, Karlsruhe Institute of Technology (KIT), Gotthard-Franz-Str. 3, 76131, Karlsruhe, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden e.V, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden e.V, Max Bergmann Center of Biomaterials, Hohe Str. 6, 01069, Dresden, Germany
- Center for Regenerative Therapies Dresden, Technical University Dresden, Fetscherstr. 105, 01307, Dresden, Germany
| | - Cornelia Lee-Thedieck
- Institute of Cell Biology and Biophysics, Leibniz University Hannover, Herrenhäuser Str. 2, 30419, Hannover, Germany
| |
Collapse
|
3
|
Sethi S, Xu T, Sarkar A, Drees C, Jacob C, Walther A. Nuclease-Resistant L-DNA Tension Probes Enable Long-Term Force Mapping of Single Cells and Cell Consortia. Angew Chem Int Ed Engl 2024:e202413983. [PMID: 39212256 DOI: 10.1002/anie.202413983] [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: 07/24/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
DNA-based tension probes with precisely programmable force responses provide important insights into cellular mechanosensing. However, their degradability in cell culture limits their use for long-term imaging, for instance, when cells migrate, divide, and differentiate. This is a critical limitation for providing insights into mechanobiology for these longer-term processes. Here, we present DNA-based tension probes that are entirely designed based on the stereoisomer of biological D-DNA, i.e., L-DNA. We demonstrate that L-DNA tension probes are essentially indestructible by nucleases and provide days-long imaging without significant loss in image quality. We also show their superiority already for short imaging times commonly used for classical D-DNA tension probes. We showcase the potential of these resilient probes to image minute movements, and for generating long term force maps of single cells and of collectively migrating cell populations.
Collapse
Affiliation(s)
- Soumya Sethi
- Life-like Materials and Systems, Department of Chemistry, University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Tao Xu
- Life-like Materials and Systems, Department of Chemistry, University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Aritra Sarkar
- Life-like Materials and Systems, Department of Chemistry, University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Christoph Drees
- Life-like Materials and Systems, Department of Chemistry, University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| | - Claire Jacob
- Department of Biology, University of Mainz, Hanns-Dieter-Hüsch-Weg 15, 55128, Mainz, Germany
| | - Andreas Walther
- Life-like Materials and Systems, Department of Chemistry, University of Mainz, Duesbergweg 10-14, 55128, Mainz, Germany
| |
Collapse
|
4
|
Rozans S, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and Controlling the Proteolytic Degradation of Cell Adhesion Peptides. ACS Biomater Sci Eng 2024; 10:4916-4926. [PMID: 38968389 PMCID: PMC11322908 DOI: 10.1021/acsbiomaterials.4c00736] [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: 04/19/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work, we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalizations to better understand how simple modifications can be used to reduce the nonspecific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types nonspecifically degraded peptides as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 h, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with the cells. We found that simple modifications to the termini could significantly reduce or completely abolish nonspecific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed nonspecific degradation. We also found that there were minimal differences in peptide degradation across cell donors and that sequences mimicking different peptides commonly used to functionalize biomaterials all had significant nonspecific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
Affiliation(s)
- Samuel
J. Rozans
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Abolfazl Salehi Moghaddam
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Yingjie Wu
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Kayleigh Atanasoff
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Liliana Nino
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - Katelyn Dunne
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| | - E. Thomas Pashuck
- Department of Bioengineering, Lehigh University, 7
Asa Drive, Suite 205, Bethlehem, PA 18015, United States
| |
Collapse
|
5
|
Rijns L, Baker MB, Dankers PYW. Using Chemistry To Recreate the Complexity of the Extracellular Matrix: Guidelines for Supramolecular Hydrogel-Cell Interactions. J Am Chem Soc 2024; 146:17539-17558. [PMID: 38888174 PMCID: PMC11229007 DOI: 10.1021/jacs.4c02980] [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: 02/28/2024] [Revised: 05/17/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Hydrogels have emerged as a promising class of extracellular matrix (ECM)-mimicking materials in regenerative medicine. Here, we briefly describe current state-of-the-art of ECM-mimicking hydrogels, ranging from natural to hybrid to completely synthetic versions, giving the prelude to the importance of supramolecular interactions to make true ECM mimics. The potential of supramolecular interactions to create ECM mimics for cell culture is illustrated through a focus on two different supramolecular hydrogel systems, both developed in our laboratories. We use some recent, significant findings to present important design principles underlying the cell-material interaction. To achieve cell spreading, we propose that slow molecular dynamics (monomer exchange within fibers) is crucial to ensure the robust incorporation of cell adhesion ligands within supramolecular fibers. Slow bulk dynamics (stress-relaxation─fiber rearrangements, τ1/2 ≈ 1000 s) is required to achieve cell spreading in soft gels (<1 kPa), while gel stiffness overrules dynamics in stiffer gels. Importantly, this resonates with the findings of others which specialize in different material types: cell spreading is impaired in case substrate relaxation occurs faster than clutch binding and focal adhesion lifetime. We conclude with discussing considerations and limitations of the supramolecular approach as well as provide a forward thinking perspective to further understand supramolecular hydrogel-cell interactions. Future work may utilize the presented guidelines underlying cell-material interactions to not only arrive at the next generation of ECM-mimicking hydrogels but also advance other fields, such as bioelectronics, opening up new opportunities for innovative applications.
Collapse
Affiliation(s)
- Laura Rijns
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Matthew B. Baker
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology
Inspired Regenerative Medicine, Maastricht
University, 6200 MD Maastricht, The Netherlands
| | - Patricia Y. W. Dankers
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
- Department
of Chemical Engineering and Chemistry, Eindhoven
University of Technology, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
6
|
Rozans SJ, Moghaddam AS, Wu Y, Atanasoff K, Nino L, Dunne K, Pashuck ET. Quantifying and controlling the proteolytic degradation of cell adhesion peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590329. [PMID: 38712239 PMCID: PMC11071418 DOI: 10.1101/2024.04.19.590329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Peptides are widely used within biomaterials to improve cell adhesion, incorporate bioactive ligands, and enable cell-mediated degradation of the matrix. While many of the peptides incorporated into biomaterials are intended to be present throughout the life of the material, their stability is not typically quantified during culture. In this work we designed a series of peptide libraries containing four different N-terminal peptide functionalizations and three C-terminal functionalization to better understand how simple modifications can be used to reduce non-specific degradation of peptides. We tested these libraries with three cell types commonly used in biomaterials research, including mesenchymal stem/stromal cells (hMSCs), endothelial cells, and macrophages, and quantified how these cell types non-specifically degraded peptide as a function of terminal amino acid and chemistry. We found that peptides in solution which contained N-terminal amines were almost entirely degraded by 48 hours, irrespective of the terminal amino acid, and that degradation occurred even at high peptide concentrations. Peptides with C-terminal carboxylic acids also had significant degradation when cultured with cells. We found that simple modifications to the termini could significantly reduce or completely abolish non-specific degradation when soluble peptides were added to cells cultured on tissue culture plastic or within hydrogel matrices, and that functionalizations which mimicked peptide conjugations to hydrogel matrices significantly slowed non-specific degradation. We also found that there were minimal differences across cell donors, and that sequences mimicking different peptides commonly-used to functionalized biomaterials all had significant non-specific degradation. Finally, we saw that there was a positive trend between RGD stability and hMSC spreading within hydrogels, indicating that improving the stability of peptides within biomaterial matrices may improve the performance of engineered matrices.
Collapse
|
7
|
Creeden JF, Sevier J, Zhang JT, Lapitsky Y, Brunicardi FC, Jin G, Nemunaitis J, Liu JY, Kalinoski A, Rao D, Liu SH. Smart exosomes enhance PDAC targeted therapy. J Control Release 2024; 368:413-429. [PMID: 38431093 DOI: 10.1016/j.jconrel.2024.02.037] [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: 11/02/2023] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 03/05/2024]
Abstract
Exosomes continue to attract interest as a promising nanocarrier drug delivery technology. They are naturally derived nanoscale extracellular vesicles with innate properties well suited to shuttle proteins, lipids, and nucleic acids between cells. Nonetheless, their clinical utility is currently limited by several major challenges, such as their inability to target tumor cells and a high proportion of clearance by the mononuclear phagocyte system (MPS) of the liver and spleen. To overcome these limitations, we developed "Smart Exosomes" that co-display RGD and CD47p110-130 through CD9 engineering (ExoSmart). The resultant ExoSmart demonstrates enhanced binding capacity to αvβ3 on pancreatic ductal adenocarcinoma (PDAC) cells, resulting in amplified cellular uptake in in vitro and in vivo models and increased chemotherapeutic efficacies. Simultaneously, ExoSmart significantly reduced liver and spleen clearance of exosomes by inhibiting macrophage phagocytosis via CD47p110-130 interaction with signal regulatory proteins (SIRPα) on macrophages. These studies demonstrate that an engineered exosome drug delivery system increases PDAC therapeutic efficacy by enhancing active PDAC targeting and prolonging circulation times, and their findings hold tremendous translational potential for cancer therapy while providing a concrete foundation for future work utilizing novel peptide-engineered exosome strategies.
Collapse
Affiliation(s)
- Justin F Creeden
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jonathan Sevier
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA
| | - Yakov Lapitsky
- Department of Chemical Engineering, University of Toledo, Toledo, OH, USA
| | - F Charles Brunicardi
- Department of Surgery, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Ge Jin
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | - Jing-Yuan Liu
- Department of Medicine, University of Toledo, Toledo, OH, USA
| | | | | | - Shi-He Liu
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
8
|
Li X, Combs JD, Salaita K, Shu X. Polarized focal adhesion kinase activity within a focal adhesion during cell migration. Nat Chem Biol 2023; 19:1458-1468. [PMID: 37349581 PMCID: PMC10732478 DOI: 10.1038/s41589-023-01353-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 05/03/2023] [Indexed: 06/24/2023]
Abstract
Focal adhesion kinase (FAK) relays integrin signaling from outside to inside cells and contributes to cell adhesion and motility. However, the spatiotemporal dynamics of FAK activity in single FAs is unclear due to the lack of a robust FAK reporter, which limits our understanding of these essential biological processes. Here we have engineered a genetically encoded FAK activity sensor, dubbed FAK-separation of phases-based activity reporter of kinase (SPARK), which visualizes endogenous FAK activity in living cells and vertebrates. Our work reveals temporal dynamics of FAK activity during FA turnover. Most importantly, our study unveils polarized FAK activity at the distal tip of newly formed single FAs in the leading edge of a migrating cell. By combining FAK-SPARK with DNA tension probes, we show that tensions applied to FAs precede FAK activation and that FAK activity is proportional to the strength of tension. These results suggest tension-induced polarized FAK activity in single FAs, advancing the mechanistic understanding of cell migration.
Collapse
Affiliation(s)
- Xiaoquan Li
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | | | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Xiaokun Shu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA.
