80 results on '"Seras-Franzoso J"'
Search Results
2. Author Reply to Peer Reviews of Cancer stem cell-derived extracellular vesicles preferentially target MHC-II– macrophages and PD1+ T cells in the tumor microenvironment
- Author
-
Pucci, F., primary, Seras-Franzoso, J., additional, Oshimori, N., additional, Claudio, N.M., additional, Ziglari, T., additional, Guo, Z., additional, and Gonzalez-Callejo, P., additional
- Published
- 2022
- Full Text
- View/download PDF
3. Cancer stem cell-derived extracellular vesicles preferentially target MHC-II– macrophages and PD1+ T cells in the tumor microenvironment
- Author
-
Gonzalez-Callejo, P., primary, Guo, Z., additional, Ziglari, T., additional, Claudio, N.M., additional, Oshimori, N., additional, Seras-Franzoso, J., additional, and Pucci, F., additional
- Published
- 2022
- Full Text
- View/download PDF
4. 0159 A new protocol for the isolation of key recombinant proteins in livestock production using lactic acid bacteria as a cell factory
- Author
-
Gifre, L., primary, Cano-Garrido, O., additional, Fàbregas, F., additional, Seras-Franzoso, J., additional, Roca, R., additional, Ferrer-Miralles, N., additional, Villaverde, A., additional, Bach, A., additional, Arís, A., additional, and Garcia-Fruitós, E., additional
- Published
- 2016
- Full Text
- View/download PDF
5. Functionalization of 3D scaffolds with protein-releasing biomaterials for intracellular delivery
- Author
-
Seras-Franzoso J, Steurer C, Roldán M, Vendrell M, Vidaurre-Agut C, Tarruella A, Saldaña L, Vilaboa N, Parera M, Elizondo E, Ratera I, Ventosa N, Veciana J, Campillo-Fernández AJ, García-Fruitós E, Vázquez E, and Villaverde A
- Subjects
3D scaffolds, Bioscaffold, Bottom-up delivery, Nanoparticles, Polylactic acid (PLA), Tissue engineering - Abstract
Appropriate combinations of mechanical and biological stimuli are required to promote proper colonization of substrate materials in regenerative medicine. In this context, 3D scaffolds formed by compatible and biodegradable materials are under continuous development in an attempt to mimic the extracellular environment of mammalian cells. We have here explored how novel 3D porous scaffolds constructed by polylactic acid, polycaprolactone or chitosan can be decorated with bacterial inclusion bodies, submicron protein particles formed by releasable functional proteins. A simple dipping-based decoration method tested here specifically favors the penetration of the functional particles deeper than 300µm from the materials' surface. The functionalized surfaces support the intracellular delivery of biologically active proteins to up to more than 80% of the colonizing cells, a process that is slightly influenced by the chemical nature of the scaffold. The combination of 3D soft scaffolds and protein-based sustained release systems (Bioscaffolds) offers promise in the fabrication of bio-inspired hybrid matrices for multifactorial control of cell proliferation in tissue engineering under complex architectonic setting-ups.
- Published
- 2013
6. Packaging protein drugs as bacterial inclusion bodies for therapeutic applications
- Author
-
Villaverde Antonio, García-Fruitós Elena, Rinas Ursula, Seras-Franzoso Joaquin, Kosoy Ana, Corchero José, and Vazquez Esther
- Subjects
Microbiology ,QR1-502 - Abstract
Abstract A growing number of insights on the biology of bacterial inclusion bodies (IBs) have revealed intriguing utilities of these protein particles. Since they combine mechanical stability and protein functionality, IBs have been already exploited in biocatalysis and explored for bottom-up topographical modification in tissue engineering. Being fully biocompatible and with tuneable bio-physical properties, IBs are currently emerging as agents for protein delivery into mammalian cells in protein-replacement cell therapies. So far, IBs formed by chaperones (heat shock protein 70, Hsp70), enzymes (catalase and dihydrofolate reductase), grow factors (leukemia inhibitory factor, LIF) and structural proteins (the cytoskeleton keratin 14) have been shown to rescue exposed cells from a spectrum of stresses and restore cell functions in absence of cytotoxicity. The natural penetrability of IBs into mammalian cells (reaching both cytoplasm and nucleus) empowers them as an unexpected platform for the controlled delivery of essentially any therapeutic polypeptide. Production of protein drugs by biopharma has been traditionally challenged by IB formation. However, a time might have arrived in which recombinant bacteria are to be engineered for the controlled packaging of therapeutic proteins as nanoparticulate materials (nanopills), for their extra- or intra-cellular release in medicine and cosmetics.
- Published
- 2012
- Full Text
- View/download PDF
7. Isolation of cell-free bacterial inclusion bodies
- Author
-
Rodríguez-Carmona Escarlata, Cano-Garrido Olivia, Seras-Franzoso Joaquin, Villaverde Antonio, and García-Fruitós Elena
- Subjects
Microbiology ,QR1-502 - Abstract
Abstract Background Bacterial inclusion bodies are submicron protein clusters usually found in recombinant bacteria that have been traditionally considered as undesirable products from protein production processes. However, being fully biocompatible, they have been recently characterized as nanoparticulate inert materials useful as scaffolds for tissue engineering, with potentially wider applicability in biomedicine and material sciences. Current protocols for inclusion body isolation from Escherichia coli usually offer between 95 to 99% of protein recovery, what in practical terms, might imply extensive bacterial cell contamination, not compatible with the use of inclusion bodies in biological interfaces. Results Using an appropriate combination of chemical and mechanical cell disruption methods we have established a convenient procedure for the recovery of bacterial inclusion bodies with undetectable levels of viable cell contamination, below 10-1 cfu/ml, keeping the particulate organization of these aggregates regarding size and protein folding features. Conclusions The application of the developed protocol allows obtaining bacterial free inclusion bodies suitable for use in mammalian cell cultures and other biological interfaces.
- Published
- 2010
- Full Text
- View/download PDF
8. Cancer stem cell-derived extracellular vesicles preferentially target MHC-II–macrophages and PD1+ T cells in the tumor microenvironment
- Author
-
Patricia Gonzalez-Callejo, Zihan Guo, Tahereh Ziglari, Natalie Marcia Claudio, Kayla Hoang Nguyen, Naoki Oshimori, Joaquim Seras-Franzoso, Ferdinando Pucci, Institut Català de la Salut, [Gonzalez-Callejo P] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. Bionanoplasmonics Group, CIC biomaGUNE, Donostia-San Sebastián, Spain. [Guo Z, Nguyen KH] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. Program in Biomedical Sciences, Oregon Health and Science University, Portland, Oregon, United States of America. [Ziglari T] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. [Claudio NM] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, Oregon, United States of America. [Oshimori N] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. Program in Biomedical Sciences, Oregon Health and Science University, Portland, Oregon, United States of America. Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, Oregon, United States of America. Department of Dermatology, Oregon Health and Science University, Portland, Oregon, United States of America. [Seras-Franzoso J] Grup de Direccionament i Alliberament Farmacològic, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. [Pucci F] Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, Oregon, United States of America. Program in Biomedical Sciences, Oregon Health and Science University, Portland, Oregon, United States of America. Department of Otolaryngology-Head and Neck Surgery, Oregon Health and Science University, Portland, Oregon, United States of America, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Coll - Càncer ,Multidisciplinary ,Neoplasms::Neoplasms::Neoplasms by Site::Head and Neck Neoplasms::Squamous Cell Carcinoma of Head and Neck [DISEASES] ,células::estructuras celulares::espacio extracelular::vesículas extracelulares [ANATOMÍA] ,Cells::Cellular Structures::Extracellular Space::Extracellular Vesicles [ANATOMY] ,Cap - Càncer ,Cèl·lules canceroses ,neoplasias::neoplasias::neoplasias por localización::neoplasias de cabeza y cuello::carcinoma de células escamosas de cabeza y cuello [ENFERMEDADES] ,células::células madre::células madre neoplásicas [ANATOMÍA] ,Cèl·lules mare ,Cells::Stem Cells::Neoplastic Stem Cells [ANATOMY] - Abstract
Cèl·lules mare del càncer; Càncers de cap i coll; Macròfags Cancer stem cells; Head and neck cancers; Macrophages Células madre del cáncer; Cánceres de cabeza y cuello; Macrófagos Immunotherapy is an approved treatment option for head and neck squamous cell carcinoma (HNSCC). However, the response rate to immune checkpoint blockade is only 13% for recurrent HNSCC, highlighting the urgent need to better understand tumor-immune interplay, with the ultimate goal of improving patient outcomes. HNSCC present high local recurrence rates and therapy resistance that can be attributed to the presence of cancer stem cells (CSC) within tumors. CSC exhibit singular properties that enable them to avoid immune detection and eradication. How CSC communicate with immune cells and which immune cell types are preferentially found within the CSC niche are still open questions. Here, we used genetic approaches to specifically label CSC-derived extracellular vesicles (EVs) and to perform Sortase-mediated in vivo proximity labeling of CSC niche cells. We identified specific immune cell subsets that were selectively targeted by EVCSC and that were found in the CSC niche. Native EVCSC preferentially targeted MHC-II–macrophages and PD1+ T cells in the tumor microenvironment, which were the same immune cell subsets enriched within the CSC niche. These observations indicate that the use of genetic technologies able to track EVs without in vitro isolation are a valuable tool to unveil the biology of native EVCSC. European Molecular Biology Organization (EMBO):Patricia Gonzalez-Callejo short-term fellowship; V Foundation for Cancer Research (VFCR):Natalie M Claudio,Ferdinando Pucci 2019-012.
