1
|
Cui L, Perini G, Minopoli A, Augello A, De Spirito M, Palmieri V, Papi M. Plant-derived extracellular vesicles release combined with systemic DOX exhibits synergistic effects in 3D bioprinted triple-negative breast cancer. Biomed Pharmacother 2024; 181:117637. [PMID: 39481327 DOI: 10.1016/j.biopha.2024.117637] [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: 08/07/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer, lacking targeted therapeutic options. Hydrogels, particularly gelatin methacrylate (GelMA), have emerged as promising materials for localized drug delivery due to their biocompatibility and tunable properties. This study investigates a dual-delivery system for enhancing the treatment efficacy of triple-negative breast cancer (TNBC) using a combination of extracellular vesicles (EVs) derived from Citrus limon L. and the chemotherapeutic drug doxorubicin (DOX). We fabricated 3D bioprinted GelMA scaffolds to achieve localized and controlled release of EVs and evaluated their synergistic effects with systemic DOX delivery on both primary and metastatic 3D TNBC models. The GelMA scaffolds, especially those with 95 % methacrylation, exhibited higher stiffness, which enhanced their sustained release. Following 48-h incubation, the combination of EVs and DOX significantly increased cytotoxicity in the primary 3D TNBC model, reducing cell viability to approximately 30 % compared to controls. This was notably more effective than treatments with DOX or EVs alone. During the extended 7-day incubation period, the combination treatment continued to show superior efficacy, with persistently high levels of ROS generation and further reduction in cell viability. In a metastatic 3D TNBC model, a significant sensitivity to the combined treatment was observed, which notably inhibited aggregate formation and migration. Importantly, EVs-embedded scaffolds promoted the proliferation of human fibroblasts, highlighting their non-toxic nature, while concurrently inhibiting TNBC cell growth. This approach provides a promising strategy to improve the treatment outcomes of TNBC by exploiting the synergistic effects of local EVs release and systemic chemotherapy.
Collapse
Affiliation(s)
- Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy
| | - Antonio Minopoli
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy
| | - Alberto Augello
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy; Istituto dei Sistemi Complessi, CNR, Via dei Taurini 19, Rome 00185, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy; Istituto dei Sistemi Complessi, CNR, Via dei Taurini 19, Rome 00185, Italy.
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, Rome 00168, Italy; Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome 00168, Italy.
| |
Collapse
|
2
|
Mungai RW, Hartman II RJ, Jolin GE, Piskorowski KW, Billiar KL. Towards a More Objective and High-throughput Spheroid Invasion Assay Quantification Method. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600893. [PMID: 39005385 PMCID: PMC11244881 DOI: 10.1101/2024.06.27.600893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Multicellular spheroids embedded in 3D hydrogels are prominent in vitro models for 3D cell invasion. Yet, quantification methods for spheroid cell invasion that are high throughput, objective and accessible are still lacking. Variations in spheroid sizes and the shapes of the cells within render it difficult to objectively assess invasion extent. The goal of this work is to develop a high-throughput quantification method of cell invasion into 3D matrices that minimizes sensitivity to initial spheroid size and cell spreading and provides precise integrative directionally-dependent metrics of invasion. By analyzing images of fluorescent cell nuclei, invasion metrics are automatically calculated at the pixel level. The initial spheroid boundary is segmented and automated calculations of the nuclear pixel distances from the initial boundary are used to compute common invasion metrics (i.e., the change in invasion area, mean distance) for the same spheroid at a later timepoint. We also introduce the area moment of inertia as an integrative metric of cell invasion that considers the invasion area as well as the pixel distances from the initial spheroid boundary. Further, we show that principal component analysis can be used to quantify the directional influence of a stimuli to invasion (e.g., due to a chemotactic gradient or contact guidance). To demonstrate the power of the analysis for cell types with different invasive potentials and the utility of this method for a variety of biological applications, the method is used to analyze the invasiveness of five different cell types. In all, implementation of this high throughput quantification method results in consistent and objective analysis of 3D multicellular spheroid invasion. We provide the analysis code in both MATLAB and Python languages as well as a GUI for ease of use for researchers with a range of computer programming skills and for applications in a variety of biological research areas such as wound healing and cancer metastasis.
