1
|
Mauro N, Calabrese G, Sciortino A, Rizzo MG, Messina F, Giammona G, Cavallaro G. Microporous Fluorescent Poly(D,L-lactide) Acid-Carbon Nanodot Scaffolds for Bone Tissue Engineering Applications. MATERIALS (BASEL, SWITZERLAND) 2024; 17:449. [PMID: 38255617 PMCID: PMC10820564 DOI: 10.3390/ma17020449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024]
Abstract
In this study, we introduce novel microporous poly(D,L-lactide) acid-carbon nanodot (PLA-CD) nanocomposite scaffolds tailored for potential applications in image-guided bone regeneration. Our primary objective was to investigate concentration-dependent structural variations and their relevance to cell growth, crucial aspects in bone regeneration. The methods employed included comprehensive characterization techniques such as DSC/TGA, FTIR, rheological, and degradation assessments, providing insights into the scaffolds' thermoplastic behavior, microstructure, and stability over time. Notably, the PLA-CD scaffolds exhibited distinct self-fluorescence, which persisted after 21 days of incubation, allowing detailed visualization in various multicolor modalities. Biocompatibility assessments were conducted by analyzing human adipose-derived stem cell (hADSC) growth on PLA-CD scaffolds, with results substantiated through cell viability and morphological analyses. hADSCs reached a cell viability of 125% and penetrated throughout the scaffold after 21 days of incubation. These findings underscore the scaffolds' potential in bone regeneration and fluorescence imaging. The multifunctional nature of the PLA-CD nanocomposite, integrating diagnostic capabilities with tunable properties, positions it as a promising candidate for advancing bone tissue engineering. Our study not only highlights key aspects of the investigation but also underscores the scaffolds' specific application in bone regeneration, providing a foundation for further research and optimization in this critical biomedical field.
Collapse
Affiliation(s)
- Nicolò Mauro
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98168 Messina, Italy; (G.C.); (M.G.R.)
| | - Alice Sciortino
- Department of Chimica e Fisica “E. Segrè”, Università Degli Studi di Palermo, Via Archirafi 36, 90123 Palermo, Italy; (A.S.); (F.M.)
| | - Maria G. Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98168 Messina, Italy; (G.C.); (M.G.R.)
| | - Fabrizio Messina
- Department of Chimica e Fisica “E. Segrè”, Università Degli Studi di Palermo, Via Archirafi 36, 90123 Palermo, Italy; (A.S.); (F.M.)
| | - Gaetano Giammona
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| | - Gennara Cavallaro
- Department of “Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche” (STEBICEF), Università Degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy; (G.G.); (G.C.)
| |
Collapse
|
2
|
Choi C, Yun E, Cha C. Emerging Technology of Nanofiber-Composite Hydrogels for Biomedical Applications. Macromol Biosci 2023; 23:e2300222. [PMID: 37530431 DOI: 10.1002/mabi.202300222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/26/2023] [Indexed: 08/03/2023]
Abstract
Hydrogels and nanofibers have been firmly established as go-to materials for various biomedical applications. They have been mostly utilized separately, rarely together, because of their distinctive attributes and shortcomings. However, the potential benefits of integrating nanofibers with hydrogels to synergistically combine their functionalities while attenuating their drawbacks are increasingly recognized. Compared to other nanocomposite materials, incorporating nanofibers into hydrogel has the distinct advantage of emulating the hierarchical structure of natural extracellular environment needed for cell and tissue culture. The most important technological aspect of developing "nanofiber-composite hydrogel" is generating nanofibers made of various polymers that are cross-linked and short enough to maintain stable dispersion in hydrated environment. In this review, recent research efforts to develop nanofiber-composite hydrogels are presented, with added emphasis on nanofiber processing techniques. Several notable examples of implementing nanofiber-composite hydrogels for biomedical applications are also introduced.