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
9
|
Zheng X, Li Y, Cui T, Yang J, Meng X, Wang H, Chen L, He J, Chen N, Meng L, Ding L, Xie R. Traceless Protein-Selective Glycan Labeling and Chemical Modification. J Am Chem Soc 2023; 145:23670-23680. [PMID: 37857274 DOI: 10.1021/jacs.3c07889] [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: 10/21/2023]
Abstract
Executing glycan editing at a molecular level not only is pivotal for the elucidation of complicated mechanisms involved in glycan-relevant biological processes but also provides a promising solution to potentiate disease therapy. However, the precision control of glycan modification or glyco-editing on a selected glycoprotein is by far a grand challenge. Of note is to preserve the intact cellular glycan landscape, which is preserved after editing events are completed. We report herein a versatile, traceless glycan modification methodology for customizing the glycoforms of targeted proteins (subtypes), by orchestrating chemical- and photoregulation in a protein-selective glycoenzymatic system. This method relies on a three-module, ligand-photocleavable linker-glycoenzyme (L-P-G) conjugate. We demonstrated that RGD- or synthetic carbohydrate ligand-containing conjugates (RPG and SPG) would not activate until after the ligand-receptor interaction is accomplished (chemical regulation). RPG and SPG can both release the glycoenzyme upon photoillumination (photoregulation). The adjustable glycoenzyme activity, combined with ligand recognition selectivity, minimizes unnecessary glycan editing perturbation, and photolytic cleavage enables precise temporal control of editing events. An altered target protein turnover and dimerization were observed in our system, emphasizing the significance of preserving the native physiological niche of a particular protein when precise modification on the carbohydrate epitope occurs.
Collapse
Affiliation(s)
- Xiaocui Zheng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Tongxiao Cui
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jing Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Xiangfeng Meng
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Nan Chen
- ChinaChomiX Biotech (Nanjing) Co., Ltd., Nanjing 210061, China
| | - Liying Meng
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| | - Ran Xie
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, China
| |
Collapse
|
10
|
Nhàn NTT, Yamada T, Yamada KH. Peptide-Based Agents for Cancer Treatment: Current Applications and Future Directions. Int J Mol Sci 2023; 24:12931. [PMID: 37629112 PMCID: PMC10454368 DOI: 10.3390/ijms241612931] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Peptide-based strategies have received an enormous amount of attention because of their specificity and applicability. Their specificity and tumor-targeting ability are applied to diagnosis and treatment for cancer patients. In this review, we will summarize recent advancements and future perspectives on peptide-based strategies for cancer treatment. The literature search was conducted to identify relevant articles for peptide-based strategies for cancer treatment. It was performed using PubMed for articles in English until June 2023. Information on clinical trials was also obtained from ClinicalTrial.gov. Given that peptide-based strategies have several advantages such as targeted delivery to the diseased area, personalized designs, relatively small sizes, and simple production process, bioactive peptides having anti-cancer activities (anti-cancer peptides or ACPs) have been tested in pre-clinical settings and clinical trials. The capability of peptides for tumor targeting is essentially useful for peptide-drug conjugates (PDCs), diagnosis, and image-guided surgery. Immunomodulation with peptide vaccines has been extensively tested in clinical trials. Despite such advantages, FDA-approved peptide agents for solid cancer are still limited. This review will provide a detailed overview of current approaches, design strategies, routes of administration, and new technological advancements. We will highlight the success and limitations of peptide-based therapies for cancer treatment.
Collapse
Affiliation(s)
- Nguyễn Thị Thanh Nhàn
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
| | - Tohru Yamada
- Department of Surgery, Division of Surgical Oncology, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Richard & Loan Hill Department of Biomedical Engineering, University of Illinois College of Engineering, Chicago, IL 60607, USA
| | - Kaori H. Yamada
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA;
- Department of Ophthalmology & Visual Sciences, University of Illinois College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
11
|
Sutherland M, Gordon A, Al-Shammari FOFO, Throup A, Cilia La Corte A, Philippou H, Shnyder SD, Patterson LH, Sheldrake HM. Synthesis and Biological Evaluation of Cyclobutane-Based β3 Integrin Antagonists: A Novel Approach to Targeting Integrins for Cancer Therapy. Cancers (Basel) 2023; 15:4023. [PMID: 37627051 PMCID: PMC10452181 DOI: 10.3390/cancers15164023] [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: 06/15/2023] [Revised: 07/25/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
The Arg-Gly-Asp (RGD)-binding family of integrin receptors, and notably the β3 subfamily, are key to multiple physiological processes involved in tissue development, cancer proliferation, and metastatic dissemination. While there is compelling preclinical evidence that both αvβ3 and αIIbβ3 are important anticancer targets, most integrin antagonists developed to target the β3 integrins are highly selective for αvβ3 or αIIbβ3. We report the design, synthesis, and biological evaluation of a new structural class of ligand-mimetic β3 integrin antagonist. These new antagonists combine a high activity against αvβ3 with a moderate affinity for αIIbβ3, providing the first evidence for a new approach to integrin targeting in cancer.
Collapse
Affiliation(s)
- Mark Sutherland
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| | - Andrew Gordon
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| | | | - Adam Throup
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| | - Amy Cilia La Corte
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Helen Philippou
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Steven D. Shnyder
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| | | | - Helen M. Sheldrake
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK
| |
Collapse
|
12
|
Zhang L, Wang P, Zhou XQ, Bretin L, Zeng X, Husiev Y, Polanco EA, Zhao G, Wijaya LS, Biver T, Le Dévédec SE, Sun W, Bonnet S. Cyclic Ruthenium-Peptide Conjugates as Integrin-Targeting Phototherapeutic Prodrugs for the Treatment of Brain Tumors. J Am Chem Soc 2023. [PMID: 37379365 DOI: 10.1021/jacs.3c04855] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
To investigate the potential of tumor-targeting photoactivated chemotherapy, a chiral ruthenium-based anticancer warhead, Λ/Δ-[Ru(Ph2phen)2(OH2)2]2+, was conjugated to the RGD-containing Ac-MRGDH-NH2 peptide by direct coordination of the M and H residues to the metal. This design afforded two diastereoisomers of a cyclic metallopeptide, Λ-[1]Cl2 and Δ-[1]Cl2. In the dark, the ruthenium-chelating peptide had a triple action. First, it prevented other biomolecules from coordinating with the metal center. Second, its hydrophilicity made [1]Cl2 amphiphilic so that it self-assembled in culture medium into nanoparticles. Third, it acted as a tumor-targeting motif by strongly binding to the integrin (Kd = 0.061 μM for the binding of Λ-[1]Cl2 to αIIbβ3), which resulted in the receptor-mediated uptake of the conjugate in vitro. Phototoxicity studies in two-dimensional (2D) monolayers of A549, U87MG, and PC-3 human cancer cell lines and U87MG three-dimensional (3D) tumor spheroids showed that the two isomers of [1]Cl2 were strongly phototoxic, with photoindexes up to 17. Mechanistic studies indicated that such phototoxicity was due to a combination of photodynamic therapy (PDT) and photoactivated chemotherapy (PACT) effects, resulting from both reactive oxygen species generation and peptide photosubstitution. Finally, in vivo studies in a subcutaneous U87MG glioblastoma mice model showed that [1]Cl2 efficiently accumulated in the tumor 12 h after injection, where green light irradiation generated a stronger tumoricidal effect than a nontargeted analogue ruthenium complex [2]Cl2. Considering the absence of systemic toxicity for the treated mice, these results demonstrate the high potential of light-sensitive integrin-targeted ruthenium-based anticancer compounds for the treatment of brain cancer in vivo.
Collapse
Affiliation(s)
- Liyan Zhang
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Peiyuan Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
- Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
| | - Xue-Quan Zhou
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Ludovic Bretin
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Xiaolong Zeng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Yurii Husiev
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Ehider A Polanco
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Gangyin Zhao
- Leiden Institute of Biology, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Lukas S Wijaya
- Leiden Academic Centre for Drug Research, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Tarita Biver
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy
| | - Sylvia E Le Dévédec
- Leiden Academic Centre for Drug Research, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian 116024, P. R. China
| | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Universiteit Leiden, Einsteinweg 55, 2333 CC Leiden, Netherlands
| |
Collapse
|
13
|
Li Y, Huo F, Chen L, Wang H, Wu J, Zhang P, Feng N, Li W, Wang L, Wang Y, Wang X, Yang X, Lu Z, Mao Y, Yan C, Ding L, Ju H. Protein-Targeted Glycan Editing on Living Cells Disrupts KRAS Signaling. Angew Chem Int Ed Engl 2023; 62:e202218148. [PMID: 37103924 DOI: 10.1002/anie.202218148] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 04/28/2023]
Abstract
The frequent mutation of KRAS oncogene in some of the most lethal human cancers has spurred incredible efforts to develop KRAS inhibitors, yet only one covalent inhibitor for the KRASG12C mutant has been approved to date. New venues to interfere with KRAS signaling are desperately needed. Here, we report a "localized oxidation-coupling" strategy to achieve protein-specific glycan editing on living cells for disrupting KRAS signaling. This glycan remodeling method exhibits excellent protein and sugar specificity and is applicable to different donor sugars and cell types. Attachment of mannotriose to the terminal galactose/N-acetyl-D-galactosamine epitopes of integrin αv β3 , a membrane receptor upstream of KRAS, blocks its binding to galectin-3, suppresses the activation of KRAS and downstream effectors, and mitigates KRAS-driven malignant phenotypes. Our work represents the first successful attempt to interfere with KRAS activity by manipulating membrane receptor glycosylation.
Collapse
Affiliation(s)
- Yiran Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Fan Huo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
| | - Liusheng Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Haiqi Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Jianzhuang Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
| | - Peiwen Zhang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Nan Feng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Wei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Lan Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Yichun Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, 211816, Nanjing, P. R. China
| | - Xiaoliang Yang
- State Key Laboratory of Coordination Chemistry and Jiangsu Key Laboratory of Advanced Organic Materials, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| | - Zhiqiang Lu
- College of Chemistry and Chemical Engineering and Henan Key Laboratory of Function-Oriented Porous Materials, Luo-yang Normal University, 471934, Luoyang, P. R. China
| | - Yang Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, 510006, Guangzhou, P. R. China
| | - Chao Yan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210023, Nanjing, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, P. R. China
| | - Lin Ding
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, P. R. China
| | - Huangxian Ju
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 210023, Nanjing, P. R. China
| |
Collapse
|
14
|
Tsai TS, Tsai IH. Full sequencing and comparison of five venom metalloproteases of Trimeresurus gracilis: The PI-enzyme is most similar to okinalysin but the PIII-enzyme is most similar to Crotalus venom enzymes. Toxicon 2023; 225:107053. [PMID: 36758773 DOI: 10.1016/j.toxicon.2023.107053] [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: 12/05/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023]
Abstract
The cDNAs encoding the Zn+2-metalloproteases (SVMPs) of Trimeresurus gracilis (abbreviated as Tgc), a pitviper endemic to Taiwan, were cloned from venom glands and sequenced. The amino-acid sequences of five novel SVMPs, including one P-III, three P-II and one P-I class enzymes, were thus deduced and subjected to BLAST-analyses. The P-III enzyme (designated as Tgc-PIII) is structurally most similar to the PIII-SVMPs of New World pitvipers but not similar to the PIII-SVMP of Ovophis okinavensis. Sequence-similarity analysis of 22 homologous PIII-SVMPs reveal three major structural subtypes of the pitviper PIII-SVMPs, which possibly have different substrate specificities. In addition, Tgc-PIII and the PI-class SVMP (named Tgc-MP) were isolated from the venom and verified by mass spectrometry. All the three deduced sequences of PII-SVMPs (Tgc-PIIs) contain an abnormal Zn+2-binding-site in their catalytic-domain, and an identical "long-disintegrin" domain. The predicted 85-residues disintegrin, gracilisin, bears high similarities to some long-disintegrins of the New-World pitvipers and salmosin3. By BLAST search and comparison, Tgc-MP is 96% similar to okinalysin, the hemorrhagic PI-SVMP of O. okinavensis, rather than any other PI-SVMPs in the databanks. Our results confirm the fast evolution of Tgc-SVMPs as well as their structural similarities to different SVMP-classes of the New-World pitvipers and of O. okinavensis, respectively. The implications of our findings are discussed along with our previous sequence comparisons of venom phospholipases A2 and ten venom serine proteases of Tgc.