Collapse
Affiliation(s)
- Rozanne W. Mungai
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA 01605
| | | | - Grace E. Jolin
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA 01605
| | - Kevin W. Piskorowski
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA 01605
| | - Kristen L. Billiar
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA 01605
| |
Collapse
|
3
|
Panigrahi A, Benicky J, Aljuhani R, Mukherjee P, Nováková Z, Bařinka C, Goldman R. Galectin-3-Binding Protein Inhibits Extracellular Heparan 6-O-Endosulfatase Sulf-2. Mol Cell Proteomics 2024; 23:100793. [PMID: 38825040 PMCID: PMC11259796 DOI: 10.1016/j.mcpro.2024.100793] [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: 01/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024] Open
Abstract
Human extracellular 6-O-endosulfatases Sulf-1 and Sulf-2 are the only enzymes that post-synthetically alter the 6-O sulfation of heparan sulfate proteoglycans (HSPG), which regulates interactions of HSPG with many proteins. Oncogenicity of Sulf-2 in different cancers has been documented, and we have shown that Sulf-2 is associated with poor survival outcomes in head and neck squamous cell carcinoma (HNSCC). Despite its importance, limited information is available on direct protein-protein interactions of the Sulf-2 protein in the tumor microenvironment. In this study, we used monoclonal antibody (mAb) affinity purification and mass spectrometry to identify galectin-3-binding protein (LG3BP) as a highly specific binding partner of Sulf-2 in the conditioned media of HNSCC cell lines. We validated their direct interaction in vitro using recombinant proteins and have shown that the chondroitin sulfate (CS) covalently bound to the Sulf-2 influences the binding to LG3BP. We confirmed the importance of the CS chain for the interaction by generating a mutant Sulf-2 protein that lacks the CS. Importantly, we have shown that the LG3BP inhibits Sulf-2 activity in vitro in a concentration-dependent manner. As a consequence, the addition of LG3BP to a spheroid cell culture inhibited the invasion of the HNSCC cells into Matrigel. Thus, Sulf-2 interaction with LG3BP may regulate the physiological activity of the Sulf-2 enzyme as well as its activity in the tumor microenvironment.
Collapse
Affiliation(s)
- Aswini Panigrahi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA; Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, District of Columbia, USA.
| | - Julius Benicky
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA; Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, District of Columbia, USA
| | - Reem Aljuhani
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| | - Pritha Mukherjee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Zora Nováková
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, District of Columbia, USA; Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, District of Columbia, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
4
|
De Spirito M, Palmieri V, Perini G, Papi M. Bridging the Gap: Integrating 3D Bioprinting and Microfluidics for Advanced Multi-Organ Models in Biomedical Research. Bioengineering (Basel) 2024; 11:664. [PMID: 39061746 PMCID: PMC11274229 DOI: 10.3390/bioengineering11070664] [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: 05/28/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Recent advancements in 3D bioprinting and microfluidic lab-on-chip systems offer promising solutions to the limitations of traditional animal models in biomedical research. Three-dimensional bioprinting enables the creation of complex, patient-specific tissue models that mimic human physiology more accurately than animal models. These 3D bioprinted tissues, when integrated with microfluidic systems, can replicate the dynamic environment of the human body, allowing for the development of multi-organ models. This integration facilitates more precise drug screening and personalized therapy development by simulating interactions between different organ systems. Such innovations not only improve predictive accuracy but also address ethical concerns associated with animal testing, aligning with the three Rs principle. Future directions include enhancing bioprinting resolution, developing advanced bioinks, and incorporating AI for optimized system design. These technologies hold the potential to revolutionize drug development, regenerative medicine, and disease modeling, leading to more effective, personalized, and humane treatments.
Collapse
Affiliation(s)
- Marco De Spirito
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Valentina Palmieri
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
- Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche, CNR, via dei Taurini 19, 00185 Rome, Italy
| | - Giordano Perini
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Massimiliano Papi
- Department of Neuroscience, Universita Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (M.D.S.); (V.P.); (G.P.)
