1
|
Kim SH, Yasunaga AB, Zhang H, Whitley KD, Li ITS. Quantitative Super-Resolution Imaging of Molecular Tension. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2408280. [PMID: 40245301 DOI: 10.1002/advs.202408280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 02/08/2025] [Indexed: 04/19/2025]
Abstract
DNA-based molecular tension probes have revolutionized the localization of mechanical events in live cells with super-resolution. However, imaging the magnitude of these forces at super-resolution has been challenging. Here, qtPAINT (quantitative tension points accumulation for imaging in nanoscale topography) is introduced as a strategy to image the magnitude of molecular tension with super-resolution accuracy. By leveraging the force-dependent dissociation kinetics of short DNA oligonucleotides on their complementary strands, tension is encoded on individual molecules through their binding kinetics. This method allowed for a quantitative analysis of these kinetics, providing a detailed reconstruction of the force magnitudes acting on each tension probe. The technique integrates a molecular-beacon PAINT imager with a hairpin molecular tension probe, achieving a force quantification range of 9-30 pN and maintaining a spatial resolution of 30-120 nm in low and high-density regions. Additionally, qtPAINT offers a temporal resolution on the order of a minute, enhancing its applicability for studying dynamic cellular processes.
Collapse
Affiliation(s)
- Seong Ho Kim
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
- Department of Chemistry and Advanced Materials, College of Natural Sciences, Gangneung-Wonju National University, Gangneung, 25457, Republic of Korea
| | - Adam B Yasunaga
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Hongyuan Zhang
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| | - Kevin D Whitley
- Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Isaac T S Li
- Department of Chemistry, University of British Columbia, Kelowna, BC, V1V 1V7, Canada
| |
Collapse
|
2
|
Chen Y, Fu T, Zou Z, Liu Y, Zhu J, Teng B, Yao K, Li H, Li J, Xie Z, He Y. Biological Reinforced Concrete for Cartilage Repair With 3D Printing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416734. [PMID: 39998315 PMCID: PMC12021066 DOI: 10.1002/advs.202416734] [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] [Received: 01/01/2025] [Revised: 02/05/2025] [Indexed: 02/26/2025]
Abstract
The development of biomimetic cartilage constructs (BCCs) with natural extracellular matrix (ECM) microenvironments and topological cues to accelerate the reconstruction of natural articular cartilage (NAC) after injury is challenging due to its complex structure, low cellular content, and less vascularity. Inspired by concrete rebar structure, a biomimetic cartilage named "biological reinforced concrete" is fabricated, with collagen fiber orientation transitioning from parallel to perpendicular, replicating the ECM microenvironments and complex construct of NAC. 3D-printed ultrafine fiber networks (UFNs) served as a degradable "biorebars", while a hybrid biohydrogel acted as "biocement". The stem cells are utilized as "bioactive aggregates". The biocement is developed by combining and screening various biohydrogels to mimic an ECM microenvironment conducive to the formation of NAC. By adjusting the fiber scale and spacing of the UFNs, the mechanical properties of the biomimetic cartilages are controlled to resemble those of NAC. Additionally, the UFNs guided the directed growth of cells and the orderly secretion of ECM. Subsequently, the developed BCCs are implanted into an osteochondral defect, and after 4 months, they successfully reconstructed the complex structure of cartilage with mechanical properties closely resembling those of NAC. The biological reinforced concrete offers a customizable and universal strategy for tissue regeneration.
Collapse
Affiliation(s)
- Yuewei Chen
- School of Mechanical EngineeringGuizhou UniversityGuiyang550025China
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu LaboratorySchool of Mechanical EngineeringZhejiang UniversityHangzhou310027China
| | - Tao Fu
- Department of Oral and Maxillofacial SurgeryThe Second Affiliated Hospital of Zhejiang University School of Medicine, School of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceHangzhouZhejiang310000China
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000China
| | - Zhongfei Zou
- School of Mechanical EngineeringGuizhou Institute of TechnologyGuiyang550003China
| | - Yanming Liu
- Department of Oral and Maxillofacial SurgeryThe Second Affiliated Hospital of Zhejiang University School of Medicine, School of Stomatology and Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceHangzhouZhejiang310000China
| | - Jianguo Zhu
- Department of UrologyGuizhou Provincial People's HospitalThe Affiliated Hospital of Guizhou UniversityGuiyangGuizhou550002China
| | - Binhong Teng
- Department of OrthodonticsThe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiang310000China
| | - Ke Yao
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu LaboratorySchool of Mechanical EngineeringZhejiang UniversityHangzhou310027China
| | - Haibin Li
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu LaboratorySchool of Mechanical EngineeringZhejiang UniversityHangzhou310027China
| | - Jiachun Li
- School of Mechanical EngineeringGuizhou UniversityGuiyang550025China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang ProvinceCancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang ProvinceHangzhou310000China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems & Liangzhu LaboratorySchool of Mechanical EngineeringZhejiang UniversityHangzhou310027China
| |
Collapse
|
3
|
Pandey V, Kundu S, Pyne A, Wang X. Live-cell imaging of single integrin tensions with minimal background fluorescence noise. Biophys J 2025; 124:1085-1094. [PMID: 39935179 PMCID: PMC11993927 DOI: 10.1016/j.bpj.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/11/2024] [Accepted: 02/04/2025] [Indexed: 02/13/2025] Open
Abstract
One powerful method for studying cell mechanobiology is to monitor receptor-mediated forces at the single-molecule level in live cells. Hairpin DNA labeled with a quencher-dye pair has been used as a tension probe (TP) to image cellular forces in real time. The TP emits fluorescence when cellular forces unfold the DNA hairpin and de-quench the dye, thereby converting the force signal into fluorescence. However, when applied to monitor cellular forces at the single-molecule level, the TP often suffers from background fluorescent spots (BFSs) due to nonquenched dyes, which interfere with molecular force imaging and analysis. In this work, we identified that the BFSs are primarily caused by missing quenchers in some TP constructs and surface-adsorbed dye-labeled DNA strands. To address these issues, we developed a double-quencher TP (dqTP) and incorporated Tween-20 treatment during surface preparation. These two simple strategies reduced the BFS level by 10-fold, significantly improving the signal/background ratio for single molecular force imaging. We demonstrated the performance of dqTP by monitoring the temporal dynamics of integrin tensions in platelets and HeLa cells, showing that single integrin tensions remain stable for at least 100 s in wild-type HeLa cells. In contrast, with vinculin knocked out, a subpopulation of integrin tensions, especially at cell peripheral regions, exhibited molecular force fluctuations with an average force duration shorter than 10 s. Overall, this work provides a convenient and practical approach to significantly reduce BFS levels on TP surfaces, offering a nearly false-signal-free platform for monitoring single-molecule forces in live cells.