Collapse
Affiliation(s)
- Cholong Choi
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Eunhye Yun
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| | - Chaenyung Cha
- Center for Programmable Matter, Department of Materials Science and Engineering, Ulsan National Institute of Science and Technology, Ulsan, 44919, Republic of Korea
| |
Collapse
|
3
|
Guidotti G, Duelen R, Bloise N, Soccio M, Gazzano M, Aluigi A, Visai L, Sampaolesi M, Lotti N. The ad hoc chemical design of random PBS-based copolymers influences the activation of cardiac differentiation while altering the HYPPO pathway target genes in hiPSCs. BIOMATERIALS ADVANCES 2023; 154:213583. [PMID: 37604040 DOI: 10.1016/j.bioadv.2023.213583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 07/23/2023] [Accepted: 08/07/2023] [Indexed: 08/23/2023]
Abstract
Cardiac tissue engineering is a cutting-edge technology aiming to replace irreversibly damaged cardiac tissue and restore contractile functionality. However, cardiac tissue engineering porous and perfusable scaffolds to enable oxygen supply in vitro and eventually promote angiogenesis in vivo are still desirable. Two fully-aliphatic random copolymers of poly(butylene succinate) (PBS), poly(butylene succinate/Pripol), P(BSBPripol), and poly(butylene/neopentyl glycol succinate), P(BSNS), containing two different subunits, neopentyl glycol and Pripol 1009, were successfully synthesized and then electrospun in tridimentional fibrous mats. The copolymers show different thermal and mechanical behaviours as result of their chemical structure. In particular, copolymerization led to a reduction in crystallinity and consequently PBS stiffness, reaching values of elastic modulus very close to those of soft tissues. Then, to check the biological suitability, human induced Pluripotent Stem Cells (hiPSCs) were directly seeded on both PBS-based copolymeric scaffolds. The results confirmed the ability of both the scaffolds to sustain cell viability and to maintain their stemness during cell expansion. Furthermore, gene expression and immunofluorescence analysis showed that P(BSBPripol) scaffold promoted an upregulation of the early cardiac progenitor and later-stage markers with a simultaneously upregulation of HYPPO pathway gene expression, crucial for mechanosensing of cardiac progenitor cells. These results suggest that the correct ad-hoc chemical design and, in turn, the mechanical properties of the matrix, such as substrate stiffness, together with surface porosity, play a critical role in regulating the behaviour of cardiac progenitors, which ultimately offers valuable insights into the development of novel bio-inspired scaffolds for cardiac tissue regeneration.
Collapse
Affiliation(s)
- Giulia Guidotti
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Robin Duelen
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Nora Bloise
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy; Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, Via Salvatore Maugeri 4, 27100 Pavia, Italy
| | - Michelina Soccio
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Via Terracini 28, 40131 Bologna, Italy
| | - Massimo Gazzano
- Organic Synthesis and Photoreactivity Institute, CNR, Via Gobetti 101, 40129 Bologna, Italy
| | - Annalisa Aluigi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Piazza del Rinascimento, 6, 61029 Urbino, (PU), Italy
| | - Livia Visai
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy; Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, Via Salvatore Maugeri 4, 27100 Pavia, Italy
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy.
| | - Nadia Lotti
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Via Terracini 28, 40131 Bologna, Italy.
| |
Collapse
|
4
|
Maggi F, Manfredi A, Carosio F, Maddalena L, Alongi J, Ferruti P, Ranucci E. Toughening Polyamidoamine Hydrogels through Covalent Grafting of Short Silk Fibers. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227808. [PMID: 36431909 PMCID: PMC9696315 DOI: 10.3390/molecules27227808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Linear amphoteric polyamidoamines (PAAs) are usually water-soluble, biodegradable and biocompatible. Crosslinked PAAs form in water hydrogels, retaining most of the favorable properties of their linear counterparts. The hydrogels prepared by the radical post-polymerization of the oligo-α,ω-bisacrylamido-terminated PAA called AGMA1, obtained by the polyaddition of 4-aminobutylguanidine (agmatine) with 2,2-bis(acrylamido)acetic acid, exhibit excellent cell-adhesion properties both in vitro and in vivo. However, due to their low mechanical strength, AGMA1 hydrogels cannot be sewn to biological tissues and need to be reinforced with fibrous materials. In this work, short silk fibers gave excellent results in this sense, proving capable of establishing covalent bonds with the PAA matrix, thanks to their lysine content, which provided amino groups capable of reacting with the terminal acrylamide groups of the AGMA1 precursor in the final crosslinking phase. Morphological analyses demonstrated that the AGMA1 matrix was intimately interconnected and adherent to the silk fibers, with neither visible holes nor empty volumes. The silk/H-AGMA1 composites were still reversibly swellable in water. In the swollen state, they could be sewn and showed no detachment between fibers and matrix and exhibited significantly improved mechanical properties compared with the plain hydrogels, particularly as regards their Young's modulus and elongation at break.
