1
|
Maremonti MI, Panzetta V, Netti PA, Causa F. HiViPore: a highly viable in-flow compression for a one-step cell mechanoporation in microfluidics to induce a free delivery of nano- macro-cargoes. J Nanobiotechnology 2024; 22:441. [PMID: 39068464 PMCID: PMC11282774 DOI: 10.1186/s12951-024-02730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Among mechanoporation techniques for intracellular delivery, microfluidic approaches succeed in high delivery efficiency and throughput. However, especially the entry of large cargoes (e.g. DNA origami, mRNAs, organic/inorganic nanoparticles) is currently impaired since it requires large cell membrane pores with the need to apply multi-step processes and high forces, dramatically reducing cell viability. RESULTS Here, HiViPore presents as a microfluidic viscoelastic contactless compression for one-step cell mechanoporation to produce large pores while preserving high cell viability. Inducing an increase of curvature at the equatorial region of cells, formation of a pore with a size of ~ 1 μm is obtained. The poration is coupled to an increase of membrane tension, measured as a raised fluorescence lifetime of 12% of a planarizable push-pull fluorescent probe (Flipper-TR) labelling the cell plasma membrane. Importantly, the local disruptions of cell membrane are transient and non-invasive, with a complete recovery of cell integrity and functions in ~ 10 min. As result, HiViPore guarantees cell viability as high as ~ 90%. In such conditions, an endocytic-free diffusion of large nanoparticles is obtained with typical size up to 500 nm and with a delivery efficiency up to 12 times higher than not-treated cells. CONCLUSIONS The proposed one-step contactless mechanoporation results in an efficient and safe approach for advancing intracellular delivery strategies. In detail, HiViPore solves the issues of low cell viability when multiple steps of poration are required to obtain large pores across the cell plasma membrane. Moreover, the compression uses a versatile, low-cost, biocompatible viscoelastic fluid, thus also optimizing the operational costs. With HiViPore, we aim to propose an easy-to-use microfluidic device to a wide range of users, involved in biomedical research, imaging techniques and nanotechnology for intracellular delivery applications in cell engineering.
Collapse
Affiliation(s)
- Maria Isabella Maremonti
- Interdisciplinary Research Centre on Biomaterials (CRIB), Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, University of Naples "Federico II", Naples, 80125, Italy
| | - Valeria Panzetta
- Interdisciplinary Research Centre on Biomaterials (CRIB), Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, University of Naples "Federico II", Naples, 80125, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Paolo Antonio Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, University of Naples "Federico II", Naples, 80125, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Naples, 80125, Italy
| | - Filippo Causa
- Interdisciplinary Research Centre on Biomaterials (CRIB), Dipartimento di Ingegneria Chimica, dei Materiali e della Produzione Industriale, University of Naples "Federico II", Naples, 80125, Italy.
| |
Collapse
|
2
|
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
|
3
|
Lu R, Yu P, Sui Y. A computational study of cell membrane damage and intracellular delivery in a cross-slot microchannel. SOFT MATTER 2024; 20:4057-4071. [PMID: 38578041 DOI: 10.1039/d4sm00047a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
We propose a three-dimensional computational framework to simulate the flow-induced cell membrane damage and the resulting enhanced intracellular mass transport in a cross-slot microchannel. We model the cell as a liquid droplet enclosed by a viscoelastic membrane and solve the cell deformation using a well-tested immersed-boundary lattice-Boltzmann method. The cell membrane damage, which is directly related to the membrane permeability, is considered using continuum damage mechanics. The transport of the diffusive solute into the cell is solved by a lattice-Boltzmann model. After validating the computational framework against several benchmark cases, we consider a cell flowing through a cross-slot microchannel, focusing on the effects of the flow strength, channel fluid viscosity and cell membrane viscosity on the membrane damage and enhanced intracellular transport. Interestingly, we find that under a comparable pressure drop across the device, for cells with low membrane viscosity, the inertial flow regime, which can be achieved by driving a low-viscosity liquid at a high speed, often leads to much larger membrane damage, compared with the high-viscosity low-speed viscous flow regime. However, the enhancement can be significantly reduced or even reversed by an increase of the cell membrane viscosity, which limits cell deformation, particularly in the inertial flow regime. Our computational framework and simulation results may guide the design and optimisation of microfluidic devices, which use cross-slot geometry to disrupt cell membranes to enhance intracellular delivery of solutes.
