1
|
Yang J, Liu Y, Li B, Li J, Yan S, Chen H. Cell elasticity measurement and sorting based on microfluidic techniques: Advances and applications. Biosens Bioelectron 2025; 271:116985. [PMID: 39642532 DOI: 10.1016/j.bios.2024.116985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Cell elasticity serves as a crucial physical biomarker that reflects changes in cellular structures and physiological states, providing key insights into cell behaviors. It links mechanical properties to biological function, highlighting its importance for understanding cell health and advancing biomedical research. Microfluidic technologies, with their capabilities for precise manipulation and high-throughput analysis, have significantly advanced the measurement of cell elasticity and elasticity-based cell sorting. This paper presents a comprehensive overview of advanced microsystems for assessing cell elasticity, discussing their advantages and limitations. The biomedical applications of elasticity-based sorting are highlighted, including cell classification, clinical diagnosis, drug screening, and stem cell differentiation prediction. The paper addresses the current challenges in the field, such as limited measurement efficiency and scalability, and explores future research directions, including the development of automated, high-throughput systems and the integration of elasticity measurements into practical biomedical applications. These advancements aim to deepen our understanding of cellular mechanics, improve diagnostic precision, and foster the development of novel therapeutic strategies. Ultimately, this work emphasizes the potential of cell elasticity as a key parameter in advancing disease diagnosis and therapeutic research.
Collapse
Affiliation(s)
- Jiahuan Yang
- School of Biomedical Engineering and Digital Health, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Yong Liu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China
| | - Bin Li
- School of Biomedical Engineering and Digital Health, Harbin Institute of Technology (Shenzhen), Shenzhen, China
| | - Jingjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, New South Wales, Australia.
| | - Sheng Yan
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, China.
| | - Huaying Chen
- School of Biomedical Engineering and Digital Health, Harbin Institute of Technology (Shenzhen), Shenzhen, China.
| |
Collapse
|
2
|
Li H, Harvey DH, Dai J, Swingle SP, Compton AM, Sugali CK, Dhamodaran K, Yao J, Lin TY, Sulchek T, Kim T, Ethier CR, Mao W. Characterization, enrichment, and computational modeling of cross-linked actin networks in trabecular meshwork cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608970. [PMID: 39229235 PMCID: PMC11370370 DOI: 10.1101/2024.08.21.608970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Purpose Cross-linked actin networks (CLANs) are prevalent in the glaucomatous trabecular meshwork (TM), yet their role in ocular hypertension remains unclear. We used a human TM cell line that spontaneously forms fluorescently-labeled CLANs (GTM3L) to explore the origin of CLANs, developed techniques to increase CLAN incidence in GMT3L cells, and computationally studied the biomechanical properties of CLAN-containing cells. Methods GTM3L cells were fluorescently sorted for viral copy number analysis. CLAN incidence was increased by (i) differential sorting of cells by adhesion, (ii) cell deswelling, and (iii) cell selection based on cell stiffness. GTM3L cells were also cultured on glass or soft hydrogel to determine substrate stiffness effects on CLAN incidence. Computational models were constructed to mimic and study the biomechanical properties of CLANs. Results All GTM3L cells had an average of 1 viral copy per cell. LifeAct-GFP expression level did not affect CLAN incidence rate, but CLAN rate was increased from ~0.28% to ~50% by a combination of adhesion selection, cell deswelling, and cell stiffness-based sorting. Further, GTM3L cells formed more CLANs on a stiff vs. a soft substrate. Computational modeling predicted that CLANs contribute to higher cell stiffness, including increased resistance of the nucleus to tensile stress when CLANs are physically linked to the nucleus. Conclusions It is possible to greatly enhance CLAN incidence in GTM3L cells. CLANs are mechanosensitive structures that affect cell biomechanical properties. Further research is needed to determine the effect of CLANs on TM biomechanics and mechanobiology as well as the etiology of CLAN formation in the TM.
Collapse
Affiliation(s)
- Haiyan Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA
| | - Devon H Harvey
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana
| | - Jiannong Dai
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana
| | - Steven P Swingle
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Anthony M Compton
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA
| | - Chenna Kesavulu Sugali
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana
| | - Kamesh Dhamodaran
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana
| | - Jing Yao
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana
| | - Tsai-Yu Lin
- Department of Medical and Molecular Genetics, Indiana University, Indianapolis, Indiana
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Taeyoon Kim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA
- Department of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA
| | - Weiming Mao
- Eugene and Marilyn Glick Eye Institute, Indiana University, Indianapolis, Indiana
- Department of Ophthalmology, Indiana University, Indianapolis, Indiana
- Department of Biochemistry & Molecular Biology, Indiana University, Indianapolis, Indiana
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, Indiana
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, Indiana
| |
Collapse
|
3
|
Iványi GT, Nemes B, Gróf I, Fekete T, Kubacková J, Tomori Z, Bánó G, Vizsnyiczai G, Kelemen L. Optically Actuated Soft Microrobot Family for Single-Cell Manipulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2401115. [PMID: 38814436 DOI: 10.1002/adma.202401115] [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: 01/22/2024] [Revised: 05/17/2024] [Indexed: 05/31/2024]
Abstract
Precisely controlled manipulation of nonadherent single cells is often a pre-requisite for their detailed investigation. Optical trapping provides a versatile means for positioning cells with submicrometer precision or measuring forces with femto-Newton resolution. A variant of the technique, called indirect optical trapping, enables single-cell manipulation with no photodamage and superior spatial control and stability by relying on optically trapped microtools biochemically bound to the cell. High-resolution 3D lithography enables to prepare such cell manipulators with any predefined shape, greatly extending the number of achievable manipulation tasks. Here, it is presented for the first time a novel family of cell manipulators that are deformable by optical tweezers and rely on their elasticity to hold cells. This provides a more straightforward approach to indirect optical trapping by avoiding biochemical functionalization for cell attachment, and consequently by enabling the manipulated cells to be released at any time. Using the photoresist Ormocomp, the deformations achievable with optical forces in the tens of pN range and present three modes of single-cell manipulation as examples to showcase the possible applications such soft microrobotic tools can offer are characterized. The applications describe here include cell collection, 3D cell imaging, and spatially and temporally controlled cell-cell interaction.
Collapse
Affiliation(s)
- Gergely T Iványi
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, 6720, Hungary
| | - Botond Nemes
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Ilona Gróf
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Tamás Fekete
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| | - Jana Kubacková
- Department of Biophysics, Institute of Experimental Physics SAS, Watsonova 47, Košice, 04001, Slovakia
| | - Zoltán Tomori
- Department of Biophysics, Institute of Experimental Physics SAS, Watsonova 47, Košice, 04001, Slovakia
| | - Gregor Bánó
- Department of Biophysics, Faculty of Science, P. J. Šafárik University in Košice, Jesenná 5, Košice, 04154, Slovakia
| | - Gaszton Vizsnyiczai
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
- Department of Biotechnology, University of Szeged, Szeged, 6720, Hungary
| | - Lóránd Kelemen
- HUN-REN Biological Research Centre, Szeged Institute of Biophysics, Temesvári krt. 62, Szeged, 6726, Hungary
| |
Collapse
|
4
|
Li SS, Xue CD, Li YJ, Chen XM, Zhao Y, Qin KR. Microfluidic characterization of single-cell biophysical properties and the applications in cancer diagnosis. Electrophoresis 2024; 45:1212-1232. [PMID: 37909658 DOI: 10.1002/elps.202300177] [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: 08/12/2023] [Revised: 09/25/2023] [Accepted: 10/16/2023] [Indexed: 11/03/2023]
Abstract
Single-cell biophysical properties play a crucial role in regulating cellular physiological states and functions, demonstrating significant potential in the fields of life sciences and clinical diagnostics. Therefore, over the last few decades, researchers have developed various detection tools to explore the relationship between the biophysical changes of biological cells and human diseases. With the rapid advancement of modern microfabrication technology, microfluidic devices have quickly emerged as a promising platform for single-cell analysis offering advantages including high-throughput, exceptional precision, and ease of manipulation. Consequently, this paper provides an overview of the recent advances in microfluidic analysis and detection systems for single-cell biophysical properties and their applications in the field of cancer. The working principles and latest research progress of single-cell biophysical property detection are first analyzed, highlighting the significance of electrical and mechanical properties. The development of data acquisition and processing methods for real-time, high-throughput, and practical applications are then discussed. Furthermore, the differences in biophysical properties between tumor and normal cells are outlined, illustrating the potential for utilizing single-cell biophysical properties for tumor cell identification, classification, and drug response assessment. Lastly, we summarize the limitations of existing microfluidic analysis and detection systems in single-cell biophysical properties, while also pointing out the prospects and future directions of their applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Shan-Shan Li
- School of Mechanical Engineering, Dalian University of Technology, Dalian, Liaoning, P. R. China
| | - Chun-Dong Xue
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, P. R. China
| | - Yong-Jiang Li
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, P. R. China
| | - Xiao-Ming Chen
- School of Optoelectronic Engineering and Instrumentation Science, Dalian University of Technology, Dalian, Liaoning, P. R. China
| | - Yan Zhao
- Department of Stomach Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, P. R. China
| | - Kai-Rong Qin
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning, P. R. China
| |
Collapse
|
5
|
Ma R, Rashid SA, Velusamy A, Deal BR, Chen W, Petrich B, Li R, Salaita K. Molecular mechanocytometry using tension-activated cell tagging. Nat Methods 2023; 20:1666-1671. [PMID: 37798479 PMCID: PMC11325290 DOI: 10.1038/s41592-023-02030-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/22/2023] [Indexed: 10/07/2023]
Abstract
Flow cytometry is used routinely to measure single-cell gene expression by staining cells with fluorescent antibodies and nucleic acids. Here, we present tension-activated cell tagging (TaCT) to label cells fluorescently based on the magnitude of molecular force transmitted through cell adhesion receptors. As a proof-of-concept, we analyzed fibroblasts and mouse platelets after TaCT using conventional flow cytometry.