Collapse
Affiliation(s)
- Vivek Pandey
- Research Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Subhankar Kundu
- Research Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Arghajit Pyne
- Research Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio
| | - Xuefeng Wang
- Research Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
4
|
Shahrokhtash A, Sivertsen MVT, Laursen SH, Sutherland DS. Nanoscale Cellular Traction Force Quantification: CRISPR-Cas12a Supercharged DNA Tension Sensors in Nanoclustered Ligand Patterns. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7339-7352. [PMID: 39868861 PMCID: PMC11803557 DOI: 10.1021/acsami.4c18358] [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] [Received: 10/23/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
High-throughput measurement of cellular traction forces at the nanoscale remains a significant challenge in mechanobiology, limiting our understanding of how cells interact with their microenvironment. Here, we present a novel technique for fabricating protein nanopatterns in standard multiwell microplate formats (96/384-wells), enabling the high-throughput quantification of cellular forces using DNA tension gauge tethers (TGTs) amplified by CRISPR-Cas12a. Our method employs sparse colloidal lithography to create nanopatterned surfaces with feature sizes ranging from sub 100 to 800 nm on transparent, planar, and fully PEGylated substrates. These surfaces allow for the orthogonal immobilization of two different proteins or biomolecules using click-chemistry, providing precise spatial control over cellular signaling cues. We demonstrate the robustness and versatility of this platform through imaging techniques, including total internal reflection fluorescence microscopy, confocal laser scanning microscopy, and high-throughput imaging. Applying this technology, we measured the early stage mechanical forces exerted by 3T3 fibroblasts across different nanoscale features, detecting forces ranging from 12 to 56 pN. By integrating the Mechano-Cas12a Assisted Tension Sensor (MCATS) system, we achieved rapid and high-throughput quantification of cellular traction forces, analyzing over 2 million cells within minutes. Our findings reveal that nanoscale clustering of integrin ligands significantly influences the mechanical responses of cells. This platform offers a powerful tool for mechanobiology research, facilitating the study of cellular forces and mechanotransduction pathways in a high-throughput manner compatible with standard cell culture systems.
Collapse
Affiliation(s)
- Ali Shahrokhtash
- Interdisciplinary
Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
- The
Centre for Cellular Signal Patterns (CellPAT), Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | | | - Sara Hvidbjerg Laursen
- Interdisciplinary
Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Duncan S. Sutherland
- Interdisciplinary
Nanoscience Center, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
- The
Centre for Cellular Signal Patterns (CellPAT), Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| |
Collapse
|
5
|
Wu J, Han B, Ai S, Wang A, Song Y, Jin M, Qu X, Wang X. Injectable double network hydrogel with adjustable stiffness for modulation of macrophage polarization. POLYMER TESTING 2025; 143:108685. [DOI: 10.1016/j.polymertesting.2024.108685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
6
|
Wijnakker JJ, van Son GJ, Krueger D, van de Wetering WJ, Lopez-Iglesias C, Schreurs R, van Rijt F, Lim S, Lin L, Peters PJ, Isberg RR, Janda CY, de Lau W, Clevers H. Integrin-activating Yersinia protein Invasin sustains long-term expansion of primary epithelial cells as 2D organoid sheets. Proc Natl Acad Sci U S A 2025; 122:e2420595121. [PMID: 39793062 PMCID: PMC11725944 DOI: 10.1073/pnas.2420595121] [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: 10/07/2024] [Accepted: 11/14/2024] [Indexed: 01/12/2025] Open
Abstract
Matrigel®/BME®, a basement membrane-like preparation, supports long-term growth of epithelial 3D organoids from adult stem cells [T. Sato et al., Nature 459, 262-265 (2009); T. Sato et al., Gastroenterology 141, 1762-1772 (2011)]. Here, we show that interaction between Matrigel's major component laminin-111 with epithelial α6β1-integrin is crucial for this process. The outer membrane protein Invasin of Yersinia is known to activate multiple integrin-β1 complexes, including integrin α6β1. A C-terminal integrin-binding fragment of Invasin, coated on culture plates, mediated gut epithelial cell adhesion. Addition of organoid growth factors allowed multipassage expansion in 2D. Polarization, junction formation, and generation of enterocytes, goblet cells, Paneth cells, and enteroendocrine cells were stable over time. Sustained expansion of other human, mouse, and even snake epithelia was accomplished under comparable conditions. The 2D "organoid sheet" format holds advantages over the 3D "in gel" format in terms of imaging, accessibility of basal and apical domains, and automation for high-throughput screening. Invasin represents a fully defined, affordable, versatile, and animal-free complement to Matrigel®/BME®.