- Istituti di Ricovero e Cura a Carattere Scientifico IRCSS, Fondazione Policlinico Universitario “A. Gemelli”, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
5
|
Cui L, Perini G, Augello A, Palmieri V, De Spirito M, Papi M. Plant-derived extracellular nanovesicles: a promising biomedical approach for effective targeting of triple negative breast cancer cells. Front Bioeng Biotechnol 2024; 12:1390708. [PMID: 38952670 PMCID: PMC11215178 DOI: 10.3389/fbioe.2024.1390708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Introduction: Triple negative breast cancer (TNBC), a highly aggressive subtype accounting for 15-20% of all breast cancer cases, faces limited treatment options often accompanied by severe side effects. In recent years, natural extracellular nanovesicles derived from plants have emerged as promising candidates for cancer therapy, given their safety profile marked by non-immunogenicity and absence of inflammatory responses. Nevertheless, the potential anti-cancer effects of Citrus limon L.-derived extracellular nanovesicles (CLENs) for breast cancer treatment is still unexplored. Methods: In this study, we investigated the anti-cancer effects of CLENs on two TNBC cell lines (4T1 and HCC-1806 cells) under growth conditions in 2D and 3D culture environments. The cellular uptake efficiency of CLENs and their internalization mechanism were evaluated in both cells using confocal microscopy. Thereafter, we assessed the effect of different concentrations of CLENs on cell viability over time using a dual approach of Calcein-AM PI live-dead assay and CellTiter-Glo bioluminescence assay. We also examined the influence of CLENs on the migratory and evasion abilities of TNBC cells through wound healing and 3D Matrigel drop evasion assays. Furthermore, Western blot analysis was employed to investigate the effects of CLENs on the phosphorylation levels of phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and extracellular signal- regulated kinase (ERK) expression. Results: We found that CLENs were internalized by the cells via endocytosis, leading to decreased cell viability, in a dose- and time-dependent manner. Additionally, the migration and evasion abilities of TNBC cells were significantly inhibited under exposed to 40 and 80 μg/mL CLENs. Furthermore, down-regulated expression levels of phosphorylated phosphoinositide 3-kinase (PI3K), protein kinase B (AKT), and extracellular signal-regulated kinase (ERK), suggesting that the inhibition of cancer cell proliferation, migration, and evasion is driven by the inhibition of the PI3K/AKT and MAPK/ERK signaling pathways. Discussion: Overall, our results demonstrate the anti-tumor efficiency of CLENs against TNBC cells, highlighting their potential as promising natural anti-cancer agents for clinical applications in cancer treatment.
Collapse
Affiliation(s)
- Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Alberto Augello
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Istituto dei Sistemi Complessi, Consiglio nazionale delle ricerche (C.N.R.), Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy
| |
Collapse
|
6
|
Santarelli G, Perini G, Salustri A, Palucci I, Rosato R, Palmieri V, Iacovelli C, Bellesi S, Sali M, Sanguinetti M, De Spirito M, Papi M, Delogu G, De Maio F. Unraveling the potential of graphene quantum dots against Mycobacterium tuberculosis infection. Front Microbiol 2024; 15:1395815. [PMID: 38774507 PMCID: PMC11107295 DOI: 10.3389/fmicb.2024.1395815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction The emergence of drug-resistant Mycobacterium tuberculosis (Mtb) strains has underscored the urgent need for novel therapeutic approaches. Carbon-based nanomaterials, such as graphene oxide (GO), have shown potential in anti-TB activities but suffer from significant toxicity issues. Methods This study explores the anti-TB potential of differently functionalized graphene quantum dots (GQDs) - non-functionalized, L-GQDs, aminated (NH2-GQDs), and carboxylated (COOH-GQDs) - alone and in combination with standard TB drugs (isoniazid, amikacin, and linezolid). Their effects were assessed in both axenic cultures and in vitro infection models. Results GQDs alone did not demonstrate direct mycobactericidal effects nor trapping activity. However, the combination of NH2-GQDs with amikacin significantly reduced CFUs in in vitro models. NH2-GQDs and COOH-GQDs also enhanced the antimicrobial activity of amikacin in infected macrophages, although L-GQDs and COOH-GQDs alone showed no significant activity. Discussion The results suggest that specific types of GQDs, particularly NH2-GQDs, can enhance the efficacy of existing anti-TB drugs. These nanoparticles might serve as effective adjuvants in anti-TB therapy by boosting drug performance and reducing bacterial counts in host cells, highlighting their potential as part of advanced drug delivery systems in tuberculosis treatment. Further investigations are needed to better understand their mechanisms and optimize their use in clinical settings.