Collapse
Affiliation(s)
- Rong Ma
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | | | | | - Brendan R Deal
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | - Wenchun Chen
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Brian Petrich
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Renhao Li
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Khalid Salaita
- Department of Chemistry, Emory University, Atlanta, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
6
|
Sun J, Huang X, Chen J, Xiang R, Ke X, Lin S, Xuan W, Liu S, Cao Z, Sun L. Recent advances in deformation-assisted microfluidic cell sorting technologies. Analyst 2023; 148:4922-4938. [PMID: 37743834 DOI: 10.1039/d3an01150j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Cell sorting is an essential prerequisite for cell research and has great value in life science and clinical studies. Among the many microfluidic cell sorting technologies, label-free methods based on the size of different cell types have been widely studied. However, the heterogeneity in size for cells of the same type and the inevitable size overlap between different types of cells would result in performance degradation in size-based sorting. To tackle such challenges, deformation-assisted technologies are receiving more attention recently. Cell deformability is an inherent biophysical marker of cells that reflects the changes in their internal structures and physiological states. It provides additional dimensional information for cell sorting besides size. Therefore, in this review, we summarize the recent advances in deformation-assisted microfluidic cell sorting technologies. According to how the deformability is characterized and the form in which the force acts, the technologies can be divided into two categories: (1) the indirect category including transit-time-based and image-based methods, and (2) the direct category including microstructure-based and hydrodynamics-based methods. Finally, the separation performance and the application scenarios of each method, the existing challenges and future outlook are discussed. Deformation-assisted microfluidic cell sorting technologies are expected to realize greater potential in the label-free analysis of cells.
Collapse
Affiliation(s)
- Jingjing Sun
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Xiwei Huang
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Jin Chen
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Rikui Xiang
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Xiang Ke
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Siru Lin
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Weipeng Xuan
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| | - Shan Liu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, China
| | - Zhen Cao
- College of Information Science and Electronic Engineering, Zhejiang University, China
| | - Lingling Sun
- Ministry of Education Key Lab of RF Circuits and Systems, Hangzhou Dianzi University, China.
| |
Collapse
|
7
|
Woodcock E, Gorelkin PV, Goff PS, Edwards CRW, Zhang Y, Korchev Y, Sviderskaya EV. Measuring Melanoma Nanomechanical Properties in Relation to Metastatic Ability and Anti-Cancer Drug Treatment Using Scanning Ion Conductance Microscopy. Cells 2023; 12:2401. [PMID: 37830615 PMCID: PMC10571876 DOI: 10.3390/cells12192401] [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: 08/31/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/14/2023] Open
Abstract
A cell's mechanical properties have been linked to cancer development, motility and metastasis and are therefore an attractive target as a universal, reliable cancer marker. For example, it has been widely published that cancer cells show a lower Young's modulus than their non-cancerous counterparts. Furthermore, the effect of anti-cancer drugs on cellular mechanics may offer a new insight into secondary mechanisms of action and drug efficiency. Scanning ion conductance microscopy (SICM) offers a nanoscale resolution, non-contact method of nanomechanical data acquisition. In this study, we used SICM to measure the nanomechanical properties of melanoma cell lines from different stages with increasing metastatic ability. Young's modulus changes following treatment with the anti-cancer drugs paclitaxel, cisplatin and dacarbazine were also measured, offering a novel perspective through the use of continuous scan mode SICM. We found that Young's modulus was inversely correlated to metastatic ability in melanoma cell lines from radial growth, vertical growth and metastatic phases. However, Young's modulus was found to be highly variable between cells and cell lines. For example, the highly metastatic cell line A375M was found to have a significantly higher Young's modulus, and this was attributed to a higher level of F-actin. Furthermore, our data following nanomechanical changes after 24 hour anti-cancer drug treatment showed that paclitaxel and cisplatin treatment significantly increased Young's modulus, attributed to an increase in microtubules. Treatment with dacarbazine saw a decrease in Young's modulus with a significantly lower F-actin corrected total cell fluorescence. Our data offer a new perspective on nanomechanical changes following drug treatment, which may be an overlooked effect. This work also highlights variations in cell nanomechanical properties between previous studies, cancer cell lines and cancer types and questions the usefulness of using nanomechanics as a diagnostic or prognostic tool.
Collapse
Affiliation(s)
- Emily Woodcock
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
| | - Peter V. Gorelkin
- Research Laboratory of Biophysics, National University of Science and Technology MISiS, Moscow 119049, Russia
| | - Philip S. Goff
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
| | | | - Yanjun Zhang
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuri Korchev
- Department of Medicine, Imperial College London, W12 0NN London, UK (Y.K.)
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Elena V. Sviderskaya
- Molecular and Clinical Sciences Research Institute, St George’s, University of London, London SW17 0RE, UK; (E.W.)
| |
Collapse
|
8
|
Daniel C, Traub F, Sachsenmaier S, Riester R, Mederake M, Konrads C, Danalache M. An exploratory study of cell stiffness as a mechanical label-free biomarker across multiple musculoskeletal sarcoma cells. BMC Cancer 2023; 23:862. [PMID: 37700272 PMCID: PMC10498616 DOI: 10.1186/s12885-023-11375-3] [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: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Cancer cells are characterized by changes in cell cytoskeletal architecture and stiffness. Despite advances in understanding the molecular mechanisms of musculoskeletal cancers, the corresponding cellular mechanical properties remain largely unexplored. The aim of this study was to investigate the changes in cellular stiffness and the associated cytoskeleton configuration alterations in various musculoskeletal cancer cells. METHODS Cell lines from five main sarcoma types of the musculoskeletal system (chondrosarcoma, osteosarcoma, Ewing sarcoma, fibrosarcoma and rhabdomyosarcoma) as well as their healthy cell counterparts (chondrocytes, osteoblasts, mesenchymal stem cells, fibroblasts, skeletal muscle cells) were subjected to cell stiffness measurements via atomic force microscopy (AFM). Biochemical and structural changes of the cytoskeleton (F-actin, β-tubulin and actin-related protein 2/3) were assessed by means of fluorescence labelling, ELISA and qPCR. RESULTS While AFM stiffness measurements showed that the majority of cancer cells (osteosarcoma, Ewing sarcoma, fibrosarcoma and rhabdomyosarcoma) were significantly less stiff than their corresponding non-malignant counterparts (p < 0.001), the chondrosarcoma cells were significant stiffer than the chondrocytes (p < 0.001). Microscopically, the distribution of F-actin differed between malignant entities and healthy counterparts: the organisation in well aligned stress fibers was disrupted in cancer cell lines and the proteins was mainly concentrated at the periphery of the cell, whereas β-tubulin had a predominantly perinuclear localization. While the F-actin content was lower in cancer cells, particularly Ewing sarcoma (p = 0.018) and Fibrosarcoma (p = 0.023), this effect was even more pronounced in the case of β-tubulin for all cancer-healthy cell duos. Interestingly, chondrosarcoma cells were characterized by a significant upregulation of β-tubulin gene expression (p = 0.005) and protein amount (p = 0.032). CONCLUSION Modifications in cellular stiffness, along with structural and compositional cytoskeleton rearrangement, constitute typical features of sarcomas cells, when compared to their healthy counterpart. Notably, whereas a decrease in stiffness is typically a feature of malignant entities, chondrosarcoma cells were stiffer than chondrocytes, with chondrosarcoma cells exhibiting a significantly upregulated β-tubulin expression. Each Sarcoma entity may have his own cellular-stiffness and cytoskeleton organisation/composition fingerprint, which in turn may be exploited for diagnostic or therapeutic purposes.
Collapse
Affiliation(s)
- Cyril Daniel
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany.
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany.
| | - Frank Traub
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedics and Traumatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55122, Mainz, Germany
| | - Saskia Sachsenmaier
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| | - Rosa Riester
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| | - Moritz Mederake
- Department of Trauma and Reconstructive Surgery, BG Clinic, University of Tübingen, 72076, Tübingen, Germany
| | - Christian Konrads
- Department of Orthopedics and Traumatology, Hanseatic Hospital Stralsund, 18437, Stralsund, Germany
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
9
|
Volmer LL, Önder CE, Volz B, Singh AR, Brucker SY, Engler T, Hartkopf AD, Koch A. Microfluidic Isolation of Disseminated Tumor Cells from the Bone Marrow of Breast Cancer Patients. Int J Mol Sci 2023; 24:13930. [PMID: 37762233 PMCID: PMC10531360 DOI: 10.3390/ijms241813930] [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: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Disseminated tumor cells (DTCs) in the bone marrow (BM) of breast cancer (BC) patients are putative precursors of metastatic disease, and their presence is associated with an adverse clinical outcome. To achieve the personalization of therapy on a clinical routine level, the characterization of DTCs and in vitro drug testing on DTCs are of great interest. Therefore, biobanking methods, as well as novel approaches to DTC isolation, need to be developed. In this study, we established a protocol for the biobanking of BM samples and evaluated a microfluidic-based separation system (Parsortix®) for the enrichment of cryopreserved DTCs. We were able to successfully isolate viable DTCs after the prior cryopreservation of BM samples. We calculated a significant increase of up to 90-fold in harvested DTCs with the proposed method compared to the current standard techniques, opening up new analysis possibilities for DTCs. Our advanced method further presents options for 3D DTC cultures, enabling the individualized testing of targeted therapies for BC patients. In conclusion, we present a novel approach for DTC enrichment, with possibilities for future clinical implications.