Collapse
Affiliation(s)
- Joost J.A.P.M. Wijnakker
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Gijs J.F. van Son
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Daniel Krueger
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | | | - Carmen Lopez-Iglesias
- The Maastricht Multimodal Imaging Institute, Maastricht University, Maastricht6229 ER, The Netherlands
| | - Robin Schreurs
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Fenna van Rijt
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Sangho Lim
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Lin Lin
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Peter J. Peters
- The Maastricht Multimodal Imaging Institute, Maastricht University, Maastricht6229 ER, The Netherlands
| | - Ralph R. Isberg
- Department of Molecular Biology and Microbiology, School of Medicine, Tufts University, Boston, MA02111
| | - Claudia Y. Janda
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Wim de Lau
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| |
Collapse
|
7
|
Mei J, Jiang XY, Tian HX, Rong DC, Song JN, Wang L, Chen YS, Wong RCB, Guo CX, Wang LS, Wang LY, Wang PY, Yin JY. Anoikis in cell fate, physiopathology, and therapeutic interventions. MedComm (Beijing) 2024; 5:e718. [PMID: 39286778 PMCID: PMC11401975 DOI: 10.1002/mco2.718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/19/2024] Open
Abstract
The extracellular matrix (ECM) governs a wide spectrum of cellular fate processes, with a particular emphasis on anoikis, an integrin-dependent form of cell death. Currently, anoikis is defined as an intrinsic apoptosis. In contrast to traditional apoptosis and necroptosis, integrin correlates ECM signaling with intracellular signaling cascades, describing the full process of anoikis. However, anoikis is frequently overlooked in physiological and pathological processes as well as traditional in vitro research models. In this review, we summarized the role of anoikis in physiological and pathological processes, spanning embryonic development, organ development, tissue repair, inflammatory responses, cardiovascular diseases, tumor metastasis, and so on. Similarly, in the realm of stem cell research focused on the functional evolution of cells, anoikis offers a potential solution to various challenges, including in vitro cell culture models, stem cell therapy, cell transplantation, and engineering applications, which are largely based on the regulation of cell fate by anoikis. More importantly, the regulatory mechanisms of anoikis based on molecular processes and ECM signaling will provide new strategies for therapeutic interventions (drug therapy and cell-based therapy) in disease. In summary, this review provides a systematic elaboration of anoikis, thus shedding light on its future research.
Collapse
Affiliation(s)
- Jie Mei
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Xue-Yao Jiang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Hui-Xiang Tian
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Ding-Chao Rong
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
| | - Jia-Nan Song
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- School of Life Sciences Westlake University Hangzhou Zhejiang China
| | - Luozixian Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Yuan-Shen Chen
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Raymond C B Wong
- Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Melbourne Victoria Australia
- Ophthalmology Department of Surgery The University of Melbourne Melbourne Victoria Australia
| | - Cheng-Xian Guo
- Center of Clinical Pharmacology the Third Xiangya Hospital Central South University Changsha Hunan China
| | - Lian-Sheng Wang
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| | - Lei-Yun Wang
- Department of Pharmacy Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology Wuhan Hubei Province China
| | - Peng-Yuan Wang
- Oujiang Laboratory Key Laboratory of Alzheimer's Disease of Zhejiang Province Institute of Aging Wenzhou Medical University Wenzhou Zhejiang China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology Xiangya Hospital, Central South University Changsha Hunan China
- Institute of Clinical Pharmacology Hunan Key Laboratory of Pharmacogenetics Central South University Changsha Hunan China
- Engineering Research Center of Applied Technology of Pharmacogenomics Ministry of Education Changsha Hunan China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital, Central South University Changsha Hunan China
| |
Collapse
|
8
|
Combs JD, Foote AK, Ogasawara H, Velusamy A, Rashid SA, Mancuso JN, Salaita K. Measuring Integrin Force Loading Rates Using a Two-Step DNA Tension Sensor. J Am Chem Soc 2024; 146:23034-23043. [PMID: 39133202 PMCID: PMC11345772 DOI: 10.1021/jacs.4c03629] [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: 03/14/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Cells apply forces to extracellular matrix (ECM) ligands through transmembrane integrin receptors: an interaction which is intimately involved in cell motility, wound healing, cancer invasion and metastasis. These small (piconewton) integrin-ECM forces have been studied by molecular tension fluorescence microscopy (MTFM), which utilizes a force-induced conformational change of a probe to detect mechanical events. MTFM has revealed the force magnitude for integrin receptors in a variety of cell models including primary cells. However, force dynamics and specifically the force loading rate (LR) have important implications in receptor signaling and adhesion formation and remain poorly characterized. Here, we develop an LR probe composed of an engineered DNA structure that undergoes two mechanical transitions at distinct force thresholds: a low force threshold at 4.7 pN (hairpin unfolding) and a high force threshold at 47 pN (duplex shearing). These transitions yield distinct fluorescence signatures observed through single-molecule fluorescence microscopy in live cells. Automated analysis of tens of thousands of events from eight cells showed that the bond lifetime of integrins that engage their ligands and transmit a force >4.7 pN decays exponentially with a τ of 45.6 s. A subset of these events mature in magnitude to >47 pN with a median loading rate of 1.1 pN s-1 and primarily localize at the periphery of the cell-substrate junction. The LR probe design is modular and can be adapted to measure force ramp rates for a broad range of mechanoreceptors and cell models, thus aiding in the study of molecular mechanotransduction in living systems.