Collapse
Affiliation(s)
- Tein-Shun Tsai
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, Taiwan
| | - Inn-Ho Tsai
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
15
|
Debnath S, Vignesh SR, Satpati P, Chatterjee S. Position of Geminal Substitution of γ Amino Acid Residues Modulates Their Ability to Form Isolated Non‐Helical C
12
β‐turn Mimics. ChemistrySelect 2023. [DOI: 10.1002/slct.202204255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Affiliation(s)
- Swapna Debnath
- Department of Chemistry Indian Institute of Technology, Guwahati Guwahati Assam India
| | - S. R. Vignesh
- Biosciences and Bioengineering Indian Institute of Technology Guwahati, Guwahati Assam India
| | - Priyadarshi Satpati
- Biosciences and Bioengineering Indian Institute of Technology Guwahati, Guwahati Assam India
| | - Sunanda Chatterjee
- Department of Chemistry Indian Institute of Technology, Guwahati Guwahati Assam India
| |
Collapse
|
16
|
Sugimoto Y, Suga T, Kato N, Umino M, Yamayoshi A, Mukai H, Kawakami S. Microfluidic Post-Insertion Method for the Efficient Preparation of PEGylated Liposomes Using High Functionality and Quality Lipids. Int J Nanomedicine 2022; 17:6675-6686. [PMID: 36597433 PMCID: PMC9805735 DOI: 10.2147/ijn.s390866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/21/2022] [Indexed: 12/29/2022] Open
Abstract
Introduction Targeted liposomes using ligand peptides have been applied to deliver therapeutic agents to the target sites. The post-insertion method is commonly used because targeted liposomes can be prepared by simple mixing of ligand peptide-lipid and liposomes. A large-scale preparation method is required for the clinical application of ligand-peptide-modified liposomes. Large-scale preparation involves an increase in volume and a change in the preparation conditions. Therefore, the physicochemical properties of liposomes may change owing to large alterations in the preparation conditions. To address this issue, we focused on a microfluidic device and developed a novel ligand peptide modification method, the microfluidic post-insertion method. Methods We used integrin αvβ3-targeted GRGDS (RGD) and cyclic RGDfK (cRGD)-modified high functionality and quality (HFQ) lipids, which we had previously developed. First, the preparation conditions of the total flow rate in the microfluidic device for modifying HFQ lipids to polyethylene glycol (PEG)-modified (PEGylated) liposomes were optimized by evaluating the physicochemical properties of the liposomes. The targeting ability of integrin αvβ3-expressing colon 26 murine colorectal carcinoma cells was evaluated by comparing the cellular association properties of the liposomes prepared by the conventional post-insertion method. Results When the RGD-HFQ lipid was modified into PEGylated liposomes by varying the total flow rate (1, 6, and 12 mL/min) of the microfluidic device, as the total flow rate increased, the polydispersity index also increased, whereas the particle size did not change. Furthermore, the RGD- and cRGD-modified PEGylated liposomes prepared at a total flow rate of 1 mL/min showed high cellular association properties equivalent to those prepared by the conventional post-insertion method. Conclusion Microfluidic post-insertion method of HFQ lipids might be useful for clinical application and large-scale preparation of targeted liposomes.
Collapse
Affiliation(s)
- Yuri Sugimoto
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Department of Chemistry of Functional Molecules, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tadaharu Suga
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Naoya Kato
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Mizuki Umino
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Asako Yamayoshi
- Department of Chemistry of Functional Molecules, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hidefumi Mukai
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan,Correspondence: Shigeru Kawakami, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8588, Japan, Tel/Fax +81 95 819 8563, Email
| |
Collapse
|
17
|
Li Y, Chen Q, Pan X, Lu W, Zhang J. New insight into the application of fluorescence platforms in tumor diagnosis: From chemical basis to clinical application. Med Res Rev 2022; 43:570-613. [PMID: 36420715 DOI: 10.1002/med.21932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 09/22/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022]
Abstract
Early and rapid diagnosis of tumors is essential for clinical treatment or management. In contrast to conventional means, bioimaging has the potential to accurately locate and diagnose tumors at an early stage. Fluorescent probe has been developed as an ideal tool to visualize tumor sites and to detect biological molecules which provides a requirement for noninvasive, real-time, precise, and specific visualization of structures and complex biochemical processes in vivo. Rencently, the development of synthetic organic chemistry and new materials have facilitated the development of near-infrared small molecular sensing platforms and nanoimaging platforms. This provides a competitive tool for various fields of bioimaging such as biological structure and function imaging, disease diagnosis, in situ at the in vivo level, and real-time dynamic imaging. This review systematically focused on the recent progress of small molecular near-infrared fluorescent probes and nano-fluorescent probes as new biomedical imaging tools in the past 3-5 years, and it covers the application of tumor biomarker sensing, tumor microenvironment imaging, and tumor vascular imaging, intraoperative guidance and as an integrated platform for diagnosis, aiming to provide guidance for researchers to design and develop future biomedical diagnostic tools.
Collapse
Affiliation(s)
- Yanchen Li
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center Xi'an Jiaotong University Xi'an China
| | - Qinhua Chen
- Department of Pharmacy Shenzhen Baoan Authentic TCM Therapy Hospital Shenzhen China
| | - Xiaoyan Pan
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center Xi'an Jiaotong University Xi'an China
| | - Wen Lu
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center Xi'an Jiaotong University Xi'an China
| | - Jie Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Health Science Center Xi'an Jiaotong University Xi'an China
| |
Collapse
|
18
|
Jain K, Kanchanawong P, Sheetz MP, Zhou X, Cai H, Changede R. Ligand functionalization of titanium nanopattern enables the analysis of cell-ligand interactions by super-resolution microscopy. Nat Protoc 2022; 17:2275-2306. [PMID: 35896742 DOI: 10.1038/s41596-022-00717-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/26/2022] [Indexed: 12/19/2022]
Abstract
The spatiotemporal aspects of early signaling events during interactions between cells and their environment dictate multiple downstream outcomes. While advances in nanopatterning techniques have allowed the isolation of these signaling events, a major limitation of conventional nanopatterning methods is its dependence on gold (Au) or related materials that plasmonically quench fluorescence and, thus, are incompatible with super-resolution fluorescence microscopy. Here we describe a novel method that integrates nanopatterning with single-molecule resolution fluorescence imaging, thus enabling mechanistic dissection of molecular-scale signaling events in conjunction with nanoscale geometry manipulation. Our method exploits nanofabricated titanium (Ti) whose oxide (TiO2) is a dielectric material with no plasmonic effects. We describe the surface chemistry for decorating specific ligands such as cyclo-RGD (arginine, glycine and aspartate: a ligand for fibronectin-binding integrins) on TiO2 nanoline and nanodot substrates, and demonstrate the ability to perform dual-color super-resolution imaging on these patterns. Ti nanofabrication is similar to other metallic materials like Au, while the functionalization of TiO2 is relatively fast, safe, economical, easy to set up with commonly available reagents, and robust against environmental parameters such as humidity. Fabrication of nanopatterns takes ~2-3 d, preparation for functionalization ~1.5-2 d, and functionalization 3 h, after which cell culture and imaging experiments can be performed. We suggest that this method may facilitate the interrogation of nanoscale geometry and force at single-molecule resolution, and should find ready applications in early detection and interpretation of physiochemical signaling events at the cell membrane in the fields of cell biology, immunology, regenerative medicine, and related fields.
Collapse
Affiliation(s)
- Kashish Jain
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.,Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.,Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, USA
| | - Xianjing Zhou
- Center for Nanoscale Materials, Argonne National Laboratory, Lemont, IL, USA
| | - Haogang Cai
- Tech4Health Institute and Department of Radiology, NYU Langone Health, New York, NY, USA.
| | - Rishita Changede
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore. .,TeOra Pte. Ltd, Singapore, Singapore.
| |
Collapse
|
19
|
Iodophenyl-conjugated closo-dodecaborate as a promising small boron molecule that binds to serum albumin and accumulates in tumor. Bioorg Med Chem Lett 2022; 72:128869. [PMID: 35772634 DOI: 10.1016/j.bmcl.2022.128869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/19/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022]
Abstract
The development of novel boron carriers applicable to various cancers is required for further expansion of boron neutron capture therapy (BNCT). In this study, we took advantage of the fact that serum albumin accumulates in tumors and developed a boron compound that interacts non-covalently with the serum albumin. 4-Iodophenylbutanamide was chosen as an albumin ligand and conjugated with closo-dodecaborate (boron-conjugated 4-iodophenylbutanamide: BC-IP). BC-IP was found to be water soluble with low cytotoxicity. The IC50 values of BC-IP were 475 µM for U87MG cells, 738 µM for HeLa cells, and > 1000 µM for A549 cells. The dissociation constant (Kd) value of BC-IP to HSA was 148 ± 8 μM, while that of disodium closo-dodecaborate (4) was > 1000 μM. Significant tumor accumulation was observed in the U87MG tumor mouse model 3 h after injection. The boron concentration in the tumor reached a maximum of 11 μgB/g at 3 h and gradually decreased to 2.4 and 2.3 μgB/g at 12 and 24 h, respectively.