Collapse
Affiliation(s)
- Léa L. Volmer
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Cansu E. Önder
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Barbara Volz
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Anjali R. Singh
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Sara Y. Brucker
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Tobias Engler
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - Andreas D. Hartkopf
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
- Department of Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| | - André Koch
- Research Institute for Women’s Health, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
10
|
Raj A, Ramirez K, Young KM, Stone N, Shankles P, Ali MNR, Compton AM, Lam W, Alexeev A, Sulchek T. Label-free microfluidic isolation of functional and viable lymphocytes from peripheral blood mononuclear cells. BIOMICROFLUIDICS 2023; 17:054102. [PMID: 37736019 PMCID: PMC10511259 DOI: 10.1063/5.0161047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/28/2023] [Indexed: 09/23/2023]
Abstract
The separation of peripheral blood mononuclear cells (PBMCs) into constituent blood cell types is a vital step to obtain immune cells for autologous cell therapies. The ability to separate PBMCs using label-free microfluidic techniques, based on differences in biomechanical properties, can have a number of benefits over other conventional techniques, including lower cost, ease of use, and avoidance of animal-derived labeling antibodies. Here, we report a microfluidic device that uses compressive diagonal ridges to separate PBMCs into highly pure samples of viable and functional lymphocytes. The technique utilizes the differences in the biophysical properties of PBMC sub-populations to direct the lymphocytes and monocytes into separate outlets. The biophysical properties of the monocytes and lymphocytes from healthy donors were first characterized using atomic force microscopy. Lymphocytes were found to be significantly stiffer than monocytes, with a mean cell stiffness of 1495 and 931 Pa, respectively. The differences in biophysical properties resulted in distinct trajectories through the microchannel terminating at different outlets, resulting in a lymphocyte sample with purity and viability both greater than 96% with no effect on the cells' ability to produce interferon gamma, a cytokine crucial for innate and adaptive immunity.
Collapse
Affiliation(s)
- Abhishek Raj
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Katily Ramirez
- School of Chemistry & Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, Georgia 30332-0400, USA
| | - Katherine M. Young
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Nicholas Stone
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Peter Shankles
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Mehdia Nadeem Rajab Ali
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Anthony Malik Compton
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | | | - Alexander Alexeev
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Todd Sulchek
- Author to whom correspondence should be addressed:
| |
Collapse
|
11
|
Basile G, Vetere A, Hu J, Ijaduola O, Zhang Y, Liu KC, Eltony AM, De Jesus DF, Fukuda K, Doherty G, Leech CA, Chepurny OG, Holz GG, Yun SH, Andersson O, Choudhary A, Wagner BK, Kulkarni RN. Excess pancreatic elastase alters acinar-β cell communication by impairing the mechano-signaling and the PAR2 pathways. Cell Metab 2023; 35:1242-1260.e9. [PMID: 37339634 PMCID: PMC10834355 DOI: 10.1016/j.cmet.2023.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/21/2023] [Accepted: 05/17/2023] [Indexed: 06/22/2023]
Abstract
Type 1 (T1D) or type 2 diabetes (T2D) are caused by a deficit of functional insulin-producing β cells. Thus, the identification of β cell trophic agents could allow the development of therapeutic strategies to counteract diabetes. The discovery of SerpinB1, an elastase inhibitor that promotes human β cell growth, prompted us to hypothesize that pancreatic elastase (PE) regulates β cell viability. Here, we report that PE is up-regulated in acinar cells and in islets from T2D patients, and negatively impacts β cell viability. Using high-throughput screening assays, we identified telaprevir as a potent PE inhibitor that can increase human and rodent β cell viability in vitro and in vivo and improve glucose tolerance in insulin-resistant mice. Phospho-antibody microarrays and single-cell RNA sequencing analysis identified PAR2 and mechano-signaling pathways as potential mediators of PE. Taken together, our work highlights PE as a potential regulator of acinar-β cell crosstalk that acts to limit β cell viability, leading to T2D.
Collapse
Affiliation(s)
- Giorgio Basile
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Amedeo Vetere
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Jiang Hu
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Oluwaseun Ijaduola
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Yi Zhang
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Ka-Cheuk Liu
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Amira M Eltony
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Dario F De Jesus
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Kazuki Fukuda
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Grace Doherty
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA
| | - Colin A Leech
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - Oleg G Chepurny
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - George G Holz
- Department of Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA; Department of Pharmacology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY 13210, USA
| | - Seok-Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA; Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Olov Andersson
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Amit Choudhary
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bridget K Wagner
- Chemical Biology and Therapeutics Science Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Rohit N Kulkarni
- Section on Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02215, USA.
| |
Collapse
|
12
|
Wu Y, Yakov S, Fu A, Yossifon G. A Magnetically and Electrically Powered Hybrid Micromotor in Conductive Solutions: Synergistic Propulsion Effects and Label-Free Cargo Transport and Sensing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204931. [PMID: 36507618 PMCID: PMC10015886 DOI: 10.1002/advs.202204931] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/31/2022] [Indexed: 06/18/2023]
Abstract
Electrically powered micro- and nanomotors are promising tools for in vitro single-cell analysis. In particular, single cells can be trapped, transported, and electroporated by a Janus particle (JP) using an externally applied electric field. However, while dielectrophoretic (DEP)-based cargo manipulation can be achieved at high-solution conductivity, electrical propulsion of these micromotors becomes ineffective at solution conductivities exceeding ≈0.3 mS cm-1 . Here, JP cargo manipulation and transport capabilities to conductive near-physiological (<6 mS cm-1 ) solutions are extended successfully by combining magnetic field-based micromotor propulsion and navigation with DEP-based manipulation of various synthetic and biological cargos. Combination of a rotating magnetic field and electric field results in enhanced micromotor mobility and steering control through tuning of the electric field frequency. In addition, the micromotor's ability of identifying apoptotic cell among viable and necrotic cells based on their dielectrophoretic difference is demonstrated, thus, enabling to analyze the apoptotic status in the single-cell samples for drug discovery, cell therapeutics, and immunotherapy. The ability to trap and transport live cells towards regions containing doxorubicin-loaded liposomes is also demonstrated. This hybrid micromotor approach for label-free trapping, transporting, and sensing of selected cells within conductive solutions opens new opportunities in drug delivery and single-cell analysis, where close-to-physiological media conditions are necessary.
Collapse
Affiliation(s)
- Yue Wu
- School of Mechanical EngineeringUniversity of Tel‐AvivTel‐Aviv69978Israel
| | - Sivan Yakov
- Faculty of Mechanical EngineeringMicro‐ and Nanofluidics LaboratoryTechnion—Israel Institute of TechnologyHaifa32000Israel
| | - Afu Fu
- Technion Integrated Cancer CenterThe Rappaport Faculty of Medicine and Research InstituteTechnion—Israel Institute of TechnologyHaifa3109602Israel
| | - Gilad Yossifon
- School of Mechanical EngineeringUniversity of Tel‐AvivTel‐Aviv69978Israel
- Faculty of Mechanical EngineeringMicro‐ and Nanofluidics LaboratoryTechnion—Israel Institute of TechnologyHaifa32000Israel
| |
Collapse
|
13
|
Murashko AV, Frolova AA, Akovantseva AA, Kotova SL, Timashev PS, Efremov YM. The cell softening as a universal indicator of cell damage during cytotoxic effects. Biochim Biophys Acta Gen Subj 2023; 1867:130348. [PMID: 36977439 DOI: 10.1016/j.bbagen.2023.130348] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/03/2023] [Accepted: 03/20/2023] [Indexed: 03/28/2023]
Abstract
Cytotoxicity assays are essential tests in studies on the safety and biocompatibility of various substances and on the efficiency of anticancer drugs. The most frequently used assays commonly require application of externally added labels and read only collective response of cells. Recent studies show that the internal biophysical parameters of cells can be associated with the cellular damage. Therefore, using atomic force microscopy, we assessed the changes in the viscoelastic parameters of cells treated with eight different common cytotoxic agents to gain a more systematic view of the occurring mechanical changes. With the robust statistical analysis to account for both the cell-level variability and the experimental reproducibility, we have found that cell softening is a common response after each treatment. More precisely, the combined changes in the viscoelastic parameters of power-law rheology model led to a significant decrease of the apparent elastic modulus. The comparison with the morphological parameters (cytoskeleton and cell shape) demonstrated a higher sensitivity of the mechanical parameters versus the morphological ones. The obtained results support the idea of cell mechanics-based cytotoxicity tests and suggest a common way of a cell responding to damaging actions by softening.
Collapse
|
14
|
Martinez-Vidal L, Chighizola M, Berardi M, Alchera E, Locatelli I, Pederzoli F, Venegoni C, Lucianò R, Milani P, Bielawski K, Salonia A, Podestà A, Alfano M. Micro-mechanical fingerprints of the rat bladder change in actinic cystitis and tumor presence. Commun Biol 2023; 6:217. [PMID: 36823431 PMCID: PMC9950451 DOI: 10.1038/s42003-023-04572-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 02/09/2023] [Indexed: 02/25/2023] Open
Abstract
Tissue mechanics determines tissue homeostasis, disease development and progression. Bladder strongly relies on its mechanical properties to perform its physiological function, but these are poorly unveiled under normal and pathological conditions. Here we characterize the mechanical fingerprints at the micro-scale level of the three tissue layers which compose the healthy bladder wall, and identify modifications associated with the onset and progression of pathological conditions (i.e., actinic cystitis and bladder cancer). We use two indentation-based instruments (an Atomic Force Microscope and a nanoindenter) and compare the micromechanical maps with a comprehensive histological analysis. We find that the healthy bladder wall is a mechanically inhomogeneous tissue, with a gradient of increasing stiffness from the urothelium to the lamina propria, which gradually decreases when reaching the muscle outer layer. Stiffening in fibrotic tissues correlate with increased deposition of dense extracellular matrix in the lamina propria. An increase in tissue compliance is observed before the onset and invasion of the tumor. By providing high resolution micromechanical investigation of each tissue layer of the bladder, we depict the intrinsic mechanical heterogeneity of the layers of a healthy bladder as compared with the mechanical properties alterations associated with either actinic cystitis or bladder tumor.