Collapse
Affiliation(s)
- Filippo Maggi
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milano, Italy
| | - Amedea Manfredi
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milano, Italy
| | - Federico Carosio
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Alessandria Campus, Viale Teresa Michel 5, 15121 Alessandria, Italy
| | - Lorenza Maddalena
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Alessandria Campus, Viale Teresa Michel 5, 15121 Alessandria, Italy
| | - Jenny Alongi
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milano, Italy
| | - Paolo Ferruti
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milano, Italy
| | - Elisabetta Ranucci
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milano, Italy
- Correspondence: ; Tel.: +39-0250314132
| |
Collapse
|
5
|
Mauro N, Giammona G, Ranucci E, Ferruti P. Synthesis of Biocompatible and Biodegradable Polyamidoamines Microgels via a Simple and Reliable Statistical Approach. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7280. [PMID: 36295345 PMCID: PMC9611214 DOI: 10.3390/ma15207280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/06/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Polyamidoamines (PAAs) are biocompatible and biodegradable polymers with a huge potential as biomaterials for pharmaceutical applications. They are obtained by the step-wise aza-Michael polyaddition of bifunctional or multifunctional amines with bisacrylamides in water. To the best of our knowledge, no synthetic protocols leading to hyperbranched PAAs as well as PAA microgels have been published so far. To fill this gap, a statistical approach was established in this work to fine-tune the aza-Michael polyaddition stoichiometry when a multifunctional co-monomer (bf) is added to a mixture of bifunctional monomers with complementary functions (a2 + b2), possibly even in presence of a monofunctional co-monomer (b1), for obtaining either microgels or hyperbranched polymers by a one-pot reaction. For this purpose, two new equations, obtained by reworking the classic Flory-Stockmayer equations, were successfully applied to the synthesis of different model systems, obtaining biocompatible microgels with tunable size distribution (200-500 nm) and properly designed end-chains in a simple and straightforward way. The same mathematical approach allowed us to empirically evaluate the actual number of active reactive functions of the co-monomers. A number of selected systems, being evaluated for their cytotoxicity in vitro, proved highly cytocompatible and, therefore, endowed with great potential for pharmaceutical and medical applications.
Collapse
Affiliation(s)
- Nicolò Mauro
- Laboratory of Biocompatible Polymers, Department of “Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche” (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Gaetano Giammona
- Laboratory of Biocompatible Polymers, Department of “Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche” (STEBICEF), University of Palermo, 90123 Palermo, Italy
| | - Elisabetta Ranucci
- Dipartimento di Chimica, Università degli Studi di Milano, 20133 Milano, Italy
| | - Paolo Ferruti
- Dipartimento di Chimica, Università degli Studi di Milano, 20133 Milano, Italy
| |
Collapse
|
6
|
Boon-In S, Theerasilp M, Crespy D. Marrying the incompatible for better: Incorporation of hydrophobic payloads in superhydrophilic hydrogels. J Colloid Interface Sci 2022; 622:75-86. [PMID: 35489103 DOI: 10.1016/j.jcis.2022.04.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/31/2023]
Abstract
HYPOTHESIS The entrapment of lyophobic in superhydrophilic hydrogels is challenging because of the intrinsic incompatibility between hydrophobic and hydrophilic molecules. To achieve such entrapment without affecting the hydrogel's formation, the electrospinning of nanodroplets or nanoparticles with a water-soluble polymer could reduce the incompatibility through the reduction of interfacial tension and the formation of a barrier film preventing coalescence or aggregation. EXPERIMENTS Nanodroplets or nanoparticles dispersion are electrospun in the presence of a hydrophilic polymer in hydrogel precursors. The dissolution of the hydrophilic nanofibers during electrospinning allows a redispersion of emulsion droplets and nanoparticles in the hydrogel's matrix. FINDINGS Superhydrophilic hydrogels with well-distributed hydrophobic nanodroplets or nanoparticles are obtained without detrimentally imparting the viscosity of hydrogel's precursors and the mechanical properties of the hydrogels. Compared with the incorporation of droplets without electrospinning, higher loadings of hydrophobic payload are achieved without premature leakage. This concept can be used to entrap hydrophobic agrochemicals, drugs, or antibacterial agents in simple hydrogels formulation.
