1
|
Renzi S, Digiacomo L, Pozzi D, Quagliarini E, Vulpis E, Giuli MV, Mancusi A, Natiello B, Pignataro MG, Canettieri G, Di Magno L, Pesce L, De Lorenzi V, Ghignoli S, Loconte L, Montone CM, Laura Capriotti A, Laganà A, Nicoletti C, Amenitsch H, Rossi M, Mura F, Parisi G, Cardarelli F, Zingoni A, Checquolo S, Caracciolo G. Structuring lipid nanoparticles, DNA, and protein corona into stealth bionanoarchitectures for in vivo gene delivery. Nat Commun 2024; 15:9119. [PMID: 39438484 PMCID: PMC11496629 DOI: 10.1038/s41467-024-53569-8] [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: 12/27/2023] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Lipid nanoparticles (LNPs) play a crucial role in addressing genetic disorders, and cancer, and combating pandemics such as COVID-19 and its variants. Yet, the ability of LNPs to effectively encapsulate large-size DNA molecules remains elusive. This is a significant limitation, as the successful delivery of large-size DNA holds immense potential for gene therapy. To address this gap, the present study focuses on the design of PEGylated LNPs, incorporating large-sized DNA, departing from traditional RNA and ionizable lipids. The resultant LNPs demonstrate a unique particle morphology. These particles were further engineered with a DNA coating and plasma proteins. This multicomponent bionanoconstruct exhibits enhanced transfection efficiency and safety in controlled laboratory settings and improved immune system evasion in in vivo tests. These findings provide valuable insights for the design and development of bionanoarchitectures for large-size DNA delivery, opening new avenues for transformative gene therapies.
Collapse
Affiliation(s)
- Serena Renzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Erica Quagliarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Vulpis
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Valeria Giuli
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Latina, Italy
| | - Angelica Mancusi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Bianca Natiello
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Gemma Pignataro
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Laura Di Magno
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Pesce
- NEST, Scuola Normale Superiore, Pisa, Italy
| | | | | | - Luisa Loconte
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | - Aldo Laganà
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Carmine Nicoletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Graz, Austria
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | - Francesco Mura
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | - Giacomo Parisi
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | | | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Latina, Italy.
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
2
|
Giulimondi F, Digiacomo L, Renzi S, Cassone C, Pirrottina A, Molfetta R, Palamà IE, Maiorano G, Gigli G, Amenitsch H, Pozzi D, Zingoni A, Caracciolo G. Optimizing Transfection Efficiency in CAR-T Cell Manufacturing through Multiple Administrations of Lipid-Based Nanoparticles. ACS APPLIED BIO MATERIALS 2024; 7:3746-3757. [PMID: 38775109 DOI: 10.1021/acsabm.4c00103] [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] [Indexed: 06/18/2024]
Abstract
The existing manufacturing protocols for CAR-T cell therapies pose notable challenges, particularly in attaining a transient transfection that endures for a significant duration. To address this gap, this study aims to formulate a transfection protocol utilizing multiple lipid-based nanoparticles (LNPs) administrations to enhance transfection efficiency (TE) to clinically relevant levels. By systematically fine-tuning and optimizing our transfection protocol through a series of iterative refinements, we have accomplished a remarkable one-order-of-magnitude augmentation in TE within the immortalized T-lymphocyte Jurkat cell line. This enhancement has been consistently observed over 2 weeks, and importantly, it has been achieved without any detrimental impact on cell viability. In the subsequent phase of our study, we aimed to optimize the gene delivery system by evaluating three lipid-based formulations tailored for DNA encapsulation using our refined protocol. These formulations encompassed two LNPs constructed from ionizable lipids and featuring systematic variations in lipid composition (iLNPs) and a cationic lipoplex (cLNP). Our findings showcased a notable standout among the three formulations, with cLNP emerging as a frontrunner for further refinement and integration into the production pipeline of CAR-T therapies. Consequently, cLNP was scrutinized for its potential to deliver CAR-encoding plasmid DNA to the HEK-293 cell line. Confocal microscopy experiments demonstrated its efficiency, revealing substantial internalization compared to iLNPs. By employing a recently developed confocal image analysis method, we substantiated that cellular entry of cLNP predominantly occurs through macropinocytosis. This mechanism leads to heightened intracellular endosomal escape and mitigates lysosomal accumulation. The successful expression of anti-CD19-CD28-CD3z, a CAR engineered to target CD19, a protein often expressed on the surface of B cells, was confirmed using a fluorescence-based assay. Overall, our results indicated the effectiveness of cLNP in gene delivery and suggested the potential of multiple administration transfection as a practical approach for refining T-cell engineering protocols in CAR therapies. Future investigations may focus on refining outcomes by adjusting transfection parameters like nucleic acid concentration, lipid-to-DNA ratio, and incubation time to achieve improved TE and increased gene expression levels.