Collapse
Affiliation(s)
- J. Dale Combs
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Alexander K. Foote
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Hiroaki Ogasawara
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Arventh Velusamy
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Sk Aysha Rashid
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | | | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
9
|
Niraula G, Pyne A, Wang X. Develop Tandem Tension Sensor to Gauge Integrin-Transmitted Molecular Forces. ACS Sens 2024; 9:3660-3670. [PMID: 38968930 PMCID: PMC11287754 DOI: 10.1021/acssensors.4c00756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/11/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
DNA-based tension sensors have innovated the imaging and calibration of mechanosensitive receptor-transmitted molecular forces, such as integrin tensions. However, these sensors mainly serve as binary reporters, only indicating if molecular forces exceed one predefined threshold. Here, we have developed tandem tension sensor (TTS), which comprises two consecutive force-sensing units, each with unique force detection thresholds and distinct fluorescence spectra, thereby enabling the quantification of molecular forces with dual reference levels. With TTS, we revealed that vinculin is not required for transmitting integrin tensions at approximately 10 pN (piconewtons) but is essential for elevating integrin tensions beyond 20 pN in focal adhesions (FAs). Such high tensions have emerged during the early stage of FA formation. TTS also successfully detected changes in integrin tensions in response to disrupted actin formation, inhibited myosin activity, and tuned substrate elasticity. We also applied TTS to examine integrin tensions in platelets and revealed two force regimes, with integrin tensions surpassing 20 pN at cell central regions and 13-20 pN integrin tensions at the cell edge. Overall, TTS, especially the construct consisting of a hairpin DNA (13 pN opening force) and a shearing DNA (20 pN opening force), stands as a valuable tool for the quantification of receptor-transmitted molecular forces within living cells.
Collapse
Affiliation(s)
- Gopal Niraula
- Department
of Physics and Astronomy, Iowa State University, Ames, Iowa 50011, United States
| | - Arghajit Pyne
- Research
Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45219, United States
| | - Xuefeng Wang
- Research
Division in Hoxworth Center, College of Medicine, University of Cincinnati, Cincinnati, Ohio 45219, United States
| |
Collapse
|
10
|
Zhou C, Zhao Y, Yang M, Yin W, Li Y, Xiao Y, Liu Y, Lang M. Diselenide-Containing Polymer Based on New Antitumor Mechanism as Efficient GSH Depletion Agent for Ferroptosis Therapy. Adv Healthc Mater 2024; 13:e2303896. [PMID: 38551494 DOI: 10.1002/adhm.202303896] [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: 11/07/2023] [Revised: 02/24/2024] [Indexed: 04/07/2024]
Abstract
Glutathione (GSH) depletion-induced ferroptosis has emerged as a promising treatment for malignant cancer. It works by inactivating glutathione peroxidase 4 (GPX4) and facilitating lipid peroxidation. However, effectively delivering inducers and depleting intracellular GSH remains challenging due to the short half-lives and high hydrophobicity of small-molecule ferroptosis inducers. These inducers often require additional carriers. Herein, diselenide-containing polymers can consume GSH to induce ferroptosis for pancreatic cancer therapy. The diselenide bonds are controllably built into the backbone of the polycarbonate with a targeting peptide CRGD (Cys-Arg-Gly-Asp), which allows for self-assembly into stable nanoparticles (denoted CRNSe) for self-delivery. Significantly, at a concentration of 12 µg mL-1, CRNSe binds to the active site cysteine of GSH resulting in a thorough depletion of GSH. In contrast, the disulfide-containing analog only causes a slight decrease in GSH level. Moreover, the depletion of GSH inactivates GPX4, ultimately inducing ferroptosis due to the accumulation of lipid peroxide in BxPC-3 cells. Both in vitro and in vivo studies have demonstrated that CRNSe exhibits potent tumor suppressive ability with few side effects on normal tissue. This study validates the anti-tumor mechanism of diselenide-containing polymers in addition to apoptosis and also provides a new strategy for inherently inducing ferroptosis in cancer therapy.