Collapse
Affiliation(s)
- Supissra Boon-In
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| | - Man Theerasilp
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| | - Daniel Crespy
- Department of Materials Science and Engineering, School of Molecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC), Rayong 21210, Thailand.
| |
Collapse
|
7
|
A Modular Composite Device of Poly(Ethylene Oxide)/Poly(Butylene Terephthalate) (PEOT/PBT) Nanofibers and Gelatin as a Dual Drug Delivery System for Local Therapy of Soft Tissue Tumors. Int J Mol Sci 2022; 23:ijms23063239. [PMID: 35328661 PMCID: PMC8948985 DOI: 10.3390/ijms23063239] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
In the clinical management of solid tumors, the possibility to successfully couple the regeneration of injured tissues with the elimination of residual tumor cells left after surgery could open doors to new therapeutic strategies. In this work, we present a composite hydrogel-electrospun nanofiber scaffold, showing a modular architecture for the delivery of two pharmaceutics with distinct release profiles, that is potentially suitable for local therapy and post-surgical treatment of solid soft tumors. The composite was obtained by coupling gelatin hydrogels to poly(ethylene oxide)/poly(butylene terephthalate) block copolymer nanofibers. Results of the scaffolds' characterization, together with the analysis of gelatin and drug release kinetics, displayed the possibility to modulate the device architecture to control the release kinetics of the drugs, also providing evidence of their activity. In vitro analyses were also performed using a human epithelioid sarcoma cell line. Furthermore, publicly available expression datasets were interrogated. Confocal imaging showcased the nontoxicity of these devices in vitro. ELISA assays confirmed a modulation of IL-10 inflammation-related cytokine supporting the role of this device in tissue repair. In silico analysis confirmed the role of IL-10 in solid tumors including 262 patients affected by sarcoma as a negative prognostic marker for overall survival. In conclusion, the developed modular composite device may provide a key-enabling technology for the treatment of soft tissue sarcoma.
Collapse
|
8
|
Kim JH, Yun JH, Song ES, Kim SU, Lee HJ, Song YS. Improvement of damaged cavernosa followed by neuron-like differentiation at injured cavernous nerve after transplantation of stem cells seeded on the PLA nanofiber in rats with cavernous nerve injury. Mol Biol Rep 2021; 48:3549-3559. [PMID: 33866496 DOI: 10.1007/s11033-021-06332-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
This study investigated the differentiation of transplanted transplanted mesenchymal stem cells MSCs into neuron-like cells, repair of erectile dysfunction (ED), and synergy of MSCs seeded to nanofibrous scaffolds with after transplantation around the injured cavernous nerve (CN) of rats. The synthesized polymer was electrospun in a rotating drum to prepare nanofiber meshes (NMs). Human MSCs were prepared and confirmed. Eight-week-old male Sprague-Dawley rats were divided into five groups of six each: group 1-sham operation; group 2-CN injury; group 3-MSCs treatment after CN injury; group 4-nanofibrous scaffold treatment after CN injury; and group 5-post-CN injury treatment combining a nanofibrous scaffold and MSCs (nano-MSCs). In the latter group, the damaged CN was instantly surrounded by an MSC-containing a nanofibrous scaffold in the aftermath of injury. Morphological analysis and immuno-histochemical staining in relation to nerves (Tuj1, NF, MAP2, MBP and peripherin), endothelium (vWF), smooth muscle (SMA), neurofilament (NF), and apoptosis (TUNEL) were performed. We evaluated the mean proportion expressed as a percentage of the ratio of muscle to collagen of penile cavernous smooth-muscle cells as well as the expression of cavernous SMA, NF, vWF, and TUNEL makers. Compared to the group free of CN injury, erectile function was markedly reduced in the group with CN injury at 2 and 4 weeks (p < 0.05). By contrast, compared to the sham operation group, erectile function was better in the group with MSC transplantation (p < 0.05). Similarly, by comparison to the group solely with hMSCs, erectile function was better in the group with nano-MSC transplantation (p < 0.05). Transplantation of MSCs demonstrated the neuronal differentiation. By contrast to MSCs on their own, neuronal differentiation was more significantly expressed in nano-MSCs. The mean proportion expressed as a percentage of the ratio of muscle to collagen of penile cavernous smooth-muscle cells, the expression of cavernous SMA, NF, vWF, and apoptosis improved in the cavernosum after transplantation. NMs showed synergy with MSCs for the repair of erectile dysfunction. Transplanted MSCs differentiated into neuron-like cells and repaired erectile dysfunction in the rats with CN injury. Transplanted MSCs increased the mean percentage of the collagen area of the caversnosum as well as the expression levels of cavernous neuronal, endothelial, smooth-muscle markers, and apoptosis.