Collapse
Affiliation(s)
- Giulia Santarelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Alessandro Salustri
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Ivana Palucci
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Roberto Rosato
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Istituto dei Sistemi Complessi, CNR, Rome, Italy
| | - Camilla Iacovelli
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Silvia Bellesi
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Michela Sali
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| | - Giovanni Delogu
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie-Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Mater Olbia Hospital, Olbia, Italy
| | - Flavio De Maio
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Rome, Italy
| |
Collapse
|
7
|
Ghaznavi H, Afzalipour R, Khoei S, Sargazi S, Shirvalilou S, Sheervalilou R. New insights into targeted therapy of glioblastoma using smart nanoparticles. Cancer Cell Int 2024; 24:160. [PMID: 38715021 PMCID: PMC11077767 DOI: 10.1186/s12935-024-03331-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024] Open
Abstract
In recent times, the intersection of nanotechnology and biomedical research has given rise to nanobiomedicine, a captivating realm that holds immense promise for revolutionizing diagnostic and therapeutic approaches in the field of cancer. This innovative fusion of biology, medicine, and nanotechnology aims to create diagnostic and therapeutic agents with enhanced safety and efficacy, particularly in the realm of theranostics for various malignancies. Diverse inorganic, organic, and hybrid organic-inorganic nanoparticles, each possessing unique properties, have been introduced into this domain. This review seeks to highlight the latest strides in targeted glioblastoma therapy by focusing on the application of inorganic smart nanoparticles. Beyond exploring the general role of nanotechnology in medical applications, this review delves into groundbreaking strategies for glioblastoma treatment, showcasing the potential of smart nanoparticles through in vitro studies, in vivo investigations, and ongoing clinical trials.
Collapse
Affiliation(s)
- Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Reza Afzalipour
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
- Department of Radiology, Faculty of Para-Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Sakine Shirvalilou
- Finetech in Medicine Research Center, Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Sheervalilou
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| |
Collapse
|
8
|
Panigrahi A, Benicky J, Aljuhani R, Mukherjee P, Nováková Z, Bařinka C, Goldman R. Galectin-3-binding protein inhibits extracellular heparan 6- O-endosulfatse Sulf-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572603. [PMID: 38187586 PMCID: PMC10769223 DOI: 10.1101/2023.12.20.572603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Human extracellular 6-O-endosulfatases Sulf-1 and Sulf-2 are the only enzymes that post-synthetically alter the 6-O sulfation of heparan sulfate proteoglycans (HSPG), which regulates interactions of HSPG with many proteins. Oncogenicity of Sulf-2 in different cancers has been documented and we have shown that Sulf-2 is associated with poor survival outcomes in head and neck squamous cell carcinoma (HNSCC). In spite of its importance, limited information is available on direct protein-protein interactions of the Sulf-2 protein in the tumor microenvironment. In this study, we used monoclonal antibody (mAb) affinity purification and mass spectrometry to identify galectin-3-binding protein (LG3BP) as a highly specific binding partner of Sulf-2 in the secretome of HNSCC cell lines. We validated their direct interaction in vitro using recombinant proteins and have shown that the chondroitin sulfate (CS) covalently bound to the Sulf-2 influences the binding to LG3BP. We confirmed importance of the CS chain for the interaction by generating a mutant Sulf-2 protein that lacks the CS. Importantly, we have shown that the LG3BP inhibits Sulf-2 activity in vitro in a concentration dependent manner. As a consequence, the addition of LG3BP to a spheroid cell culture inhibited invasion of the HNSCC cells into Matrigel. Thus, Sulf-2 interaction with LG3BP has functional relevance, and may regulate physiological activity of the Sulf-2 enzyme as well as its activity in the tumor microenvironment.
Collapse
Affiliation(s)
- Aswini Panigrahi
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC 20057, USA
| | - Julius Benicky
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC 20057, USA
| | - Reem Aljuhani
- Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| | - Pritha Mukherjee
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Zora Nováková
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Cyril Bařinka
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech Republic
| | - Radoslav Goldman
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
- Clinical and Translational Glycoscience Research Center, Georgetown University, Washington, DC 20057, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
9
|
Swidan MM, Essa BM, Sakr TM. Pristine/folate-functionalized graphene oxide as two intrinsically radioiodinated nano-theranostics: self/dual in vivo targeting comparative study. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00157-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
Background
Nanomedicine offers great potentials for theranostic studies via providing higher efficacy and safety levels. This work aimed to develop and evaluate a new nanoplatform as a tumor theranostic probe.