- Published
- 2023
9. Editorial: High added-value nanoparticles: Rethinking and recycling cell protein waste
- Author
-
Unzueta Elorza, Ugutz, Sanchez, Julieta M., Garcia-Fruitos, Elena, Seras-Franzoso, Joaquin, Universitat Autònoma de Barcelona, Producció Animal, Producció de Remugants, Institut Català de la Salut, [Unzueta U] Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain. Josep Carreras Leukaemia Research Institute (IJC, Campus Sant Pau), Barcelona, Spain. CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. [Sanchez JM] CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET- Universidad Nacional de Córdoba, ICTA, FCEFyN, UNC, Córdoba, Argentina. [Garcia-Fruitós E] Department of Ruminant Production, Institute of Agrifood Research and Technology (IRTA), Caldes de Montbui, Spain. [Seras-Franzoso J] CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. Grup de Recerca en Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Farmacologia ,Technology, Industry, and Agriculture::Manufactured Materials::Nanostructures::Nanoparticles [TECHNOLOGY, INDUSTRY, AND AGRICULTURE] ,Histology ,Nanopartícules ,disciplinas de las ciencias naturales::disciplinas de las ciencias biológicas::farmacología [DISCIPLINAS Y OCUPACIONES] ,Biomedical Engineering ,tecnología, industria y agricultura::productos manufacturados::nanoestructuras::nanopartículas [TECNOLOGÍA, INDUSTRIA Y AGRICULTURA] ,Bioengineering ,Nanobiotechnlogy ,Virus-based nanomaterials ,Inclusion bodies (IBs) ,Green biotechnology ,Biomedicine ,Cost-eficàcia ,Extracellular vesicles (EVs) ,epidemiología y bioestadística::epidemiología::usos de la epidemiología::análisis de coste-eficiencia [SALUD PÚBLICA] ,Epidemiology and Biostatistics::Epidemiology::Uses of Epidemiology::Cost Efficiency Analysis [PUBLIC HEALTH] ,Natural Science Disciplines::Biological Science Disciplines::Pharmacology [DISCIPLINES AND OCCUPATIONS] ,Biotechnology - Abstract
Extracellular vesicles; Biomedicine; Nanobiotechnlogy Vesículas extracelulares; Biomedicina; Nanobiotecnología Vesícules extracel·lulars; Biomedicina; Nanobiotecnologia
- Published
- 2022
- Full Text
- View/download PDF
10. Selectively Targeting Breast Cancer Stem Cells By 8-Quinolinol and Niclosamide
- Author
-
Cámara-Sánchez, Patricia, Díaz Riascos, Zamira Vanessa, García Aranda, Natalia, Gener, Petra, Seras-Franzoso, Joaquin, Giani-Alonso, Micaela, Royo, Miriam, Vázquez Gómez, Esther, Schwartz, Simon, Abasolo, Ibane, Universitat Autònoma de Barcelona. Institut de Biotecnologia i de Biomedicina \\'Vicent Villar Palasí\\', European Commission, Institut Català de la Salut, [Cámara-Sánchez P, Gener P, Seras-Franzoso J, Schwartz S Jr] Grup de Direccionament i Alliberament Farmacològic, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. [Díaz-Riascos ZV, García-Aranda N, Abasolo I] Grup de Direccionament i Alliberament Farmacològic, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. Àrea de Validació Funcional i Investigació Preclínica (FVPR), Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Giani-Alonso M] Grup de Direccionament i Alliberament Farmacològic, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Cèl·lules canceroses - Proliferació ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Triple Negative Breast Neoplasms ,Cells::Stem Cells::Neoplastic Stem Cells [ANATOMY] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,behavioral disciplines and activities ,Catalysis ,Quimioteràpia combinada ,8-quinolinol ,Inorganic Chemistry ,Mice ,triple negative breast cancer ,cancer stem cells ,niclosamide ,combination therapy ,Breast cancer ,Cell Line, Tumor ,terapéutica::farmacoterapia::farmacoterapia combinada [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Medicine ,Animals ,Humans ,Triple negative breast cancer ,Physical and Theoretical Chemistry ,Combination therapy ,Molecular Biology ,Spectroscopy ,Niclosamide ,Cell Proliferation ,business.industry ,Cancer stem cells ,Organic Chemistry ,Therapeutics::Drug Therapy::Drug Therapy, Combination [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,General Medicine ,medicine.disease ,Oxyquinoline ,Computer Science Applications ,neoplasias::neoplasias por localización::neoplasias de la mama::neoplasias de mama triple negativos [ENFERMEDADES] ,Mama - Càncer - Tractament ,Neoplasms::Neoplasms by Site::Breast Neoplasms::Triple Negative Breast Neoplasms [DISEASES] ,Cancer research ,Neoplastic Stem Cells ,Stem cell ,células::células madre::células madre neoplásicas [ANATOMÍA] ,business ,Cèl·lules mare ,8-Quinolinol ,medicine.drug - Abstract
Cancer maintenance, metastatic dissemination and drug resistance are sustained by cancer stem cells (CSCs). Triple negative breast cancer (TNBC) is the breast cancer subtype with the highest number of CSCs and the poorest prognosis. Here, we aimed to identify potential drugs targeting CSCs to be further employed in combination with standard chemotherapy in TNBC treatment. The anti-CSC efficacy of up to 17 small drugs was tested in TNBC cell lines using cell viability assays on differentiated cancer cells and CSCs. Then, the effect of 2 selected drugs (8-quinolinol -8Q- and niclosamide -NCS-) in the cancer stemness features were evaluated using mammosphere growth, cell invasion, migration and anchorage-independent growth assays. Changes in the expression of stemness genes after 8Q or NCS treatment were also evaluated. Moreover, the potential synergism of 8Q and NCS with PTX on CSC proliferation and stemness-related signaling pathways was evaluated using TNBC cell lines, CSC-reporter sublines, and CSC-enriched mammospheres. Finally, the efficacy of NCS in combination with PTX was analyzed in vivo using an orthotopic mouse model of MDA-MB-231 cells. Among all tested drug candidates, 8Q and NCS showed remarkable specific anti-CSC activity in terms of CSC viability, migration, invasion and anchorage independent growth reduction in vitro. Moreover, specific 8Q/PTX and NCS/PTX ratios at which both drugs displayed a synergistic effect in different TNBC cell lines were identified. The sole use of PTX increased the relative presence of CSCs in TNBC cells, whereas the combination of 8Q and NCS counteracted this pro-CSC activity of PTX while significantly reducing cell viability. In vivo, the combination of NCS with PTX reduced tumor growth and limited the dissemination of the disease by reducing circulating tumor cells and the incidence of lung metastasis. The combination of 8Q and NCS with PTX at established ratios inhibits both the proliferation of differentiated cancer cells and the viability of CSCs, paving the way for more efficacious TNBC treatments., This work was supported by the Instituto de Salud Carlos III (ISCiii), through Networking Research Center on Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), an initiative that also counts with the assistance from the European Regional Development Fund (ERDF), specifically in the PENTRI-2 Project and by the “Fundació Marató TV3” (337/C/2013) to I.A., M.R. and E.V. Our laboratories were also supported by the Fondo de Investigaciones Sanitarias (FIS, grants PI20/1474 to S.S.J. and PI18/00871 and PI21/00936), co-financed by the ERDF and the 2017-SGR-638 of the Catalan Government to S.S.J. and EvoNano Project (GA800983), funded by European Union’s Horizon 2020 FET Open Programme. N.G.-A. was supported by grants from Pla Estratègic de Recerca i Innovació en Salut (PERIS) of Catalonia (SLT006/17/00270 270).
- Published
- 2021
- Full Text
- View/download PDF
11. Extracellular vesicles from recombinant cell factories improve the activity and efficacy of enzymes defective in lysosomal storage disorders
- Author
-
Rosa Mendoza, Zamira V. Díaz-Riascos, Sandra Mancilla, Lorenzo Albertazzi, Natalia García-Aranda, Ana Boullosa, Antonio Villaverde, Monica Mandaña, Patricia González, Ibane Abasolo, Anna Rosell, Guillem Pintos-Morell, José Luis Corchero, Josefina Casas, Simó Schwartz, Alba Grayston, Joaquin Seras-Franzoso, Roger Riera, Elena García-Fruitós, Marc Moltó-Abad, Institut Català de la Salut, [Seras-Franzoso J, González P, Schwartz S Jr] Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain. [Díaz-Riascos ZV, García-Aranda N, Mandaña M, Boullosa A, Mancilla S, Abasolo I] Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain. Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Corchero JL] Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain. Institut de Biotecnologia i de Biomedicina (IBB) and Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra, Spain. [Grayston A, Rosell A] Neurovascular Research Laboratory, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Moltó-Abad M, Pintos-Morell G] Drug Delivery & Targeting, CIBBIM-Nanomedicine, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Division of Rare Diseases, Reference Center for Hereditary Metabolic Disorders (CSUR, XUEC, MetabERN, and CIBER-ER). Vall d’Hebron Hospital Universitari, Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Male ,0301 basic medicine ,Hydrolases ,Cell ,Sanfilippo syndrome ,Otros calificadores::Otros calificadores::Otros calificadores::/enzimología [Otros calificadores] ,law.invention ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,law ,Cells::Cellular Structures::Extracellular Space::Extracellular Vesicles [ANATOMY] ,Cloning, Molecular ,Research Articles ,Mice, Knockout ,chemistry.chemical_classification ,Trihexosylceramides ,Brain ,Enzyme replacement therapy ,Recombinant Proteins ,Cell biology ,N-sulfoglucosamine sulfohydrolase ,medicine.anatomical_structure ,Metabolisme - Trastorns ,030220 oncology & carcinogenesis ,Recombinant DNA ,Pharmaceutical Vehicles ,Research Article ,Histology ,Globotriaosylceramide ,CHO Cells ,Nutritional and Metabolic Diseases::Metabolic Diseases::Metabolism, Inborn Errors::Lysosomal Storage Diseases [DISEASES] ,Lysosomal storage disorders ,Enzims extracel·lulars - Ús terapèutic ,lysosomal storage disorders ,Extracellular Vesicles ,03 medical and health sciences ,Cricetulus ,In vivo ,medicine ,Animals ,Humans ,Alpha-galactosidase A ,enfermedades nutricionales y metabólicas::enfermedades metabólicas::alteraciones congénitas del metabolismo::enfermedades por almacenamiento lisosómico [ENFERMEDADES] ,Fabry disease ,QH573-671 ,alpha‐galactosidase A ,células::estructuras celulares::espacio extracelular::vesículas extracelulares [ANATOMÍA] ,N‐sulfoglucosamine sulfohydrolase ,Cell Biology ,medicine.disease ,Lysosomal Storage Diseases ,Mice, Inbred C57BL ,HEK293 Cells ,030104 developmental biology ,Enzyme ,chemistry ,alpha-Galactosidase ,Drug delivery ,Lysosomes ,Cytology ,Other subheadings::Other subheadings::Other subheadings::/enzymology [Other subheadings] - Abstract
In the present study the use of extracellular vesicles (EVs) as vehicles for therapeutic enzymes in lysosomal storage disorders was explored. EVs were isolated from mammalian cells overexpressing alpha‐galactosidase A (GLA) or N‐sulfoglucosamine sulfohydrolase (SGSH) enzymes, defective in Fabry and Sanfilippo A diseases, respectively. Direct purification of EVs from cell supernatants was found to be a simple and efficient method to obtain highly active GLA and SGSH proteins, even after EV lyophilization. Likewise, EVs carrying GLA (EV‐GLA) were rapidly uptaken and reached the lysosomes in cellular models of Fabry disease, restoring lysosomal functionality much more efficiently than the recombinant enzyme in clinical use. In vivo, EVs were well tolerated and distributed among all main organs, including the brain. DiR‐labelled EVs were localized in brain parenchyma 1 h after intra‐arterial (internal carotid artery) or intravenous (tail vein) administrations. Moreover, a single intravenous administration of EV‐GLA was able to reduce globotriaosylceramide (Gb3) substrate levels in clinically relevant tissues, such kidneys and brain. Overall, our results demonstrate that EVs from cells overexpressing lysosomal enzymes act as natural protein delivery systems, improving the activity and the efficacy of the recombinant proteins and facilitating their access to organs neglected by conventional enzyme replacement therapies., This study has been supported by ISCIII (PI18_00871 co‐founded by Fondo Europeo de Desarrollo Regional (FEDER)), and CIBER‐BBN (EXPLORE) granted to IA. Different CIBER‐BBN units of ICTS ‘NANBIOSIS’ have participated in this work (https://www.nanbiosis.es/platform-units/), more specifically the U1/Protein Production Platform for protein purification, Unit 6 for NTA analysis and TFF purification and U20/FVPR for in vivo assays.