Collapse
Affiliation(s)
- Chen Zhou
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuhao Zhao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, 200030, China
| | - Mao Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, 200030, China
| | - Wang Yin
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yongsheng Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, 200030, China
| | - Yan Xiao
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai, 200030, China
| | - Meidong Lang
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
11
|
Li J, Jo MH, Yan J, Hall T, Lee J, López-Sánchez U, Yan S, Ha T, Springer TA. Ligand binding initiates single-molecule integrin conformational activation. Cell 2024; 187:2990-3005.e17. [PMID: 38772370 PMCID: PMC11162317 DOI: 10.1016/j.cell.2024.04.049] [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: 10/27/2023] [Revised: 02/21/2024] [Accepted: 04/30/2024] [Indexed: 05/23/2024]
Abstract
Integrins link the extracellular environment to the actin cytoskeleton in cell migration and adhesiveness. Rapid coordination between events outside and inside the cell is essential. Single-molecule fluorescence dynamics show that ligand binding to the bent-closed integrin conformation, which predominates on cell surfaces, is followed within milliseconds by two concerted changes, leg extension and headpiece opening, to give the high-affinity integrin conformation. The extended-closed integrin conformation is not an intermediate but can be directly accessed from the extended-open conformation and provides a pathway for ligand dissociation. In contrast to ligand, talin, which links the integrin β-subunit cytoplasmic domain to the actin cytoskeleton, modestly stabilizes but does not induce extension or opening. Integrin activation is thus initiated by outside-in signaling and followed by inside-out signaling. Our results further imply that talin binding is insufficient for inside-out integrin activation and that tensile force transmission through the ligand-integrin-talin-actin cytoskeleton complex is required.
Collapse
Affiliation(s)
- Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Myung Hyun Jo
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Taylor Hall
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Joon Lee
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Uriel López-Sánchez
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Sophia Yan
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Newton South High School, Newton, MA 02459, USA
| | - Taekjip Ha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA.
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Jo MH, Meneses P, Yang O, Carcamo CC, Pangeni S, Ha T. Determination of single-molecule loading rate during mechanotransduction in cell adhesion. Science 2024; 383:1374-1379. [PMID: 38513010 PMCID: PMC10977658 DOI: 10.1126/science.adk6921] [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: 09/06/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024]
Abstract
Cells connect with their environment through surface receptors and use physical tension in receptor-ligand bonds for various cellular processes. Single-molecule techniques have revealed bond strength by measuring "rupture force," but it has long been recognized that rupture force is dependent on loading rate-how quickly force is ramped up. Thus, the physiological loading rate needs to be measured to reveal the mechanical strength of individual bonds in their functional context. We have developed an overstretching tension sensor (OTS) to allow more accurate force measurement in physiological conditions with single-molecule detection sensitivity even in mechanically active regions. We used serially connected OTSs to show that the integrin loading rate ranged from 0.5 to 4 piconewtons per second and was about three times higher in leukocytes than in epithelial cells.
Collapse
Affiliation(s)
- Myung Hyun Jo
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Paul Meneses
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Olivia Yang
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Claudia C. Carcamo
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sushil Pangeni
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Taekjip Ha
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
13
|
Combs JD, Foote AK, Ogasawara H, Velusamy A, Rashid SA, Mancuso JN, Salaita K. Measuring integrin force loading rates using a two-step DNA tension sensor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585042. [PMID: 38558970 PMCID: PMC10980004 DOI: 10.1101/2024.03.15.585042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Cells apply forces to extracellular matrix (ECM) ligands through transmembrane integrin receptors: an interaction which is intimately involved in cell motility, wound healing, cancer invasion and metastasis. These small (pN) forces exerted by cells have been studied by molecular tension fluorescence microscopy (MTFM), which utilizes a force-induced conformational change of a probe to detect mechanical events. MTFM has revealed the force magnitude for integrins receptors in a variety of cell models including primary cells. However, force dynamics and specifically the force loading rate (LR) have important implications in receptor signaling and adhesion formation and remain poorly characterized. Here, we develop a LR probe which is comprised of an engineered DNA structures that undergoes two mechanical transitions at distinct force thresholds: a low force threshold at 4.7 pN corresponding to hairpin unfolding and a high force threshold at 56 pN triggered through duplex shearing. These transitions yield distinct fluorescence signatures observed through single-molecule fluorescence microscopy in live-cells. Automated analysis of tens of thousands of events from 8 cells showed that the bond lifetime of integrins that engage their ligands and transmit a force >4.7 pN decays exponentially with a τ of 45.6 sec. A small subset of these events (<10%) mature in magnitude to >56pN with a median loading rate of 1.3 pNs-1 with these mechanical ramp events localizing at the periphery of the cell-substrate junction. Importantly, the LR probe design is modular and can be adapted to measure force ramp rates for a broad range of mechanoreceptors and cell models, thus aiding in the study of mechanotransduction.