Collapse
Affiliation(s)
- Francesca Giulimondi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Serena Renzi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Chiara Cassone
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Andrea Pirrottina
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | | | - Gabriele Maiorano
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, Lecce 73100, Italy
| | - Giuseppe Gigli
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, Lecce 73100, Italy
- Department of Medicine, University of Salento, Arnesano street c/o Campus Ecotekne, Lecce 73100, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Graz 8010, Austria
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, V.le Regina Elena 291, Rome 00161, Italy
| |
Collapse
|
3
|
Kettmayer C, Gratton E, Estrada LC. Comparison of MSD analysis from single particle tracking with MSD from images. Getting the best of both worlds. Methods Appl Fluoresc 2023; 12:015001. [PMID: 37751748 DOI: 10.1088/2050-6120/acfd7e] [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: 05/18/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Fluorescence microscopy can provide valuable information about cell interior dynamics. Particularly, mean squared displacement (MSD) analysis is widely used to characterize proteins and sub-cellular structures' mobility providing the laws of molecular diffusion. The MSD curve is traditionally extracted from individual trajectories recorded by single-particle tracking-based techniques. More recently, image correlation methods like iMSD have been shown capable of providing averaged dynamic information directly from images, without the need for isolation and localization of individual particles. iMSD is a powerful technique that has been successfully applied to many different biological problems, over a wide spatial and temporal scales. The aim of this work is to review and compare these two well-established methodologies and their performance in different situations, to give an insight on how to make the most out of their unique characteristics. We show the analysis of the same datasets by the two methods. Regardless of the experimental differences in the input data for MSD or iMSD analysis, our results show that the two approaches can address equivalent questions for free diffusing systems. We focused on studying a range of diffusion coefficients between D = 0.001μm2s-1and D = 0.1μm2s-1, where we verified that the equivalence is maintained even for the case of isolated particles. This opens new opportunities for studying intracellular dynamics using equipment commonly available in any biophysical laboratory.
Collapse
Affiliation(s)
- Constanza Kettmayer
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física. Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Física de Buenos Aires (IFIBA). Buenos Aires, Argentina
| | - Enrico Gratton
- Laboratory for Fluorescence Dynamics, Department of Biomedical Engineering, University of California, Irvine, CA, United States of America
| | - Laura C Estrada
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Física. Buenos Aires, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Física de Buenos Aires (IFIBA). Buenos Aires, Argentina
| |
Collapse
|
4
|
Digiacomo L, Renzi S, Quagliarini E, Pozzi D, Amenitsch H, Ferri G, Pesce L, De Lorenzi V, Matteoli G, Cardarelli F, Caracciolo G. Investigating the mechanism of action of DNA-loaded PEGylated lipid nanoparticles. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102697. [PMID: 37507061 DOI: 10.1016/j.nano.2023.102697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
PEGylated lipid nanoparticles (LNPs) are commonly used to deliver bioactive molecules, but the role of PEGylation in DNA-loaded LNP interactions at the cellular and subcellular levels remains poorly understood. In this study, we investigated the mechanism of action of DNA-loaded PEGylated LNPs using gene reporter technologies, dynamic light scattering (DLS), synchrotron small angle X-ray scattering (SAXS), and fluorescence confocal microscopy (FCS). We found that PEG has no significant impact on the size or nanostructure of DNA LNPs but reduces their zeta potential and interaction with anionic cell membranes. PEGylation increases the structural stability of LNPs and results in lower DNA unloading. FCS experiments revealed that PEGylated LNPs are internalized intact inside cells and largely shuttled to lysosomes, while unPEGylated LNPs undergo massive destabilization on the plasma membrane. These findings can inform the design, optimization, and validation of DNA-loaded LNPs for gene delivery and vaccine development.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Serena Renzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, 8010 Graz, Austria