Collapse
|
20
|
Makino A, Ueda M, Uematsu Y, Ohora T, Ohtani T, Miyagawa S, Fujibayashi Y, Okazawa H, Tokunaga Y, Kiyono Y. Development of Low Molecular Weight Ligands for Integrin α<sub>v</sub>β<sub>3</sub>. Chem Pharm Bull (Tokyo) 2022; 70:293-299. [DOI: 10.1248/cpb.c21-01085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Akira Makino
- Biomedical Imaging Research Center, University of Fukui
| | - Masahiro Ueda
- Graduate School of Engineering, Materials Science and Engineering, University of Fukui
| | - Yoshitaka Uematsu
- Graduate School of Engineering, Materials Science and Engineering, University of Fukui
| | - Takuya Ohora
- Graduate School of Engineering, Materials Science and Engineering, University of Fukui
| | - Takayuki Ohtani
- Graduate School of Engineering, Materials Science and Engineering, University of Fukui
| | - Shinobu Miyagawa
- Graduate School of Engineering, Materials Science and Engineering, University of Fukui
| | | | | | - Yuji Tokunaga
- Research and Education Program for Life Science, University of Fukui
| | - Yasushi Kiyono
- Research and Education Program for Life Science, University of Fukui
| |
Collapse
|
21
|
Feasibility of 125I-RGD uptake as a marker of angiogenesis after myocardial infarction. Ann Nucl Med 2021; 36:235-243. [PMID: 34837162 DOI: 10.1007/s12149-021-01695-4] [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: 08/10/2021] [Accepted: 11/10/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE Angiogenesis is an important process facilitating the healing process after myocardial infarction. 125I-RGD imaging may be a promising candidate to image angiogenesis but may also detect inflammation. METHODS Left coronary artery was occluded for 30 min, followed by reperfusion in a rat model (n = 31). One, 3, 7 and 14 days, 1 and 2 months later, Triple-tracer autoradiography was performed. 125I-RGD (1.5 MBq) and 201Tl (15 MBq) were injected at 80 and 10 min before sacrifice. Left coronary artery was reoccluded and 99mTc-MIBI (150-180 MBq) was injected 1 min before sacrifice to verify the area at risk. Angiogenesis and macrophage infiltration were evaluated by immunohistochemical analysis with anti-alpha-smooth muscle actin and anti-CD68, respectively. RESULTS 125I-RGD uptake ratio in the area at risk was weak at day 3 (1.23 ± 0.23 but increased markedly and peaked at day 7 (2.27 ± 0.37) followed by a gradual reduction until 1 and 2 months later (1.93 ± 0.16 at 1 month, 1.58 ± 0.15 at 2 month). In the immunohistochemical analysis, copious staining of anti-CD68 cells was observed, with anti-SMA cells stained only minimally at day 3. The number of anti-CD68 cells was decreased significantly at day 7 but largely absent at 1 month. Anti-SMA positive cells peaked at day 7 and reduced gradually until 1 month. CONCLUSIONS Myocardial 125I-RGD uptake reflects angiogenesis rather than inflammation after myocardial infarction.
Collapse
|
22
|
Benli-Hoppe T, Göl Öztürk Ş, Öztürk Ö, Berger S, Wagner E, Yazdi M. Transferrin Receptor Targeted Polyplexes Completely Comprised of Sequence-Defined Components. Macromol Rapid Commun 2021; 43:e2100602. [PMID: 34713524 DOI: 10.1002/marc.202100602] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/20/2021] [Indexed: 11/08/2022]
Abstract
Human transferrin protein (Tf) modified polyplexes have already displayed encouraging potential for receptor-mediated nucleic acid delivery into tumors. The use of a blood-derived targeting protein and polydisperse macromolecular cationic subunits however presents a practical challenge for pharmaceutical grade production. Here, Tf receptor (TfR) targeted small interfering RNA (siRNA) polyplexes are designed that are completely composed of synthetic, monodisperse, and sequence-defined subunits generated by solid-phase supported synthesis. An optimized cationizable lipo-oligoaminoamide (lipo-OAA) is used for siRNA core polyplex formation, and a retro-enantio peptide (reTfR) attached via a monodisperse polyethylene glycol (PEG) spacer via click chemistry is applied for targeting. Improved gene silencing is demonstrated in TfR-expressing KB and DU145 cells. Analogous plasmid DNA (pDNA) polyplexes are successfully used for receptor-mediated gene delivery in TfR-rich K562 cells and Neuro2a cells. Six lipo-OAAs differing in their lipidic domain and redox-sensitive attachment of lipid residues are tested in order to evaluate the impact of core polyplex stability on receptor-dependent gene transfer.
Collapse
Affiliation(s)
- Teoman Benli-Hoppe
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Şurhan Göl Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Özgür Öztürk
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Simone Berger
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Center for Drug Research, and Center for Nanoscience, Ludwig-Maximilians-Universität Munich, Butenandtstrasse 5-13, Munich, 81377, Germany
| |
Collapse
|
23
|
Xia Y, To J, Chan N, Hu S, Liew HT, Balamkundu S, Zhang X, Lescar J, Bhattacharjya S, Tam JP, Liu C. N
γ
‐Hydroxyasparagine: A Multifunctional Unnatural Amino Acid That is a Good P1 Substrate of Asparaginyl Peptide Ligases. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202108125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Yiyin Xia
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Janet To
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Ning‐Yu Chan
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Side Hu
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Heng Tai Liew
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Seetharamsing Balamkundu
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
- Singapore-MIT Alliance for Research and Technology Singapore 138602 Singapore
| | - Xiaohong Zhang
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Julien Lescar
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - James P. Tam
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| | - Chuan‐Fa Liu
- School of Biological Sciences Nanyang Technological University 60 Nanyang Drive Singapore 637551 Singapore
| |
Collapse
|
24
|
Xia Y, To J, Chan NY, Hu S, Liew HT, Balamkundu S, Zhang X, Lescar J, Bhattacharjya S, Tam JP, Liu CF. N γ -Hydroxyasparagine: A Multifunctional Unnatural Amino Acid That is a Good P1 Substrate of Asparaginyl Peptide Ligases. Angew Chem Int Ed Engl 2021; 60:22207-22211. [PMID: 34396662 DOI: 10.1002/anie.202108125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Indexed: 11/10/2022]
Abstract
Peptidyl asparaginyl ligases (PALs) are powerful tools for peptide macrocyclization. Herein, we report that a derivative of Asn, namely Nγ -hydroxyasparagine or Asn(OH), is an unnatural P1 substrate of PALs. By Asn(OH)-mediated cyclization, we prepared cyclic peptides as new matrix metalloproteinase 2 (MMP2) inhibitors displaying the hydroxamic acid moiety of Asn(OH) as the key pharmacophore. The most potent cyclic peptide (Ki =2.8±0.5 nM) was built on the hyperstable tetracyclic scaffold of rhesus theta defensin-1. The Asn(OH) residue in the cyclized peptides can also be readily oxidized to Asp. By this approach, we synthesized several bioactive Asp-containing cyclic peptides (MCoTI-II, kB2, SFTI, and integrin-targeting RGD peptides) that are otherwise difficult targets for PAL-catalyzed cyclization owing to unfavorable kinetics of the P1-Asp substrates. This study demonstrates that substrate engineering is a useful strategy to expand the application of PAL ligation in the synthesis of therapeutic cyclic peptides.
Collapse
Affiliation(s)
- Yiyin Xia
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Janet To
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Ning-Yu Chan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Side Hu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Heng Tai Liew
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Seetharamsing Balamkundu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.,Singapore-MIT Alliance for Research and Technology, Singapore, 138602, Singapore
| | - Xiaohong Zhang
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Surajit Bhattacharjya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - James P Tam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Chuan-Fa Liu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| |
Collapse
|
25
|
Ahmadi P, Muguruma K, Chang TC, Tamura S, Tsubokura K, Egawa Y, Suzuki T, Dohmae N, Nakao Y, Tanaka K. In vivo metal-catalyzed SeCT therapy by a proapoptotic peptide. Chem Sci 2021; 12:12266-12273. [PMID: 34603656 PMCID: PMC8480321 DOI: 10.1039/d1sc01784e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023] Open
Abstract
Selective cell tagging (SeCT) therapy is a strategy for labeling a targeted cell with certain chemical moieties via a catalytic chemical transformation in order to elicit a therapeutic effect. Herein, we report a cancer therapy based on targeted cell surface tagging with proapoptotic peptides (Ac-GGKLFG-X; X = reactive group) that induce apoptosis when attached to the cell surface. Using either Au-catalyzed amidation or Ru-catalyzed alkylation, these proapoptotic peptides showed excellent therapeutic effects both in vitro and in vivo. In particular, co-treatment with proapoptotic peptide and the carrier-Ru complex significantly and synergistically inhibited tumor growth and prolonged survival rate of tumor-bearing mice after only a single injection. This is the first report of Ru catalyst application in vivo, and this approach could be used in SeCT for cancer therapy.
Collapse
Affiliation(s)
- Peni Ahmadi
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Kyohei Muguruma
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama Meguro Tokyo 152-8552 Japan
| | - Tsung-Che Chang
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Satoru Tamura
- Department of Medicinal and Organic Chemistry, School of Pharmacy, Iwate Medical University Yahaba Iwate 028-3694 Japan
| | - Kazuki Tsubokura
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Yasuko Egawa
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Takehiro Suzuki
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Naoshi Dohmae
- Biomolecular Characterization Unit, RIKEN Center for Sustainable Resource Science 2-1 Hirosawa Wako Saitama 351-0198 Japan
| | - Yoichi Nakao
- School of Advanced Science and Engineering, Department of Chemistry and Biochemistry, Waseda University 3-4-1 Okubo Shinjuku Tokyo 169-8555 Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry, RIKEN Cluster for Pioneering Research 2-1 Hirosawa Wako Saitama 351-0198 Japan
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology 2-12-1 Ookayama Meguro Tokyo 152-8552 Japan
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University 18 Kremlyovskaya Street Kazan 420008 Russia
| |
Collapse
|
26
|
Shimizu Y, Ando M, Iikuni S, Watanabe H, Ono M. Development of a hydroxamamide-based bifunctional chelating agent to prepare technetium-99m-labeled bivalent ligand probes. Sci Rep 2021; 11:18714. [PMID: 34548586 PMCID: PMC8455562 DOI: 10.1038/s41598-021-98235-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/31/2021] [Indexed: 11/17/2022] Open
Abstract
Hydroxamamide (Ham) is a thiol-free chelating agent that forms technetium-99m (99mTc)-complexes with a metal-to-ligand ratio of 1:2 under moderate reaction conditions. Therefore, Ham-based chelating agents will produce 99mTc-labeled compounds with a bivalent targeting scaffold. For their universal usage, we developed a novel Ham-based bifunctional chelating agent, “Ham-Mal”, with a maleimide group that can easily conjugate with a thiol group, for to preparing 99mTc-labeled bivalent ligand probes. Ham-Mal was synthesized by a four-step reaction, and then reacted with cysteine or c(RGDfC) to produce Ham-Cys or Ham-RGD. These precursors were reacted with 99mTcO4- for 10 min under room temperature to obtain 99mTc-(Ham-Cys)2 and 99mTc -(Ham-RGD)2. The cellular uptake level of 99mTc-(Ham-RGD)2 by U87MG (high Integrin ɑvβ3 expression) cells was significantly higher than that by PC3 (low Integrin ɑvβ3 expression) cells at 60 min after the incubation, and the uptake was significantly suppressed by pre-treatment for 15 min with excess c(RGDfK) peptide. In the in vivo study with U87MG/PC3 dual xenografted BALB/c-nu mice, the radioactivity of U87MG tumor tissue was significantly higher than that of PC3 tumor tissue at 360 min after the administration of 99mTc-(Ham-RGD)2. These results suggest Ham-Mal may have potential as a bifunctional chelating agent for 99mTc-labeled bivalent ligand probes.