Collapse
Affiliation(s)
- Laura Martinez-Vidal
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - M Chighizola
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy
| | - M Berardi
- Optics11, Amsterdam, The Netherlands
- LaserLab, Department of Physics and Astronomy, VU University, Amsterdam, The Netherlands
| | - E Alchera
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - I Locatelli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - F Pederzoli
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - C Venegoni
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - R Lucianò
- Pathology Unit, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
| | - P Milani
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy
| | | | - A Salonia
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy
- Università Vita-Salute San Raffaele, Via Olgettina, 60, Milan, 20132, Italy
| | - A Podestà
- C.I.Ma.I.Na and Dipartimento di Fisica "Aldo Pontremoli", Università degli Studi di Milano, Milan, 20133, Italy.
| | - M Alfano
- Division of Experimental Oncology/Unit of Urology, IRCCS Ospedale San Raffaele, Milan, 20132, Italy.
| |
Collapse
|
15
|
Wubshet NH, Liu AP. Methods to mechanically perturb and characterize GUV-based minimal cell models. Comput Struct Biotechnol J 2022; 21:550-562. [PMID: 36659916 PMCID: PMC9816913 DOI: 10.1016/j.csbj.2022.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/15/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Cells shield organelles and the cytosol via an active boundary predominantly made of phospholipids and membrane proteins, yet allowing communication between the intracellular and extracellular environment. Micron-sized liposome compartments commonly known as giant unilamellar vesicles (GUVs) are used to model the cell membrane and encapsulate biological materials and processes in a cell-like confinement. In the field of bottom-up synthetic biology, many have utilized GUVs as substrates to study various biological processes such as protein-lipid interactions, cytoskeletal assembly, and dynamics of protein synthesis. Like cells, it is ideal that GUVs are also mechanically durable and able to stay intact when the inner and outer environment changes. As a result, studies have demonstrated approaches to tune the mechanical properties of GUVs by modulating membrane composition and lumenal material property. In this context, there have been many different methods developed to test the mechanical properties of GUVs. In this review, we will survey various perturbation techniques employed to mechanically characterize GUVs.
Collapse
Affiliation(s)
- Nadab H. Wubshet
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Allen P. Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biophysics, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Kiwanuka M, Higgins G, Ngcobo S, Nagawa J, Lang DM, Zaman MH, Davies NH, Franz T. Effect of paclitaxel treatment on cellular mechanics and morphology of human oesophageal squamous cell carcinoma in 2D and 3D environments. Integr Biol (Camb) 2022; 14:zyac013. [PMID: 36244059 PMCID: PMC9585394 DOI: 10.1093/intbio/zyac013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 08/14/2022] [Accepted: 09/10/2022] [Indexed: 06/16/2023]
Abstract
During chemotherapy, structural and mechanical changes in malignant cells have been observed in several cancers, including leukaemia and pancreatic and prostate cancer. Such cellular changes may act as physical biomarkers for chemoresistance and cancer recurrence. This study aimed to determine how exposure to paclitaxel affects the intracellular stiffness of human oesophageal cancer of South African origin in vitro. A human oesophageal squamous cell carcinoma cell line WHCO1 was cultured on glass substrates (2D) and in collagen gels (3D) and exposed to paclitaxel for up to 48 h. Cellular morphology and stiffness were assessed with confocal microscopy, visually aided morpho-phenotyping image recognition and mitochondrial particle tracking microrheology at 24 and 48 h. In the 2D environment, the intracellular stiffness was higher for the paclitaxel-treated than for untreated cells at 24 and 48 h. In the 3D environment, the paclitaxel-treated cells were stiffer than the untreated cells at 24 h, but no statistically significant differences in stiffness were observed at 48 h. In 2D, paclitaxel-treated cells were significantly larger at 24 and 48 h and more circular at 24 but not at 48 h than the untreated controls. In 3D, there were no significant morphological differences between treated and untreated cells. The distribution of cell shapes was not significantly different across the different treatment conditions in 2D and 3D environments. Future studies with patient-derived primary cancer cells and prolonged drug exposure will help identify physical cellular biomarkers to detect chemoresistance onset and assess therapy effectiveness in oesophageal cancer patients.
Collapse
Affiliation(s)
- Martin Kiwanuka
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Ghodeejah Higgins
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Silindile Ngcobo
- Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, University of Cape Town, Observatory, South Africa
| | - Juliet Nagawa
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Dirk M Lang
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Neil H Davies
- Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, University of Cape Town, Observatory, South Africa
| | - Thomas Franz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
- Bioengineering Science Research Group, Engineering Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
17
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
18
|
Dempsey ME, Chickering GR, González-Cruz RD, Fonseca VC, Darling EM. Discovery of surface biomarkers for cell mechanophenotype via an intracellular protein-based enrichment strategy. Cell Mol Life Sci 2022; 79:320. [PMID: 35622146 PMCID: PMC9239330 DOI: 10.1007/s00018-022-04351-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Cellular mechanophenotype is often a defining characteristic of conditions like cancer malignancy/metastasis, cardiovascular disease, lung and liver fibrosis, and stem cell differentiation. However, acquiring living cells based on mechanophenotype is challenging for conventional cell sorters due to a lack of biomarkers. In this study, we demonstrate a workflow for surface protein discovery associated with cellular mechanophenotype. We sorted heterogeneous adipose-derived stem/stromal cells (ASCs) into groups with low vs. high lamin A/C, an intracellular protein linked to whole-cell mechanophenotype. Proteomic data of enriched groups identified surface protein candidates as potential biochemical proxies for ASC mechanophenotype. Select surface biomarkers were used for live-cell enrichment, with subsequent single-cell mechanical testing and lineage-specific differentiation. Ultimately, we identified CD44 to have a strong inverse correlation with whole-cell elastic modulus, with CD44lo cells exhibiting moduli three times greater than that of CD44hi cells. Functionally, these stiff and soft ASCs showed enhanced osteogenic and adipogenic differentiation potential, respectively. The described workflow can be replicated for any phenotype with a known correlated intracellular protein, allowing for the acquisition of live cells for further characterization, diagnostics, or therapeutics.
Collapse
Affiliation(s)
- Megan E Dempsey
- Center for Biomedical Engineering, Brown University, Providence, RI, 02912, USA
| | | | | | - Vera C Fonseca
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
| | - Eric M Darling
- Center for Biomedical Engineering, Brown University, Providence, RI, 02912, USA.
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA.
- School of Engineering, Brown University, Providence, RI, 02912, USA.
- Department of Orthopaedics, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
19
|
|
20
|
Young KM, Shankles PG, Chen T, Ahkee K, Bules S, Sulchek T. Scaling microfluidic throughput with flow-balanced manifolds to simply control devices with multiple inlets and outlets. BIOMICROFLUIDICS 2022; 16:034104. [PMID: 35600502 PMCID: PMC9118023 DOI: 10.1063/5.0080510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/02/2022] [Indexed: 05/18/2023]
Abstract
Microfluidics can bring unique functionalities to cell processing, but the small channel dimensions often limit the throughput for cell processing that prevents scaling necessary for key applications. While processing throughput can be improved by increasing cell concentration or flow rate, an excessive number or velocity of cells can result in device failure. Designing parallel channels can linearly increase the throughput by channel number, but for microfluidic devices with multiple inlets and outlets, the design of the channel architecture with parallel channels can result in intractable numbers of inlets and outlets. We demonstrate an approach to use multiple parallel channels for complex microfluidic designs that uses a second manifold layer to connect three inlets and five outlets per channel in a manner that balances flow properties through each channel. The flow balancing in the individual microfluidic channels was accomplished through a combination of analytical and finite element analysis modeling. Volumetric flow and cell flow velocity were measured in each multiplexed channel to validate these models. We demonstrate eight-channel operation of a label-free mechanical separation device that retains the accuracy of a single channel separation. Using the parallelized device and a model biomechanical cell system for sorting of cells based on their viability, we processed over 16 × 106 cells total over three replicates at a rate of 5.3 × 106 cells per hour. Thus, parallelization of complex microfluidics with a flow-balanced manifold system can enable higher throughput processing with the same number of inlet and outlet channels to control.
Collapse
Affiliation(s)
- Katherine M. Young
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Peter G. Shankles
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Theresa Chen
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Kelly Ahkee
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Sydney Bules
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Todd Sulchek
- Author to whom correspondence should be addressed:. Phone: (404) 385-1887
| |
Collapse
|
21
|
Choi G, Tang Z, Guan W. Microfluidic high-throughput single-cell mechanotyping: Devices and
applications. NANOTECHNOLOGY AND PRECISION ENGINEERING 2021. [DOI: 10.1063/10.0006042] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Gihoon Choi
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| | - Zifan Tang
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| | - Weihua Guan
- Department of Electrical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802,
USA
| |
Collapse
|
22
|
Chrit FE, Raj A, Young KM, Stone NE, Shankles PG, Lokireddy K, Flowers C, Waller EK, Alexeev A, Sulchek T. Microfluidic Platform to Transduce Cell Viability to Distinct Flow Pathways for High-Accuracy Sensing. ACS Sens 2021; 6:3789-3799. [PMID: 34546721 DOI: 10.1021/acssensors.1c01770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mechanical properties of cells such as stiffness can act as biomarkers to sort or detect cell functional properties such as viability. In this study, we report the use of a microfluidic device as a high-sensitivity sensor that transduces cell biomechanics to cell separation to accurately detect viability. Cell populations are flowed and deflected at a number of skew ridges such that deflection per ridge, cell-ridge interaction time, and cell size can all be used as sensor inputs to accurately determine the cell state. The angle of the ridges was evaluated to optimize the differences in cell translation between viable and nonviable cells while allowing continuous flow. In the first mode of operation, we flowed viable and nonviable cells through the device and conducted a sensitivity analysis by recording the cell's total deflection as a binary classifier that differentiates viable from nonviable cells. The performance of the sensor was assessed using an area under the curve (AUC) analysis to be 0.97. By including additional sensor inputs in the second mode of operation, we conducted a principal component analysis (PCA) to further improve the identification of the cell state by clustering populations with little overlap between viable and nonviable cells. We therefore found that microfluidic separation devices can be used to efficiently sort cells and accurately sense viability in a label-free manner.