| | - Gianmarco Ferri
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | - Luca Pesce
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | | | - Giulia Matteoli
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | | | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
5
|
McClure JE, Li Z. Capturing membrane structure and function in lattice Boltzmann models. Phys Rev E 2023; 107:024408. [PMID: 36932594 DOI: 10.1103/physreve.107.024408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
We develop a mesoscopic approach to model the nonequilibrium behavior of membranes at the cellular scale. Relying on lattice Boltzmann methods, we develop a solution procedure to recover the Nernst-Planck equations and Gauss's law. A general closure rule is developed to describe mass transport across the membrane, which is able to account for protein-mediated diffusion based on a coarse-grained representation. We demonstrate that our model is able to recover the Goldman equation from first principles and show that hyperpolarization occurs when membrane charging dynamics are controlled by multiple relaxation timescales. The approach provides a promising way to characterize non-equilibrium behaviors that arise due to the role of membranes in mediating transport based on realistic three-dimensional cell geometries.
Collapse
Affiliation(s)
- James E McClure
- National Security Institute and Center for Soft Matter and Biological Physics Virginia Polytechnic and State University and Blacksburg, Virginia, 24060, USA
| | - Zhe Li
- Research School of Physics, The Australian National University, Canberra, 2601, Australia
| |
Collapse
|
6
|
Cui L, Renzi S, Quagliarini E, Digiacomo L, Amenitsch H, Masuelli L, Bei R, Ferri G, Cardarelli F, Wang J, Amici A, Pozzi D, Marchini C, Caracciolo G. Efficient Delivery of DNA Using Lipid Nanoparticles. Pharmaceutics 2022; 14:1698. [PMID: 36015328 PMCID: PMC9416266 DOI: 10.3390/pharmaceutics14081698] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/24/2022] [Accepted: 07/28/2022] [Indexed: 11/22/2022] Open
Abstract
DNA vaccination has been extensively studied as a promising strategy for tumor treatment. Despite the efforts, the therapeutic efficacy of DNA vaccines has been limited by their intrinsic poor cellular internalization. Electroporation, which is based on the application of a controlled electric field to enhance DNA penetration into cells, has been the method of choice to produce acceptable levels of gene transfer in vivo. However, this method may cause cell damage or rupture, non-specific targeting, and even degradation of pDNA. Skin irritation, muscle contractions, pain, alterations in skin structure, and irreversible cell damage have been frequently reported. To overcome these limitations, in this work, we use a microfluidic platform to generate DNA-loaded lipid nanoparticles (LNPs) which are then characterized by a combination of dynamic light scattering (DLS), synchrotron small-angle X-ray scattering (SAXS), and transmission electron microscopy (TEM). Despite the clinical successes obtained by LNPs for mRNA and siRNA delivery, little is known about LNPs encapsulating bulkier DNA molecules, the clinical application of which remains challenging. For in vitro screening, LNPs were administered to human embryonic kidney 293 (HEK-293) and Chinese hamster ovary (CHO) cell lines and ranked for their transfection efficiency (TE) and cytotoxicity. The LNP formulation exhibiting the highest TE and the lowest cytotoxicity was then tested for the delivery of the DNA vaccine pVAX-hECTM targeting the human neoantigen HER2, an oncoprotein overexpressed in several cancer types. Using fluorescence-activated cell sorting (FACS), immunofluorescence assays and fluorescence confocal microscopy (FCS), we proved that pVAX-hECTM-loaded LNPs produce massive expression of the HER2 antigen on the cell membrane of HEK-293 cells. Our results provide new insights into the structure-activity relationship of DNA-loaded LNPs and pave the way for the access of this gene delivery technology to preclinical studies.