- Published
- 2021
12. Pivotal role of AKT2 during dynamic phenotypic change of breast cancer stem cells
- Author
-
[Gener P, Seras-Franzoso J, Perez A, Pindado LA, Casas G] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Rafael D] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. [Arango D] Investigació Biomèdica en Tumors de l'Aparell Digestiu, CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Fernández Y] Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Díaz-Riascos Z] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Abasolo I, Schwartz S] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain and Hospital Universitari Vall d'Hebron
- Subjects
Serina-proteases ,Enzimas y Coenzimas::Enzimas::Transferasas::Fosfotransferasas::Fosfotransferasas (Aceptor de Grupo Alcohol)::Proteínas Quinasas::Proteínas Serina-Treonina Quinasas::Proteína Serina-Treonina Quinasas de Interacción con Receptores::Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor [COMPUESTOS QUÍMICOS Y DROGAS] ,Enzymes and Coenzymes::Enzymes::Transferases::Phosphotransferases::Phosphotransferases (Alcohol Group Acceptor)::Protein Kinases::Protein-Serine-Threonine Kinases::Receptor-Interacting Protein Serine-Threonine Kinases::Receptor-Interacting Protein Serine-Threonine Kinase 2 [CHEMICALS AND DRUGS] ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,Mama - Tumors ,Cèl·lules canceroses ,Neoplasias::Neoplasias por Localización::Neoplasias de la Mama [ENFERMEDADES] ,células::células madre::células madre neoplásicas [ANATOMÍA] ,Cells::Stem Cells::Neoplastic Stem Cells [ANATOMY] - Published
- 2021
13. Targeting antitumoral proteins to breast cancer by local administration of functional inclusion bodies
- Author
-
[Pesarrodona M] Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain. CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. [Jauset T, Beaulieu ME] Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Peptomyc S.L. Edifici Cellex, Barcelona, Spain. [Díaz-Riascos ZV, Mancilla S, Fernández Y, Abasolo I] CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. CIBBIM-Nanomedicina Àrea de Validació Funcional i Estudis Preclínics, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona Spain. CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. [Sánchez-Chardi A] Departament de Biologia Evolutiva Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona Spain. [Seras-Franzoso J, Baltà-Foix R] CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Schwartz Jr S] CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Soucek L] Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Peptomyc S.L. Edifici Cellex, Barcelona, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain. Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain and Hospital Universitari Vall d'Hebron
- Subjects
Mama - Càncer ,Proteïnes recombinants ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,células::estructuras celulares::cuerpos de inclusión [ANATOMÍA] ,Neoplasias::Neoplasias por Localización::Neoplasias de la Mama [ENFERMEDADES] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,Cells::Cellular Structures::Inclusion Bodies [ANATOMY] ,Medicaments antineoplàstics - Published
- 2021
14. Sterilization Procedure for Temperature-Sensitive Hydrogels Loaded with Silver Nanoparticles for Clinical Applications
- Author
-
Institut Català de la Salut, [Rafael D] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. [Andrade F] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. [Martinez-Trucharte F] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Basas J] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Seras-Franzoso J] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Palau M] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Gomis X] Laboratori de Resistència Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. [Pérez-Burgos M] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Blanco A] Servei de Cirurgia Ortopèdica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. [López-Fernández A] Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Vélez R] Servei de Cirurgia Ortopèdica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. [Abasolo I] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. [Aguirre M] Servei de Cirugia Ortopedica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Gavaldà J] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Schwartz S Jr] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain., and Hospital Universitari Vall d'Hebron
- Subjects
Col·loides ,Complex Mixtures::Colloids::Gels::Hydrogels [CHEMICALS AND DRUGS] ,Other subheadings::Other subheadings::/prevention & control [Other subheadings] ,Mezclas Complejas::Coloides::Geles::Hidrogeles [COMPUESTOS QUÍMICOS Y DROGAS] ,Otros calificadores::Otros calificadores::/prevención & control [Otros calificadores] ,Other subheadings::/methods [Other subheadings] ,Environment and Public Health::Public Health::Public Health Practice::Communicable Disease Control::Infection Control::Sterilization [HEALTH CARE] ,Bacterial Infections and Mycoses::Infection [DISEASES] ,Otros calificadores::/métodos [Otros calificadores] ,ambiente y salud pública::salud pública::práctica de la salud pública::control de enfermedades transmisibles::control de infecciones::esterilización [ATENCIÓN DE SALUD] ,Esterilització ,Infecció - Prevenció ,Infecciones Bacterianas y Micosis::Infección [ENFERMEDADES] - Published
- 2021
15. Perspectives of nano-carrier drug delivery systems to overcome cancer drug resistance in the clinics
- Author
-
Fernanda Andrade, Ibane Abasolo, Petra Gener, Anna Ulldemolins, Joaquin Seras-Franzoso, Simó Schwartz, Diana Rafael, Institut Català de la Salut, [Ulldemolins A, Seras-Franzoso J] Grup de Distribució i Orientació de Fàrmacs, Centre de Recerca en Biologia Molecular i Bioquímica per a Nanomedicina (CIBBIMNanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. [Andrade F, Rafael D] Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain. [Abasolo I, Gener P, Schwartz Jr S] Grup de Distribució i Orientació de Fàrmacs, Centre de Recerca en Biologia Molecular i Bioquímica per a Nanomedicina (CIBBIMNanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain. Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza 50009, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
fenómenos fisiológicos::fenómenos farmacológicos y toxicológicos::fenómenos farmacológicos::resistencia a medicamentos [FENÓMENOS Y PROCESOS] ,business.industry ,Cancer drugs ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,Nanotechnology ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Neoplasms [DISEASES] ,neoplasias [ENFERMEDADES] ,disciplinas de las ciencias naturales::nanotecnología::nanomedicina [DISCIPLINAS Y OCUPACIONES] ,Nano ,Drug delivery ,Nanomedicina ,Physiological Phenomena::Pharmacological and Toxicological Phenomena::Pharmacological Phenomena::Drug Resistance [PHENOMENA AND PROCESSES] ,Natural Science Disciplines::Nanotechnology::Nanomedicine [DISCIPLINES AND OCCUPATIONS] ,Nanomedicine ,Medicine ,business ,Càncer - Quimioteràpia - Complicacions ,Resistència als medicaments - Abstract
Sistemes d'administració de fàrmacs; Resistència; Tractament del càncer Sistemas de administración de fármacos; Resistencia; Tratamiento del cancer Drug delivery systems; Resistance; Cancer treatment Advanced cancer is still considered an incurable disease because of its metastatic spread to distal organs and progressive gain of chemoresistance. Even though considerable treatment progress and more effective therapies have been achieved over the past years, recurrence in the long-term and undesired side effects are still the main drawbacks of current clinical protocols. Moreover, a majority of chemotherapeutic drugs are highly hydrophobic and need to be diluted in organic solvents, which cause high toxicity, in order to reach effective therapeutic dose. These limitations of conventional cancer therapies prompted the use of nanomedicine, the medical application of nanotechnology, to provide more effective and safer cancer treatment. Potential of nanomedicines to overcome resistance, ameliorate solubility, improve pharmacological profile, and reduce adverse effects of chemotherapeutical drugs is thus highly regarded. Their use in the clinical setting has increased over the last decade. Among the various existing nanosystems, nanoparticles have the ability to transform conventional medicine by reducing the adverse effects and providing a controlled release of therapeutic agents. Also, their small size facilitates the intracellular uptake. Here, we provide a closer review of clinical prospects and mechanisms of action of nanomedicines to overcome drug resistance. The significance of specific targeting towards cancer cells is debated as well.
- Published
- 2020
16. Pivotal Role of AKT2 during Dynamic Phenotypic Change of Breast Cancer Stem Cells
- Author
-
Simó Schwartz, Joaquin Seras-Franzoso, Anna Paradís Pérez, Ibane Abasolo, Petra Gener, Diego Arango, Luis Alamo Pindado, Yolanda Fernández, Glòria Casas, Diana Rafael, Zamira V. Díaz-Riascos, [Gener P, Seras-Franzoso J, Perez A, Pindado LA, Casas G] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Rafael D] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. [Arango D] Investigació Biomèdica en Tumors de l'Aparell Digestiu, CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Fernández Y] Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Díaz-Riascos Z] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. [Abasolo I, Schwartz S] Direccionament i alliberament farmacològic, Nanomedicina Oncologia molecular (CIBBIM-Nanomedicina), Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain. Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. Àrea de Validació Funcional i Estudis Preclínics (FVPR), CIBBIM-Nanomedicina, Vall d’Hebron Institut de Recerca (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
0301 basic medicine ,Cancer Research ,Small interfering RNA ,Serina-proteases ,dynamic phenotype ,Enzymes and Coenzymes::Enzymes::Transferases::Phosphotransferases::Phosphotransferases (Alcohol Group Acceptor)::Protein Kinases::Protein-Serine-Threonine Kinases::Receptor-Interacting Protein Serine-Threonine Kinases::Receptor-Interacting Protein Serine-Threonine Kinase 2 [CHEMICALS AND DRUGS] ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,Epithelial-to-mesenchymal transition (EMT) ,neoplasias::neoplasias por localización::neoplasias de la mama [ENFERMEDADES] ,AKT2 ,Biology ,Cells::Stem Cells::Neoplastic Stem Cells [ANATOMY] ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Circulating tumor cell ,Breast cancer ,Cancer stem cell ,Cancer stem cells (CSC) ,Mama - Tumors ,medicine ,cancer stem cells (CSC) ,Dynamic phenotype ,Mesenchymal stem cell ,Cancer ,AKT2 targeting ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,030104 developmental biology ,Oncology ,enzimas y coenzimas::enzimas::transferasas::fosfotransferasas::fosfotransferasas (grupo alcohol aceptor)::proteína cinasas::proteína-serina-treonina cinasas::proteínas serina-treonina cinasas de interacción con receptores::proteína serina-treonina cinasa 2 de interacción con receptor [COMPUESTOS QUÍMICOS Y DROGAS] ,030220 oncology & carcinogenesis ,Cancer research ,Cèl·lules canceroses ,células::células madre::células madre neoplásicas [ANATOMÍA] ,Stem cell ,epithelial-to-mesenchymal transition (EMT) - Abstract
Cancer stem cells (CSC); Dynamic phenotype; Epithelial-to-mesenchymal transition (EMT) Células madre cancerosas (CSC); Fenotipo dinámico; Transición epitelial a mesenquimal (EMT) Cèl·lules mare canceroses (CSC); Fenotip dinàmic; Transició epitelial a mesenquimal (EMT) Therapeutic resistance seen in aggressive forms of breast cancer remains challenging for current treatments. More than half of the patients suffer from a disease relapse, most of them with distant metastases. Cancer maintenance, resistance to therapy, and metastatic disease seem to be sustained by the presence of cancer stem cells (CSC) within a tumor. The difficulty in targeting this subpopulation derives from their dynamic interconversion process, where CSC can differentiate to non-CSC, which in turn de-differentiate into cells with CSC properties. Using fluorescent CSC models driven by the expression of ALDH1A 1(aldehyde dehydrogenase 1A1), we confirmed this dynamic phenotypic change in MDA-MB-231 breast cancer cells and to identify Serine/Threonine Kinase 2 (AKT2) as an important player in the process. To confirm the central role of AKT2, we silenced AKT2 expression via small interfering RNA and using a chemical inhibitor (CCT128930), in both CSC and non-CSC from different cancer cell lines. Our results revealed that AKT2 inhibition effectively prevents non-CSC reversion through mesenchymal to epithelial transition, reducing invasion and colony formation ability of both, non-CSC and CSC. Further, AKT2 inhibition reduced CSC survival in low attachment conditions. Interestingly, in orthotopic tumor mouse models, high expression levels of AKT2 were detected in circulating tumor cells (CTC). These findings suggest AKT2 as a promising target for future anti-cancer therapies at three important levels: (i) Epithelial-to-mesenchymal transition (EMT) reversion and maintenance of CSC subpopulation in primary tumors, (ii) reduction of CTC and the likelihood of metastatic spread, and (iii) prevention of tumor recurrence through inhibition of CSC tumorigenic and metastatic potential. This work was funded by Fondo de Investigaciones Sanitarias (FIS) from ISCIII, Spanish ministry of Economy and Competitiveness, grant PI17/02242 co-financed by The European Regional Development Fund (FEDER); AC15/00092 grant (Target4Cancer project) from Euro-NanoMed II and PENTRI project, financed by Marato TV3, and EvoNano project, funded by European Union's Horizon 2020 FET Open programme under grant agreement. No. 800983. JSR was supported by a post-doctoral grant from Asociacion Espanola Contra el Cancer (AECC).
- Published
- 2019
17. Sterilization Procedure for Temperature-Sensitive Hydrogels Loaded with Silver Nanoparticles for Clinical Applications
- Author
-
Joaquin Seras-Franzoso, Alberto Blanco, Simó Schwartz, Fernanda Andrade, Marc Pérez-Burgos, Roberto Vélez, Alba López-Fernández, Ibane Abasolo, Francesc Martinez-Trucharte, Marta Palau, Diana Rafael, Jana Basas, Marius Aguirre, Joan Gavaldà, Xavier Gomis, Instituto de Investigação e Inovação em Saúde, Institut Català de la Salut, [Rafael D] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. [Andrade F] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal. [Martinez-Trucharte F] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Basas J] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Seras-Franzoso J] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Palau M] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Gomis X] Laboratori de Resistència Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Servei de Malalties Infeccioses, Hospital Universitari Vall d'Hebron, Barcelona, Spain. [Pérez-Burgos M] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Blanco A] Servei de Cirurgia Ortopèdica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. [López-Fernández A] Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Vélez R] Servei de Cirurgia Ortopèdica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Grup en Enginyeria tissular musculoesquelètica, Vall d’Hebron Institut de Recerca, Barcelona, Spain. [Abasolo I] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain. [Aguirre M] Servei de Cirugia Ortopedica, Hospital Universitari Vall d'Hebron, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Gavaldà J] Laboratori de Resistencia Antimicrobiana, Vall d’Hebron Institut de Recerca, Barcelona, Spain. Spanish Network for Research in Infectious Diseases (REIPI RD16/0016/0003), Instituto de Salud Carlos III, Madrid, Spain. [Schwartz S Jr] Grup Drug Delivery & Targeting (DDT), Vall d’Hebron Institut de Recerca, Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. Networking Research Centre for Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid, Spain., and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Otros calificadores::Otros calificadores::/prevención & control [Otros calificadores] ,silver nanoparticles ,Materials science ,XDR strains ,Other subheadings::/methods [Other subheadings] ,Sterilization procedure ,General Chemical Engineering ,Post-operative infecions ,02 engineering and technology ,Chirurgic applications ,Poloxamer ,Post-operative infections ,Silver nanoparticle ,mezclas complejas::coloides::geles::hidrogeles [COMPUESTOS QUÍMICOS Y DROGAS] ,Autoclave ,Complex Mixtures::Colloids::Gels::Hydrogels [CHEMICALS AND DRUGS] ,Other subheadings::Other subheadings::/prevention & control [Other subheadings] ,03 medical and health sciences ,anti-bacterial agents ,Otros calificadores::/métodos [Otros calificadores] ,Anti-bacterial agents ,General Materials Science ,hydrogels ,Col·loides ,0303 health sciences ,030306 microbiology ,chirurgic applications ,Sterilization ,infecciones bacterianas y micosis::infección [ENFERMEDADES] ,Hydrogels ,sterilization ,Sterilization (microbiology) ,021001 nanoscience & nanotechnology ,Antimicrobial ,Environment and Public Health::Public Health::Public Health Practice::Communicable Disease Control::Infection Control::Sterilization [HEALTH CARE] ,Bacterial Infections and Mycoses::Infection [DISEASES] ,Silver nanopaticles ,ambiente y salud pública::salud pública::práctica de la salud pública::control de enfermedades transmisibles::control de infecciones::esterilización [ATENCIÓN DE SALUD] ,Esterilització ,Drug delivery ,Self-healing hydrogels ,Temperature sensitive ,Silver nanoparticles ,Infecció - Prevenció ,0210 nano-technology ,poloxamer ,post-operative infections ,Biomedical engineering - Abstract
Hydrogels (HG) have recognized benefits as drug delivery platforms for biomedical applications. Their high sensitivity to sterilization processes is however one of the greatest challenges regarding their clinical translation. Concerning infection diseases, prevention of post-operatory related infections is crucial to ensure appropriate patient recovery and good clinical outcomes. Silver nanoparticles (AgNPs) have shown good antimicrobial properties but sustained release at the right place is required. Thus, we produced and characterized thermo-sensitive HG based on Pluronic®, F127 loaded with AgNPs (HG-AgNPs) and their integrity and functionality after sterilization by dry-heat and autoclave methods were carefully assessed. The quality attributes of HG-AgNPs were seriously affected by dry-heat methods but not by autoclaving methods, which allowed to ensure the required sterility. Also, direct sterilization of the final HG-AgNPs product proved more effective than of the raw material, allowing simpler production procedures in non-sterile conditions. The mechanical properties were assessed in post mortem rat models and the HG-AgNPs were tested for its antimicrobial properties in vitro using extremely drug-resistant (XDR) clinical strains. The produced HG-AgNPs prove to be versatile, easy produced and cost-effective products, with activity against XDR strains and an adequate gelation time and spreadability features and optimal for in situ biomedical applications.