Results
Carboxyl-functionalized graphene oxide nanosheets (FGO) was well synthesized from graphite powder and then conjugated with folic acid to act as a targeted nano-probe. Full characterization and in vitro cytotoxicity evaluation were conducted; besides, in vivo bio-evaluation was attained via intrinsic radioiodination approach in both normal and tumor-bearing Albino mice. The results indicated that FGO as well as conjugated graphene oxide nanosheets (CGO) are comparatively non-toxic to normal cells even at higher concentrations. Pharmacokinetics of FGO and CGO showed intensive and selective uptake in the tumor sites where CGO showed high T/NT of 7.27 that was 4 folds of FGO at 1 h post injection. Additionally, radioiodinated-CGO (ICGO) had declared a superior prominence over the previously published tumor targeted GO radiotracers regarding the physicochemical properties pertaining ability and tumor accumulation behavior.
Conclusions
In conclusion, ICGO can be used as a selective tumor targeting agent for cancer theranosis with aid of I-131 that has a maximum beta and gamma energies of 606.3 and 364.5 keV, respectively.
Collapse
|
10
|
Perini G, Palmieri V, Friggeri G, Augello A, De Spirito M, Papi M. Carboxylated graphene quantum dots-mediated photothermal therapy enhances drug-membrane permeability, ROS production, and the immune system recruitment on 3D glioblastoma models. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00168-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
AbstractGraphene quantum dots (GQDs) are biocompatible nanoparticles employed in biomedical field, thanks to their size and photophysical properties. GQDs have shown the capability to cross biological barriers, including the blood–brain barrier, which makes them promising agents for brain diseases therapy. It has been shown that surface-functionalized GQDs enhance membrane fluidity and intracellular uptake, exerting a synergistic effect with antitumor drugs at subtherapeutic doses. Here, we tested GQDs effects in combination with chemotherapeutic agents doxorubicin and temozolomide, on a complex 3D spheroid model of glioblastoma. We observed that the capability of GQDs to absorb and convert near-infrared light into heat is a key factor in membrane permeability enhancement on 3D model. This non-invasive therapeutic strategy named photothermal therapy (PTT), combined to chemotherapy at subtherapeutic doses, significantly increased the effect of antitumor drugs by reducing tumor growth and viability. Furthermore, the increase in membrane permeability due to GQDs-mediated PTT enhanced the release of reactive oxygen species with strong migration of the immune system towards irradiated cancer spheroids. Our data indicate that the increase in membrane permeability can enhance the efficacy of antitumor drugs at subtherapeutic doses against glioblastoma, reducing side effects, and directing immune response, ultimately improving quality of life for patients.
Collapse
|
11
|
Pierangeli D, Perini G, Palmieri V, Grecco I, Friggeri G, De Spirito M, Papi M, DelRe E, Conti C. Extreme transport of light in spheroids of tumor cells. Nat Commun 2023; 14:4662. [PMID: 37537177 PMCID: PMC10400595 DOI: 10.1038/s41467-023-40379-7] [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: 07/01/2022] [Accepted: 07/14/2023] [Indexed: 08/05/2023] Open
Abstract
Extreme waves are intense and unexpected wavepackets ubiquitous in complex systems. In optics, these rogue waves are promising as robust and noise-resistant beams for probing and manipulating the underlying material. Localizing large optical power is crucial especially in biomedical systems, where, however, extremely intense beams have not yet been observed. We here discover that tumor-cell spheroids manifest optical rogue waves when illuminated by randomly modulated laser beams. The intensity of light transmitted through bio-printed three-dimensional tumor models follows a signature Weibull statistical distribution, where extreme events correspond to spatially-localized optical modes propagating within the cell network. Experiments varying the input beam power and size indicate that the rogue waves have a nonlinear origin. We show that these nonlinear optical filaments form high-transmission channels with enhanced transmission. They deliver large optical power through the tumor spheroid, and can be exploited to achieve a local temperature increase controlled by the input wave shape. Our findings shed light on optical propagation in biological aggregates and demonstrate how nonlinear extreme event formation allows light concentration in deep tissues, paving the way to using rogue waves in biomedical applications, such as light-activated therapies.
Collapse
Affiliation(s)
- Davide Pierangeli
- Institute for Complex Systems, National Research Council, Rome, 00185, Italy.