Collapse
Affiliation(s)
- Lishan Cui
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Serena Renzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, 8010 Graz, Austria
| | - Laura Masuelli
- Department of Experimental Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
| | - Gianmarco Ferri
- National Enterprise for NanoScience and NanoTechnology (NEST), Scuola Normale Superiore, 56127 Pisa, Italy
| | - Francesco Cardarelli
- National Enterprise for NanoScience and NanoTechnology (NEST), Scuola Normale Superiore, 56127 Pisa, Italy
| | - Junbiao Wang
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Augusto Amici
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Cristina Marchini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| |
Collapse
|
7
|
Ferri G, Pesce L, Tesi M, Marchetti P, Cardarelli F. β-Cell Pathophysiology: A Review of Advanced Optical Microscopy Applications. Int J Mol Sci 2021; 22:ijms222312820. [PMID: 34884624 PMCID: PMC8657725 DOI: 10.3390/ijms222312820] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 11/30/2022] Open
Abstract
β-cells convert glucose (input) resulting in the controlled release of insulin (output), which in turn has the role to maintain glucose homeostasis. β-cell function is regulated by a complex interplay between the metabolic processing of the input, its transformation into second-messenger signals, and final mobilization of insulin-containing granules towards secretion of the output. Failure at any level in this process marks β-cell dysfunction in diabetes, thus making β-cells obvious potential targets for therapeutic purposes. Addressing quantitatively β-cell (dys)function at the molecular level in living samples requires probing simultaneously the spatial and temporal dimensions at the proper resolution. To this aim, an increasing amount of research efforts are exploiting the potentiality of biophysical techniques. In particular, using excitation light in the visible/infrared range, a number of optical-microscopy-based approaches have been tailored to the study of β-cell-(dys)function at the molecular level, either in label-free mode (i.e., exploiting intrinsic autofluorescence of cells) or by the use of organic/genetically-encoded fluorescent probes. Here, relevant examples from the literature are reviewed and discussed. Based on this, new potential lines of development in the field are drawn.
Collapse
Affiliation(s)
- Gianmarco Ferri
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
| | - Luca Pesce
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
| | - Marta Tesi
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, 56127 Pisa, Italy; (M.T.); (P.M.)
| | - Piero Marchetti
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, 56127 Pisa, Italy; (M.T.); (P.M.)
| | - Francesco Cardarelli
- NEST Laboratory, Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
- Correspondence:
| |
Collapse
|
8
|
Ferri G, Tesi M, Pesce L, Bugliani M, Grano F, Occhipinti M, Suleiman M, De Luca C, Marselli L, Marchetti P, Cardarelli F. Spatiotemporal Correlation Spectroscopy Reveals a Protective Effect of Peptide-Based GLP-1 Receptor Agonism against Lipotoxicity on Insulin Granule Dynamics in Primary Human β-Cells. Pharmaceutics 2021; 13:pharmaceutics13091403. [PMID: 34575477 PMCID: PMC8464798 DOI: 10.3390/pharmaceutics13091403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/18/2021] [Accepted: 09/02/2021] [Indexed: 12/25/2022] Open
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are being used for the treatment of type 2 diabetes (T2D) and may have beneficial effects on the pancreatic β-cells. Here, we evaluated the effects of GLP-1R agonism on insulin secretory granule (ISG) dynamics in primary β-cells isolated from human islets exposed to palmitate-induced lipotoxic stress. Islets cells were exposed for 48 h to 0.5 mM palmitate (hereafter, ‘Palm’) with or without the addition of a GLP-1 agonist, namely 10 nM exendin-4 (hereafter, ‘Ex-4’). Dissociated cells were first transfected with syncollin-EGFP in order to fluorescently mark the ISGs. Then, by applying a recently established spatiotemporal correlation spectroscopy technique, the average structural (i.e., size) and dynamic (i.e., the local diffusivity and mode of motion) properties of ISGs are extracted from a calculated imaging-derived Mean Square Displacement (iMSD) trace. Besides defining the structural/dynamic fingerprint of ISGs in human cells for the first time, iMSD analysis allowed to probe fingerprint variations under selected conditions: namely, it was shown that Palm affects ISGs dynamics in response to acute glucose stimulation by abolishing the ISGs mobilization typically imparted by glucose and, concomitantly, by reducing the extent of ISGs active/directed intracellular movement. By contrast, co-treatment with Ex-4 normalizes ISG dynamics, i.e., re-establish ISG mobilization and ability to perform active transport in response to glucose stimulation. These observations were correlated with standard glucose-stimulated insulin secretion (GSIS), which resulted in being reduced in cells exposed to Palm but preserved in cells concomitantly exposed to 10 nM Ex-4. Our data support the idea that GLP-1R agonism may exert its beneficial effect on human β-cells under metabolic stress by maintaining ISGs’ proper intracellular dynamics.