Collapse
Affiliation(s)
- J. Dale Combs
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | | | | | - Arventh Velusamy
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Sk Aysha Rashid
- Department of Chemistry, 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, GA 30322, USA
| |
Collapse
|
14
|
He T, Giacomini D, Tolomelli A, Baiula M, Gentilucci L. Conjecturing about Small-Molecule Agonists and Antagonists of α4β1 Integrin: From Mechanistic Insight to Potential Therapeutic Applications. Biomedicines 2024; 12:316. [PMID: 38397918 PMCID: PMC10887150 DOI: 10.3390/biomedicines12020316] [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: 12/21/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Integrins are heterodimeric cell-surface receptors that regulate cell-cell adhesion and cellular functions through bidirectional signaling. On the other hand, anomalous trafficking of integrins is also implicated in severe pathologies as cancer, thrombosis, inflammation, allergies, and multiple sclerosis. For this reason, they are attractive candidates as drug targets. However, despite promising preclinical data, several anti-integrin drugs failed in late-stage clinical trials for chronic indications, with paradoxical side effects. One possible reason is that, at low concentration, ligands proposed as antagonists may also act as partial agonists. Hence, the comprehension of the specific structural features for ligands' agonism or antagonism is currently of the utmost interest. For α4β1 integrin, the situation is particularly obscure because neither the crystallographic nor the cryo-EM structures are known. In addition, very few potent and selective agonists are available for investigating the mechanism at the basis of the receptor activation. In this account, we discuss the physiological role of α4β1 integrin and the related pathologies, and review the few agonists. Finally, we speculate on plausible models to explain agonism vs. antagonism by comparison with RGD-binding integrins and by analysis of computational simulations performed with homology or hybrid receptor structures.
Collapse
Affiliation(s)
- Tingting He
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, Ue4, 40129 Bologna, Italy; (T.H.); (D.G.); (A.T.)
| | - Daria Giacomini
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, Ue4, 40129 Bologna, Italy; (T.H.); (D.G.); (A.T.)
| | - Alessandra Tolomelli
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, Ue4, 40129 Bologna, Italy; (T.H.); (D.G.); (A.T.)
| | - Monica Baiula
- Department of Pharmacology and Biotechnology (FABIT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy;
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Gobetti 83, Ue4, 40129 Bologna, Italy; (T.H.); (D.G.); (A.T.)
- Health Sciences & Technologies (HST) CIRI, University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano Emilia, Italy
| |
Collapse
|
15
|
Hao Y, Yan J, Fraser C, Jiang A, Anuganti M, Zhang R, Lloyd K, Jardine J, Coppola J, Meijers R, Li J, Springer TA. Synthetic integrin antibodies discovered by yeast display reveal αV subunit pairing preferences with β subunits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.26.577394. [PMID: 38328192 PMCID: PMC10849667 DOI: 10.1101/2024.01.26.577394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Eight of the 24 integrin heterodimers bind to the tripeptide Arg-Gly-Asp (RGD) motif in their extracellular ligands, and play essential roles in cell adhesion, migration, and homeostasis. Despite similarity in recognizing the RGD motif and some redundancy, these integrins can selectively recognize RGD-containing ligands including fibronectin, vitronectin, fibrinogen, nephronectin and the prodomain of the transforming growth factors to fulfill specific functions in cellular processes. Subtype-specific antibodies against RGD-binding integrins are desirable for investigating their specific functions. In this study, we discovered 11 antibodies that exhibit high specificity and affinity towards integrins αVβ3, αVβ5, αVβ6, αVβ8, and α5β1 from a synthetic yeast-displayed Fab library. Of these, 6 are function-blocking antibodies containing an R(G/L/T) D motif in their CDR3 sequences. We report antibody binding specificity, kinetics, and binding affinity for purified integrin ectodomains as well as intact integrins on the cell surface. We further employed these antibodies to reveal binding preferences of the αV subunit for its 5 β-subunit partners: β6=β8>β3>β1=β5.
Collapse
Affiliation(s)
- Yuxin Hao
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Courtney Fraser
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Murali Anuganti
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Roushu Zhang
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Kenneth Lloyd
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Joseph Jardine
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Jessica Coppola
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Rob Meijers
- Institute for Protein Innovation, Harvard Institutes of Medicine, 4 Blackfan Circle, Room 921, Boston, MA 02115
| | - Jing Li
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Department of Pediatrics, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Hao Y, Yan J, Fraser C, Jiang A, Anuganti M, Zhang R, Lloyd K, Jardine J, Coppola J, Meijers R, Li J, Springer TA. Synthetic integrin antibodies discovered by yeast display reveal αV subunit pairing preferences with β subunits. MAbs 2024; 16:2365891. [PMID: 38889315 PMCID: PMC11188837 DOI: 10.1080/19420862.2024.2365891] [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: 02/16/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
Integrins are cell surface receptors that mediate the interactions of cells with their surroundings and play essential roles in cell adhesion, migration, and homeostasis. Eight of the 24 integrins bind to the tripeptide Arg-Gly-Asp (RGD) motif in their extracellular ligands, comprising the RGD-binding integrin subfamily. Despite similarity in recognizing the RGD motif and some redundancy, these integrins can selectively recognize RGD-containing ligands to fulfill specific functions in cellular processes. Antibodies against individual RGD-binding integrins are desirable for investigating their specific functions, and were selected here from a synthetic yeast-displayed Fab library. We discovered 11 antibodies that exhibit high specificity and affinity toward their target integrins, i.e. αVβ3, αVβ5, αVβ6, αVβ8, and α5β1. Of these, six are function-blocking antibodies and contain a ligand-mimetic R(G/L/T)D motif in their CDR3 sequences. We report antibody-binding specificity, kinetics, and binding affinity for purified integrin ectodomains, as well as intact integrins on the cell surface. We further used these antibodies to reveal binding preferences of the αV subunit for its 5 β-subunit partners: β6 = β8 > β3 > β1 = β5.