- Published
- 2019
18. Targeting antitumoral proteins to breast cancer by local administration of functional inclusion bodies
- Author
-
Pesarrodona, M., Pesarrodona, Mireia, Jauset, Toni, Díaz-Riascos, Zamira V., Sánchez-Chardi, Alejandro, Beaulieu, Marie Eve, Seras-Franzoso, Joaquin, Sánchez-García, Laura, Baltà-Foix, Ricardo, Mancilla, Sandra, Fernández, Yolanda, Rinas, Ursula, Schwartz, Simó, Soucek, Laura, Villaverde, Antonio, Abasolo, Ibane, Vázquez, Esther, [Pesarrodona M] Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, Spain. CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. [Jauset T, Beaulieu ME] Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Peptomyc S.L. Edifici Cellex, Barcelona, Spain. [Díaz-Riascos ZV, Mancilla S, Fernández Y, Abasolo I] CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. CIBBIM-Nanomedicina Àrea de Validació Funcional i Estudis Preclínics, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona Spain. CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. [Sánchez-Chardi A] Departament de Biologia Evolutiva Ecologia i Ciències Ambientals, Facultat de Biologia, Universitat de Barcelona, Barcelona Spain. [Seras-Franzoso J, Baltà-Foix R] CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Schwartz Jr S] CIBER de Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain. CIBBIM-Nanomedicina Direccionament i Alliberament Farmacològic, Vall d'Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Barcelona, Spain. [Soucek L] Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain. Peptomyc S.L. Edifici Cellex, Barcelona, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain. Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, Bellaterra, Spain, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, and HZI,Helmholtz-Zentrum für Infektionsforschung GmbH, Inhoffenstr. 7,38124 Braunschweig, Germany.
- Subjects
Cancer therapy ,General Chemical Engineering ,Neoplasms::Neoplasms by Site::Breast Neoplasms [DISEASES] ,neoplasias::neoplasias por localización::neoplasias de la mama [ENFERMEDADES] ,General Physics and Astronomy ,Medicine (miscellaneous) ,Diseases ,inclusion bodies ,Biofabrication ,02 engineering and technology ,01 natural sciences ,Inclusion bodies ,law.invention ,Medicaments antineoplàstics ,Mama - Càncer ,law ,General Materials Science ,lcsh:Science ,Controlled drug delivery ,Recombinant proteins ,Functional amyloids ,Targeted drug delivery ,Full Paper ,biology ,Chemistry ,General Engineering ,Full Papers ,021001 nanoscience & nanotechnology ,3. Good health ,Human form models ,functional amyloids ,Recombinant DNA ,cancer therapy ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,0210 nano-technology ,ddc:624 ,010402 general chemistry ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Protein drug release ,Breast cancer ,Proteïnes recombinants ,medicine ,biofabrication ,CD44 ,Cancer ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,células::estructuras celulares::cuerpos de inclusión [ANATOMÍA] ,Neoplasias::Neoplasias por Localización::Neoplasias de la Mama [ENFERMEDADES] ,medicine.disease ,0104 chemical sciences ,Protein materials ,biology.protein ,Cancer research ,lcsh:Q ,Protein drugs ,protein drug release ,Cells::Cellular Structures::Inclusion Bodies [ANATOMY] ,ddc:600 ,Human breast - Abstract
Biofabrication; Cancer therapy; Functional amyloids Biofabricación; Terapia contra el cáncer; Amiloides funcionales Biofabricació; Teràpia contra el càncer; Amiloides funcionals Two structurally and functionally unrelated proteins, namely Omomyc and p31, are engineered as CD44-targeted inclusion bodies produced in recombinant bacteria. In this unusual particulate form, both types of protein materials selectively penetrate and kill CD44+ tumor cells in culture, and upon local administration, promote destruction of tumoral tissue in orthotropic mouse models of human breast cancer. These findings support the concept of bacterial inclusion bodies as versatile protein materials suitable for application in chronic diseases that, like cancer, can benefit from a local slow release of therapeutic proteins. This study has been supported by La Fundacio Marato TV3 and NanoCanTri (CIBER-BBN) to E.V. and I.A., and partially by ISCIII (PI15/00272 and PI1702242 co-founded by Fondo Europeo de Desarrollo Regional (FEDER), to E.V. and S.S., respectively), and Agencia Estatal de Investigacion (AEI) and FEDER (BIO2016-76063-R, AEI/FEDER, UE), AGAUR (2017SGR-229) and CIBER-BBN (VENOM4CANCER) granted to A.V. Protein production and DLS have been partially performed by the ICTS "NANBIOSIS," more specifically by the Protein Production Platform of CIBER-BBN/IBB () and the Biomaterial Processing and Nanostructuring Unit (), respectively. Biodistribution and immunohistochemistry assays were performed at the ICTS "NANBIOSIS," specifically by U20/FVPR (). L.S.-G. was supported by predoctoral fellowship from AGAUR (2018FI_B2_00051). L.S. was supported by the European Research Council (CoG #617473) and the Instituto de Salud Carlos III (FIS #PI16/01224). J.S.-F. was supported by an AECC post-doctoral fellowship. A.V. received an ICREA ACADEMIA award
19. 3D Bioprinted Tumor-Stroma Models of Triple-Negative Breast Cancer Stem Cells for Preclinical Targeted Therapy Evaluation.
- Author
-
González-Callejo P, García-Astrain C, Herrero-Ruiz A, Henriksen-Lacey M, Seras-Franzoso J, Abasolo I, and Liz-Marzán LM
- Subjects
- Humans, Female, Tumor Microenvironment drug effects, Cell Line, Tumor, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents therapeutic use, Stromal Cells drug effects, Stromal Cells pathology, Stromal Cells metabolism, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells pathology, Neoplastic Stem Cells metabolism, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms drug therapy, Bioprinting, Printing, Three-Dimensional
- Abstract
Breast cancer stem cells (CSCs) play a pivotal role in therapy resistance and tumor relapse, emphasizing the need for reliable in vitro models that recapitulate the complexity of the CSC tumor microenvironment to accelerate drug discovery. We present a bioprinted breast CSC tumor-stroma model incorporating triple-negative breast CSCs (TNB-CSCs) and stromal cells (human breast fibroblasts), within a breast-derived decellularized extracellular matrix bioink. Comparison of molecular signatures in this model with different clinical subtypes of bioprinted tumor-stroma models unveils a unique molecular profile for artificial CSC tumor models. We additionally demonstrate that the model can recapitulate the invasive potential of TNB-CSC. Surface-enhanced Raman scattering imaging allowed us to monitor the invasive potential of tumor cells in deep z -axis planes, thereby overcoming the depth-imaging limitations of confocal fluorescence microscopy. As a proof-of-concept application, we conducted high-throughput drug testing analysis to assess the efficacy of CSC-targeted therapy in combination with conventional chemotherapeutic compounds. The results highlight the usefulness of tumor-stroma models as a promising drug-screening platform, providing insights into therapeutic efficacy against CSC populations resistant to conventional therapies.
- Published
- 2024
- Full Text
- View/download PDF
20. The Nanotechnology-Based Approaches against Kirsten Rat Sarcoma-Mutated Cancers.
- Author
-
Andrade F, German-Cortés J, Montero S, Carcavilla P, Baranda-Martínez-Abascal D, Moltó-Abad M, Seras-Franzoso J, Díaz-Riascos ZV, Rafael D, and Abasolo I
- Abstract
Kirsten rat sarcoma (KRAS) is a small GTPase which acts as a molecular switch to regulate several cell biological processes including cell survival, proliferation, and differentiation. Alterations in KRAS have been found in 25% of all human cancers, with pancreatic cancer (90%), colorectal cancer (45%), and lung cancer (35%) being the types of cancer with the highest mutation rates. KRAS oncogenic mutations are not only responsible for malignant cell transformation and tumor development but also related to poor prognosis, low survival rate, and resistance to chemotherapy. Although different strategies have been developed to specifically target this oncoprotein over the last few decades, almost all of them have failed, relying on the current therapeutic solutions to target proteins involved in the KRAS pathway using chemical or gene therapy. Nanomedicine can certainly bring a solution for the lack of specificity and effectiveness of anti-KRAS therapy. Therefore, nanoparticles of different natures are being developed to improve the therapeutic index of drugs, genetic material, and/or biomolecules and to allow their delivery specifically into the cells of interest. The present work aims to summarize the most recent advances related to the use of nanotechnology for the development of new therapeutic strategies against KRAS-mutated cancers.
- Published
- 2023
- Full Text
- View/download PDF
21. Extracellular vesicles secreted by triple-negative breast cancer stem cells trigger premetastatic niche remodeling and metastatic growth in the lungs.
- Author
-
González-Callejo P, Gener P, Díaz-Riascos ZV, Conti S, Cámara-Sánchez P, Riera R, Mancilla S, García-Gabilondo M, Peg V, Arango D, Rosell A, Labernadie A, Trepat X, Albertazzi L, Schwartz S Jr, Seras-Franzoso J, and Abasolo I
- Subjects
- Humans, Cell Line, Tumor, Neoplastic Stem Cells metabolism, Lung pathology, Tumor Microenvironment, Triple Negative Breast Neoplasms pathology, Extracellular Vesicles pathology
- Abstract
Tumor secreted extracellular vesicles (EVs) are potent intercellular signaling platforms. They are responsible for the accommodation of the premetastatic niche (PMN) to support cancer cell engraftment and metastatic growth. However, complex cancer cell composition within the tumor increases also the heterogeneity among cancer secreted EVs subsets, a functional diversity that has been poorly explored. This phenomenon is particularly relevant in highly plastic and heterogenous triple-negative breast cancer (TNBC), in which a significant representation of malignant cancer stem cells (CSCs) is displayed. Herein, we selectively isolated and characterized EVs from CSC or differentiated cancer cells (DCC; EVs
CSC and EVsDCC , respectively) from the MDA-MB-231 TNBC cell line. Our results showed that EVsCSC and EVsDCC contain distinct bioactive cargos and therefore elicit a differential effect on stromal cells in the TME. Specifically, EVsDCC activated secretory cancer associated fibroblasts (CAFs), triggering IL-6/IL-8 signaling and sustaining CSC phenotype maintenance. Complementarily, EVsCSC promoted the activation of α-SMA+ myofibroblastic CAFs subpopulations and increased the endothelial remodeling, enhancing the invasive potential of TNBC cells in vitro and in vivo. In addition, solely the EVsCSC mediated signaling prompted the transformation of healthy lungs into receptive niches able to support metastatic growth of breast cancer cells., (© 2023 The Authors. International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC.)- Published
- 2023
- Full Text
- View/download PDF
22. Intracellular Delivery of Anti-Kirsten Rat Sarcoma Antibodies Mediated by Polymeric Micelles Exerts Strong In Vitro and In Vivo Anti-Tumorigenic Activity in Kirsten Rat Sarcoma-Mutated Cancers.