Collapse
Affiliation(s)
- Gianmarco Ferri
- Laboratorio NEST-Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
| | - Marta Tesi
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Luca Pesce
- Laboratorio NEST-Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Francesca Grano
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Margherita Occhipinti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Carmela De Luca
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56127 Pisa, Italy; (M.T.); (M.B.); (F.G.); (M.O.); (M.S.); (C.D.L.); (L.M.); (P.M.)
| | - Francesco Cardarelli
- Laboratorio NEST-Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy; (G.F.); (L.P.)
- Correspondence:
| |
Collapse
|
9
|
Pham TD. Fuzzy Recurrence Exponents of Subcellular-Nanostructure Dynamics in Time-lapse Confocal Imaging. IEEE Trans Nanobioscience 2021; 20:497-506. [PMID: 34398761 DOI: 10.1109/tnb.2021.3105533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Studying the dynamics of nanostructures in the intracellular space is important because it allows gaining insights into the mechanism of complex biological functions of organelles. Understanding such dynamical processes can contribute to the development of nanomedicine for the diagnosis and treatment of many diseases caused by the interaction of multiple genes and environmental factors. Here a quantitative measure of spatial-temporal dynamics of nanostructures within a cell line in the context of nonlinear dynamics is introduced, where early endosomes, late endosomes, and lysosomes recorded by time-lapse confocal imaging are examined. The mathematical derivation of the proposed technique is based on the concept of recurrence dynamics and sequential rate of change over time. The quantification introduced as fuzzy recurrence exponents can be generalized for characterizing the dynamics of experimental evolutions in other nanostructures of living cells captured under the optical microscope.
Collapse
|
10
|
Optimizing the Efficiency of a Cytocompatible Carbon-Dots-Based FRET Platform and Its Application as a Riboflavin Sensor in Beverages. NANOMATERIALS 2021; 11:nano11081981. [PMID: 34443812 PMCID: PMC8399497 DOI: 10.3390/nano11081981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/19/2022]
Abstract
In this work, the Förster resonance energy transfer (FRET) between carbon dots (CDs) as energy donors and riboflavin (RF) as an energy acceptor was optimized and the main parameters that characterize the FRET process were determined. The results were successfully applied in the development of an ultrasensitive ratiometric fluorescent sensor for the selective and sensitive determination of RF in different beverages. Water-soluble CDs with a high quantum yield (54%) were synthesized by a facile and direct microwave-assisted technique. The CDs were characterized by transmission electron microscopy (TEM), Fourier transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), Zeta potential, and UV-visible and molecular fluorescence spectroscopy. The study of the FRET process at two donor concentrations showed that the energy transfer efficiency decreases as the donor concentration increases, confirming its dependence on the acceptor:donor ratio in nanoparticle-based systems. The results show the importance of optimizing the FRET process conditions to improve the corresponding output signal. The variation in the ratiometric signal with the concentration of RF showed linearity in a concentration range of 0 to 11 µM with R2 = 0.9973 and a detection limit of 0.025 µM. The developed nanosensor showed good selectivity over other possible types of interference. The sensor was then applied for the determination of RF in beverage samples using the standard addition method with recoveries between 96% and 106%. Preliminary cytocompatibility tests carried out with breast cancer cells (MDA-MB-231) revealed the nanosensor to be cytocompatible in its working concentration regime, even after long incubation times with cells. Altogether, the developed RF determination method was found to be fast, low-cost, highly sensitive, and selective and can be extended to other samples of interest in the biological and food sectors. Moreover, thanks to its long-lasting cytocompatibility, the developed platform can also be envisaged for other applications of biological interest, such as intracellular sensing and staining for live cell microscopy.