- Physics Department, Sapienza University of Rome, Rome, 00185, Italy.
| | - Giordano Perini
- Neuroscience Department, University Cattolica del Sacro Cuore, Rome, 00168, Italy
- IRCSS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, 00168, Italy
| | - Valentina Palmieri
- Institute for Complex Systems, National Research Council, Rome, 00185, Italy
- Neuroscience Department, University Cattolica del Sacro Cuore, Rome, 00168, Italy
| | - Ivana Grecco
- Physics Department, Sapienza University of Rome, Rome, 00185, Italy
| | - Ginevra Friggeri
- Neuroscience Department, University Cattolica del Sacro Cuore, Rome, 00168, Italy
- IRCSS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, 00168, Italy
| | - Marco De Spirito
- Neuroscience Department, University Cattolica del Sacro Cuore, Rome, 00168, Italy
- IRCSS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, 00168, Italy
| | - Massimiliano Papi
- Neuroscience Department, University Cattolica del Sacro Cuore, Rome, 00168, Italy.
- IRCSS, Fondazione Policlinico Universitario Agostino Gemelli, Rome, 00168, Italy.
| | - Eugenio DelRe
- Physics Department, Sapienza University of Rome, Rome, 00185, Italy
| | - Claudio Conti
- Physics Department, Sapienza University of Rome, Rome, 00185, Italy
| |
Collapse
|
12
|
Bhaloo A, Nguyen S, Lee BH, Valimukhametova A, Gonzalez-Rodriguez R, Sottile O, Dorsky A, Naumov AV. Doped Graphene Quantum Dots as Biocompatible Radical Scavenging Agents. Antioxidants (Basel) 2023; 12:1536. [PMID: 37627531 PMCID: PMC10451549 DOI: 10.3390/antiox12081536] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 08/27/2023] Open
Abstract
Oxidative stress is proven to be a leading factor in a multitude of adverse conditions, from Alzheimer's disease to cancer. Thus, developing effective radical scavenging agents to eliminate reactive oxygen species (ROS) driving many oxidative processes has become critical. In addition to conventional antioxidants, nanoscale structures and metal-organic complexes have recently shown promising potential for radical scavenging. To design an optimal nanoscale ROS scavenging agent, we have synthesized ten types of biocompatible graphene quantum dots (GQDs) augmented with various metal dopants. The radical scavenging abilities of these novel metal-doped GQD structures were, for the first time, assessed via the DPPH, KMnO4, and RHB (Rhodamine B protectant) assays. While all metal-doped GQDs consistently demonstrate antioxidant properties higher than the undoped cores, aluminum-doped GQDs exhibit 60-95% radical scavenging ability of ascorbic acid positive control. Tm-doped GQDs match the radical scavenging properties of ascorbic acid in the KMnO4 assay. All doped GQD structures possess fluorescence imaging capabilities that enable their tracking in vitro, ensuring their successful cellular internalization. Given such multifunctionality, biocompatible doped GQD antioxidants can become prospective candidates for multimodal therapeutics, including the reduction of ROS with concomitant imaging and therapeutic delivery to cancer tumors.
Collapse
Affiliation(s)
- Adam Bhaloo
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | - Steven Nguyen
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | - Bong Han Lee
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | - Alina Valimukhametova
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | | | - Olivia Sottile
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | - Abby Dorsky
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| | - Anton V. Naumov
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX 76129, USA; (A.B.); (S.N.); (B.H.L.); (A.V.); (O.S.); (A.D.)