- Author
-
Rafael D, Montero S, Carcavilla P, Andrade F, German-Cortés J, Diaz-Riascos ZV, Seras-Franzoso J, Llaguno M, Fernández B, Pereira A, Duran-Lara EF, Schwartz S Jr, and Abasolo I
- Subjects
- Animals, Mice, Carcinogenesis, Cell Proliferation, Micelles, Mutation, Polymers pharmacology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Proto-Oncogene Proteins p21(ras) pharmacology, Intracellular Space, Colorectal Neoplasms pathology, Neoplasms
- Abstract
The Kirsten rat sarcoma viral oncogene (KRAS) is one of the most well-known proto-oncogenes, frequently mutated in pancreatic and colorectal cancers, among others. We hypothesized that the intracellular delivery of anti-KRAS antibodies (KRAS-Ab) with biodegradable polymeric micelles (PM) would block the overactivation of the KRAS-associated cascades and revert the effect of its mutation. To this end, PM-containing KRAS-Ab (PM-KRAS) were obtained using Pluronic F127. The feasibility of using PM for antibody encapsulation as well as the conformational change of the polymer and its intermolecular interactions with the antibodies was studied, for the first time, using in silico modeling. In vitro , encapsulation of KRAS-Ab allowed their intracellular delivery in different pancreatic and colorectal cancer cell lines. Interestingly, PM-KRAS promoted a high proliferation impairment in regular cultures of KRAS-mutated HCT116 and MIA PaCa-2 cells, whereas the effect was neglectable in non-mutated or KRAS-independent HCT-8 and PANC-1 cancer cells, respectively. Additionally, PM-KRAS induced a remarkable inhibition of the colony formation ability in low-attachment conditions in KRAS-mutated cells. In vivo, when compared with the vehicle, the intravenous administration of PM-KRAS significantly reduced tumor volume growth in HCT116 subcutaneous tumor-bearing mice. Analysis of the KRAS-mediated cascade in cell cultures and tumor samples showed that the effect of PM-KRAS was mediated by a significant reduction of the ERK phosphorylation and a decrease in expression in the stemness-related genes. Altogether, these results unprecedently demonstrate that the delivery of KRAS-Ab mediated by PM can safely and effectively reduce the tumorigenicity and the stemness properties of KRAS-dependent cells, thus bringing up new possibilities to reach undruggable intracellular targets.
- Published
- 2023
- Full Text
- View/download PDF
23. Cancer stem cell-derived extracellular vesicles preferentially target MHC-II-macrophages and PD1+ T cells in the tumor microenvironment.
- Author
-
Gonzalez-Callejo P, Guo Z, Ziglari T, Claudio NM, Nguyen KH, Oshimori N, Seras-Franzoso J, and Pucci F
- Subjects
- Humans, Squamous Cell Carcinoma of Head and Neck pathology, T-Lymphocytes pathology, Tumor Microenvironment, Cell Line, Tumor, Neoplasm Recurrence, Local pathology, Neoplastic Stem Cells metabolism, Carcinoma, Squamous Cell pathology, Head and Neck Neoplasms pathology, Extracellular Vesicles pathology
- Abstract
Immunotherapy is an approved treatment option for head and neck squamous cell carcinoma (HNSCC). However, the response rate to immune checkpoint blockade is only 13% for recurrent HNSCC, highlighting the urgent need to better understand tumor-immune interplay, with the ultimate goal of improving patient outcomes. HNSCC present high local recurrence rates and therapy resistance that can be attributed to the presence of cancer stem cells (CSC) within tumors. CSC exhibit singular properties that enable them to avoid immune detection and eradication. How CSC communicate with immune cells and which immune cell types are preferentially found within the CSC niche are still open questions. Here, we used genetic approaches to specifically label CSC-derived extracellular vesicles (EVs) and to perform Sortase-mediated in vivo proximity labeling of CSC niche cells. We identified specific immune cell subsets that were selectively targeted by EVCSC and that were found in the CSC niche. Native EVCSC preferentially targeted MHC-II-macrophages and PD1+ T cells in the tumor microenvironment, which were the same immune cell subsets enriched within the CSC niche. These observations indicate that the use of genetic technologies able to track EVs without in vitro isolation are a valuable tool to unveil the biology of native EVCSC., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Gonzalez-Callejo et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
24. Selectively Targeting Breast Cancer Stem Cells by 8-Quinolinol and Niclosamide.
- Author
-
Cámara-Sánchez P, Díaz-Riascos ZV, García-Aranda N, Gener P, Seras-Franzoso J, Giani-Alonso M, Royo M, Vázquez E, Schwartz S Jr, and Abasolo I
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation, Humans, Mice, Neoplastic Stem Cells metabolism, Niclosamide pharmacology, Niclosamide therapeutic use, Oxyquinoline, Triple Negative Breast Neoplasms pathology
- Abstract
Cancer maintenance, metastatic dissemination and drug resistance are sustained by cancer stem cells (CSCs). Triple negative breast cancer (TNBC) is the breast cancer subtype with the highest number of CSCs and the poorest prognosis. Here, we aimed to identify potential drugs targeting CSCs to be further employed in combination with standard chemotherapy in TNBC treatment. The anti-CSC efficacy of up to 17 small drugs was tested in TNBC cell lines using cell viability assays on differentiated cancer cells and CSCs. Then, the effect of 2 selected drugs (8-quinolinol -8Q- and niclosamide -NCS-) in the cancer stemness features were evaluated using mammosphere growth, cell invasion, migration and anchorage-independent growth assays. Changes in the expression of stemness genes after 8Q or NCS treatment were also evaluated. Moreover, the potential synergism of 8Q and NCS with PTX on CSC proliferation and stemness-related signaling pathways was evaluated using TNBC cell lines, CSC-reporter sublines, and CSC-enriched mammospheres. Finally, the efficacy of NCS in combination with PTX was analyzed in vivo using an orthotopic mouse model of MDA-MB-231 cells. Among all tested drug candidates, 8Q and NCS showed remarkable specific anti-CSC activity in terms of CSC viability, migration, invasion and anchorage independent growth reduction in vitro. Moreover, specific 8Q/PTX and NCS/PTX ratios at which both drugs displayed a synergistic effect in different TNBC cell lines were identified. The sole use of PTX increased the relative presence of CSCs in TNBC cells, whereas the combination of 8Q and NCS counteracted this pro-CSC activity of PTX while significantly reducing cell viability. In vivo, the combination of NCS with PTX reduced tumor growth and limited the dissemination of the disease by reducing circulating tumor cells and the incidence of lung metastasis. The combination of 8Q and NCS with PTX at established ratios inhibits both the proliferation of differentiated cancer cells and the viability of CSCs, paving the way for more efficacious TNBC treatments.
- Published
- 2022
- Full Text
- View/download PDF
25. Purification of Inclusion Bodies Produced in Bacteria and Yeast.
- Author
-
Seras-Franzoso J, Cano-Garrido O, Peternel S, Arís A, and Garcia-Fruitós E
- Subjects
- Bacteria metabolism, Biotechnology, Pichia metabolism, Recombinant Proteins metabolism, Inclusion Bodies metabolism, Saccharomyces cerevisiae
- Abstract
Purification of inclusion bodies (IBs) is gaining importance due to the raising of novel applications for these submicron particulate protein clusters, with potential uses in the biomedical and biotechnological fields among others. Here, we present five optimized methods to purify IBs adapting classical procedures to the material nature, as well as the requirements of the producer cell (Gram-negative bacteria, Gram-positive bacteria, or yeast) and the IB final application., (© 2022. Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
26. Polylactide, Processed by a Foaming Method Using Compressed Freon R134a, for Tissue Engineering.
- Author
-
Aguado M, Saldaña L, Pérez Del Río E, Guasch J, Parera M, Córdoba A, Seras-Franzoso J, Cano-Garrido O, Vázquez E, Villaverde A, Veciana J, Ratera I, Vilaboa N, and Ventosa N
- Abstract
Fabricating polymeric scaffolds using cost-effective manufacturing processes is still challenging. Gas foaming techniques using supercritical carbon dioxide (scCO
2 ) have attracted attention for producing synthetic polymer matrices; however, the high-pressure requirements are often a technological barrier for its widespread use. Compressed 1,1,1,2-tetrafluoroethane, known as Freon R134a, offers advantages over CO2 in manufacturing processes in terms of lower pressure and temperature conditions and the use of low-cost equipment. Here, we report for the first time the use of Freon R134a for generating porous polymer matrices, specifically polylactide (PLA). PLA scaffolds processed with Freon R134a exhibited larger pore sizes, and total porosity, and appropriate mechanical properties compared with those achieved by scCO2 processing. PLGA scaffolds processed with Freon R134a were highly porous and showed a relatively fragile structure. Human mesenchymal stem cells (MSCs) attached to PLA scaffolds processed with Freon R134a, and their metabolic activity increased during culturing. In addition, MSCs displayed spread morphology on the PLA scaffolds processed with Freon R134a, with a well-organized actin cytoskeleton and a dense matrix of fibronectin fibrils. Functionalization of Freon R134a-processed PLA scaffolds with protein nanoparticles, used as bioactive factors, enhanced the scaffolds' cytocompatibility. These findings indicate that gas foaming using compressed Freon R134a could represent a cost-effective and environmentally friendly fabrication technology to produce polymeric scaffolds for tissue engineering approaches.- Published
- 2021
- Full Text
- View/download PDF
27. Pluronic F127 micelles improve the stability and enhance the anticancer stem cell efficacy of citral in breast cancer.
- Author
-
Abu-Serie MM, Andrade F, Cámara-Sánchez P, Seras-Franzoso J, Rafael D, Díaz-Riascos ZV, Gener P, Abasolo I, and Schwartz S Jr
- Subjects
- Acyclic Monoterpenes, Cell Line, Tumor, Female, Humans, Neoplastic Stem Cells, Poloxamer, Breast Neoplasms drug therapy, Micelles
- Abstract
Aim: Improving the stability and anti-cancer stem cell (CSC) activity of citral, a natural ALDH1A inhibitor. Materials & methods: Citral-loaded micelles (CLM) were obtained using Pluronic
® F127 and its efficacy tested on the growth of four breast cancer cell lines. The impact of the CLM on the growth and functional hallmarks of breast CSCs were also evaluated using mammosphere and CSC reporter cell lines. Results: CLM improved the stability and growth inhibitory effects of citral. Importantly, CLM fully blocking the stemness features of CSCs (self-renewal, differentiation and migration) and in combination with paclitaxel CLM sensitized breast cancer cells to the chemotherapy. Conclusion: Targeting CSCs with CLM could improve the treatment of advanced breast cancer in combination with the standard chemotherapy.- Published
- 2021
- Full Text
- View/download PDF
28. Hyaluronic acid (HA)-coated naproxen-nanoparticles selectively target breast cancer stem cells through COX-independent pathways.
- Author
-
Espinosa-Cano E, Huerta-Madroñal M, Cámara-Sánchez P, Seras-Franzoso J, Schwartz S Jr, Abasolo I, San Román J, and Aguilar MR
- Subjects
- Cell Line, Tumor, Endothelial Cells, Glycogen Synthase Kinase 3 beta, Humans, Hyaluronan Receptors, Hyaluronic Acid, Naproxen pharmacology, Neoplastic Stem Cells, Antineoplastic Agents pharmacology, Breast Neoplasms drug therapy, Nanoparticles
- Abstract
Cytotoxic chemotherapy continues to be the main therapeutic option for patients with metastatic breast cancer. Several studies have reported a significant association between chronic inflammation, carcinogenesis and the presence of cancer stem cells (CSC). We hypothesized that the use of non-steroidal anti-inflammatory drugs targeted to the CSC population could help reducing tumor progression and dissemination in otherwise hard to treat metastatic breast cancer. Within this study cationic naproxen (NAP)-bearing polymeric nanoparticles (NPs) were obtained by self-assembly and they were coated with hyaluronic acid (HA) via electrostatic interaction. HA-coated and uncoated NAP-bearing NPs with different sizes were produced by changing the ionic strength of the aqueous preparation solutions (i.e. 300 and 350 nm or 100 and 130 nm in diameter, respectively). HA-NPs were fully characterized in terms of physicochemical parameters and biological response in cancer cells, macrophages and endothelial cells. Our results revealed that HA-coating of NPs provided a better control in NAP release and improved their hemocompatibility, while ensuring a strong CSC-targeting in MCF-7 breast cancer cells. Furthermore, the best polymeric NPs formulation significantly (p < 0.001) reduced MCF-7 cells viability when compared to free drug (i.e. 45 ± 6% for S-HA-NPs and 87 ± 10% for free NAP) by p53-dependent induction of apoptosis; and the migration of these cell line was also significantly (p < 0.01) reduced by the nano-formulated NAP (i.e. 76.4% of open wound for S-HA-NPs and 61.6% of open wound for NAP). This increased anti-cancer activity of HA-NAP-NPs might be related to the induction of apoptosis through alterations of the GSK-3β-related COX-independent pathway. Overall, these findings suggest that the HA-NAP-NPs have the potential to improve the treatment of advanced breast cancer by increasing the anti-proliferative effect of NAP within the CSC subpopulation., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