Collapse
Affiliation(s)
- Ruixin Lu
- School of Mechanical Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK.
| | - Peng Yu
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yi Sui
- School of Engineering and Materials Science, Queen Mary University of London, London E1 4NS, UK.
| |
Collapse
|
4
|
Kim H, Kim S, Lim H, Chung AJ. Expanding CAR-T cell immunotherapy horizons through microfluidics. LAB ON A CHIP 2024; 24:1088-1120. [PMID: 38174732 DOI: 10.1039/d3lc00622k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapies have revolutionized cancer treatment, particularly in hematological malignancies. However, their application to solid tumors is limited, and they face challenges in safety, scalability, and cost. To enhance current CAR-T cell therapies, the integration of microfluidic technologies, harnessing their inherent advantages, such as reduced sample consumption, simplicity in operation, cost-effectiveness, automation, and high scalability, has emerged as a powerful solution. This review provides a comprehensive overview of the step-by-step manufacturing process of CAR-T cells, identifies existing difficulties at each production stage, and discusses the successful implementation of microfluidics and related technologies in addressing these challenges. Furthermore, this review investigates the potential of microfluidics-based methodologies in advancing cell-based therapy across various applications, including solid tumors, next-generation CAR constructs, T-cell receptors, and the development of allogeneic "off-the-shelf" CAR products.
Collapse
Affiliation(s)
- Hyelee Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Suyeon Kim
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Hyunjung Lim
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
| | - Aram J Chung
- Department of Bioengineering, Korea University, 02841 Seoul, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, 02841 Seoul, Republic of Korea.
- School of Biomedical Engineering, Korea University, 02841 Seoul, Republic of Korea.
- MxT Biotech, 04785 Seoul, Republic of Korea
| |
Collapse
|
5
|
Teer L, Yaddanapudi K, Chen J. Biophysical Control of the Glioblastoma Immunosuppressive Microenvironment: Opportunities for Immunotherapy. Bioengineering (Basel) 2024; 11:93. [PMID: 38247970 PMCID: PMC10813491 DOI: 10.3390/bioengineering11010093] [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: 11/01/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
GBM is the most aggressive and common form of primary brain cancer with a dismal prognosis. Current GBM treatments have not improved patient survival, due to the propensity for tumor cell adaptation and immune evasion, leading to a persistent progression of the disease. In recent years, the tumor microenvironment (TME) has been identified as a critical regulator of these pro-tumorigenic changes, providing a complex array of biomolecular and biophysical signals that facilitate evasion strategies by modulating tumor cells, stromal cells, and immune populations. Efforts to unravel these complex TME interactions are necessary to improve GBM therapy. Immunotherapy is a promising treatment strategy that utilizes a patient's own immune system for tumor eradication and has exhibited exciting results in many cancer types; however, the highly immunosuppressive interactions between the immune cell populations and the GBM TME continue to present challenges. In order to elucidate these interactions, novel bioengineering models are being employed to decipher the mechanisms of immunologically "cold" GBMs. Additionally, these data are being leveraged to develop cell engineering strategies to bolster immunotherapy efficacy. This review presents an in-depth analysis of the biophysical interactions of the GBM TME and immune cell populations as well as the systems used to elucidate the underlying immunosuppressive mechanisms for improving current therapies.
