1
|
Papini G, Furini G, Matteucci M, Biemmi V, Casieri V, Di Lascio N, Milano G, Chincoli LR, Faita F, Barile L, Lionetti V. Cardiomyocyte-targeting exosomes from sulforaphane-treated fibroblasts affords cardioprotection in infarcted rats. J Transl Med 2023; 21:313. [PMID: 37161563 PMCID: PMC10169450 DOI: 10.1186/s12967-023-04155-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/25/2023] [Indexed: 05/11/2023] Open
Abstract
BACKGROUND Exosomes (EXOs), tiny extracellular vesicles that facilitate cell-cell communication, are being explored as a heart failure treatment, although the features of the cell source restrict their efficacy. Fibroblasts the most prevalent non-myocyte heart cells, release poor cardioprotective EXOs. A noninvasive method for manufacturing fibroblast-derived exosomes (F-EXOs) that target cardiomyocytes and slow cardiac remodeling is expected. As a cardioprotective isothiocyanate, sulforaphane (SFN)-induced F-EXOs (SFN-F-EXOs) should recapitulate its anti-remodeling properties. METHODS Exosomes from low-dose SFN (3 μM/7 days)-treated NIH/3T3 murine cells were examined for number, size, and protein composition. Fluorescence microscopy, RT-qPCR, and western blot assessed cell size, oxidative stress, AcH4 levels, hypertrophic gene expression, and caspase-3 activation in angiotensin II (AngII)-stressed HL-1 murine cardiomyocytes 12 h-treated with various EXOs. The uptake of fluorescently-labeled EXOs was also measured in cardiomyocytes. The cardiac function of infarcted male Wistar rats intramyocardially injected with different EXOs (1·1012) was examined by echocardiography. Left ventricular infarct size, hypertrophy, and capillary density were measured. RESULTS Sustained treatment of NIH/3T3 with non-toxic SFN concentration significantly enhances the release of CD81 + EXOs rich in TSG101 (Tumor susceptibility gene 101) and Hsp70 (Heat Shock Protein 70), and containing maspin, an endogenous histone deacetylase 1 inhibitor. SFN-F-EXOs counteract angiotensin II (AngII)-induced hypertrophy and apoptosis in murine HL-1 cardiomyocytes enhancing SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a) levels more effectively than F-EXOs. In stressed cardiomyocytes, SFN-F-EXOs boost AcH4 levels by 30% (p < 0.05) and significantly reduce oxidative stress more than F-EXOs. Fluorescence microscopy showed that mouse cardiomyocytes take in SFN-F-EXOs ~ threefold more than F-EXOs. Compared to vehicle-injected infarcted hearts, SFN-F-EXOs reduce hypertrophy, scar size, and improve contractility. CONCLUSIONS Long-term low-dose SFN treatment of fibroblasts enhances the release of anti-remodeling cardiomyocyte-targeted F-EXOs, which effectively prevent the onset of HF. The proposed method opens a new avenue for large-scale production of cardioprotective exosomes for clinical application using allogeneic fibroblasts.
Collapse
Affiliation(s)
- Gaia Papini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giulia Furini
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy
| | - Marco Matteucci
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Vanessa Biemmi
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Valentina Casieri
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Nicole Di Lascio
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
| | - Giuseppina Milano
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lucia Rosa Chincoli
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Lucio Barile
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Svizzera Italiana, 6900, Lugano, Switzerland
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, The Interdisciplinary Research Center "Health Science", Scuola Superiore Sant'Anna, Via G. Moruzzi, 1, 56124, Pisa, Italy.
- Anesthesiology and Intensive Care Medicine, UOSVD, Fondazione Toscana G. Monasterio, Pisa, Italy.