Collapse
Affiliation(s)
- Fatima Ezahra Chrit
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Abhishek Raj
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Katherine M. Young
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia 30332, United States
| | - Nicholas E. Stone
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Peter G. Shankles
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Kesiharjun Lokireddy
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Christopher Flowers
- Winship Cancer Institute, Emory School of Medicine, 1365 Clifton NE Road, Atlanta, Georgia 30322, United States
| | - Edmund K. Waller
- Winship Cancer Institute, Emory School of Medicine, 1365 Clifton NE Road, Atlanta, Georgia 30322, United States
| | - Alexander Alexeev
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Todd Sulchek
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
23
|
Stone NE, Raj A, Young KM, DeLuca AP, Chrit FE, Tucker BA, Alexeev A, McDonald J, Benigno BB, Sulchek T. Label-free microfluidic enrichment of cancer cells from non-cancer cells in ascites. Sci Rep 2021; 11:18032. [PMID: 34504124 PMCID: PMC8429413 DOI: 10.1038/s41598-021-96862-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/13/2021] [Indexed: 11/18/2022] Open
Abstract
The isolation of a patient's metastatic cancer cells is the first, enabling step toward treatment of that patient using modern personalized medicine techniques. Whereas traditional standard-of-care approaches select treatments for cancer patients based on the histological classification of cancerous tissue at the time of diagnosis, personalized medicine techniques leverage molecular and functional analysis of a patient's own cancer cells to select treatments with the highest likelihood of being effective. Unfortunately, the pure populations of cancer cells required for these analyses can be difficult to acquire, given that metastatic cancer cells typically reside in fluid containing many different cell populations. Detection and analyses of cancer cells therefore require separation from these contaminating cells. Conventional cell sorting approaches such as Fluorescence Activated Cell Sorting or Magnetic Activated Cell Sorting rely on the presence of distinct surface markers on cells of interest which may not be known nor exist for cancer applications. In this work, we present a microfluidic platform capable of label-free enrichment of tumor cells from the ascites fluid of ovarian cancer patients. This approach sorts cells based on differences in biomechanical properties, and therefore does not require any labeling or other pre-sort interference with the cells. The method is also useful in the cases when specific surface markers do not exist for cells of interest. In model ovarian cancer cell lines, the method was used to separate invasive subtypes from less invasive subtypes with an enrichment of ~ sixfold. In ascites specimens from ovarian cancer patients, we found the enrichment protocol resulted in an improved purity of P53 mutant cells indicative of the presence of ovarian cancer cells. We believe that this technology could enable the application of personalized medicine based on analysis of liquid biopsy patient specimens, such as ascites from ovarian cancer patients, for quick evaluation of metastatic disease progression and determination of patient-specific treatment.
Collapse
Affiliation(s)
- Nicholas E Stone
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Abhishek Raj
- Department of Mechanical Engineering, Indian Institute of Technology Patna, Bihar, 801103, India
| | - Katherine M Young
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Adam P DeLuca
- Department of Ophthalmology and Visual Science, Carver College of Medicine, Institute for Vision Research, University of Iowa, Iowa City, IA, 52242, USA
| | - Fatima Ezahra Chrit
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Budd A Tucker
- Department of Ophthalmology and Visual Science, Carver College of Medicine, Institute for Vision Research, University of Iowa, Iowa City, IA, 52242, USA
| | - Alexander Alexeev
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - John McDonald
- School of Biology, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | | | - Todd Sulchek
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
24
|
Šimoliūnas E, Ivanauskienė I, Bagdzevičiūtė L, Rinkūnaitė I, Alksnė M, Baltriukienė D. Surface stiffness depended gingival mesenchymal stem cell sensitivity to oxidative stress. Free Radic Biol Med 2021; 169:62-73. [PMID: 33862162 DOI: 10.1016/j.freeradbiomed.2021.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/16/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in the fields of cell therapy and tissue engineering, due to their wide spectrum of differentiation potential, immunomodulation function and ongoing oxidative stress (OS) reduction. Nevertheless, OS impact is often overlooked in these research fields. It is not only responsible for the induction and development of many ailments, e.g., diabetes, lung fibrosis, and cancer, moreover, OS causes stem cell death and senescence during cell therapy and tissue engineering practices. As MSCs are used to treat various tissues, they interact with different tissue-specific mechanical environments, thus it is important to understand how the mechanical environment impacts MSC sensitivity to OS. In this work, for the first time, as known to the authors, it was shown that gingival MSCs (GMSCs) sensitivity to OS depends on the stiffness of the surface, on which the cells are grown. Furthermore, the activity and expression of mitogen activated protein kinases ERK, JNK, and p38 were surface stiffness dependent. GMSCs isolated from intermediate/stiff gingiva tissue (~20 kPa) have shown the best proliferative and survival properties, then grown on the stiffest tissues mimicking polyacrylamide hydrogels (40 kPa). Therefore, MSC source might determine their sensitivity to OS in different stiffness environments and should be accounted when developing a treatment strategy.
Collapse
Affiliation(s)
- Egidijus Šimoliūnas
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania.
| | - Indrė Ivanauskienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Lina Bagdzevičiūtė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Ieva Rinkūnaitė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Milda Alksnė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Daiva Baltriukienė
- Department of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
25
|
Cermeño EA, O'Melia MJ, Han WM, Veith A, Barber G, Huang EH, Thomas SN, García AJ. Hydrodynamic shear-based purification of cancer cells with enhanced tumorigenic potential. Integr Biol (Camb) 2021; 12:1-11. [PMID: 31965190 DOI: 10.1093/intbio/zyz038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 11/13/2022]
Abstract
Tumor-initiating cells (TICs), a subpopulation of cancerous cells with high tumorigenic potential and stem-cell-like properties, drive tumor progression and are resistant to conventional therapies. Identification and isolation of TICs are limited by their low frequency and lack of robust markers. Here, we characterize the heterogeneous adhesive properties of a panel of human and murine cancer cells and demonstrate differences in adhesion strength among cells, which exhibit TIC properties and those that do not. These differences in adhesion strength were exploited to rapidly (~10 min) and efficiently isolate cancerous cells with increased tumorigenic potential in a label-free manner by use of a microfluidic technology. Isolated murine and human cancer cells gave rise to larger tumors with increased growth rate and higher frequency in both immunocompetent and immunocompromised mice, respectively. This rapid and label-free TIC isolation technology has the potential to be a valuable tool for facilitating research into TIC biology and the development of more efficient diagnostics and cancer therapies.
Collapse
Affiliation(s)
- Efraín A Cermeño
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meghan J O'Melia
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,Coulter Department of Biomedical Engineering, Georgia Tech/Emory, Atlanta, GA, USA
| | - Woojin M Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Austin Veith
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Graham Barber
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Emina H Huang
- Lerner Research Institute, Department of Cancer Biology, Department of Colorectal Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Susan N Thomas
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
26
|
Makowiecka A, Mazurkiewicz E, Mrówczyńska E, Malek N, Battistella A, Lazzarino M, Nowak D, Mazur AJ. Changes in Biomechanical Properties of A375 Cells Due to the Silencing of TMSB4X Expression Are Not Directly Correlated with Alterations in Their Stemness Features. Cells 2021; 10:cells10040769. [PMID: 33807338 PMCID: PMC8067020 DOI: 10.3390/cells10040769] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 12/27/2022] Open
Abstract
Thymosin β4 (Tβ4) is a small, 44-amino acid polypeptide. It has been implicated in multiple processes, including cell movement, angiogenesis, and stemness. Previously, we reported that melanoma cell lines differ in Tβ4 levels. Studies on stable clones with silenced TMSB4X expression showed that Tβ4 impacted adhesion and epithelial-mesenchymal transition progression. Here, we show that the cells with silenced TMSB4X expression exhibited altered actin cytoskeleton’s organization and subcellular relocalization of two intermediate filament proteins: Nestin and Vimentin. The rearrangement of the cell cytoskeleton resulted in changes in the cells’ topology, height, and stiffness defined by Young’s modulus. Simultaneously, only for some A375 clones with a lowered Tβ4 level, we observed a decreased ability to initiate colony formation in soft agar, tumor formation in vivo, and alterations in Nanog’s expression level transcription factor regulating stemness. Thus, we show for the first time that in A375 cells, biomechanical properties are not directly coupled to stemness features, and this cell line is phenotypically heterogeneous.
Collapse
Affiliation(s)
- Aleksandra Makowiecka
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
- Correspondence: or (A.M.); (A.J.M.); Tel.: +48-71-375-6206 (A.J.M.)
| | - Ewa Mazurkiewicz
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Ewa Mrówczyńska
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Natalia Malek
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Alice Battistella
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
| | - Marco Lazzarino
- Istituto Officina dei Materiali-National Research Council, I-34149 Trieste, Italy; (A.B.); (M.L.)
| | - Dorota Nowak
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
| | - Antonina Joanna Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, 50-383 Wrocław, Poland; (E.M.); (E.M.); (N.M.); (D.N.)