29. Nanotechnology-based approaches for treating lysosomal storage disorders, a focus on Fabry disease.
- Author
-
Abasolo I, Seras-Franzoso J, Moltó-Abad M, Díaz-Riascos V, Corchero JL, Pintos-Morell G, and Schwartz S Jr
- Subjects
- Humans, Lysosomes, Enzyme Replacement Therapy, Fabry Disease drug therapy, Lysosomal Storage Diseases drug therapy, Nanomedicine
- Abstract
Lysosomal storage disorders (LSDs) are a group of rare diseases in which the defect of a lysosomal protein results in a pathogenic accumulation of nonmetabolized products within the cells. The main treatment for LSDs is enzyme replacement therapy (ERT), consisting in the exogenous administration a recombinant protein to replace the defective one. Although several diseases such as Gaucher, Fabry, and Pompe are treated following this approach, ERT is limited to LSDs without severe neuronal affectation because recombinant enzymes do not cross the blood-brain barrier. Moreover, ERT shows additional drawbacks, including enzyme low half-life, poor bioavailability, and immunogenic responses. In this scenario, nanotechnology-based drug delivery systems (DDS) have been proposed as solution to overcome these limitations and improve the efficacy of ERT. The present review summarizes distinct approaches followed by our group and collaborators on the use of DDS for restoring lysosomal enzymes in disease-affected cells. During the last decade, we have been exploring different synthetic nanoparticles, from electrolytic complexes, to liposomes and aggresomes, for the delivery of α-galactosidase A (GLA) enzyme. Studies were mainly conducted on Fabry disease models, but results can be also extrapolated to other LSDs, as well as to other diseases treated with alternative therapeutic proteins. The advantages and disadvantages of different DDS, the difficulties from working with very labile and highly glycosylated enzymes and the relevance of using appropriate targeting moieties is thoroughly discussed. Finally, the use of natural DDS, namely extracellular vesicles (EVs) is also introduced. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies., (© 2020 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
30. Perspectives of nano-carrier drug delivery systems to overcome cancer drug resistance in the clinics.
- Author
-
Ulldemolins A, Seras-Franzoso J, Andrade F, Rafael D, Abasolo I, Gener P, and Schwartz S Jr
- Abstract
Advanced cancer is still considered an incurable disease because of its metastatic spread to distal organs and progressive gain of chemoresistance. Even though considerable treatment progress and more effective therapies have been achieved over the past years, recurrence in the long-term and undesired side effects are still the main drawbacks of current clinical protocols. Moreover, a majority of chemotherapeutic drugs are highly hydrophobic and need to be diluted in organic solvents, which cause high toxicity, in order to reach effective therapeutic dose. These limitations of conventional cancer therapies prompted the use of nanomedicine, the medical application of nanotechnology, to provide more effective and safer cancer treatment. Potential of nanomedicines to overcome resistance, ameliorate solubility, improve pharmacological profile, and reduce adverse effects of chemotherapeutical drugs is thus highly regarded. Their use in the clinical setting has increased over the last decade. Among the various existing nanosystems, nanoparticles have the ability to transform conventional medicine by reducing the adverse effects and providing a controlled release of therapeutic agents. Also, their small size facilitates the intracellular uptake. Here, we provide a closer review of clinical prospects and mechanisms of action of nanomedicines to overcome drug resistance. The significance of specific targeting towards cancer cells is debated as well., Competing Interests: All authors declared that there are no conflicts of interest., (© The Author(s) 2021.)
- Published
- 2021
- Full Text
- View/download PDF
31. Polymeric micelles targeted against CD44v6 receptor increase niclosamide efficacy against colorectal cancer stem cells and reduce circulating tumor cells in vivo.
- Author
-
Andrade F, Rafael D, Vilar-Hernández M, Montero S, Martínez-Trucharte F, Seras-Franzoso J, Díaz-Riascos ZV, Boullosa A, García-Aranda N, Cámara-Sánchez P, Arango D, Nestor M, Abasolo I, Sarmento B, and Schwartz S Jr
- Subjects
- Cell Line, Tumor, Humans, Hyaluronan Receptors, Micelles, Neoplastic Stem Cells, Niclosamide, Colorectal Neoplasms drug therapy, Neoplastic Cells, Circulating
- Abstract
Colorectal cancer (CRC) is a highly prevalent disease worldwide. Patient survival is hampered by tumor relapse and the appearance of drug-resistant metastases, which are sustained by the presence of cancer stem cells (CSC). Specific delivery of anti-CSC chemotherapeutic drugs to tumors by using targeted drug delivery systems that can also target CSC sub-population might substantially improve current clinical outcomes. CD44v6 is a robust biomarker for advanced CRC and CSC, due to its functional role in tumorigenesis and cancer initiation process. Here, we show that CD44v6-targeted polymeric micelles (PM) loaded with niclosamide (NCS), a drug against CSC, is a good therapeutic strategy against colorectal CSC and circulating tumor cells (CTC) in vivo. HCT116 cells were sorted according to their CD44v6 receptor expression into CD44v6+ (high) and CDv44v6- (low) subpopulations. Accordingly, CD44v6+ cells presented stemness properties, such as overexpression of defined stemness markers (ALDH1A1, CD44v3 and CXCR4) and high capacity to form colonspheres in low attachment conditions. NCS-loaded PM functionalized with an antibody fragment against CD44v6 (Fab-CD44v6) presented adequate size, charge, and encapsulation efficiency. In addition, Fab-CD44v6 significantly increased PM internalization in CD44v6+ cells. Further, encapsulation of NCS improved its effectiveness in vitro, particularly against colonspheres, and allowed to increase its intravenous dosage in vivo by increasing the amount of NCS able to be administered without causing toxicity. Remarkably, functionalized PM accumulate in tumors and significantly reduce CTC in vivo. In conclusion, CD44v6 targeted PM meet the essential conditions to become an efficient anti-CSC therapy., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
32. Extracellular vesicles from recombinant cell factories improve the activity and efficacy of enzymes defective in lysosomal storage disorders.
- Author
-
Seras-Franzoso J, Díaz-Riascos ZV, Corchero JL, González P, García-Aranda N, Mandaña M, Riera R, Boullosa A, Mancilla S, Grayston A, Moltó-Abad M, Garcia-Fruitós E, Mendoza R, Pintos-Morell G, Albertazzi L, Rosell A, Casas J, Villaverde A, Schwartz S Jr, and Abasolo I
- Subjects
- Animals, Brain metabolism, CHO Cells, Cloning, Molecular, Cricetulus, Fabry Disease enzymology, Fabry Disease therapy, HEK293 Cells, Humans, Hydrolases metabolism, Lysosomal Storage Diseases enzymology, Lysosomes, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Recombinant Proteins administration & dosage, Recombinant Proteins metabolism, Recombinant Proteins therapeutic use, Trihexosylceramides metabolism, alpha-Galactosidase metabolism, Extracellular Vesicles metabolism, Extracellular Vesicles transplantation, Lysosomal Storage Diseases therapy, Pharmaceutical Vehicles metabolism
- Abstract
In the present study the use of extracellular vesicles (EVs) as vehicles for therapeutic enzymes in lysosomal storage disorders was explored. EVs were isolated from mammalian cells overexpressing alpha-galactosidase A (GLA) or N-sulfoglucosamine sulfohydrolase (SGSH) enzymes, defective in Fabry and Sanfilippo A diseases, respectively. Direct purification of EVs from cell supernatants was found to be a simple and efficient method to obtain highly active GLA and SGSH proteins, even after EV lyophilization. Likewise, EVs carrying GLA (EV-GLA) were rapidly uptaken and reached the lysosomes in cellular models of Fabry disease, restoring lysosomal functionality much more efficiently than the recombinant enzyme in clinical use. In vivo, EVs were well tolerated and distributed among all main organs, including the brain. DiR-labelled EVs were localized in brain parenchyma 1 h after intra-arterial (internal carotid artery) or intravenous (tail vein) administrations. Moreover, a single intravenous administration of EV-GLA was able to reduce globotriaosylceramide (Gb3) substrate levels in clinically relevant tissues, such kidneys and brain. Overall, our results demonstrate that EVs from cells overexpressing lysosomal enzymes act as natural protein delivery systems, improving the activity and the efficacy of the recombinant proteins and facilitating their access to organs neglected by conventional enzyme replacement therapies., Competing Interests: Joaquin Seras‐Franzoso, José Luis Corchero, Simó Schwartz Jr and Ibane Abasolo are co‐applicants of a patent describing the use of engineered EV for the producing highly active enzymes (P201930056, 24/01/2019)., (© 2021 The Authors. Journal of Extracellular Vesicles published by Wiley Periodicals, LLC on behalf of the International Society for Extracellular Vesicles.)
- Published
- 2021
- Full Text
- View/download PDF
33. The potential of nanomedicine to alter cancer stem cell dynamics: the impact of extracellular vesicles.
- Author
-
Gener P, Gonzalez Callejo P, Seras-Franzoso J, Andrade F, Rafael D, Abasolo I, and Schwartz S Jr
- Subjects
- Humans, Nanomedicine, Neoplastic Stem Cells, Tumor Microenvironment, Extracellular Vesicles, Neoplasms therapy
- Abstract
The presence of highly resistant cancer stem cells (CSCs) within tumors as drivers of metastatic spread has been commonly accepted. Nonetheless, the likelihood of its dynamic phenotype has been strongly discussed. Importantly, intratumoral cell-to-cell communication seems to act as the main regulatory mechanism of CSC reversion. Today, new strategies for cancer treatment focusing into modulating tumor cell intercommunication and the possibility to modulate the composition of the tumor microenvironment are being explored. In this review, we summarize the literature describing the phenomenon of CSC reversion and the factors known to influence this phenotypic switch. Furthermore, we will discuss the possible role of nanomedicine toward altering this reversion, and to influence the tumor microenvironment composition and the metastatic spread of the disease.
- Published
- 2020
- Full Text
- View/download PDF
34. Extracellular Vesicles as Drug Delivery Systems in Cancer.
- Author
-
Hernandez-Oller L, Seras-Franzoso J, Andrade F, Rafael D, Abasolo I, Gener P, and Schwartz S Jr
- Abstract
Within tumors, Cancer Stem Cell (CSC) subpopulation has an important role in maintaining growth and dissemination while preserving high resistance against current treatments. It has been shown that, when CSCs are eliminated, the surrounding Differentiated Cancer Cells (DCCs) may reverse their phenotype and gain CSC-like features to preserve tumor progression and ensure tumor survival. This strongly suggests the existence of paracrine communication within tumor cells. It is evidenced that the molecular crosstalk is at least partly mediated by Extracellular Vesicles (EVs), which are cell-derived membranous nanoparticles that contain and transport complex molecules that can affect and modify the biological behavior of distal cells and their molecular background. This ability of directional transport of small molecules prospects EVs as natural Drug Delivery Systems (DDS). EVs present inherent homing abilities and are less immunogenic than synthetic nanoparticles, in general. Currently, strong efforts are focused into the development and improvement of EV-based DDS. Even though EV-DDS have already reached early phases in clinical trials, their clinical application is still far from commercialization since protocols for EVs loading, modification and isolation need to be standardized for large-scale production. Here, we summarized recent knowledge regarding the use of EVs as natural DDS against CSCs and cancer resistance.
- Published
- 2020
- Full Text
- View/download PDF
35. Intracellular Delivery of Anti-SMC2 Antibodies against Cancer Stem Cells.
- Author
-
Montero S, Seras-Franzoso J, Andrade F, Martinez-Trucharte F, Vilar-Hernández M, Quesada M, Xandri H, Arango D, Abasolo I, Rafael D, and Schwartz S Jr
- Abstract
Structural maintenance of chromosomes protein 2 (SMC2) is a central component of the condensin complex involved in DNA supercoiling, an essential process for embryonic stem cell survival. SMC2 over-expression has been related with tumorigenesis and cancer malignancy and its inhibition is regarded as a potential therapeutic strategy even though no drugs are currently available. Here, we propose to inhibit SMC2 by intracellular delivery of specific antibodies against the SMC2 protein. This strategy aims to reduce cancer malignancy by targeting cancer stem cells (CSC), the tumoral subpopulation responsible of tumor recurrence and metastasis. In order to prevent degradation and improve cellular internalization, anti-SMC2 antibodies (Ab-SMC2) were delivered by polymeric micelles (PM) based on Pluronic
® F127 amphiphilic polymers. Importantly, scaffolding the Ab-SMC2 onto nanoparticles allowed its cellular internalization and highly increased its efficacy in terms of cytotoxicity and inhibition of tumorsphere formation in MDA-MB-231 and HCT116 breast and colon cancer cell lines, respectively. Moreover, in the case of the HCT116 cell line G1, cell-cycle arrest was also observed. In contrast, no effects from free Ab-SMC2 were detected in any case. Further, combination therapy of anti-SMC2 micelles with paclitaxel (PTX) and 5-Fluorouracil (5-FU) was also explored. For this, PTX and 5-FU were respectively loaded into an anti-SMC2 decorated PM. The efficacy of both encapsulated drugs was higher than their free forms in both the HCT116 and MDA-MB-231 cell lines. Remarkably, micelles loaded with Ab-SMC2 and PTX showed the highest efficacy in terms of inhibition of tumorsphere formation in HCT116 cells. Accordingly, our data clearly suggest an effective intracellular release of antibodies targeting SMC2 in these cell models and, further, strong cytotoxicity against CSC, alone and in combined treatments with Standard-of-Care drugs., Competing Interests: The authors declare no conflict of interest. A patent on the use of SMC2 as biomarker of colorectal cancer was issued by S.S.J. in 2006.- Published