Collapse
Affiliation(s)
- Yuxin Hao
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jiabin Yan
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Courtney Fraser
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | | | - Roushu Zhang
- Institute for Protein Innovation, Boston, MA, USA
| | | | | | | | - Rob Meijers
- Institute for Protein Innovation, Boston, MA, USA
| | - Jing Li
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy A. Springer
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Abstract
Multicellular organisms generate tissues of diverse shapes and functions from cells and extracellular matrices. Their adhesion molecules mediate cell-cell and cell-matrix interactions, which not only play crucial roles in maintaining tissue integrity but also serve as key regulators of tissue morphogenesis. Cells constantly probe their environment to make decisions: They integrate chemical and mechanical information from the environment via diffusible ligand- or adhesion-based signaling to decide whether to release specific signaling molecules or enzymes, to divide or differentiate, to move away or stay, or even whether to live or die. These decisions in turn modify their environment, including the chemical nature and mechanical properties of the extracellular matrix. Tissue morphology is the physical manifestation of the remodeling of cells and matrices by their historical biochemical and biophysical landscapes. We review our understanding of matrix and adhesion molecules in tissue morphogenesis, with an emphasis on key physical interactions that drive morphogenesis.
Collapse
Affiliation(s)
- Di Wu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA;
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| |
Collapse
|
18
|
Kim Y, Tram LTH, Kim KA, Kim BC. Defining Integrin Tension Required for Chemotaxis of Metastatic Breast Cancer Cells in Confinement. Adv Healthc Mater 2023; 12:e2202747. [PMID: 37256848 DOI: 10.1002/adhm.202202747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 05/21/2023] [Indexed: 06/02/2023]
Abstract
Cancer metastasis is affected by chemical factors and physical cues. From cell adhesion to migration, mechanical tension applied to integrin expresses on the cell membrane and physical confinement significantly regulates cancer cell behaviors. Despite the physical interplay between integrins in cells and ligands in the tumor microenvironment, quantitative analysis of integrin tension during cancer cell migration in microconfined spaces remains elusive owing to the limited experimental tools. Herein, a platform termed microconfinement tension gauge tether to monitor spatial integrin tension with single-molecule precision by analyzing the epithelial-growth-factor-induced chemotaxis of metastatic human breast cancer cells in microfluidic channels is developed. The results reveal that the metastatic cancer cells exert the strongest integrin tension in the range of 54-100 pN at the leading edges of cells during chemokinetic migration on a planar surface, while the cells exert the strongest integrin tension exceeding 100 pN at the cell rear when entering microconfinement. Further analysis demonstrates that cells undergo mesenchymal migration under high integrin tension and less confinement, which is converted to amoeboid migration under low integrin tension or high confinement. In summary, the results identify a basic mechanism underlying the mechanical interactions between integrin tension and microenvironment that determines cancer invasion and metastasis.
Collapse
Affiliation(s)
- Young Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Le Thi Hong Tram
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Kyung Ah Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Byoung Choul Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| |
Collapse
|
19
|
Rashid SA, Dong Y, Ogasawara H, Vierengel M, Essien ME, Salaita K. All-Covalent Nuclease-Resistant and Hydrogel-Tethered DNA Hairpin Probes Map pN Cell Traction Forces. ACS APPLIED MATERIALS & INTERFACES 2023; 15:33362-33372. [PMID: 37409737 PMCID: PMC10360067 DOI: 10.1021/acsami.3c04826] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/15/2023] [Indexed: 07/07/2023]
Abstract
Cells sense and respond to the physical properties of their environment through receptor-mediated signaling, a process known as mechanotransduction, which can modulate critical cellular functions such as proliferation, differentiation, and survival. At the molecular level, cell adhesion receptors, such as integrins, transmit piconewton (pN)-scale forces to the extracellular matrix, and the magnitude of the force plays a critical role in cell signaling. The most sensitive approach to measuring integrin forces involves DNA hairpin-based sensors, which are used to quantify and map forces in living cells. Despite the broad use of DNA hairpin sensors to study a variety of mechanotransduction processes, these sensors are typically anchored to rigid glass slides, which are orders of magnitude stiffer than the extracellular matrix and hence modulate native biological responses. Here, we have developed nuclease-resistant DNA hairpin probes that are all covalently tethered to PEG hydrogels to image cell traction forces on physiologically relevant substrate stiffness. Using HeLa cells as a model cell line, we show that the molecular forces transmitted by integrins are highly sensitive to the bulk modulus of the substrate, and cells cultured on the 6 and 13 kPa gels produced a greater number of hairpin unfolding events compared to the 2 kPa substrates. Tension signals are spatially colocalized with pY118-paxillin, confirming focal adhesion-mediated probe opening. Additionally, we found that integrin forces are greater than 5.8 pN but less than 19 pN on 13 kPa gels. This work provides a general strategy to integrate molecular tension probes into hydrogels, which can better mimic in vivo mechanotransduction.