| |
Collapse
|
2
|
Sola F, Canonico B, Montanari M, Volpe A, Barattini C, Pellegrino C, Cesarini E, Guescini M, Battistelli M, Ortolani C, Ventola A, Papa S. Uptake and Intracellular Trafficking Studies of Multiple Dye-Doped Core-Shell Silica Nanoparticles in Lymphoid and Myeloid Cells. Nanotechnol Sci Appl 2021; 14:29-48. [PMID: 33727804 PMCID: PMC7954439 DOI: 10.2147/nsa.s290867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/22/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction Since most biologically active macromolecules are natural nanostructures, operating in the same scale of biomolecules gives the great advantage to enhance the interaction with cellular components. Noteworthy efforts in nanotechnology, particularly in biomedical and pharmaceutical fields, have propelled a high number of studies on the biological effects of nanomaterials. Moreover, the determination of specific physicochemical properties of nanomaterials is crucial for the evaluation and design of novel safe and efficient therapeutics and diagnostic tools. In this in vitro study, we report a physicochemical characterisation of fluorescent silica nanoparticles (NPs), interacting with biological models (U937 and PBMC cells), describing the specific triggered biologic response. Methods Flow Cytometric and Confocal analyses are the main method platforms. However TEM, NTA, DLS, and chemical procedures to synthesize NPs were employed. Results NTB700 NPs, employed in this study, are fluorescent core-shell silica nanoparticles, synthesized through a micelle-assisted method, where the fluorescence energy transfer process, known as FRET, occurs at a high efficiency rate. Using flow cytometry and confocal microscopy, we observed that NTB700 NP uptake seemed to be a rapid, concentration-, energy- and cell type-dependent process, which did not induce significant cytotoxic effects. We did not observe a preferred route of internalization, although their size and the possible aggregated state could influence their extrusion. At this level of analysis, our investigation focuses on lysosome and mitochondria pathways, highlighting that both are involved in NP co-localization. Despite the main mitochondria localization, NPs did not induce a significant increase of intracellular ROS, known inductors of apoptosis, during the time course of analyses. Finally, both lymphoid and myeloid cells are able to release NPs, essential to their biosafety. Discussion These data allow to consider NTB700 NPs a promising platform for future development of a multifunctional system, by combining imaging and localized therapeutic applications in a unique tool.
Collapse
Affiliation(s)
- Federica Sola
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy.,AcZon Srl, Monte San Pietro, BO, 40050, Italy
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Mariele Montanari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | | | - Chiara Barattini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy.,AcZon Srl, Monte San Pietro, BO, 40050, Italy
| | | | - Erica Cesarini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Michele Guescini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Michela Battistelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | - Claudio Ortolani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| | | | - Stefano Papa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
| |
Collapse
|
3
|
Xu D, Wan Y, Li Z, Wang C, Zou Q, Du C, Wang Y. Tailorable hierarchical structures of biomimetic hydroxyapatite micro/nano particles promoting endocytosis and osteogenic differentiation of stem cells. Biomater Sci 2020; 8:3286-3300. [PMID: 32490486 DOI: 10.1039/d0bm00443j] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hydroxyapatite (HA) micro/nano particles show great promise as artificial bone and dental substitutes, or drug carrier systems. However, the precise regulation of hydroxyapatite micro/nano particles with controllable physicochemical properties (such as hierarchical structure, particle size, potential and crystallinity) is still a challenge. Furthermore, the effects of different hierarchical structures on biological responses have been rarely reported. Herein, the HA particles with a precisely tailored micro/nano hierarchical structure have been developed using an elaborate biomimetic synthesis technology. Three representative particles, namely, micro/nano needle-like HA particles, micro/nano rod-like HA particles, and micro/nano flake-like HA particles, were featured to evaluate their biological responses to stem cells. The pore structure facilitated the adsorption of serum adhesive proteins, which together with the unique hierarchical architecture of micro/nano flake-like HA particles remarkably promoted the endocytosis efficiency in a concentration-dependent manner. The qRT-PCR together with RNA-seq and western blot analyses showed that micro/nano flake-like HA particles more significantly up-regulated the expression of genes and production of proteins related to osteogenic differentiation among the three particles through the activated ERK/MAPK signalling pathway. RNA-seq further revealed a complex mechanism of cell interface events, suggesting that the hierarchical architecture of HA particles is of crucial importance for the regulation of actin cytoskeleton involved in the modulation of cell adhesion which positively stimulated osteogenic differentiation of stem cells. Moreover, the endocytosis of particles into lysosomes resulted in an increase in the intracellular Ca2+ levels, which activated possible intracellular Ca2+-mediated signaling cascades (Ras/cAMP/Rap1/MAPK signaling pathways) related to osteogenic differentiation of stem cells. Our findings shed light on the effects of different hierarchical structures of HA particles on stem cell differentiation and contribute to the optimal design of implant materials.
Collapse
Affiliation(s)
- Dong Xu
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou 510641, PR China.