- 2020
- Full Text
- View/download PDF
36. The Biological Potential Hidden in Inclusion Bodies.
- Author
-
Gifre-Renom L, Seras-Franzoso J, Rafael D, Andrade F, Cano-Garrido O, Martinez-Trucharte F, Ugarte-Berzal E, Martens E, Boon L, Villaverde A, Opdenakker G, Schwartz S Jr, Arís A, and Garcia-Fruitós E
- Abstract
Inclusion bodies (IBs) are protein nanoclusters obtained during recombinant protein production processes, and several studies have demonstrated their potential as biomaterials for therapeutic protein delivery. Nevertheless, IBs have been, so far, exclusively sifted by their biological activity in vitro to be considered in further protein-based treatments in vivo. Matrix metalloproteinase-9 (MMP-9) protein, which has an important role facilitating the migration of immune cells, was used as model protein. The MMP-9 IBs were compared with their soluble counterpart and with MMP-9 encapsulated in polymeric-based micelles (PM) through ionic and covalent binding. The soluble MMP-9 and the MMP-9-ionic PM showed the highest activity values in vitro. IBs showed the lowest activity values in vitro, but the specific activity evolution in 50% bovine serum at room temperature proved that they were the most stable format. The data obtained with the use of an air-pouch mouse model showed that MMP-9 IBs presented the highest in vivo activity compared to the soluble MMP-9, which was associated only to a low and a transitory peak of activity. These results demonstrated that the in vivo performance is the addition of many parameters that did not always correlate with the in vitro behavior of the protein of interest, becoming especially relevant at evaluating the potential of IBs as a protein-based nanomaterial for therapeutic purposes.
- Published
- 2020
- Full Text
- View/download PDF
37. Engineering a Nanostructured Nucleolin-Binding Peptide for Intracellular Drug Delivery in Triple-Negative Breast Cancer Stem Cells.
- Author
-
Pesarrodona M, Sánchez-García L, Seras-Franzoso J, Sánchez-Chardi A, Baltá-Foix R, Cámara-Sánchez P, Gener P, Jara JJ, Pulido D, Serna N, Schwartz S Jr, Royo M, Villaverde A, Abasolo I, and Vazquez E
- Subjects
- ADP Ribose Transferases chemistry, ADP Ribose Transferases pharmacology, Antineoplastic Agents chemistry, Antineoplastic Agents metabolism, Antineoplastic Agents pharmacology, Bacterial Toxins chemistry, Bacterial Toxins pharmacology, Cell Line, Tumor, Cell Survival drug effects, Exotoxins chemistry, Exotoxins pharmacology, Female, Humans, Neoplastic Stem Cells cytology, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Peptides metabolism, Peptides pharmacology, Pseudomonas aeruginosa metabolism, Receptors, Cell Surface chemistry, Receptors, Cell Surface metabolism, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Virulence Factors chemistry, Virulence Factors pharmacology, Pseudomonas aeruginosa Exotoxin A, Drug Carriers chemistry, Nanostructures chemistry, Peptides chemistry
- Abstract
Five peptide ligands of four different cell surface receptors (nucleolin, CXCR1, CMKLR1, and CD44v6) have been evaluated as targeting moieties for triple-negative human breast cancers. Among them, the peptide F3, derived from phage display, promotes the fast and efficient internalization of a genetically fused green fluorescent protein (GFP) inside MDA-MB-231 cancer stem cells in a specific receptor-dependent fashion. The further engineering of this protein into the modular construct F3-RK-GFP-H6 and the subsequent construct F3-RK-PE24-H6 resulted in self-assembling polypeptides that organize as discrete and regular nanoparticles. These materials, 15-20 nm in size, show enhanced nucleolin-dependent cell penetrability. We show that the F3-RK-PE24-H6, based on the Pseudomonas aeruginosa exotoxin A (PE24) as a core functional domain, is highly cytotoxic over target cells. The combination of F3, the cationic peptide (RK)
n , and the toxin domain PE24 in such unusual presentation appears as a promising approach to cell-targeted drug carriers in breast cancers and addresses selective drug delivery in otherwise difficult-to-treat triple-negative breast cancers.- Published
- 2020
- Full Text
- View/download PDF
38. Zileuton™ loaded in polymer micelles effectively reduce breast cancer circulating tumor cells and intratumoral cancer stem cells.
- Author
-
Gener P, Montero S, Xandri-Monje H, Díaz-Riascos ZV, Rafael D, Andrade F, Martínez-Trucharte F, González P, Seras-Franzoso J, Manzano A, Arango D, Sayós J, Abasolo I, and Schwartz S Jr
- Subjects
- Animals, Female, Humans, Hydroxyurea chemistry, Hydroxyurea pharmacology, MCF-7 Cells, Mice, Mice, Inbred NOD, Mice, SCID, Xenograft Model Antitumor Assays, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, Hydroxyurea analogs & derivatives, Micelles, Neoplastic Cells, Circulating metabolism, Neoplastic Cells, Circulating pathology, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology
- Abstract
Tumor recurrence, metastatic spread and progressive gain of chemo-resistance of advanced cancers are sustained by the presence of cancer stem cells (CSCs) within the tumor. Targeted therapies with the aim to eradicate these cells are thus highly regarded. However, often the use of new anti-cancer therapies is hampered by pharmacokinetic demands. Drug delivery through nanoparticles has great potential to increase efficacy and reduce toxicity and adverse effects. However, its production has to be based on intelligent design. Likewise, we developed polymeric nanoparticles loaded with Zileuton™, a potent inhibitor of cancer stem cells (CSCs), which was chosen based on high throughput screening. Its great potential for CSCs treatment was subsequently demonstrated in in vitro and in in vivo CSC fluorescent models. Encapsulated Zileuton™ reduces amount of CSCs within the tumor and effectively blocks the circulating tumor cells (CTCs) in the blood stream and metastatic spread., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. High-Throughput Cell Motility Studies on Surface-Bound Protein Nanoparticles with Diverse Structural and Compositional Characteristics.
- Author
-
Tatkiewicz WI, Seras-Franzoso J, García-Fruitós E, Vazquez E, Kyvik AR, Ventosa N, Guasch J, Villaverde A, Veciana J, and Ratera I
- Abstract
Eighty areas with different structural and compositional characteristics made of bacterial inclusion bodies formed by the fibroblast growth factor (FGF-IBs) were simultaneously patterned on a glass surface with an evaporation-assisted method that relies on the coffee-drop effect. The resulting surface patterned with these protein nanoparticles enabled to perform a high-throughput study of the motility of NIH-3T3 fibroblasts under different conditions including the gradient steepness, particle concentrations, and area widths of patterned FGF-IBs, using for the data analysis a methodology that includes "heat maps". From this analysis, we observed that gradients of concentrations of surface-bound FGF-IBs stimulate the total cell movement but do not affect the total net distances traveled by cells. Moreover, cells tend to move toward an optimal intermediate FGF-IB concentration (i.e., cells seeded on areas with high IB concentrations moved toward areas with lower concentrations and vice versa, reaching the optimal concentration). Additionally, a higher motility was obtained when cells were deposited on narrow and highly concentrated areas with IBs. FGF-IBs can be therefore used to enhance and guide cell migration, confirming that the decoration of surfaces with such IB-like protein nanoparticles is a promising platform for regenerative medicine and tissue engineering.
- Published
- 2019
- Full Text
- View/download PDF
40. Pivotal Role of AKT2 during Dynamic Phenotypic Change of Breast Cancer Stem Cells.
- Author
-
Gener P, Rafael D, Seras-Franzoso J, Perez A, Pindado LA, Casas G, Arango D, Fernández Y, Díaz-Riascos ZV, Abasolo I, and Schwartz S Jr
- Abstract
Therapeutic resistance seen in aggressive forms of breast cancer remains challenging for current treatments. More than half of the patients suffer from a disease relapse, most of them with distant metastases. Cancer maintenance, resistance to therapy, and metastatic disease seem to be sustained by the presence of cancer stem cells (CSC) within a tumor. The difficulty in targeting this subpopulation derives from their dynamic interconversion process, where CSC can differentiate to non-CSC, which in turn de-differentiate into cells with CSC properties. Using fluorescent CSC models driven by the expression of ALDH1A 1 (aldehyde dehydrogenase 1A1), we confirmed this dynamic phenotypic change in MDA-MB-231 breast cancer cells and to identify Serine/Threonine Kinase 2 (AKT2) as an important player in the process. To confirm the central role of AKT2, we silenced AKT2 expression via small interfering RNA and using a chemical inhibitor (CCT128930), in both CSC and non-CSC from different cancer cell lines. Our results revealed that AKT2 inhibition effectively prevents non-CSC reversion through mesenchymal to epithelial transition, reducing invasion and colony formation ability of both, non-CSC and CSC. Further, AKT2 inhibition reduced CSC survival in low attachment conditions. Interestingly, in orthotopic tumor mouse models, high expression levels of AKT2 were detected in circulating tumor cells (CTC). These findings suggest AKT2 as a promising target for future anti-cancer therapies at three important levels: (i) Epithelial-to-mesenchymal transition (EMT) reversion and maintenance of CSC subpopulation in primary tumors, (ii) reduction of CTC and the likelihood of metastatic spread, and (iii) prevention of tumor recurrence through inhibition of CSC tumorigenic and metastatic potential., Competing Interests: The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.
- Published
- 2019
- Full Text
- View/download PDF
41. Targeting Antitumoral Proteins to Breast Cancer by Local Administration of Functional Inclusion Bodies.
- Author
-
Pesarrodona M, Jauset T, Díaz-Riascos ZV, Sánchez-Chardi A, Beaulieu ME, Seras-Franzoso J, Sánchez-García L, Baltà-Foix R, Mancilla S, Fernández Y, Rinas U, Schwartz S Jr, Soucek L, Villaverde A, Abasolo I, and Vázquez E
- Abstract
Two structurally and functionally unrelated proteins, namely Omomyc and p31, are engineered as CD44-targeted inclusion bodies produced in recombinant bacteria. In this unusual particulate form, both types of protein materials selectively penetrate and kill CD44
+ tumor cells in culture, and upon local administration, promote destruction of tumoral tissue in orthotropic mouse models of human breast cancer. These findings support the concept of bacterial inclusion bodies as versatile protein materials suitable for application in chronic diseases that, like cancer, can benefit from a local slow release of therapeutic proteins., Competing Interests: T.J. is an employee of Peptomyc S.L.; L.S. and M.E.B. are co‐founders and shareholders of the same company.- Published
- 2019
- Full Text
- View/download PDF
42. Sterilization Procedure for Temperature-Sensitive Hydrogels Loaded with Silver Nanoparticles for Clinical Applications.