Collapse
|
11
|
Ferri G, Tesi M, Massarelli F, Marselli L, Marchetti P, Cardarelli F. Metabolic response of Insulinoma 1E cells to glucose stimulation studied by fluorescence lifetime imaging. FASEB Bioadv 2020; 2:409-418. [PMID: 32676581 PMCID: PMC7354695 DOI: 10.1096/fba.2020-00014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/01/2020] [Accepted: 05/21/2020] [Indexed: 01/02/2023] Open
Abstract
A cascade of highly regulated biochemical processes connects glucose stimulation to insulin secretion in specialized cells of mammalian pancreas, the β-cells. Given the importance of this process for systemic glucose homeostasis, noninvasive and fast strategies capable to monitor the response to glucose in living cells are highly desirable. Here, we use the phasor-based approach to Fluorescence Lifetime IMaging (FLIM) microscopy to quantify the ratio between protein-bound and free Nicotinamide adenine dinucleotide (phosphate) species in their reduced form (NAD(P)H), and the Insulinoma cell line INS-1E as a β-like cellular model. Phasor-FLIM analysis shows that the bound/free ratio of NAD(P)H species increases upon pulsed glucose stimulation. Such response is impaired by 48-hours preincubation of cells under hyperglycemic conditions. Phasor-FLIM concomitantly monitors the appearance of long-lifetime species (LLS) as characteristic products of hyperglycemia-induced oxidative stress.
Collapse
Affiliation(s)
| | - Marta Tesi
- Department of Clinical and Experimental MedicineIslet Cell LaboratoryUniversity of PisaPisaItaly
| | | | - Lorella Marselli
- Department of Clinical and Experimental MedicineIslet Cell LaboratoryUniversity of PisaPisaItaly
| | - Piero Marchetti
- Department of Clinical and Experimental MedicineIslet Cell LaboratoryUniversity of PisaPisaItaly
| | | |
Collapse
|
12
|
Durso W, Martins M, Marchetti L, Cremisi F, Luin S, Cardarelli F. Lysosome Dynamic Properties during Neuronal Stem Cell Differentiation Studied by Spatiotemporal Fluctuation Spectroscopy and Organelle Tracking. Int J Mol Sci 2020; 21:ijms21093397. [PMID: 32403391 PMCID: PMC7247004 DOI: 10.3390/ijms21093397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 01/20/2023] Open
Abstract
We investigated lysosome dynamics during neuronal stem cell (NSC) differentiation by two quantitative and complementary biophysical methods based on fluorescence: imaging-derived mean square displacement (iMSD) and single-particle tracking (SPT). The former extracts the average dynamics and size of the whole population of moving lysosomes directly from imaging, with no need to calculate single trajectories; the latter resolves the finest heterogeneities and dynamic features at the single-lysosome level, which are lost in the iMSD analysis. In brief, iMSD analysis reveals that, from a structural point of view, lysosomes decrement in size during NSC differentiation, from 1 μm average diameter in the embryonic cells to approximately 500 nm diameter in the fully differentiated cells. Concomitantly, iMSD analysis highlights modification of key dynamic parameters, such as the average local organelle diffusivity and anomalous coefficient, which may parallel cytoskeleton remodeling during the differentiation process. From average to local, SPT allows mapping heterogeneous dynamic responses of single lysosomes in different districts of the cells. For instance, a dramatic decrease of lysosomal transport in the soma is followed by a rapid increase of transport in the projections at specific time points during neuronal differentiation, an observation compatible with the hypothesis that lysosomal active mobilization shifts from the soma to the newborn projections. Our combined results provide new insight into the lysosome size and dynamics regulation throughout NSC differentiation, supporting new functions proposed for this organelle.