| | | | | | | | | | | | | |
Collapse
|
4
|
Varadaraj A, Jenkins LM, Singh P, Chanda A, Snider J, Lee NY, Amsalem-Zafran AR, Ehrlich M, Henis YI, Mythreye K. TGF-β triggers rapid fibrillogenesis via a novel TβRII-dependent fibronectin-trafficking mechanism. Mol Biol Cell 2017; 28:1195-1207. [PMID: 28298487 PMCID: PMC5415016 DOI: 10.1091/mbc.e16-08-0601] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 02/02/2023] Open
Abstract
There is increased recycling of soluble fibronectin from the cell surface for fibrillogenesis. This recycling is regulated by TGF-β in a transcription- and SMAD-independent manner via specific TβRII and integrin α5β1 interactions. The recycling of fibronectin is Rab11 dependent and is required for TGF-β–induced cell migration. Fibronectin (FN) is a critical regulator of extracellular matrix (ECM) remodeling through its availability and stepwise polymerization for fibrillogenesis. Availability of FN is regulated by its synthesis and turnover, and fibrillogenesis is a multistep, integrin-dependent process essential for cell migration, proliferation, and tissue function. Transforming growth factor β (TGF-β) is an established regulator of ECM remodeling via transcriptional control of ECM proteins. Here we show that TGF-β, through increased FN trafficking in a transcription- and SMAD-independent manner, is a direct and rapid inducer of the fibrillogenesis required for TGF-β–induced cell migration. Whereas TGF-β signaling is dispensable for rapid fibrillogenesis, stable interactions between the cytoplasmic domain of the type II TGF-β receptor (TβRII) and the FN receptor (α5β1 integrin) are required. We find that, in response to TGF-β, cell surface–internalized FN is not degraded by the lysosome but instead undergoes recycling and incorporation into fibrils, a process dependent on TβRII. These findings are the first to show direct use of trafficked and recycled FN for fibrillogenesis, with a striking role for TGF-β in this process. Given the significant physiological consequences associated with FN availability and polymerization, our findings provide new insights into the regulation of fibrillogenesis for cellular homeostasis.
Collapse
Affiliation(s)
- Archana Varadaraj
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Laura M Jenkins
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Priyanka Singh
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Anindya Chanda
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29201
| | - John Snider
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - N Y Lee
- Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208 .,Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208
| |
Collapse
|
5
|
Peñaloza JP, Márquez-Miranda V, Cabaña-Brunod M, Reyes-Ramírez R, Llancalahuen FM, Vilos C, Maldonado-Biermann F, Velásquez LA, Fuentes JA, González-Nilo FD, Rodríguez-Díaz M, Otero C. Intracellular trafficking and cellular uptake mechanism of PHBV nanoparticles for targeted delivery in epithelial cell lines. J Nanobiotechnology 2017; 15:1. [PMID: 28049488 PMCID: PMC5210312 DOI: 10.1186/s12951-016-0241-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/09/2016] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Nanotechnology is a science that involves imaging, measurement, modeling and a manipulation of matter at the nanometric scale. One application of this technology is drug delivery systems based on nanoparticles obtained from natural or synthetic sources. An example of these systems is synthetized from poly(3-hydroxybutyrate-co-3-hydroxyvalerate), which is a biodegradable, biocompatible and a low production cost polymer. The aim of this work was to investigate the uptake mechanism of PHBV nanoparticles in two different epithelial cell lines (HeLa and SKOV-3). RESULTS As a first step, we characterized size, shape and surface charge of nanoparticles using dynamic light scattering and transmission electron microscopy. Intracellular incorporation was evaluated through flow cytometry and fluorescence microscopy using intracellular markers. We concluded that cellular uptake mechanism is carried out in a time, concentration and energy dependent way. Our results showed that nanoparticle uptake displays a cell-specific pattern, since we have observed different colocalization in two different cell lines. In HeLa (Cervical cancer cells) this process may occur via classical endocytosis pathway and some internalization via caveolin-dependent was also observed, whereas in SKOV-3 (Ovarian cancer cells) these patterns were not observed. Rearrangement of actin filaments showed differential nanoparticle internalization patterns for HeLa and SKOV-3. Additionally, final fate of nanoparticles was also determined, showing that in both cell lines, nanoparticles ended up in lysosomes but at different times, where they are finally degraded, thereby releasing their contents. CONCLUSIONS Our results, provide novel insight about PHBV nanoparticles internalization suggesting that for develop a proper drug delivery system is critical understand the uptake mechanism.
Collapse
Affiliation(s)
- Juan P. Peñaloza
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
- Escuela de Bioquímica, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Valeria Márquez-Miranda
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Echaurren #183, 8370071 Santiago, Chile
| | - Mauricio Cabaña-Brunod
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
- Escuela de Bioquímica, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Rodrigo Reyes-Ramírez
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
- Escuela de Bioquímica, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Felipe M. Llancalahuen
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
- Escuela de Bioquímica, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Cristian Vilos
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Echaurren #183, 8370071 Santiago, Chile
| | | | - Luis A. Velásquez
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Juan A. Fuentes
- Laboratorio de Genética y Patógenesis Bacteriana, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Santiago, Chile
| | - Fernando D. González-Nilo
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Echaurren #183, 8370071 Santiago, Chile
| | - Maité Rodríguez-Díaz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Carolina Otero
- Center for Integrative Medicine and Innovative Science, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| |
Collapse
|
6
|
Khan TA, Wang X, Maynard JA. Inclusion of an RGD Motif Alters Invasin Integrin-Binding Affinity and Specificity. Biochemistry 2016; 55:2078-90. [DOI: 10.1021/acs.biochem.5b01243] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Tarik A. Khan
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Xianzhe Wang
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer A. Maynard
- Departments of Chemical Engineering and ‡Biochemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|