Collapse
Affiliation(s)
- Yoichi Shimizu
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan. .,Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoin-kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Masato Ando
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Shimpei Iikuni
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Hiroyuki Watanabe
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Masahiro Ono
- Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29, Yoshida Shimoadachi-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
27
|
Duan Y, Glazier R, Bazrafshan A, Hu Y, Rashid SA, Petrich BG, Ke Y, Salaita K. Mechanically Triggered Hybridization Chain Reaction. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107660] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Yuxin Duan
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | | | - Yuesong Hu
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | - Sk Aysha Rashid
- Department of Chemistry Emory University Atlanta GA 30322 USA
| | | | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| | - Khalid Salaita
- Department of Chemistry Emory University Atlanta GA 30322 USA
- Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University Atlanta GA 30322 USA
| |
Collapse
|
28
|
Duan Y, Glazier R, Bazrafshan A, Hu Y, Rashid SA, Petrich BG, Ke Y, Salaita K. Mechanically Triggered Hybridization Chain Reaction. Angew Chem Int Ed Engl 2021; 60:19974-19981. [PMID: 34242462 PMCID: PMC8390435 DOI: 10.1002/anie.202107660] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Indexed: 01/16/2023]
Abstract
Cells transmit piconewton forces to receptors to mediate processes such as migration and immune recognition. A major challenge in quantifying such forces is the sparsity of cell mechanical events. Accordingly, molecular tension is typically quantified with high resolution fluorescence microscopy, which hinders widespread adoption and application. Here, we report a mechanically triggered hybridization chain reaction (mechano-HCR) that allows chemical amplification of mechanical events. The amplification is triggered when a cell receptor mechanically denatures a duplex revealing a cryptic initiator to activate the HCR reaction in situ. Importantly, mechano-HCR enables direct readout of pN forces using a plate reader. We leverage this capability and measured mechano-IC50 for aspirin, Y-27632, and eptifibatide. Given that cell mechanical phenotypes are of clinical importance, mechano-HCR may offer a convenient route for drug discovery, personalized medicine, and disease diagnosis.
Collapse
Affiliation(s)
- Yuxin Duan
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Roxanne Glazier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | | | - Yuesong Hu
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Sk Aysha Rashid
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Brian G Petrich
- Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
29
|
Sun T, Wei L, Tian H, Zhan W, Ma H, Nie D, Wang S, Chen X, Tang G. Novel PET/CT tracers for targeted imaging of membrane receptors to evaluate cardiomyocyte apoptosis and tissue repair process in a rat model of myocardial infarction. Apoptosis 2021; 26:460-473. [PMID: 34185202 DOI: 10.1007/s10495-021-01681-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to employ novel tracers PET imaging approach to define the time course and intensity of myocardial repair after apoptosis and to correlate the imaging signal to immunohistochemical staining in myocardial infarction (MI). We designed novel αVβ3-targeted and radio-functionalized tracers for detection of apoptosis in H9C2 cells and myocardial tissue. MI rats were imaged with [18F]FDG, [18F]ANP-Cin or [18F]ANP-RGD2 using a small-animal PET/CT device. Rats were sacrificed, and tissue samples from viable and injured myocardial areas were sectioned for TUNEL assay and histology. The uncorrected radiochemical yield of [18F]ANP-Cin and [18F]ANP-RGD2 were 41.3 ± 5.4% and 21.17 ± 4.7%, respectively. Two tracers meet many criteria for cardiac imaging, including high stability, high binding, no toxicity, fast renal clearance and excellent biodistribution in rat models. The uptake of [18F]ANP-Cin was significantly higher on the 1st and 3rd day than the 7th or 28th day after MI induction, a timeframe associated with increased cardiomyocyte apoptosis. Higher uptake of [18F]ANP-Cin was observed in MI rats than in N-acetylcysteine (NAC)-treated rats on the 3rd days. In contrast with [18F]ANP-Cin, no hot-spots was observed with [18F]ANP-RGD2 on the 1st day and more hot-spots was observed from the 3rd day to the 7th day, then less on the 28th days in the high apoptotic site. There was no uptake of [18F]FDG in or around the apoptotic region. On the 7th day the uptake of [18F]ANP-RGD2 was higher in NAC-treated rats than MI rats. [18F]ANP-Cin and [18F]ANP-RGD2 are superior to [18F]FDG for PET/CT imaging for evaluation of cardiomyocyte apoptosis and tissue repair processes in the MI rats.
Collapse
Affiliation(s)
- Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Wanlin Zhan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hui Ma
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shilin Wang
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xin Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ganghua Tang
- Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
30
|
Huang Y, Gu B, Salles-Crawley II, Taylor KA, Yu L, Ren J, Liu X, Emerson M, Longstaff C, Hughes AD, Thom SA, Xu XY, Chen R. Fibrinogen-mimicking, multiarm nanovesicles for human thrombus-specific delivery of tissue plasminogen activator and targeted thrombolytic therapy. SCIENCE ADVANCES 2021; 7:7/23/eabf9033. [PMID: 34078604 PMCID: PMC8172176 DOI: 10.1126/sciadv.abf9033] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/14/2021] [Indexed: 05/03/2023]
Abstract
Clinical use of tissue plasminogen activator (tPA) in thrombolytic therapy is limited by its short circulation time and hemorrhagic side effects. Inspired by fibrinogen binding to activated platelets, we report a fibrinogen-mimicking, multiarm nanovesicle for thrombus-specific tPA delivery and targeted thrombolysis. This biomimetic system is based on the lipid nanovesicle coated with polyethylene glycol (PEG) terminally conjugated with a cyclic RGD (cRGD) peptide. Our experiments with human blood demonstrated its highly selective binding to activated platelets and efficient tPA release at a thrombus site under both static and physiological flow conditions. Its clot dissolution time in a microfluidic system was comparable to that of free tPA. Furthermore, we report a purpose-built computational model capable of simulating targeted thrombolysis of the tPA-loaded nanovesicle and with a potential in predicting the dynamics of thrombolysis in physiologically realistic scenarios. This combined experimental and computational work presents a promising platform for development of thrombolytic nanomedicines.
Collapse
Affiliation(s)
- Yu Huang
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Boram Gu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
- School of Chemical Engineering, Chonnam National University, Gwangju, Republic of Korea
| | - Isabelle I Salles-Crawley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kirk A Taylor
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Li Yu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Jie Ren
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Xuhan Liu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Michael Emerson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Colin Longstaff
- Biotherapeutics Section, National Institute for Biological Standards and Control, South Mimms, Herts, UK
| | - Alun D Hughes
- Institute of Cardiovascular Science, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at University College London, London, UK
| | - Simon A Thom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK.
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK.
| |
Collapse
|
31
|
Hong YT, Teo JY, Jeon H, Kong H. Shear-Resistant, Biological Tethering of Nanostimulators for Enhanced Therapeutic Cell Paracrine Factor Secretion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17276-17288. [PMID: 33830733 PMCID: PMC10440850 DOI: 10.1021/acsami.1c01520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mesenchymal stromal cells (MSCs) secreting multiple growth factors and immunomodulatory cytokines are promising for regenerative medicine. To further enhance their secretory activity, efforts have emerged to tether nanosized carriers of secretory stimuli, named nanostimulators, to the MSC surface by forming nonchemical bonds. Despite some successes, there is a great need to improve the retention of nanostimulators during transport through a syringe needle, where high shear stress exerted on the cell surface separates them. To this end, we hypothesize that poly(lactic-co-glycolic acid)-block-hyaluronic acid (PLGA-HA) conjugated with integrin-binding RGD peptides, denoted PLGA-HA-RGD, can form nanostimulators that remain on the cell surface stably during the injection. The resulting HA-CD44 and RGD-integrin bonds would synergistically increase the adhesion strength of nanostimulators. Interestingly, nanostimulators prepared with PLGA-HA-RGD show 3- to 6-fold higher retention than those made with PLGA-HA. Therefore, the PLGA-HA-RGD nanostimulators induced MSCs to secrete 1.5-fold higher vascular endothelial growth factors and a 1.2-fold higher tissue inhibitor of matrix metalloproteinase-1 as compared to PLGA-HA nanostimulators. Consequently, MSCs tethered with PLGA-HA-RGD nanostimulators served to stimulate endothelial cell activities to form a blood vessel-like endothelial lumen with increased length and number of junctions. The nanostimulator design strategy would also be broadly applicable to regulate, protect, and home a broad array of therapeutic or immune cells by tethering carriers with bioactive molecules of interest.
Collapse
Affiliation(s)
- Yu-Tong Hong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jye Yng Teo
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore
| | - Hojeong Jeon
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 136-701, Republic of Korea
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
32
|
Ashokkumar P, Collot M, Klymchenko AS. Fluorogenic Squaraine Dendrimers for Background-Free Imaging of Integrin Receptors in Cancer Cells. Chemistry 2021; 27:6795-6803. [PMID: 33567148 DOI: 10.1002/chem.202100480] [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: 02/07/2021] [Indexed: 11/06/2022]
Abstract
To overcome the limited brightness of existing fluorogenic molecular probes for biomolecular targets, we introduce a concept of fluorogenic dendrimer probe, which undergoes polarity-dependent switching due to intramolecular aggregation-caused quenching of its fluorophores. Based on a rational design of dendrimers with four and eight squaraine dyes, we found that octamer bearing dyes through a sufficiently long PEG(8) linker displays >400-fold fluorescence enhancement from water to non-polar dioxane. High extinction coefficient (≈2,300,000 m-1 cm-1 ) resulted from eight squaraine dyes and quantum yield (≈25 %) make this octamer the brightest environment-sensitive fluorogenic molecule reported to date. Its conjugate with cyclic RGD used at low concentration (3 nm) enables integrin-specific fluorescence imaging of cancer cells with high signal-to-background ratio. The developed dendrimer probe is a "golden middle" between molecular probes and nanoparticles, combining small size, turn-on response and high brightness, important for bioimaging.
Collapse
Affiliation(s)
- Pichandi Ashokkumar
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, 67401, Illkirch, France.,Department of Bioelectronics and Biosensors, Alagappa University, Karaikudi, 630 004, Tamil Nadu, India
| | - Mayeul Collot
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, 67401, Illkirch, France
| | - Andrey S Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Faculté de Pharmacie, Université de Strasbourg, 74, Route du Rhin, 67401, Illkirch, France
| |
Collapse
|
33
|
Vong K, Tahara T, Urano S, Nasibullin I, Tsubokura K, Nakao Y, Kurbangalieva A, Onoe H, Watanabe Y, Tanaka K. Disrupting tumor onset and growth via selective cell tagging (SeCT) therapy. SCIENCE ADVANCES 2021; 7:7/17/eabg4038. [PMID: 33893089 PMCID: PMC8064634 DOI: 10.1126/sciadv.abg4038] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/08/2021] [Indexed: 05/16/2023]
Abstract
This study presents the early framework of selective cell tagging (SeCT) therapy, which is the concept of preferentially labeling specific cells in vivo with chemical moieties that can elicit a therapeutic response. Using glycosylated artificial metalloenzyme (GArM)-based protein labeling, this study reports two separate functional strategies. In one approach, early tumor onset can be suppressed by tagging cancer cells in living mice with an integrin-blocking cyclic-Arg-Gly-Asp (cRGD) moiety, thereby disrupting cell adhesion onto the extracellular matrix. In another approach, tumor growth in mice can be reduced by tagging with a cytotoxic doxorubicin moiety. Subsequent cell death occurs following internalization and drug release. Overall, experiments have shown that mouse populations receiving the mixture of SeCT labeling reagents exhibited a significant delay/reduction in tumor onset and growth compared with controls. Highlighting its adaptability, this work represents a foundational step for further development of SeCT therapy and its potential therapeutic applications.