Collapse
Affiliation(s)
- Jae Heon Kim
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea.,Department of Microbiology, Soonchunhyang University School of Medicine, Cheonan, Republic of Korea
| | - Jong Hyun Yun
- Department of Urology, Soonchunhyang University School of Medicine, Gumi, Republic of Korea
| | - Eun Seop Song
- Korea Medical Dispute Mediation and Abitration Agency, Seoul, Republic of Korea
| | - Seung U Kim
- Division of Neurology, Department of Medicine, UBC Hospital, University of British Columbia, Vancouver, Canada
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea. .,Research Institute, e-Biogen Inc, Seoul, Republic of Korea.
| | - Yun Seob Song
- Department of Urology, Soonchunhyang University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Marcioni M, Alongi J, Ranucci E, Malinconico M, Laurienzo P, Ferruti P, Manfredi A. Semi-Crystalline Hydrophobic Polyamidoamines: A New Family of Technological Materials? Polymers (Basel) 2021; 13:polym13071018. [PMID: 33806055 PMCID: PMC8036605 DOI: 10.3390/polym13071018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/20/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
The hitherto known polyamidoamines (PAAs) are not suitable as structural materials because they are usually water-soluble or swellable in water. This paper deals with the synthesis and characterization of semi-crystalline hydrophobic PAAs (H-PAAs) by combining different bis-sec-amines with bis-acrylamides obtained from C6–C12 bis-prim-amines. H-PAAs were initially obtained in a solution of benzyl alcohol, a solvent suitable for both monomers and polymers. Their number average molecular weights, M¯n, which were determined with 1H-NMR by evaluating the percentage of their terminal units, varied from 6000 to >10,000. The solubility, thermal properties, ignitability and water resistance of H-PAAs were determined. They were soluble in organic solvents, semi-crystalline and thermally stable. The most promising ones were also prepared using a bulk process, which has never been previously reported for PAA synthesis. In the form of films, these H-PAAs were apparently unaffected by water. The films underwent tensile and wettability tests. They showed similar Young moduli (260–263 MPa), whereas the maximum stress and the stress at break depended on the number of methylene groups of the starting bis-acrylamides. Their wettability was somewhat higher than that of common Nylons. Interestingly, none of the H-PAAs considered, either as films or powders, ignited after prolonged exposure to a methane flame.
Collapse
Affiliation(s)
- Massimo Marcioni
- Dipartimento di Chimica, Università Degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy; (M.M.); (J.A.); (E.R.)
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, Alessandria Campus, Viale T. Michel, 15121 Alessandria, Italy
| | - Jenny Alongi
- Dipartimento di Chimica, Università Degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy; (M.M.); (J.A.); (E.R.)
| | - Elisabetta Ranucci
- Dipartimento di Chimica, Università Degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy; (M.M.); (J.A.); (E.R.)
| | - Mario Malinconico
- Istituto Polimeri, Compositi e Biomateriali (IPCB), Consiglio Nazionale Delle Ricerche, via Campi Flegrei 34, 80078 Pozzuoli, Italy; (M.M.); (P.L.)
| | - Paola Laurienzo
- Istituto Polimeri, Compositi e Biomateriali (IPCB), Consiglio Nazionale Delle Ricerche, via Campi Flegrei 34, 80078 Pozzuoli, Italy; (M.M.); (P.L.)
| | - Paolo Ferruti
- Dipartimento di Chimica, Università Degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy; (M.M.); (J.A.); (E.R.)