- Correspondence: or (A.M.); (A.J.M.); Tel.: +48-71-375-6206 (A.J.M.)
| |
Collapse
|
27
|
Stone NE, Voigt AP, Cooke JA, Giacalone JC, Hanasoge S, Mullins RF, Tucker BA, Sulchek T. Label-free microfluidic enrichment of photoreceptor cells. Exp Eye Res 2020; 199:108166. [PMID: 32771499 DOI: 10.1016/j.exer.2020.108166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/20/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022]
Abstract
Inherited retinal degenerative disorders such as retinitis pigmentosa and Usher syndrome are characterized by progressive death of photoreceptor cells. To restore vision to patients blinded by these diseases, a stem cell-based photoreceptor cell replacement strategy will likely be required. Although retinal stem cell differentiation protocols suitable for generating photoreceptor cells exist, they often yield a rather heterogenous mixture of cell types. To enrich the donor cell population for one or a few cell types, scientists have traditionally relied upon the use of antibody-based selection approaches. However, these strategies are quite labor intensive and require animal derived reagents and equipment that are not well suited to current good manufacturing practices (cGMP). The purpose of this study was to develop and evaluate a microfluidic cell sorting device capable of exploiting the physical and mechanical differences between retinal cell types to enrich specific donor cell populations such as Retinal Pigment Epithelial (RPE) cells and photoreceptor cells. Using this device, we were able to separate a mixture of RPE and iPSC-derived photoreceptor precursor cell lines into two substantially enriched fractions. The enrichment factor of the RPE fraction was 2 and that of the photoreceptor precursor cell fraction was 2.7. Similarly, when human retina, obtained from 3 independent donors, was dissociated and passed through the sorting device, the heterogeneous mixture could be reliably sorted into RPE and photoreceptor cell rich fractions. In summary, microfluidic cell sorting is a promising approach for antibody free enrichment of retinal cell populations.
Collapse
Affiliation(s)
- Nicholas E Stone
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Andrew P Voigt
- Institute for Vision Research, Department of Ophthalmology and Visual Science, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jessica A Cooke
- Institute for Vision Research, Department of Ophthalmology and Visual Science, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Joseph C Giacalone
- Institute for Vision Research, Department of Ophthalmology and Visual Science, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Srinivas Hanasoge
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Robert F Mullins
- Institute for Vision Research, Department of Ophthalmology and Visual Science, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Budd A Tucker
- Institute for Vision Research, Department of Ophthalmology and Visual Science, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Todd Sulchek
- The George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
28
|
Microfluidic Chip based direct triple antibody immunoassay for monitoring patient comparative response to leukemia treatment. Biomed Microdevices 2020; 22:48. [PMID: 32661698 DOI: 10.1007/s10544-020-00503-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We report a time and cost-efficient microfluidic chip for screening the leukemia cells having three specific antigens. In this method, the target blast cells are double sorted with immunomagnetic beads and captured by the 3rd antibody immobilized on the gold surface in a microfluidic chip. The captured blast cells in the chip were imaged using a bright-field optical microscope and images were analyzed to quantify the cells. First sorting was performed with nano size immunomagnetic beads and followed by 2nd sorting where micron size immunomagnetic beads were used. The low-cost microfluidic platform is made of PMMA and glass including micro size gold pads. The developed microfluidic platform was optimized with cultured B type lymphoblast cells and tested with the samples of leukemia patients. The 8 bone marrow samples of 4 leukemia patients on the initial diagnosis and on the 15th day after the start of the chemotherapy treatment were tested both with the developed microfluidic platform and the flow cytometry. A 99% statistical agreement between the two methods shows that the microfluidic chip is able to monitor the decrease in the number of blast cells due to the chemotherapy. The experiments with the patient samples demonstrate that the developed system can perform relative measurements and have a potential to monitor the patient response to the applied therapy and to enable personalized dose adjustment.
Collapse
|
29
|
Islam M, Raj A, McFarland B, Brink HM, Ciciliano J, Fay M, Myers DR, Flowers C, Waller EK, Lam W, Alexeev A, Sulchek T. Stiffness based enrichment of leukemia cells using microfluidics. APL Bioeng 2020; 4:036101. [PMID: 32637856 PMCID: PMC7332299 DOI: 10.1063/1.5143436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 06/08/2020] [Indexed: 01/06/2023] Open
Abstract
To improve the survival rate of cancer patients, new diagnosis strategies are necessary to detect lower levels of cancer cells before and after treatment regimens. The scarcity of diseased cells, particularly in residual disease after treatment, demands highly sensitive detection approaches or the ability to enrich the diseased cells in relation to normal cells. We report a label-free microfluidic approach to enrich leukemia cells from healthy cells using inherent differences in cell biophysical properties. The microfluidic device consists of a channel with an array of diagonal ridges that recurrently compress and translate flowing cells in proportion to cell stiffness. Using devices optimized for acute T cell leukemia model Jurkat, the stiffer white blood cells were translated orthogonally to the channel length, while softer leukemia cells followed hydrodynamic flow. The device enriched Jurkat leukemia cells from white blood cells with an enrichment factor of over 760. The sensitivity, specificity, and accuracy of the device were found to be >0.8. The values of sensitivity and specificity could be adjusted by selecting one or multiple outlets for analysis. We demonstrate that low levels of Jurkat leukemia cells (1 in 104 white blood cells) could be more quickly detected using flow cytometry by using the stiffness sorting pre-enrichment. In a second mode of operation, the device was implemented to sort resistive leukemia cells from both drug-sensitive leukemia cells and normal white blood cells. Therefore, microfluidic biomechanical sorting can be a useful tool to enrich leukemia cells that may improve downstream analyses.
Collapse
Affiliation(s)
- Muhymin Islam
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Abhishek Raj
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | - Brynn McFarland
- The School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Hannah Maxine Brink
- The School of Biological Sciences, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Jordan Ciciliano
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Meredith Fay
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - David Richard Myers
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Christopher Flowers
- Winship Cancer Institute, Emory School of Medicine, 1365 Clifton NE Rd.: Atlanta, Georgia 30322, USA
| | - Edmund K Waller
- Winship Cancer Institute, Emory School of Medicine, 1365 Clifton NE Rd.: Atlanta, Georgia 30322, USA
| | - Wilbur Lam
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, Georgia 30332-0535, USA
| | - Alexander Alexeev
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, Georgia 30332-0405, USA
| | | |
Collapse
|
30
|
Chen Z, Zhu Y, Xu D, Alam MM, Shui L, Chen H. Cell elasticity measurement using a microfluidic device with real-time pressure feedback. LAB ON A CHIP 2020; 20:2343-2353. [PMID: 32463051 DOI: 10.1039/d0lc00092b] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The study of cell elasticity provides new insights into not only cell biology but also disease diagnosis based on cell mechanical state variation. Microfluidic technologies have made noticeable progress in studying cell deformation with capabilities of high throughput and automation. This paper reports the development of a novel microfluidic system to precisely measure the elasticity of cells having large deformation in a constriction channel. It integrated i) a separation unit to isolate rod- or flake-shaped particles that might block the constriction channel to increase the measurement throughput and ii) a pressure feedback system precisely detecting the pressure drop inducing the deformation of each cell. The fluid dynamics of the separation unit was modeled to understand the separation mechanism before the experimental determination of separation efficiency. Afterward, the pressure system was characterized to demonstrate its sensitivity and reproducibility in measuring the subtle pressure drop along a constriction channel. Finally, the microfluidic system was employed to study the stiffness of both K562 and endothelial cells. The cell protrusion and pressure drop were employed to calculate the mechanical properties based on a power-law rheology model describing the viscoelastic behaviors of cells. Both the stiffness and the fluidity of K562 and endothelial cells were consistent with those in previous studies. The system has remarkable application potential in the precise evaluation of cell mechanical properties.
Collapse
Affiliation(s)
- Zhenlin Chen
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen, Shenzhen 518055, China.
| | | | | | | | | | | |
Collapse
|
31
|
Integrating the Tumor Microenvironment into Cancer Therapy. Cancers (Basel) 2020; 12:cancers12061677. [PMID: 32599891 PMCID: PMC7352326 DOI: 10.3390/cancers12061677] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Tumor progression is mediated by reciprocal interaction between tumor cells and their surrounding tumor microenvironment (TME), which among other factors encompasses the extracellular milieu, immune cells, fibroblasts, and the vascular system. However, the complexity of cancer goes beyond the local interaction of tumor cells with their microenvironment. We are on the path to understanding cancer from a systemic viewpoint where the host macroenvironment also plays a crucial role in determining tumor progression. Indeed, growing evidence is emerging on the impact of the gut microbiota, metabolism, biomechanics, and the neuroimmunological axis on cancer. Thus, external factors capable of influencing the entire body system, such as emotional stress, surgery, or psychosocial factors, must be taken into consideration for enhanced management and treatment of cancer patients. In this article, we review prognostic and predictive biomarkers, as well as their potential evaluation and quantitative analysis. Our overarching aim is to open up new fields of study and intervention possibilities, within the framework of an integral vision of cancer as a functional tissue with the capacity to respond to different non-cytotoxic factors, hormonal, immunological, and mechanical forces, and others inducing stroma and tumor reprogramming.
Collapse
|
32
|
Kelp G, Li J, Lu J, DiNapoli N, Delgado R, Liu C, Fan D, Dutta-Gupta S, Shvets G. Infrared spectroscopy of live cells from a flowing solution using electrically-biased plasmonic metasurfaces. LAB ON A CHIP 2020; 20:2136-2153. [PMID: 32406430 DOI: 10.1039/c9lc01054h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Spectral cytopathology (SCP) is a promising label-free technique for diagnosing diseases and monitoring therapeutic outcomes using FTIR spectroscopy. In most cases, cells must be immobilized on a substrate prior to spectroscopic interrogation. This creates significant limitations for high throughput phenotypic whole-cell analysis, especially for the non-adherent cells. Here we demonstrate how metasurface-enhanced infrared reflection spectroscopy (MEIRS) can be applied to a continuous flow of live cell solution by applying AC voltage to metallic metasurfaces. By integrating metasurfaces with microfluidic delivery channels and attracting the cells to the metasurface via dielectrophoretic (DEP) force, we collect the infrared spectra of cells in real time within a minute, and correlate the spectra with simultaneously acquired images of the attracted cells. The resulting DEP-MEIRS technique paves the way for rapid SCP of complex cell-containing body fluids with low cell concentrations, and for the development of a wide range of label-free liquid biopsies.