Collapse
Affiliation(s)
- Sk Aysha Rashid
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Hiroaki Ogasawara
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Maia Vierengel
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Mark Edoho Essien
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
20
|
Jeong S, Shin W, Park M, Lee JU, Lim Y, Noh K, Lee JH, Jun YW, Kwak M, Cheon J. Hydrogel Magnetomechanical Actuator Nanoparticles for Wireless Remote Control of Mechanosignaling In Vivo. NANO LETTERS 2023; 23:5227-5235. [PMID: 37192537 PMCID: PMC10614426 DOI: 10.1021/acs.nanolett.3c01207] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
As a new enabling nanotechnology tool for wireless, target-specific, and long-distance stimulation of mechanoreceptors in vivo, here we present a hydrogel magnetomechanical actuator (h-MMA) nanoparticle. To allow both deep-tissue penetration of input signals and efficient force generation, h-MMA integrates a two-step transduction mechanism that converts magnetic anisotropic energy to thermal energy within its magnetic core (i.e., Zn0.4Fe2.6O4 nanoparticle cluster) and then to mechanical energy to induce the surrounding polymer (i.e., pNiPMAm) shell contraction, finally delivering forces to activate targeted mechanoreceptors. We show that h-MMAs enable on-demand modulation of Notch signaling in both fluorescence reporter cell lines and a xenograft mouse model, demonstrating its utility as a powerful in vivo perturbation approach for mechanobiology interrogation in a minimally invasive and untethered manner.
Collapse
Affiliation(s)
- Sumin Jeong
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Wookjin Shin
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Mansoo Park
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jung-uk Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
| | - Yongjun Lim
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
| | - Kunwoo Noh
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Hyun Lee
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Young-wook Jun
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
- Department of Otolaryngology, University of California, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, USA
- Helen Diller Family Cancer Comprehensive Center (HDFCCC), University of California, San Francisco, CA, USA
| | - Minsuk Kwak
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Jinwoo Cheon
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), A dvanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
21
|
Zhang W, Lu W, Yu Q, Liu X, Jiang H. Upregulated desmin/integrin β1/MAPK axis promotes elastic cartilage regeneration with increased ECM mechanical strength. Int J Biol Sci 2023; 19:2740-2755. [PMID: 37324935 PMCID: PMC10266073 DOI: 10.7150/ijbs.83024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Elastic cartilage tissue engineering is promising for providing available scaffolds for plastic reconstructive surgery. The insufficient mechanical strength of regenerative tissue and scarce resources of reparative cells are two obstacles for the preparation of tissue-engineered elastic cartilage scaffolds. Auricular chondrocytes are important reparative cells for elastic cartilage tissue engineering, but resources are scarce. Identifying auricular chondrocytes with enhanced capability of elastic cartilage formation is conducive to reducing the damage to donor sites by decreasing the demand on native tissue isolation. Based on the biochemical and biomechanical differences in native auricular cartilage, we found that auricular chondrocytes with upregulated desmin expressed more integrin β1, forming a stronger interaction with the substrate. Meanwhile, activated MAPK pathway was found in auricular chondrocytes highly expressing desmin. When desmin was knocked down, the chondrogenesis and mechanical sensitivity of chondrocytes were both impaired, and the MAPK pathway was downregulated. Finally, auricular chondrocytes highly expressing desmin regenerated more elastic cartilage with increased ECM mechanical strength. Therefore, desmin/integrin β1/MAPK signaling can not only serve as a selection standard but also a manipulation target of auricular chondrocytes to promote elastic cartilage regeneration.
Collapse
Affiliation(s)
| | | | | | - Xia Liu
- ✉ Corresponding authors: Xia Liu, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, PR China. E-mail: . Haiyue Jiang, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, PR China. E-mail:
| | - Haiyue Jiang
- ✉ Corresponding authors: Xia Liu, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, PR China. E-mail: . Haiyue Jiang, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, PR China. E-mail:
| |
Collapse
|
22
|
Pawlak MR, Smiley AT, Ramirez MP, Kelly MD, Shamsan GA, Anderson SM, Smeester BA, Largaespada DA, Odde DJ, Gordon WR. RAD-TGTs: high-throughput measurement of cellular mechanotype via rupture and delivery of DNA tension probes. Nat Commun 2023; 14:2468. [PMID: 37117218 PMCID: PMC10147940 DOI: 10.1038/s41467-023-38157-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Mechanical forces drive critical cellular processes that are reflected in mechanical phenotypes, or mechanotypes, of cells and their microenvironment. We present here "Rupture And Deliver" Tension Gauge Tethers (RAD-TGTs) in which flow cytometry is used to record the mechanical history of thousands of cells exerting forces on their surroundings via their propensity to rupture immobilized DNA duplex tension probes. We demonstrate that RAD-TGTs recapitulate prior DNA tension probe studies while also yielding a gain of fluorescence in the force-generating cell that is detectable by flow cytometry. Furthermore, the rupture propensity is altered following disruption of the cytoskeleton using drugs or CRISPR-knockout of mechanosensing proteins. Importantly, RAD-TGTs can differentiate distinct mechanotypes among mixed populations of cells. We also establish oligo rupture and delivery can be measured via DNA sequencing. RAD-TGTs provide a facile and powerful assay to enable high-throughput mechanotype profiling, which could find various applications, for example, in combination with CRISPR screens and -omics analysis.
Collapse
Affiliation(s)
- Matthew R Pawlak
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Adam T Smiley
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Maria Paz Ramirez
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Marcus D Kelly
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ghaidan A Shamsan
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Sarah M Anderson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | | | | | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Wendy R Gordon
- Departments of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|