Collapse
Affiliation(s)
- William Durso
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
| | - Manuella Martins
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Laura Marchetti
- Center for Nanotechnology Innovation@NEST (CNI@NEST), Piazza San Silvestro 12, 56126 Pisa, Italy;
| | - Federico Cremisi
- Bio@SNS Laboratory—Scuola Normale Superiore, via G. Moruzzi, 1, 56126 Pisa, Italy; (M.M.); (F.C.)
| | - Stefano Luin
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- NEST, Istituto Nanoscienze, CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
- Correspondence: (S.L.); (F.C.)
| | - Francesco Cardarelli
- NEST Laboratory—Scuola Normale Superiore, Piazza San Silvestro 12, 56127 Pisa, Italy;
- Correspondence: (S.L.); (F.C.)
| |
Collapse
|
13
|
A mechanistic explanation of the inhibitory role of the protein corona on liposomal gene expression. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183159. [PMID: 31857070 DOI: 10.1016/j.bbamem.2019.183159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022]
Abstract
The past three decades have witnessed fast advances in the use of cationic liposome-DNA complexes (lipoplexes) for gene delivery applications. However, no lipoplex formulation has reached into the clinical practice so far. The primary drawback limiting clinical use of lipoplexes is the lack of mechanistic understanding of their low transfection efficiency (TE) in vivo. In physiological environments, lipoplexes are coated by a protein corona (PC) that mediates the interactions with the cell machinery. Here we show that the formation of PC can change the interactions of multicomponent (MC) lipoplexes with our cell model (i.e., HeLa). At the highest lipoplex concentration, the formation of PC can reduce the TE of MC lipoplexes from 60% to <5%. Combining dynamic light scattering and synchrotron small-angle X-ray scattering (SAXS), we clarify that the formation of PC modifies physical-chemical properties of MC lipoplexes so as to affect their TE. Moreover, we examined single transfection barriers by a combination of fluorescence-activated cell sorting, single-cell real-time fluorescence confocal microscopy, and synchrotron SAXS. We demonstrate that PC formation has the ability to modify the relative contribution of caveolae-mediated endocytosis and macropinocytosis in lipoplexes uptake, in favor of the latter, increasing accumulation of PC-decorated lipoplexes into degradative lysosomal compartments. Finally, we report evidences that PC reduces the structural stability of lipoplexes against solubilization by cellular lipids, likely favoring premature DNA release and cytosolic digestion by DNAase. These combined effects revealed here offer a comprehensive mechanistic explanation on the reason behind reduction in gene expression of MC lipoplexes.