Collapse
Affiliation(s)
- Kenward Vong
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Tsuyoshi Tahara
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Sayaka Urano
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
| | - Igor Nasibullin
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Kazuki Tsubokura
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
- School of Advanced Science and Engineering, Department of Chemistry and Biochemistry, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Yoichi Nakao
- School of Advanced Science and Engineering, Department of Chemistry and Biochemistry, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Almira Kurbangalieva
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
| | - Hirotaka Onoe
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Yasuyoshi Watanabe
- RIKEN Center for Life Science Technologies, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Katsunori Tanaka
- Biofunctional Synthetic Chemistry Laboratory, RIKEN Cluster for Pioneering Research, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan.
- GlycoTargeting Research Laboratory, RIKEN Baton Zone Program, 2-1 Hirosawa, Wako-shi, Saitama 351-0198, Japan
- Biofunctional Chemistry Laboratory, A. Butlerov Institute of Chemistry, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia
- Department of Chemical Science and Engineering, School of Materials and Chemical Technology, Tokyo Institute of Technology, 2-12-1 O-okayama, Meguro-ku, Tokyo 152-8552, Japan
| |
Collapse
|
34
|
Banerjee S, Szepes M, Dibbert N, Rios-Camacho JC, Kirschning A, Gruh I, Dräger G. Dextran-based scaffolds for in-situ hydrogelation: Use for next generation of bioartificial cardiac tissues. Carbohydr Polym 2021; 262:117924. [PMID: 33838803 DOI: 10.1016/j.carbpol.2021.117924] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/13/2021] [Accepted: 03/05/2021] [Indexed: 10/21/2022]
Abstract
In pursuit of a chemically-defined matrix for in vitro cardiac tissue generation, we present dextran (Dex)-derived hydrogels as matrices suitable for bioartificial cardiac tissues (BCT). The dextran hydrogels were generated in situ by using hydrazone formation as the crosslinking reaction. Material properties were flexibly adjusted, by varying the degrees of derivatization and the molecular weight of dextran used. Furthermore, to modulate dextran's bioactivity, cyclic pentapeptide RGD was coupled to its backbone. BCTs were generated by using a blend of modified dextran and human collagen (hColI) in combination with induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) and fibroblasts. These hColI + Dex blends with or without RGD supported tissue formation and functional maturation of CMs. Contraction forces (hColI + Dex-RGD: 0.27 ± 0.02 mN; hColI + Dex: 0.26 ± 0.01 mN) and frequencies were comparable to published constructs. Thus, we could demonstrate that, independent of the presence of RGD, our covalently linked dextran hydrogels are a promising matrix for building cardiac grafts.
Collapse
Affiliation(s)
- Samhita Banerjee
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Monika Szepes
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Nick Dibbert
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Julio-Cesar Rios-Camacho
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Andreas Kirschning
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany
| | - Ina Gruh
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Gerald Dräger
- Institute for Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167 Hannover, Germany.
| |
Collapse
|
35
|
Ludwig BS, Tomassi S, Di Maro S, Di Leva FS, Benge A, Reichart F, Nieberler M, Kühn FE, Kessler H, Marinelli L, Reuning U, Kossatz S. The organometallic ferrocene exhibits amplified anti-tumor activity by targeted delivery via highly selective ligands to αvβ3, αvβ6, or α5β1 integrins. Biomaterials 2021; 271:120754. [PMID: 33756215 DOI: 10.1016/j.biomaterials.2021.120754] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/17/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022]
Abstract
High levels of reactive oxygen species (ROS) in tumors have been shown to exert anti-tumor activity, leading to the concept of ROS induction as therapeutic strategy. The organometallic compound ferrocene (Fc) generates ROS through a reversible one-electron oxidation. Incorporation of Fc into a tumor-targeting, bioactive molecule can enhance its therapeutic activity and enable tumor specific delivery. Therefore, we conjugated Fc to five synthetic, Arg-Gly-Asp (RGD)-based integrin binding ligands to enable targeting of the cell adhesion and signaling receptor integrin subtypes αvβ3, α5β1, or αvβ6, which are overexpressed in various, distinct tumors. We designed and synthesized a library of integrin-ligand-ferrocene (ILF) derivatives and showed that ILF conjugates maintained the high integrin affinity and selectivity of their parent ligands. A thorough biological characterization allowed us to identify the two most promising ligands, an αvβ3 (L2b) and an αvβ6 (L3b) targeting ILF, which displayed selective integrin-dependent cell uptake and pronounced ferrocene-mediated anti-tumor effects in vitro, along with increased ROS production and DNA damage. Hence, ILFs are promising candidates for the selective, tumor-targeted delivery of ferrocene to maximize its anti-cancer efficacy and minimize systemic toxicity, thereby improving the therapeutic window of ferrocene compared to currently used non-selective anti-cancer drugs.
Collapse
Affiliation(s)
- Beatrice Stefanie Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum rechts der Isar, Technical University Munich, Munich, Germany; Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Stefano Tomassi
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Salvatore Di Maro
- Università degli Studi della Campania "Luigi Vanvitelli", Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Caserta, Italy
| | | | - Anke Benge
- Department of Obstetrics and Gynecology, Clinical Research Unit, University Hospital Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Florian Reichart
- Institute for Advanced Study, Department of Chemistry, Technical University Munich, Garching, Germany
| | - Markus Nieberler
- Department of Oral and Maxillofacial Surgery, University Hospital Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Fritz E Kühn
- Molecular Catalysis, Catalysis Research Center, Technical University Munich, Munich, Germany; Department of Chemistry, Technical University Munich, Munich, Germany
| | - Horst Kessler
- Institute for Advanced Study, Department of Chemistry, Technical University Munich, Garching, Germany
| | - Luciana Marinelli
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Ute Reuning
- Department of Obstetrics and Gynecology, Clinical Research Unit, University Hospital Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum rechts der Isar, Technical University Munich, Munich, Germany; Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany; Department of Chemistry, Technical University Munich, Munich, Germany.
| |
Collapse
|
36
|
Bowen J, Schneible J, Bacon K, Labar C, Menegatti S, Rao BM. Screening of Yeast Display Libraries of Enzymatically Treated Peptides to Discover Macrocyclic Peptide Ligands. Int J Mol Sci 2021; 22:ijms22041634. [PMID: 33562883 PMCID: PMC7915732 DOI: 10.3390/ijms22041634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/12/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
We present the construction and screening of yeast display libraries of post-translationally modified peptides wherein site-selective enzymatic treatment of linear peptides is achieved using bacterial transglutaminase. To this end, we developed two alternative routes, namely (i) yeast display of linear peptides followed by treatment with recombinant transglutaminase in solution; or (ii) intracellular co-expression of linear peptides and transglutaminase to achieve peptide modification in the endoplasmic reticulum prior to yeast surface display. The efficiency of peptide modification was evaluated via orthogonal detection of epitope tags integrated in the yeast-displayed peptides by flow cytometry, and via comparative cleavage of putative cyclic vs. linear peptides by tobacco etch virus (TEV) protease. Subsequently, yeast display libraries of transglutaminase-treated peptides were screened to isolate binders to the N-terminal region of the Yes-Associated Protein (YAP) and its WW domains using magnetic selection and fluorescence activated cell sorting (FACS). The identified peptide cyclo[E-LYLAYPAH-K] featured a KD of 1.75 μM for YAP and 0.68 μM for the WW domains of YAP as well as high binding selectivity against albumin and lysozyme. These results demonstrate the usefulness of enzyme-mediated cyclization in screening combinatorial libraries to identify cyclic peptide binders.
Collapse
Affiliation(s)
- John Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27695, USA; (J.B.); (J.S.); (K.B.)
| | - John Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27695, USA; (J.B.); (J.S.); (K.B.)
| | - Kaitlyn Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27695, USA; (J.B.); (J.S.); (K.B.)
| | - Collin Labar
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC 27695, USA;
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27695, USA; (J.B.); (J.S.); (K.B.)
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
- Correspondence: (S.M.); (B.M.R.)
| | - Balaji M. Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way, Raleigh, NC 27695, USA; (J.B.); (J.S.); (K.B.)
- Biomanufacturing Training and Education Center (BTEC), North Carolina State University, 850 Oval Dr, Raleigh, NC 27606, USA
- Correspondence: (S.M.); (B.M.R.)
| |
Collapse
|
37
|
Kye M, Zhang Z, Lim Y. Self‐assembling cyclic peptide‐oligonucleotide conjugates: Synthetic strategies and the effect of cyclic topology on self‐assembly and base pairing. Pept Sci (Hoboken) 2020. [DOI: 10.1002/pep2.24193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Mahnseok Kye
- Department of Materials Science & Engineering Yonsei University Seoul South Korea
| | - Zhihao Zhang
- Department of Materials Science & Engineering Yonsei University Seoul South Korea
| | - Yong‐beom Lim
- Department of Materials Science & Engineering Yonsei University Seoul South Korea
| |
Collapse
|
38
|
Sefkow-Werner J, Machillot P, Sales A, Castro-Ramirez E, Degardin M, Boturyn D, Cavalcanti-Adam EA, Albiges-Rizo C, Picart C, Migliorini E. Heparan sulfate co-immobilized with cRGD ligands and BMP2 on biomimetic platforms promotes BMP2-mediated osteogenic differentiation. Acta Biomater 2020; 114:90-103. [PMID: 32673751 DOI: 10.1016/j.actbio.2020.07.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/27/2022]
Abstract
The chemical and physical properties of the extracellular matrix (ECM) are known to be fundamental for regulating growth factor bioactivity. The role of heparan sulfate (HS), a glycosaminoglycan, and of cell adhesion proteins (containing the cyclic RGD (cRGD) ligands) on bone morphogenetic protein 2 (BMP2)-mediated osteogenic differentiation has not been fully explored. In particular, it is not known whether and how their effects can be potentiated when they are presented in controlled close proximity, as in the ECM. Here, we developed streptavidin platforms to mimic selective aspects of the in vivo presentation of cRGD, HS and BMP2, with a nanoscale-control of their surface density and orientation to study cell adhesion and osteogenic differentiation. We showed that whereas a controlled increase in cRGD surface concentration upregulated BMP2 signaling due to β3 integrin recruitment, silencing either β1 or β3 integrins negatively affected BMP2-mediated phosphorylation of SMAD1/5/9 and alkaline phosphatase expression. Furthermore, the presence of adsorbed BMP2 promoted cellular adhesion at very low cRGD concentrations. Finally, we proved that HS co-immobilized with cRGD both sustained BMP2 signaling and enhanced osteogenic differentiation compared to BMP2 directly immobilized on streptavidin, even with a low cRGD surface concentration. Altogether, our results show that HS facilitated and sustained the synergy between BMP2 and integrin pathways and that the co-immobilization of HS and cRGD peptides optimised BMP2-mediated osteogenic differentiation. Statement of significance The growth factor BMP2 is used to treat large bone defects. Previous studies have shown that the presentation of BMP2 via extracellular matrix molecules, such as heparan sulfate (HS), can upregulate BMP2 signaling. The potential advantages of dose reduction and local specificity have stimulated interest in further investigations into biomimetic approaches. We designed a streptavidin model surface eligible for immobilizing tunable amounts of molecules from the extracellular space, such as HS, adhesion motifs (cyclic RGD) and BMP2. By studying cellular adhesion, BMP2 bioactivity and its osteogenic potential we reveal the combined effect of integrins, HS and BMP2, which contribute in answering fundamental questions regarding cell-matrix interaction.