Collapse
Affiliation(s)
- Landon Teer
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| | - Kavitha Yaddanapudi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
- Immuno-Oncology Program, Brown Cancer Center, Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Joseph Chen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| |
Collapse
|
6
|
Sevenler D, Toner M. High throughput intracellular delivery by viscoelastic mechanoporation. Nat Commun 2024; 15:115. [PMID: 38167490 PMCID: PMC10762167 DOI: 10.1038/s41467-023-44447-w] [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: 03/22/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Brief pulses of electric field (electroporation) and/or tensile stress (mechanoporation) have been used to reversibly permeabilize the plasma membrane of mammalian cells and deliver materials to the cytosol. However, electroporation can be harmful to cells, while efficient mechanoporation strategies have not been scalable due to the use of narrow constrictions or needles which are susceptible to clogging. Here we report a high throughput approach to mechanoporation in which the plasma membrane is stretched and reversibly permeabilized by viscoelastic fluid forces within a microfluidic chip without surface contact. Biomolecules are delivered directly to the cytosol within seconds at a throughput exceeding 250 million cells per minute. Viscoelastic mechanoporation is compatible with a variety of biomolecules including proteins, RNA, and CRISPR-Cas9 ribonucleoprotein complexes, as well as a range of cell types including HEK293T cells and primary T cells. Altogether, viscoelastic mechanoporation appears feasible for contact-free permeabilization and delivery of biomolecules to mammalian cells ex vivo.
Collapse
Affiliation(s)
- Derin Sevenler
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Shriners Children's, Boston, MA, 02114, USA.
| |
Collapse
|
7
|
Zhuang C, Gould JE, Enninful A, Shao S, Mak M. Biophysical and mechanobiological considerations for T-cell-based immunotherapy. Trends Pharmacol Sci 2023; 44:366-378. [PMID: 37172572 PMCID: PMC10188210 DOI: 10.1016/j.tips.2023.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/24/2023] [Accepted: 03/24/2023] [Indexed: 05/15/2023]
Abstract
Immunotherapies modulate the body's defense system to treat cancer. While these therapies have shown efficacy against multiple types of cancer, patient response rates are limited, and the off-target effects can be severe. Typical approaches in developing immunotherapies tend to focus on antigen targeting and molecular signaling, while overlooking biophysical and mechanobiological effects. Immune cells and tumor cells are both responsive to biophysical cues, which are prominent in the tumor microenvironment. Recent studies have shown that mechanosensing - including through Piezo1, adhesions, and Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) - influences tumor-immune interactions and immunotherapeutic efficacy. Furthermore, biophysical methods such as fluidic systems and mechanoactivation schemes can improve the controllability and manufacturing of engineered T cells, with potential for increasing therapeutic efficacy and specificity. This review focuses on leveraging advances in immune biophysics and mechanobiology toward improving chimeric antigen receptor (CAR) T-cell and anti-programmed cell death protein 1 (anti-PD-1) therapies.
Collapse
Affiliation(s)
- Chuzhi Zhuang
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Jared E Gould
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Archibald Enninful
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Stephanie Shao
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA.
| |
Collapse
|
8
|
Sevenler D, Toner M. High throughput intracellular delivery by viscoelastic mechanoporation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.24.538131. [PMID: 37163007 PMCID: PMC10168280 DOI: 10.1101/2023.04.24.538131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Brief and intense electric fields (electroporation) and/or tensile stresses (mechanoporation) have been used to temporarily permeabilize the plasma membrane of mammalian cells for the purpose of delivering materials to the cytosol. However, electroporation can be harmful to cells, while efficient mechanoporation strategies have not been scalable due to the use of narrow constrictions or needles which are susceptible to clogging. Here we report a method of mechanoporation in which cells were stretched and permeabilized by viscoelastic flow forces without surface contact. Inertio-elastic cell focusing aligned cells to the center of the device, avoiding direct contact with walls and enabling efficient (95%) intracellular delivery to over 200 million cells per minute. Functional biomolecules such as proteins, RNA, and ribonucleoprotein complexes were successfully delivered to Jurkat cells. Efficient intracellular delivery to HEK293T cells and primary activated T cells was also demonstrated. Contact-free mechanoporation using viscoelastic fluid forces appears to be feasible method for efficient and high throughput intracellular delivery of biomolecules to mammalian cells ex vivo.
Collapse
Affiliation(s)
- Derin Sevenler
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Shriners Hospitals for Children, Boston, MA, USA
| |
Collapse
|