Collapse
|
14
|
Ferri G, Digiacomo L, Lavagnino Z, Occhipinti M, Bugliani M, Cappello V, Caracciolo G, Marchetti P, Piston DW, Cardarelli F. Insulin secretory granules labelled with phogrin-fluorescent proteins show alterations in size, mobility and responsiveness to glucose stimulation in living β-cells. Sci Rep 2019; 9:2890. [PMID: 30814595 PMCID: PMC6393586 DOI: 10.1038/s41598-019-39329-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/15/2019] [Indexed: 11/13/2022] Open
Abstract
The intracellular life of insulin secretory granules (ISGs) from biogenesis to secretion depends on their structural (e.g. size) and dynamic (e.g. diffusivity, mode of motion) properties. Thus, it would be useful to have rapid and robust measurements of such parameters in living β-cells. To provide such measurements, we have developed a fast spatiotemporal fluctuation spectroscopy. We calculate an imaging-derived Mean Squared Displacement (iMSD), which simultaneously provides the size, average diffusivity, and anomalous coefficient of ISGs, without the need to extract individual trajectories. Clustering of structural and dynamic quantities in a multidimensional parametric space defines the ISGs' properties for different conditions. First, we create a reference using INS-1E cells expressing proinsulin fused to a fluorescent protein (FP) under basal culture conditions and validate our analysis by testing well-established stimuli, such as glucose intake, cytoskeleton disruption, or cholesterol overload. After, we investigate the effect of FP-tagged ISG protein markers on the structural and dynamic properties of the granule. While iMSD analysis produces similar results for most of the lumenal markers, the transmembrane marker phogrin-FP shows a clearly altered result. Phogrin overexpression induces a substantial granule enlargement and higher mobility, together with a partial de-polymerization of the actin cytoskeleton, and reduced cell responsiveness to glucose stimulation. Our data suggest a more careful interpretation of many previous ISG-based reports in living β-cells. The presented data pave the way to high-throughput cell-based screening of ISG structure and dynamics under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Gianmarco Ferri
- NEST - Scuola Normale Superiore, Istituto Nanoscienze - CNR (CNR-NANO), Pisa, Italy
- Nanoscopy, Nanophysics, Istituto Italiano di Tecnologia, via Morego 30, 16163, Genoa, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, "La Sapienza" University of Rome, Rome, Italy
| | - Zeno Lavagnino
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Margherita Occhipinti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | | | - Giulio Caracciolo
- Department of Molecular Medicine, "La Sapienza" University of Rome, Rome, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Francesco Cardarelli
- NEST - Scuola Normale Superiore, Istituto Nanoscienze - CNR (CNR-NANO), Pisa, Italy.
| |
Collapse
|
15
|
Durso W, D'Autilia F, Amodeo R, Marchetti L, Cardarelli F. Probing labeling-induced lysosome alterations in living cells by imaging-derived mean squared displacement analysis. Biochem Biophys Res Commun 2018; 503:2704-2709. [PMID: 30100062 DOI: 10.1016/j.bbrc.2018.08.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 08/03/2018] [Indexed: 10/28/2022]
Abstract
Lysosomes are not merely degradative organelles but play a central role in nutrient sensing, metabolism and cell-growth regulation. Our ability to study their function in living cells strictly relies on the use of lysosome-specific fluorescent probes tailored to optical microscopy applications. Still, no report thus far quantitatively analyzed the effect of labeling strategies/procedures on lysosome properties in live cells. We tackle this issue by a recently developed spatiotemporal fluctuation spectroscopy strategy that extracts structural (size) and dynamic (diffusion) properties directly from imaging, with no a-priori knowledge of the system. We highlight hitherto neglected alterations of lysosome properties upon labeling. In particular, we demonstrate that Lipofectamine reagents, used to transiently express lysosome markers fused to fluorescent proteins (FPs) (e.g. LAMP1-FP or CD63-FP), irreversibly alter the organelle structural identity, inducing a ∼2-fold increase of lysosome average size. The organelle structural identity is preserved, instead, if electroporation or Effectene are used as transfection strategies, provided that the expression levels of the recombinant protein marker are kept low. This latter condition can be achieved also by generating cell lines stably expressing the desired FP-tagged marker. Reported results call into question the interpretation of a massive amount of data collected so far using fluorescent protein markers and suggest useful guidelines for future studies.
Collapse
Affiliation(s)
- William Durso
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Pisa, Italy; NEST - Scuola Normale Superiore, Istituto Nanoscienze - CNR (CNR-NANO), Pisa, Italy
| | - Francesca D'Autilia
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Pisa, Italy
| | - Rosy Amodeo
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Pisa, Italy; NEST - Scuola Normale Superiore, Istituto Nanoscienze - CNR (CNR-NANO), Pisa, Italy
| | - Laura Marchetti
- Center for Nanotechnology Innovation@NEST, Istituto Italiano di Tecnologia, Pisa, Italy
| | - Francesco Cardarelli
- NEST - Scuola Normale Superiore, Istituto Nanoscienze - CNR (CNR-NANO), Pisa, Italy.
| |
Collapse
|