Collapse
|
39
|
Cyclic RGD-Functionalized closo-Dodecaborate Albumin Conjugates as Integrin Targeting Boron Carriers for Neutron Capture Therapy. Mol Pharm 2020; 17:3740-3747. [PMID: 32845640 DOI: 10.1021/acs.molpharmaceut.0c00478] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cyclic RGD (cRGD) peptide-conjugated boronated albumin was developed to direct toward integrin αvβ3, which overexpresses on many cancer cells. A stepwise conjugation of c[RGDfK(Mal)] and maleimide-conjugated closo-dodecaborate (MID) to bovine serum albumin (BSA) afforded cRGD-MID-BSA, which was noncytotoxic toward both U87MG and A549 cells. As compared with l-BPA, selective antitumor activity of cRGD-MID-BSA toward U87MG cells overexpressing integrin αvβ3 was identified after thermal neutron irradiation. In vivo fluorescence live imaging of Cy5-conjugated cRGD-MID-BSA and MID-BSA revealed that both cRGD-MID-BSA and MID-BSA similarly reached the maximum accumulation during 8-12 h after injection. The selective accumulation and retention of Cy5-cRGD-MID-BSA was more pronounced than Cy5-MID-BSA after 24 h. An in vivo boron neutron capture therapy (BNCT) study revealed that the cRGD peptide ligand combination enhanced accumulation of MID-BSA into tumor cells in U87MG xenograft models. The significant tumor growth suppression was observed in U87MG xenograft models at a dose of 7.5 mg [10B]/kg after neutron irradiation.
Collapse
|
40
|
Goyal R, Jerath G, Chandrasekharan A, Kumar TRS, Ramakrishnan V. Peptide-based delivery vectors with pre-defined geometrical locks. RSC Med Chem 2020; 11:1303-1313. [PMID: 34095842 DOI: 10.1039/d0md00229a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Design of peptide-based targeted delivery vectors with attributes of specificity and selective cellular targeting by fixing their topology and resulting electrostatic fingerprint is the objective of this study. We formulated our peptide design platform by utilizing the possibilities of side-chain induced geometric restrictions in a typical peptide molecule. Conceptually, we locked the conformation of the RGD/NGR motif of tumor homing peptides (THPs) by mutating glycine in these motifs with d-proline and tailed the peptides with a syndiotactic amphipathic segment for cellular penetration. The designed peptides were synthesized, characterized, and tested in vitro on various cell lines, including breast cancer (MDA-MB-231), cervical cancer (HeLa), osteosarcoma (U2-OS) and non-cancer mammary epithelial cells (MCF-10A), by flow cytometry and confocal microscopy. The results showed differential cellular uptake in different cell types, as a result of the distinct electrostatic fingerprint encoded in their design. The uptake of serum pre-treated peptides by cells reveals the retention of peptide activity even after the incubation with serum. In addition, peptide-methotrexate (MTX) conjugates compared to the methotrexate drug showed enhanced apoptotic cell death in MTX-resistant MDA-MB-231 cells, indicating the increase in MTX bioavailability.
Collapse
Affiliation(s)
- Ruchika Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Gaurav Jerath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| | - Aneesh Chandrasekharan
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram-695014 Kerala India
| | - T R Santhosh Kumar
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram-695014 Kerala India
| | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati Guwahati-781039 Assam India
| |
Collapse
|
41
|
Jwad R, Weissberger D, Hunter L. Strategies for Fine-Tuning the Conformations of Cyclic Peptides. Chem Rev 2020; 120:9743-9789. [PMID: 32786420 DOI: 10.1021/acs.chemrev.0c00013] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cyclic peptides are promising scaffolds for drug development, attributable in part to their increased conformational order compared to linear peptides. However, when optimizing the target-binding or pharmacokinetic properties of cyclic peptides, it is frequently necessary to "fine-tune" their conformations, e.g., by imposing greater rigidity, by subtly altering certain side chain vectors, or by adjusting the global shape of the macrocycle. This review systematically examines the various types of structural modifications that can be made to cyclic peptides in order to achieve such conformational control.
Collapse
Affiliation(s)
- Rasha Jwad
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Daniel Weissberger
- School of Chemistry, University of New South Wales (UNSW) Sydney, New South Wales 2052, Australia
| | - Luke Hunter
- School of Chemistry, University of New South Wales (UNSW) Sydney, New South Wales 2052, Australia
| |
Collapse
|
42
|
Karimi F, Thombare VJ, Hutton CA, O'Connor AJ, Qiao GG, Heath DE. Biomaterials functionalized with nanoclusters of integrin- and syndecan-binding ligands improve cell adhesion and mechanosensing under shear flow conditions. J Biomed Mater Res A 2020; 109:313-325. [PMID: 32490581 DOI: 10.1002/jbm.a.37024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022]
Abstract
We have engineered biomaterials that display nanoclusters of ligands that bind both integrin and syndecan-4 cell receptors. These surfaces regulate cell behaviors under static conditions including adhesion, spreading, actin stress fiber formation, and migration. The syndecan-4 receptors are also critical mediators of cellular mechanotransduction. In this contribution we assess whether this novel class of materials can regulate the response of cells to applied mechanical stimulation, using the shear stress imparted by laminar fluid flow as a model stimulus. Specifically, we assess endothelial cell detachment due to flow, cell alignment due to flow, and cell adhesion from the flowing fluid. A high degree of cell retention was observed on surfaces containing integrin-binding ligands or a mixed population of integrin- and syndecan-binding ligands. However, the presence of both ligand types was necessary for the cells to align in the direction of flow. These results imply that integrin engagement is necessary for adhesion strength, but engagement of both receptor types aids in appropriate mechanotransduction. Additionally, it was found that surfaces functionalized with both ligand types were able to scavenge a larger number of cells from flow, and to do so at a faster rate, compared to surfaces functionalized with only integrin- or syndecan-binding ligands. These results show that interfaces functionalized with both integrin- and syndecan-binding ligands regulate a significant range of biophysical cell behaviors in response to shear stress.
Collapse
Affiliation(s)
- Fatemeh Karimi
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia.,Polymer Science Group, Department of Chemical Engineering, Particulate Fluid Processing Centre, University of Melbourne, Parkville, Victoria, Australia.,Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
| | - Varsha Jagannath Thombare
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Craig A Hutton
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Victoria, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Greg G Qiao
- Polymer Science Group, Department of Chemical Engineering, Particulate Fluid Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel E Heath
- Department of Biomedical Engineering, Particulate Fluids Processing Centre, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
43
|
Kawakami S, Suga T. [Development of Nano-DDS Carriers for Control of Spatial Distribution Using Multi-color Deep Imaging]. YAKUGAKU ZASSHI 2020; 140:633-640. [PMID: 32378663 DOI: 10.1248/yakushi.19-00218-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Because active-targeted liposomes are very complex formulations, quality characteristics of functional lipids have not been defined yet, and this is a major obstacle in clinical application of active targeted liposomes. We have developed high functionality and quality (HFQ) lipids, which define quality characteristics of functional lipids for clinical drug delivery system (DDS) applications. Because HFQ lipids are designed to enable facile and rapid functionalization of DDS carrier by simple and one-step mixing, we are expanding applications for not only liposomes but also exosomes and cells. Recently, we developed multi-color deep imaging by tissue clearing for analysis of spatial distribution of DDS in various tissues. Nanocarriers are usually non-uniformly distributed in solid tumors because of their heterogeneity. Especially, in refractory cancer such as pancreatic cancer, the presence of collagen and blood vessels greatly affects intra-tumor distribution of DDS carrier. Therefore information on spatial relations between the tissue structure and DDS carrier is important to regulate precisely intra-tumor distribution of DDS carrier. Recently, our group has established multi-color deep imaging to analyze spatial distribution of stromal collagen, liposomes, and blood vessels in pancreatic tumor tissue. In this review, we present recent research in developing HFQ lipids. Moreover, current status of research on DDS for pancreatic cancer treatment is reviewed.
Collapse
Affiliation(s)
- Shigeru Kawakami
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| | - Tadaharu Suga
- Department of Pharmaceutical Informatics, Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
44
|
Yang J, Yang J, Wang H, Wang J, Xiong J, Qiao C, Ran C. An atom-economical design of PET tracer for imaging α vβ 3 integrin via utilizing the three-in-one function of 64Copper. Chem Commun (Camb) 2020; 56:1788-1791. [PMID: 31960841 DOI: 10.1039/c9cc08690k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, αvβ3 integrin in U87 tumor cells was imaged with a 64Cu-peptidic probe, in which the linear peptide GHRGDHG is used as a pre-ligand, while 64Cu bears three functional roles that include generation of the PET signal, coordination with two GH moieties of the pre-ligand, and cyclizing the linear pre-ligand into an active cyclic-RGD form (termed as 64Cu-Cyclo-RGD) for αvβ3 integrin.