Collapse
Affiliation(s)
- Glen Kelp
- Department of Physics, University of Texas at Austin, Austin, Texas 78712, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Chen Z, Jiang K, Zou Z, Luo X, Lim CT, Wen C. High-throughput and label-free isolation of senescent murine mesenchymal stem cells. BIOMICROFLUIDICS 2020; 14:034106. [PMID: 32477445 PMCID: PMC7244328 DOI: 10.1063/5.0011925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023]
Abstract
Under internal or external insults such as aging and oxidative stresses, cells are induced into a senescent state and stop cellular division permanently. As senescent cells (SnCs) accumulate, the regeneration capacity of biological tissue would be compromised, which has been found to be associated with a plethora of age-related disorders. Therefore, isolating SnCs becomes necessary. To address the lack of effective surface markers for SnCs isolation, a label-free microfluidic device was proposed in this paper, in which a spiral microchannel was deployed to isolate SnCs based on their size differences. We adopted a well-received cellular senescence model by exerting excessive oxidative stress to murine mesenchymal stem cells. This model was then validated through a series of SnCs characterizations including size measurement, p16INK4a expression level, senescence-associated beta-galactosidase, and doubling time. The senescence chip demonstrated an efficiency of 75% and viability over 85% at a flow rate of 5 ml/min. The average cell size from the inner outlet was 5 μm larger than that from the outer outlet. The isolated cells had a sixfold higher p16INK4a expression level. Overall, the chip had an area under curve of 0.719 in the receiver operating characteristic analysis, showing decent performance in sorting SnCs. By having the ability to perform size-based sorting at a high flow rate, such a microfluidic device can provide high-throughput and label-free isolation of SnCs. To further improve the isolation performance, the device can be modified to introduce additional physical biomarkers of SnCs such as stiffness. This device poses a good potential in purification for cytotherapy or estimation of biological age.
Collapse
Affiliation(s)
- Zhengkun Chen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Kuan Jiang
- Mechanobiology Institute, National University of Singapore, Singapore 117411
| | - Zhou Zou
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaohe Luo
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | | | - Chunyi Wen
- Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
34
|
Affiliation(s)
- Malgorzata A. Witek
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
- Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Ian M. Freed
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
| | - Steven A. Soper
- Department of Chemistry, The University of Kansas, Lawrence, Kansas 66044, United States
- Center of Biomodular Multiscale Systems for Precision Medicine, The University of Kansas, Lawrence, Kansas 66044, United States
- Department of Mechanical Engineering, The University of Kansas, Lawrence, Kansas 66044, United States
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66044, United States
| |
Collapse
|
35
|
Liu A, Yu T, Young K, Stone N, Hanasoge S, Kirby TJ, Varadarajan V, Colonna N, Liu J, Raj A, Lammerding J, Alexeev A, Sulchek T. Cell Mechanical and Physiological Behavior in the Regime of Rapid Mechanical Compressions that Lead to Cell Volume Change. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903857. [PMID: 31782912 PMCID: PMC7012384 DOI: 10.1002/smll.201903857] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/05/2019] [Indexed: 04/14/2023]
Abstract
Cells respond to mechanical forces by deforming in accordance with viscoelastic solid behavior. Studies of microscale cell deformation observed by high speed video microscopy have elucidated a new cell behavior in which sufficiently rapid mechanical compression of cells can lead to transient cell volume loss and then recovery. This work has discovered that the resulting volume exchange between the cell interior and the surrounding fluid can be utilized for efficient, convective delivery of large macromolecules (2000 kDa) to the cell interior. However, many fundamental questions remain about this cell behavior, including the range of deformation time scales that result in cell volume loss and the physiological effects experienced by the cell. In this study, a relationship is established between cell viscoelastic properties and the inertial forces imposed on the cell that serves as a predictor of cell volume loss across human cell types. It is determined that cells maintain nuclear envelope integrity and demonstrate low protein loss after the volume exchange process. These results define a highly controlled cell volume exchange mechanism for intracellular delivery of large macromolecules that maintains cell viability and function for invaluable downstream research and clinical applications.
Collapse
Affiliation(s)
- Anna Liu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Tong Yu
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Katherine Young
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Nicholas Stone
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Srinivas Hanasoge
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Tyler J. Kirby
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Vikram Varadarajan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Nicholas Colonna
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0535, USA
| | - Janet Liu
- Aragon High School, 900 Alameda de las Pulgas, San Mateo, CA, 94402, USA
| | - Abhishek Raj
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Jan Lammerding
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14853, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, 14853, USA
| | - Alexander Alexeev
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| | - Todd Sulchek
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA, 30332-0405, USA
| |
Collapse
|
36
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
37
|
Zhou J, Mukherjee P, Gao H, Luan Q, Papautsky I. Label-free microfluidic sorting of microparticles. APL Bioeng 2019; 3:041504. [PMID: 31832577 PMCID: PMC6906121 DOI: 10.1063/1.5120501] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Massive growth of the microfluidics field has triggered numerous advances in focusing, separating, ordering, concentrating, and mixing of microparticles. Microfluidic systems capable of performing these functions are rapidly finding applications in industrial, environmental, and biomedical fields. Passive and label-free methods are one of the major categories of such systems that have received enormous attention owing to device operational simplicity and low costs. With new platforms continuously being proposed, our aim here is to provide an updated overview of the state of the art for passive label-free microparticle separation, with emphasis on performance and operational conditions. In addition to the now common separation approaches using Newtonian flows, such as deterministic lateral displacement, pinched flow fractionation, cross-flow filtration, hydrodynamic filtration, and inertial microfluidics, we also discuss separation approaches using non-Newtonian, viscoelastic flow. We then highlight the newly emerging approach based on shear-induced diffusion, which enables direct processing of complex samples such as untreated whole blood. Finally, we hope that an improved understanding of label-free passive sorting approaches can lead to sophisticated and useful platforms toward automation in industrial, environmental, and biomedical fields.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Prithviraj Mukherjee
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Hua Gao
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Qiyue Luan
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Ian Papautsky
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
38
|
Wang K, Xue Y, Peng Y, Pang X, Zhang Y, Ruiz-Ortega LI, Tian Y, Ngan AHW, Tang B. Elastic modulus and migration capability of drug treated leukemia cells K562. Biochem Biophys Res Commun 2019; 516:177-182. [PMID: 31204049 DOI: 10.1016/j.bbrc.2019.06.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 01/25/2023]
Abstract
Leukemia is a commonly seen disease caused by abnormal differentiation of hematopoietic stem cells and blasting in bone marrow. Despite drugs are used to treat the disease clinically, the influence of these drugs on leukemia cells' biomechanical properties, which are closely related to complications like leukostasis or infiltration, is still unclear. Due to non-adherent and viscoelastic nature of leukemia cells, accurate measurement of their elastic modulus is still a challenging issue. In this study, we adopted rate-jump method together with optical tweezers indentation to accurately measure elastic modulus of leukemia cells K562 after phorbol 12-myristate 13-acetate (PMA), all-trans retinoic acid (ATRA), Cytoxan (CTX), and Dexamethasone (DEX) treatment, respectively. We found that compared to control sample, K562 cells treated by PMA showed nearly a threefold increase in elastic modulus. Transwell experiment results suggested that the K562 cells treated with PMA have the lowest migration capability. Besides, it was shown that the cytoskeleton protein gene α-tubulin and vimentin have a significant increase in expression after PMA treatment by qPCR. The results indicate that PMA has a significant influence on protein expression, stiffness, and migration ability of the leukemia cell K562, and may also play an important role in the leukostasis in leukemia.
Collapse
Affiliation(s)
- Kui Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China; Department of Mechanical Engineering, University of Hong Kong, Hong Kong, PR China
| | - Yuntian Xue
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Ying Peng
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Xiangchao Pang
- College of Materials Science and Engineering, Central South University of Forestry and Technology, Changsha, 410004, PR China
| | - Yuanjun Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - L I Ruiz-Ortega
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - Ye Tian
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China
| | - A H W Ngan
- Department of Mechanical Engineering, University of Hong Kong, Hong Kong, PR China
| | - Bin Tang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, PR China; Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment, PR China.
| |
Collapse
|
39
|
Aljaghtham MS, Liu ZL, Guo JJ, He J, Celik E. Numerical simulations of cell flow and trapping within microfluidic channels for stiffness based cell isolation. J Biomech 2019; 85:43-49. [PMID: 30655079 DOI: 10.1016/j.jbiomech.2019.01.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 11/10/2018] [Accepted: 01/03/2019] [Indexed: 11/18/2022]
Abstract
Analysis of rare cells in heterogenous mixtures is proven to be beneficial for regenerative medicine, cancer treatment and prenatal diagnostics. Scarcity of these cells, however, makes the isolation process extremely challenging. Efficiency in cell isolation is still low and therefore, novel cell isolation strategies with new biomarkers need exploration. In this study, we investigated the feasibility of using the mechanical stiffness difference to detect and isolate the rare cells from the surrounding cells without labelling them. Fluid and solid mechanics simulations have shown that cell isolation can be performed at high efficiency using stiffness-based isolation. Accuracy of the numerical simulations is established using microfluidic flow chamber experiments.