| |
Collapse
|
13
|
Boltman T, Meyer M, Ekpo O. Diagnostic and Therapeutic Approaches for Glioblastoma and Neuroblastoma Cancers Using Chlorotoxin Nanoparticles. Cancers (Basel) 2023; 15:3388. [PMID: 37444498 DOI: 10.3390/cancers15133388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 07/15/2023] Open
Abstract
Glioblastoma multiforme (GB) and high-risk neuroblastoma (NB) are known to have poor therapeutic outcomes. As for most cancers, chemotherapy and radiotherapy are the current mainstay treatments for GB and NB. However, the known limitations of systemic toxicity, drug resistance, poor targeted delivery, and inability to access the blood-brain barrier (BBB), make these treatments less satisfactory. Other treatment options have been investigated in many studies in the literature, especially nutraceutical and naturopathic products, most of which have also been reported to be poorly effective against these cancer types. This necessitates the development of treatment strategies with the potential to cross the BBB and specifically target cancer cells. Compounds that target the endopeptidase, matrix metalloproteinase 2 (MMP-2), have been reported to offer therapeutic insights for GB and NB since MMP-2 is known to be over-expressed in these cancers and plays significant roles in such physiological processes as angiogenesis, metastasis, and cellular invasion. Chlorotoxin (CTX) is a promising 36-amino acid peptide isolated from the venom of the deathstalker scorpion, Leiurus quinquestriatus, demonstrating high selectivity and binding affinity to a broad-spectrum of cancers, especially GB and NB through specific molecular targets, including MMP-2. The favorable characteristics of nanoparticles (NPs) such as their small sizes, large surface area for active targeting, BBB permeability, etc. make CTX-functionalized NPs (CTX-NPs) promising diagnostic and therapeutic applications for addressing the many challenges associated with these cancers. CTX-NPs may function by improving diffusion through the BBB, enabling increased localization of chemotherapeutic and genotherapeutic drugs to diseased cells specifically, enhancing imaging modalities such as magnetic resonance imaging (MRI), single-photon emission computed tomography (SPECT), optical imaging techniques, image-guided surgery, as well as improving the sensitization of radio-resistant cells to radiotherapy treatment. This review discusses the characteristics of GB and NB cancers, related treatment challenges as well as the potential of CTX and its functionalized NP formulations as targeting systems for diagnostic, therapeutic, and theranostic purposes. It also provides insights into the potential mechanisms through which CTX crosses the BBB to bind cancer cells and provides suggestions for the development and application of novel CTX-based formulations for the diagnosis and treatment of GB and NB in the future.
Collapse
Affiliation(s)
- Taahirah Boltman
- Department of Medical Biosciences, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Mervin Meyer
- Department of Science and Innovation/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Robert Sobukwe Road, Bellville, Cape Town 7535, South Africa
| | - Okobi Ekpo
- Department of Anatomy and Cellular Biology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
14
|
Cancer Cell Models for the Development of Anti-Cancer Drugs. Int J Mol Sci 2022; 23:ijms232214457. [PMID: 36430932 PMCID: PMC9699439 DOI: 10.3390/ijms232214457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
In the multi-factorial etiology of organ-site cancers by suspect human chemical carcinogens, oncogenic virus, activation of RAS, Myc and HER-2 oncogenes, inactivation of TP53, RB and APC tumor suppressor genes represent early-occurring genetic events [...].
Collapse
|
15
|
Quantum Dots Mediated Imaging and Phototherapy in Cancer Spheroid Models: State of the Art and Perspectives. Pharmaceutics 2022; 14:pharmaceutics14102136. [PMID: 36297571 PMCID: PMC9611360 DOI: 10.3390/pharmaceutics14102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Abstract
Quantum Dots (QDs) are fluorescent nanoparticles known for their exceptional optical properties, i.e., high fluorescence emission, photostability, narrow emission spectrum, and broad excitation wavelength. These properties make QDs an exciting choice for bioimaging applications, notably in cancer imaging. Challenges lie in their ability to specifically label targeted cells. Numerous studies have been carried out with QDs coupled to various ligands like peptides, antibodies, aptamers, etc., to achieve efficient targeting. Most studies were conducted in vitro with two-dimensional cell monolayers (n = 8902) before evolving towards more sophisticated models. Three-dimensional multicellular tumor models better recapitulate in vivo conditions by mimicking cell-to-cell and cell-matrix interactions. To date, only few studies (n = 34) were conducted in 3D in vitro models such as spheroids, whereas these models could better represent QDs behavior in tumors compared to monolayers. Thus, the purpose of this review is to present a state of the art on the studies conducted with Quantum Dots on spheroid models for imaging and phototherapy purposes.
Collapse
|
16
|
Perini G, Rosenkranz A, Friggeri G, Zambrano D, Rosa E, Augello A, Palmieri V, De Spirito M, Papi M. Advanced usage of Ti3C2Tx MXenes for photothermal therapy on different 3D breast cancer models. Biomed Pharmacother 2022; 153:113496. [DOI: 10.1016/j.biopha.2022.113496] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/21/2022] [Accepted: 07/27/2022] [Indexed: 12/12/2022] Open
|