Collapse
Affiliation(s)
- Jing Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China. and College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, China
| | - Jian Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China. and School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Huan Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China.
| | - Junfeng Wang
- Gorden Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China
| | - Jianping Xiong
- Program in Structural Biology, Nephrology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China
| | - Chunhua Qiao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215006, China
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, China.
| |
Collapse
|
45
|
Wang Q, Onuma K, Liu C, Wong H, Bloom MS, Elliott EE, Cao RR, Hu N, Lingampalli N, Sharpe O, Zhao X, Sohn DH, Lepus CM, Sokolove J, Mao R, Cisar CT, Raghu H, Chu CR, Giori NJ, Willingham SB, Prohaska SS, Cheng Z, Weissman IL, Robinson WH. Dysregulated integrin αVβ3 and CD47 signaling promotes joint inflammation, cartilage breakdown, and progression of osteoarthritis. JCI Insight 2019; 4:128616. [PMID: 31534047 PMCID: PMC6795293 DOI: 10.1172/jci.insight.128616] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the leading cause of joint failure, yet the underlying mechanisms remain elusive, and no approved therapies that slow progression exist. Dysregulated integrin function was previously implicated in OA pathogenesis. However, the roles of integrin αVβ3 and the integrin-associated receptor CD47 in OA remain largely unknown. Here, transcriptomic and proteomic analyses of human and murine osteoarthritic tissues revealed dysregulated expression of αVβ3, CD47, and their ligands. Using genetically deficient mice and pharmacologic inhibitors, we showed that αVβ3, CD47, and the downstream signaling molecules Fyn and FAK are crucial to OA pathogenesis. MicroPET/CT imaging of a mouse model showed elevated ligand-binding capacities of integrin αVβ3 and CD47 in osteoarthritic joints. Further, our in vitro studies demonstrated that chondrocyte breakdown products, derived from articular cartilage of individuals with OA, induced αVβ3/CD47-dependent expression of inflammatory and degradative mediators, and revealed the downstream signaling network. Our findings identify a central role for dysregulated αVβ3 and CD47 signaling in OA pathogenesis and suggest that activation of αVβ3 and CD47 signaling in many articular cell types contributes to inflammation and joint destruction in OA. Thus, the data presented here provide a rationale for targeting αVβ3, CD47, and their signaling pathways as a disease-modifying therapy.
Collapse
Affiliation(s)
- Qian Wang
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Kazuhiro Onuma
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Changhao Liu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Eileen E. Elliott
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Richard R.L. Cao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nick Hu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Xiaoyan Zhao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Dong Hyun Sohn
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do, South Korea
| | - Christin M. Lepus
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Jeremy Sokolove
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Rong Mao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Cecilia T. Cisar
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Harini Raghu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Constance R. Chu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Nicholas J. Giori
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Stephen B. Willingham
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Susan S. Prohaska
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
46
|
Park SE, Sajid MI, Parang K, Tiwari RK. Cyclic Cell-Penetrating Peptides as Efficient Intracellular Drug Delivery Tools. Mol Pharm 2019; 16:3727-3743. [PMID: 31329448 DOI: 10.1021/acs.molpharmaceut.9b00633] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cyclic cell-penetrating peptides are relatively a newer class of peptides that have a huge potential for the intracellular delivery of therapeutic agents aimed at treating challenging ailments like multidrug-resistant bacterial diseases, cancer, and HIV infection. Cell-penetrating peptides (CPPs) have been extensively explored as intracellular delivery vehicles; however, they have some inherent limitations like poor stability, endosomal entrapment, toxicity, and suboptimal cell penetration. Owing to their favorable properties that avoid these limitations, cyclic CPPs can provide a good alternative to linear CPPs. Several Reviews have been published in the past decade that cover CPPs and cyclic peptides independently. To the best of our knowledge, this is one of the first Reviews that covers cyclic CPPs comprehensively in the light of studies published so far. In this Review, we have detailed examples of cyclic CPPs, their structures, and cyclization strategies followed by a detailed account of their advantages over their linear counterparts. A hot area in cyclic CPPs is the exploration of cell-penetration mechanisms; this Review highlights this topic in detail. Finally, we will review the applications of cyclic CPPs, followed by conclusions and future prospects.
Collapse
Affiliation(s)
- Shang Eun Park
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences , Chapman University School of Pharmacy , Harry and Diane Rinker Health Science Campus, Irvine , California 92618 , United States
| | - Muhammad Imran Sajid
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences , Chapman University School of Pharmacy , Harry and Diane Rinker Health Science Campus, Irvine , California 92618 , United States.,Faculty of Pharmacy , University of Central Punjab , Lahore 54000 , Pakistan
| | - Keykavous Parang
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences , Chapman University School of Pharmacy , Harry and Diane Rinker Health Science Campus, Irvine , California 92618 , United States
| | - Rakesh Kumar Tiwari
- Center for Targeted Drug Delivery, Department of Biomedical and Pharmaceutical Sciences , Chapman University School of Pharmacy , Harry and Diane Rinker Health Science Campus, Irvine , California 92618 , United States
| |
Collapse
|
47
|
Siitonen R, Peuhu E, Autio A, Liljenbäck H, Mattila E, Metsälä O, Käkelä M, Saanijoki T, Dijkgraaf I, Jalkanen S, Ivaska J, Roivainen A. 68Ga-DOTA-E[c(RGDfK)] 2 PET Imaging of SHARPIN-Regulated Integrin Activity in Mice. J Nucl Med 2019; 60:1380-1387. [PMID: 30850498 DOI: 10.2967/jnumed.118.222026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/04/2019] [Indexed: 12/26/2022] Open
Abstract
Shank-associated RH domain-interacting protein (SHARPIN) is a cytosolic protein that plays a key role in activation of nuclear factor κ-light-chain enhancer of activated B cells and regulation of inflammation. Furthermore, SHARPIN controls integrin-dependent cell adhesion and migration in several normal and malignant cell types, and loss of SHARPIN correlates with increased integrin activity in mice. Arginyl-glycyl-aspartic acid (RGD), a cell adhesion tripeptide motif, is an integrin recognition sequence that facilitates PET imaging of integrin upregulation during tumor angiogenesis. We hypothesized that increased integrin activity due to loss of SHARPIN protein would affect the uptake of αvβ3-selective cyclic, dimeric peptide 68Ga-DOTA-E[c(RGDfK)]2, where E[c(RGDfk)]2 = glutamic acid-[cyclo(arginyl-glycyl-aspartic acid-D-phenylalanine-lysine)], both in several tissue types and in the tumor microenvironment. To test this hypothesis, we used RGD-based in vivo PET imaging to evaluate wild-type (wt) and SHARPIN-deficient mice (Sharpin cpdm , where cpdm = chronic proliferative dermatitis in mice) with and without melanoma tumor allografts. Methods: Sharpin cpdm mice with spontaneous null mutation in the Sharpin gene and their wt littermates with or without B16-F10-luc melanoma tumors were studied by in vivo imaging and ex vivo measurements with cyclic-RGD peptide 68Ga-DOTA-E[c(RGDfK)]2 After the last 68Ga-DOTA-E[c(RGDfK)]2 peptide PET/CT, tumors were cut into cryosections for autoradiography, histology, and immunohistochemistry. Results: The ex vivo uptake of 68Ga-DOTA-E[c(RGDfK)]2 in the mouse skin and tumor was significantly higher in Sharpin cpdm mice than in wt mice. B16-F10-luc tumors were detected 4 d after inoculation, without differences in volume or blood flow between the mouse strains. PET imaging with 68Ga-DOTA-E[c(RGDfK)]2 peptide at day 10 after inoculation revealed significantly higher uptake in the tumors transplanted into Sharpin cpdm mice than in wt mice. Furthermore, tumor vascularization was increased in the Sharpin cpdm mice. Conclusion: Sharpin cpdm mice demonstrated increased integrin activity and vascularization in B16-F10-luc melanoma tumors, as demonstrated by RGD-based in vivo PET imaging. These data indicate that SHARPIN, a protein previously associated with increased cancer growth and metastasis, may also have important regulatory roles in controlling the tumor microenvironment.
Collapse
Affiliation(s)
| | - Emilia Peuhu
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Anu Autio
- Turku PET Centre, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Elina Mattila
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Ingrid Dijkgraaf
- Department of Biochemistry, University of Maastricht, Maastricht, the Netherlands
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Biochemistry, University of Turku, Turku, Finland; and
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland .,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| |
Collapse
|
48
|
Cesar PHS, Braga MA, Trento MVC, Menaldo DL, Marcussi S. Snake Venom Disintegrins: An Overview of their Interaction with Integrins. Curr Drug Targets 2019; 20:465-477. [DOI: 10.2174/1389450119666181022154737] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Disintegrins are non-enzymatic proteins that interfere on cell–cell interactions and signal transduction, contributing to the toxicity of snake venoms and play an essential role in envenomations. Most of their pharmacological and toxic effects are the result of the interaction of these molecules with cell surface ligands, which has been widely described and studied. These proteins may act on platelets, leading to hemorrhage, and may also induce apoptosis and cytotoxicity, which highlights a high pharmacological potential for the development of thrombolytic and antitumor agents. Additionally, these molecules interfere with the functions of integrins by altering various cellular processes such as migration, adhesion and proliferation. This review gathers information on functional characteristics of disintegrins isolated from snake venoms, emphasizing a comprehensive view of the possibility of direct use of these molecules in the development of new drugs, or even indirectly as structural models.
Collapse
Affiliation(s)
- Pedro Henrique Souza Cesar
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Mariana Aparecida Braga
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Marcus Vinicius Cardoso Trento
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Danilo Luccas Menaldo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo (FCFRP-USP), Ribeirão Preto-SP, Brazil
| | - Silvana Marcussi
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| |
Collapse
|
49
|
Bicho D, Ajami S, Liu C, Reis RL, Oliveira JM. Peptide-biofunctionalization of biomaterials for osteochondral tissue regeneration in early stage osteoarthritis: challenges and opportunities. J Mater Chem B 2019; 7:1027-1044. [DOI: 10.1039/c8tb03173h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis is a degenerative joint disease characterized by the progressive deterioration of articular cartilage, synovial inflammation and changes in periarticular and subchondral bone, being a leading cause of disability.
Collapse
Affiliation(s)
- D. Bicho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - S. Ajami
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - C. Liu
- Institute of Orthopaedics and Musculo-Skeletal Sci, University College London, Royal National Orthopaedic Hospital
- Stanmore
- UK
| | - R. L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| | - J. M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra
- Guimarães
- Portugal
- ICVS/3B's – PT Government Associate Laboratory
- Braga/Guimarães
| |
Collapse
|
50
|
Abstract
The functionalization of nanoparticles with specific receptor ligands enables their accumulation in targeted tissues and can be used therapeutically to transport drugs or for diagnostic purposes (Parveen et al., Nanomedicine 8:147-166, 2012). We could recently show that targeting endothelial cells in retinal and choroidal capillaries can be realized even under physiological conditions using quantum dots as model nanoparticles functionalized with an integrin-binding peptide (Pollinger et al., Proc Natl Acad Sci 110:6115-6120, 2013). Even though the chemistry that we used was well described in the literature and may be considered standard for the purpose, there are a number of preparation steps that are delicate and deserve special attention. It is, therefore, our intention to describe step by step the critical methods of ligand immobilization on quantum dot surfaces to facilitate the reader to reproduce our work. Here we describe the chemical modification of quantum dots with c(RGDfC) as targeting peptide that allows the resulting modified nanoparticles to adhere to endothelial cells also in the retinal tissue. We illustrate the properties of the resulting particles by showing some of the in vitro results from our previous studies. Doing so, we concomitantly encourage the reader to check particles intended for targeting cells in vivo first by extensive in vitro analysis of particle interaction with cells by the means of flow cytometry and confocal microscopy to confirm the successful functionalization. Only then the application of functionalized quantum dots into the systemic circulation of mice led to the desired localization of nanoparticles in the retinal and choroidal blood vessels (Pollinger et al., Proc Natl Acad Sci 110:6115-6120, 2013).
Collapse
Affiliation(s)
- Alexandra Haunberger
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|