Collapse
Affiliation(s)
- Mutabe S Aljaghtham
- Department of Mechanical and Aerospace Engineering, University of Miami, USA
| | - Zixiang L Liu
- Department Mechanical Engineering, Georgia Institute of Technology, USA
| | - Jing J Guo
- Department of Physics, Florida International University, USA
| | - Jin He
- Department of Physics, Florida International University, USA
| | - Emrah Celik
- Department of Mechanical and Aerospace Engineering, University of Miami, USA.
| |
Collapse
|
40
|
Viscoelastic Separation and Concentration of Fungi from Blood for Highly Sensitive Molecular Diagnostics. Sci Rep 2019; 9:3067. [PMID: 30816161 PMCID: PMC6395622 DOI: 10.1038/s41598-019-39175-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/18/2019] [Indexed: 12/30/2022] Open
Abstract
Isolation and concentration of fungi in the blood improves sensitivity of the polymerase chain reaction (PCR) method to detect fungi in blood. This study demonstrates a sheathless, continuous separation and concentration method of candida cells using a viscoelastic fluid that enables rapid detection of rare candida cells by PCR analysis. To validate device performance using a viscoelastic fluid, flow characteristics of 2 μm particles were estimated at different flow rates. Additionally, a mixture of 2 μm and 13 μm particles was successfully separated based on size difference at 100 μl/min. Candida cells were successfully separated from the white blood cells (WBCs) with a separation efficiency of 99.1% and concentrated approximately 9.9-fold at the center outlet compared to the initial concentration (~2.5 × 107 cells/ml). Sequential 1st and 2nd concentration processes were used to increase the final number of candida cells to ~2.3 × 109 cells/ml, which was concentrated ~92-fold. Finally, despite the undetectable initial concentration of 101 CFU/ml, removal of WBCs and the additional buffer solution enabled the quantitative reverse transcription (RT)-PCR detection of candida cells after the 1st concentration (Ct = 31.43) and the 2nd concentration process (Ct = 29.30).
Collapse
|
41
|
Kelp G, Arju N, Lee A, Esquivel E, Delgado R, Yu Y, Dutta-Gupta S, Sokolov K, Shvets G. Application of metasurface-enhanced infra-red spectroscopy to distinguish between normal and cancerous cell types. Analyst 2019; 144:1115-1127. [PMID: 30702730 PMCID: PMC6437688 DOI: 10.1039/c8an01433g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Fourier transform infrared (FTIR) spectra of biological cells can reveal clinically important information about cells' composition, including their normal or cancerous status. The recently emerged diagnostic technique of spectral cytopathology (SCP) combines FTIR with multivariate statistical analysis to detect cell abnormalities, differentiate between cell types, and monitor disease progression. We demonstrate a new variant of SCP, a metasurface-enhanced infrared reflection spectroscopic cytopathology (MEIRSC) that utilises judiciously designed plasmonic metasurfaces to localize and enhance the evanescent field near the cell's membrane, and to carry out spectroscopic interrogations of the cells attached to the metasurface using reflected infrared light. Our findings indicate that the MEIRSC approach enables us to differentiate between normal and cancerous human colon cells. The sensitivity of MEIRSC is such that a very small (about 50 nm deep) portion of the cell can yield valuable diagnostic information.
Collapse
Affiliation(s)
- G Kelp
- Department of Physics, University of Texas at Austin, Austin, Texas 78712, USA and School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA.
| | - N Arju
- Department of Physics, University of Texas at Austin, Austin, Texas 78712, USA
| | - A Lee
- Department of Imaging Physics, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA and Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - E Esquivel
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - R Delgado
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA.
| | - Y Yu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA.
| | - S Dutta-Gupta
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA. and Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology Hyderabad, Hyderabad, Telangana 502285, India
| | - K Sokolov
- Department of Imaging Physics, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA and Department of Biomedical Engineering, University of Texas at Austin, Austin, TX 78712, USA
| | - G Shvets
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
42
|
Biophysical properties of cells for cancer diagnosis. J Biomech 2019; 86:1-7. [PMID: 30803699 DOI: 10.1016/j.jbiomech.2019.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/23/2018] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
Biophysical properties associated with the microenvironment of a tumor has been recognized as an important modulator for cell behaviour and function. Particularly, tissue rigidity is important during tumor carcinogenesis as it affects the tumor's ability to metastasis. Multiple downstream pathways are affected with a difference in rigidity of the extracellular matrix. The insight into tumor mechanosignalling represents a promising field that may lead to novel approaches for cancer diagnostics. Measurement of rigidity of the extracellular matrix or the tissue is a potential diagnostics approach for cancer detection. Altered extracellular matrix states persist for a long period of time and have lower heterogeneity compared to protein or genetic markers, therefore are more reliable as biomarkers. On the other hand, measurement of different kinase associated proteins or transcripts provide an early insight into potential transition of cells towards metastasis. Co-localization of transcriptional factors like YAP/TAZ provide an insight to determine if the cells are undergoing metastatic changes. This review explains the unique biophysical properties of the tumor microenvironment that present the potential targets for the diagnosis of cancer.
Collapse
|
43
|
Singaraju AB, Bahl D, Stevens LL. Brillouin Light Scattering: Development of a Near Century-Old Technique for Characterizing the Mechanical Properties of Materials. AAPS PharmSciTech 2019; 20:109. [PMID: 30746575 DOI: 10.1208/s12249-019-1311-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/15/2019] [Indexed: 11/30/2022] Open
Abstract
Brillouin light scattering (BLS), a technique theoretically described nearly a century back by the French physicist Léon Brillouin in 1922, is a light-scattering method for determining the mechanical properties of materials. This inelastic scattering method is described by the Bragg diffraction of light from a propagating fluctuation in the local dielectric. These fluctuations arise spontaneously from thermally populated sound waves intrinsic to all materials, and thus BLS may be broadly applied to transparent samples of any phase. This review begins with a brief historical overview of the development of BLS, from its theoretical prediction to the current state of the art, and notes specific technological advancements that enabled the development of BLS. Despite the broad utility of BLS, no commercial spectrometer is currently available for purchase, but rather individual components are assembled to suit a specific application. Central to any BLS spectrometer is the interferometer, and its performance characteristics-scanning or non-scanning, multi-passing, and stabilization-are critical considerations for spectrometer design. Consistent with any light-scattering method, the frequency shift is a key observable in BLS, and we summarize the connection of this measurement to evaluate the mechanical properties of materials. With emphasis toward pharmaceutical materials analysis, we introduce the traditional BLS approach for single-crystal elasticity, and this is followed by a discussion of more recent developments in powder BLS. We conclude our review with a perspective on future developments in BLS that may enable BLS as a novel addition to the current catalog of process analytical technologies.
Collapse
|
44
|
Dalili A, Samiei E, Hoorfar M. A review of sorting, separation and isolation of cells and microbeads for biomedical applications: microfluidic approaches. Analyst 2019; 144:87-113. [DOI: 10.1039/c8an01061g] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We have reviewed the microfluidic approaches for cell/particle isolation and sorting, and extensively explained the mechanism behind each method.
Collapse
Affiliation(s)
- Arash Dalili
- The University of British
- School of Engineering
- Kelowna
- Canada V1 V 1 V7
| | - Ehsan Samiei
- University of Victoria
- Department of Mechanical Engineering
- Victoria
- Canada
| | - Mina Hoorfar
- The University of British
- School of Engineering
- Kelowna
- Canada V1 V 1 V7
| |
Collapse
|
45
|
Almabadi HM, Nagesh PKB, Sahay P, Bhandari S, Eckstein EC, Jaggi M, Chauhan SC, Yallapu MM, Pradhan P. Optical study of chemotherapy efficiency in cancer treatment via intracellular structural disorder analysis using partial wave spectroscopy. JOURNAL OF BIOPHOTONICS 2018; 11:e201800056. [PMID: 29869394 DOI: 10.1002/jbio.201800056] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/01/2018] [Indexed: 06/08/2023]
Abstract
As cancer progresses, macromolecules, such as DNA, RNA or lipids, inside cells undergo spatial structural rearrangements and alterations. Mesoscopic light transport-based optical partial wave spectroscopy (PWS) was recently introduced to quantify changes in the nanoscale structural disorder in biological cells. The PWS measurement is performed using a parameter termed as "disorder strength" (L d ), which represents the degree of nanoscale structural disorder inside the cells. It was shown that cancerous cells have higher disorder strength than normal cells. In this work, we first used the PWS to analyze the hierarchy of different types of prostate cancer cells, namely, C4-2, DU-145 and PC-3, by quantifying their average disorder strengths. Results expectedly showed that L d values increases in accordance with the increasing aggressiveness/tumorigenicity levels of these cells. Using the L d parameter, we then analyzed the chemoresistance properties of these prostate cancer cells to docetaxel drug compared to their chemosensitivity. Results show that chemoresistant cancer cells have increased L d values, that is, higher disorder strength, relative to chemosensitive cancer cells. Thus, use of the L d metric can be effective in determining the efficacy of particular chemotherapy.
Collapse
Affiliation(s)
- Huda M Almabadi
- Department of Physics and Materials Science, BioNanoPhotonics Laboratory, University of Memphis, Memphis, Tennessee
- Biomedical Engineering, University of Memphis, Memphis, Tennessee
| | - Prashanth K B Nagesh
- Department of Pharmaceutical Sciences and the Center for Cancer Research, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Peeyush Sahay
- Department of Physics and Materials Science, BioNanoPhotonics Laboratory, University of Memphis, Memphis, Tennessee
| | - Shiva Bhandari
- Department of Physics and Materials Science, BioNanoPhotonics Laboratory, University of Memphis, Memphis, Tennessee
| | | | - Meena Jaggi
- Department of Pharmaceutical Sciences and the Center for Cancer Research, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and the Center for Cancer Research, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences and the Center for Cancer Research, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Prabhakar Pradhan
- Department of Physics and Materials Science, BioNanoPhotonics Laboratory, University of Memphis, Memphis, Tennessee
| |
Collapse
|
46
|
Affiliation(s)
- Gongchen Sun
- School of Chemical & Biomolecular Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| | - Hang Lu
- School of Chemical & Biomolecular Engineering , Georgia Institute of Technology , Atlanta , Georgia 30332 , United States
| |
Collapse
|