- Author
-
Rafael D, Andrade F, Martinez-Trucharte F, Basas J, Seras-Franzoso J, Palau M, Gomis X, Pérez-Burgos M, Blanco A, López-Fernández A, Vélez R, Abasolo I, Aguirre M, Gavaldà J, and Schwartz S Jr
- Abstract
Hydrogels (HG) have recognized benefits as drug delivery platforms for biomedical applications. Their high sensitivity to sterilization processes is however one of the greatest challenges regarding their clinical translation. Concerning infection diseases, prevention of post-operatory related infections is crucial to ensure appropriate patient recovery and good clinical outcomes. Silver nanoparticles (AgNPs) have shown good antimicrobial properties but sustained release at the right place is required. Thus, we produced and characterized thermo-sensitive HG based on Pluronic
® F127 loaded with AgNPs (HG-AgNPs) and their integrity and functionality after sterilization by dry-heat and autoclave methods were carefully assessed. The quality attributes of HG-AgNPs were seriously affected by dry-heat methods but not by autoclaving methods, which allowed to ensure the required sterility. Also, direct sterilization of the final HG-AgNPs product proved more effective than of the raw material, allowing simpler production procedures in non-sterile conditions. The mechanical properties were assessed in post mortem rat models and the HG-AgNPs were tested for its antimicrobial properties in vitro using extremely drug-resistant (XDR) clinical strains. The produced HG-AgNPs prove to be versatile, easy produced and cost-effective products, with activity against XDR strains and an adequate gelation time and spreadability features and optimal for in situ biomedical applications.- Published
- 2019
- Full Text
- View/download PDF
43. AKT2 siRNA delivery with amphiphilic-based polymeric micelles show efficacy against cancer stem cells.
- Author
-
Rafael D, Gener P, Andrade F, Seras-Franzoso J, Montero S, Fernández Y, Hidalgo M, Arango D, Sayós J, Florindo HF, Abasolo I, Schwartz S Jr, and Videira M
- Subjects
- Antineoplastic Agents chemistry, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Cell Line, Tumor, Drug Delivery Systems methods, Female, Gene Transfer Techniques, Humans, MCF-7 Cells, Micelles, Nanomedicine methods, Poloxamer chemistry, Polyethyleneimine chemistry, RNA Interference drug effects, Antineoplastic Agents pharmacology, Neoplastic Stem Cells drug effects, Polymers chemistry, Proto-Oncogene Proteins c-akt genetics, RNA, Small Interfering genetics
- Abstract
Development of RNA interference-based therapies with appropriate therapeutic window remains a challenge for advanced cancers. Because cancer stem cells (CSC) are responsible of sustaining the metastatic spread of the disease to distal organs and the progressive gain of resistance of advanced cancers, new anticancer therapies should be validated specifically for this subpopulation of cells. A new amphihilic-based gene delivery system that combines Pluronic
® F127 micelles with polyplexes spontaneously formed by electrostatic interaction between anionic siRNA and cationic polyethylenimine (PEI) 10K, was designed (PM). Resultant PM gather the requirements for an efficient and safe transport of siRNA in terms of its physicochemical characteristics, internalization capacity, toxicity profile and silencing efficacy. PM were loaded with a siRNA against AKT2, an important oncogene involved in breast cancer tumorigenesis, with a special role in CSC malignancy. Efficacy of siAKT2-PM was validated in CSC isolated from two breast cancer cell lines: MCF-7 and Triple Negative MDA-MB-231 corresponding to an aggressive subtype of breast cancer. In both cases, we observed significant reduction on cell invasion capacity and strong inhibition of mammosphere formation after treatment. These results prompt AKT2 inhibition as a powerful therapeutic target against CSC and pave the way to the appearance of more effective nanomedicine-based gene therapies aimed to prevent CSC-related tumor recurrence.- Published
- 2018
- Full Text
- View/download PDF
44. Dynamism, Sensitivity, and Consequences of Mesenchymal and Stem-Like Phenotype of Cancer Cells.
- Author
-
Gener P, Seras-Franzoso J, Callejo PG, Andrade F, Rafael D, Martínez F, Montero S, Arango D, Sayós J, Abasolo I, and Schwartz S Jr
- Abstract
There are remarkable similarities in the description of cancer stem cells (CSCs) and cancer cells with mesenchymal phenotype. Both cell types are highly tumorigenic, resistant against common anticancer treatment, and thought to cause metastatic growth. Moreover, cancer cells are able to switch between CSC and non-CSC phenotypes and vice versa, to ensure the necessary balance within the tumor. Likewise, cancer cells can switch between epithelial and mesenchymal phenotypes via well-described transition (EMT/MET) that is thought to be crucial for tumor propagation. In this review, we discuss whether, and to which extend, the CSCs and mesenchymal cancer cells are overlapping phenomena in terms of mechanisms, origin, and implication for cancer treatment. As well, we describe the dynamism of both phenotypes and involvement of the tumor microenvironment in CSC reversion and in EMT.
- Published
- 2018
- Full Text
- View/download PDF
45. Surface-Bound Gradient Deposition of Protein Nanoparticles for Cell Motility Studies.
- Author
-
Tatkiewicz WI, Seras-Franzoso J, Garcia-Fruitós E, Vazquez E, Kyvik AR, Guasch J, Villaverde A, Veciana J, and Ratera I
- Subjects
- Cell Movement, Nanoparticles
- Abstract
A versatile evaporation-assisted methodology based on the coffee-drop effect is described to deposit nanoparticles on surfaces, obtaining for the first time patterned gradients of protein nanoparticles (pNPs) by using a simple custom-made device. Fully controllable patterns with specific periodicities consisting of stripes with different widths and distinct nanoparticle concentration as well as gradients can be produced over large areas (∼10 cm
2 ) in a fast (up to 10 mm2 /min), reproducible, and cost-effective manner using an operational protocol optimized by an evolutionary algorithm. The developed method opens the possibility to decorate surfaces "a-la-carte" with pNPs enabling different categories of high-throughput studies on cell motility.- Published
- 2018
- Full Text
- View/download PDF
46. Release of targeted protein nanoparticles from functional bacterial amyloids: A death star-like approach.
- Author
-
Unzueta U, Cespedes MV, Sala R, Alamo P, Sánchez-Chardi A, Pesarrodona M, Sánchez-García L, Cano-Garrido O, Villaverde A, Vázquez E, Mangues R, and Seras-Franzoso J
- Subjects
- Amyloid metabolism, Animals, Bacteria metabolism, Delayed-Action Preparations, Drug Liberation, Female, Humans, Inclusion Bodies metabolism, Mice, Mice, Nude, Proteins chemistry, Receptors, CXCR4 metabolism, Tissue Distribution, Xenograft Model Antitumor Assays, Colorectal Neoplasms metabolism, Drug Delivery Systems, Nanoparticles, Proteins administration & dosage
- Abstract
Sustained release of drug delivery systems (DDS) has the capacity to increase cancer treatment efficiency in terms of drug dosage reduction and subsequent decrease of deleterious side effects. In this regard, many biomaterials are being investigated but none offers morphometric and functional plasticity and versatility comparable to protein-based nanoparticles (pNPs). Here we describe a new DDS by which pNPs are fabricated as bacterial inclusion bodies (IB), that can be easily isolated, subcutaneously injected and used as reservoirs for the sustained release of targeted pNPs. Our approach combines the high performance of pNP, regarding specific cell targeting and biodistribution with the IB supramolecular organization, stability and cost effectiveness. This renders a platform able to provide a sustained source of CXCR4-targeted pNPs that selectively accumulate in tumor cells in a CXCR4
+ colorectal cancer xenograft model. In addition, the proposed system could be potentially adapted to any other protein construct offering a plethora of possible new therapeutic applications in nanomedicine., (Copyright © 2018 Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
47. Bacterial Inclusion Bodies: Discovering Their Better Half.
- Author
-
Rinas U, Garcia-Fruitós E, Corchero JL, Vázquez E, Seras-Franzoso J, and Villaverde A
- Subjects
- Bacteria chemistry, Inclusion Bodies chemistry, Bacteria metabolism, Inclusion Bodies metabolism
- Abstract
Bacterial inclusion bodies (IBs) are functional, non-toxic amyloids occurring in recombinant bacteria showing analogies with secretory granules of the mammalian endocrine system. The scientific interest in these mesoscale protein aggregates has been historically masked by their status as a hurdle in recombinant protein production. However, progressive understanding of how the cell handles the quality of recombinant polypeptides and the main features of their intriguing molecular organization has stimulated the interest in inclusion bodies and spurred their use in diverse technological fields. The engineering and tailoring of IBs as functional protein particles for materials science and biomedicine is a good example of how formerly undesired bacterial byproducts can be rediscovered as promising functional materials for a broad spectrum of applications., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
48. Engineering tumor cell targeting in nanoscale amyloidal materials.
- Author
-
Unzueta U, Seras-Franzoso J, Céspedes MV, Saccardo P, Cortés F, Rueda F, Garcia-Fruitós E, Ferrer-Miralles N, Mangues R, Vázquez E, and Villaverde A
- Subjects
- Amyloid metabolism, Cell Line, Tumor, Drug Delivery Systems methods, Escherichia coli metabolism, Green Fluorescent Proteins chemistry, Green Fluorescent Proteins metabolism, HeLa Cells, Humans, Inclusion Bodies metabolism, Peptides administration & dosage, Peptides chemistry, Peptides metabolism, Receptors, CXCR4 administration & dosage, Receptors, CXCR4 chemistry, Receptors, CXCR4 metabolism, Recombinant Fusion Proteins administration & dosage, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Amyloid administration & dosage, Amyloid chemistry, Inclusion Bodies chemistry, Nanostructures administration & dosage, Nanostructures chemistry, Neoplasms drug therapy
- Abstract
Bacterial inclusion bodies are non-toxic, mechanically stable and functional protein amyloids within the nanoscale size range that are able to naturally penetrate into mammalian cells, where they deliver the embedded protein in a functional form. The potential use of inclusion bodies in protein delivery or protein replacement therapies is strongly impaired by the absence of specificity in cell binding and penetration, thus preventing targeting. To address this issue, we have here explored whether the genetic fusion of two tumor-homing peptides, the CXCR4 ligands R9 and T22, to an inclusion body-forming green fluorescent protein (GFP), would keep the interaction potential and the functionality of the fused peptides and then confer CXCR4 specificity in cell binding and further uptake of the materials. The fusion proteins have been well produced in Escherichia coli in their full-length form, keeping the potential for fluorescence emission of the partner GFP. By using specific inhibitors of CXCR4 binding, we have demonstrated that the engineered protein particles are able to penetrate CXCR4
+ cells, in a receptor-mediated way, without toxicity or visible cytopathic effects, proving the availability of the peptide ligands on the surface of inclusion bodies. Since no further modification is required upon their purification, the biological production of genetically targeted inclusion bodies opens a plethora of cost-effective possibilities in the tissue-specific intracellular transfer of functional proteins through the use of structurally and functionally tailored soft materials.- Published
- 2017
- Full Text
- View/download PDF
49. Nanostructured recombinant cytokines: A highly stable alternative to short-lived prophylactics.
- Author
-
Torrealba D, Parra D, Seras-Franzoso J, Vallejos-Vidal E, Yero D, Gibert I, Villaverde A, Garcia-Fruitós E, and Roher N
- Subjects
- Animals, Chemokine CCL4 chemistry, Cytokines administration & dosage, Cytokines chemistry, Drug Stability, Hydrogen-Ion Concentration, Kinetics, Nanostructures chemistry, Pseudomonas Infections immunology, Pseudomonas Infections pathology, Recombinant Proteins chemistry, Recombinant Proteins genetics, Treatment Outcome, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha immunology, Zebrafish, Chemokine CCL4 administration & dosage, Nanostructures administration & dosage, Protein Engineering methods, Pseudomonas Infections prevention & control, Recombinant Proteins adverse effects, Tumor Necrosis Factor-alpha administration & dosage
- Abstract
Cytokines have been widely used as adjuvants and therapeutic agents in treatments of human diseases. Despite their recognized potential as drugs, the medical use of cytokines has considerable drawbacks, mainly related to their low stability and short half-life. Such intrinsic limitations imply the administration of high doses, often prompting toxicity, undesirable side effects and greater production costs. Here, we describe a new category of mechanically stable nanostructured cytokines (TNFα and CCL4/MIP-1β) that resist harsh physicochemical conditions in vitro (pH and temperature), while maintaining functionality. These bio-functional materials are produced in recombinant cell factories through cost-effective and fully scalable processes. Notably, we demonstrate their prophylactic potential in vivo showing they protect zebrafish from a lethal infection by Pseudomonas aeruginosa., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
50. Bacterial mimetics of endocrine secretory granules as immobilized in vivo depots for functional protein drugs.
- Author
-
Céspedes MV, Fernández Y, Unzueta U, Mendoza R, Seras-Franzoso J, Sánchez-Chardi A, Álamo P, Toledo-Rubio V, Ferrer-Miralles N, Vázquez E, Schwartz S, Abasolo I, Corchero JL, Mangues R, and Villaverde A
- Subjects
- Amyloid metabolism, Amyloidogenic Proteins metabolism, Animals, Apoptosis drug effects, Biomimetics methods, Cell Line, Tumor, Female, HT29 Cells, Humans, Inclusion Bodies metabolism, Mice, Mice, Nude, Nanostructures administration & dosage, Bacteria metabolism, Cytoplasmic Granules metabolism, Endocrine System metabolism, Pharmaceutical Preparations metabolism, Secretory Vesicles metabolism
- Abstract
In the human endocrine system many protein hormones including urotensin, glucagon, obestatin, bombesin and secretin, among others, are supplied from amyloidal secretory granules. These granules form part of the so called functional amyloids, which within the whole aggregome appear to be more abundant than formerly believed. Bacterial inclusion bodies (IBs) are non-toxic, nanostructured functional amyloids whose biological fabrication can be tailored to render materials with defined biophysical properties. Since under physiological conditions they steadily release their building block protein in a soluble and functional form, IBs are considered as mimetics of endocrine secretory granules. We have explored here if the in vivo implantation of functional IBs in a given tissue would represent a stable local source of functional protein. Upon intratumoral injection of bacterial IBs formed by a potent protein ligand of CXCR4 we have observed high stability and prevalence of the material in absence of toxicity, accompanied by apoptosis of CXCR4
+ cells and tumor ablation. Then, the local immobilization of bacterial amyloids formed by therapeutic proteins in tumors or other tissues might represent a promising strategy for a sustained local delivery of protein drugs by mimicking the functional amyloidal architecture of the mammals' endocrine system.- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.