- Correspondence: (P.F.); (A.M.); Tel.: +39-02-50314128 (P.F.); +39-02-50314181 (A.M.)
| | - Amedea Manfredi
- Dipartimento di Chimica, Università Degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy; (M.M.); (J.A.); (E.R.)
- Correspondence: (P.F.); (A.M.); Tel.: +39-02-50314128 (P.F.); +39-02-50314181 (A.M.)
| |
Collapse
|
10
|
Jia Y, Sciutto G, Mazzeo R, Samorì C, Focarete ML, Prati S, Gualandi C. Organogel Coupled with Microstructured Electrospun Polymeric Nonwovens for the Effective Cleaning of Sensitive Surfaces. ACS APPLIED MATERIALS & INTERFACES 2020; 12:39620-39629. [PMID: 32820898 PMCID: PMC8009474 DOI: 10.1021/acsami.0c09543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/07/2020] [Indexed: 05/21/2023]
Abstract
Hydrogels and organogels are widely used as cleaning materials, especially when a controlled solvent release is necessary to prevent substrate damage. This situation is often encountered in the personal care and electronic components fields and represents a challenge in restoration, where the removal of a thin layer of aged varnish from a painting may compromise the integrity of the painting itself. There is an urgent need for new and effective cleaning materials capable of controlling and limiting the use of solvents, achieving at the same time high cleaning efficacy. In this paper, new sandwich-like composites that fully address these requirements are developed by using an organogel (poly(3-hydroxybutyrate) + γ-valerolactone) in the core and two external layers of electrospun nonwovens made of continuous submicrometric fibers produced by electrospinning (either poly(vinyl alcohol) or polyamide 6,6). The new composite materials exhibit an extremely efficient cleaning action that results in the complete elimination of the varnish layer with a minimal amount of solvent adsorbed by the painting layer after the treatment. This demonstrates that the combined materials exert a superficial action that is of utmost importance to safeguard the painting. Moreover, we found that the electrospun nonwoven layers act as mechanically reinforcement components, greatly improving the bending resistance of organogels and their handling. The characterization of these innovative cleaning materials allowed us to propose a mechanism to explain their action: electrospun fibers play the leading role by slowing down the diffusion of the solvent and by conferring to the entire composite a microstructured rough superficial morphology, enabling to achieve outstanding cleaning performance.
Collapse
Affiliation(s)
- Yiming Jia
- Department
of Chemistry “G. Ciamician”, Microchemistry and Microscopy
Art Diagnostic Laboratory (M2ADL), University
of Bologna, Via Guaccimanni 42, 48121 Ravenna, Italy
- Chongqing
Cultural Heritage Research Institute, 400013 Chongqing, China
| | - Giorgia Sciutto
- Department
of Chemistry “G. Ciamician”, Microchemistry and Microscopy
Art Diagnostic Laboratory (M2ADL), University
of Bologna, Via Guaccimanni 42, 48121 Ravenna, Italy
| | - Rocco Mazzeo
- Department
of Chemistry “G. Ciamician”, Microchemistry and Microscopy
Art Diagnostic Laboratory (M2ADL), University
of Bologna, Via Guaccimanni 42, 48121 Ravenna, Italy
| | - Chiara Samorì
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Sant’Alberto 163, 48123 Ravenna, Italy
| | - Maria Letizia Focarete
- Department
of Chemistry “Giacomo Ciamician” and INSTM UdR of Bologna, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Health
Sciences & Technologies (HST) CIRI, University of Bologna, Via Tolara di Sopra 41/E, 40064 Ozzano Emilia Bologna, Italy
| | - Silvia Prati
- Department
of Chemistry “G. Ciamician”, Microchemistry and Microscopy
Art Diagnostic Laboratory (M2ADL), University
of Bologna, Via Guaccimanni 42, 48121 Ravenna, Italy
| | - Chiara Gualandi
- Department
of Chemistry “Giacomo Ciamician” and INSTM UdR of Bologna, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
- Interdepartmental
Center for Industrial Research on Advanced Applications in Mechanical
Engineering and Materials Technology, CIRI-MAM, University of Bologna, Viale Risorgimento, 2, 40136 Bologna, Italy
| |
Collapse
|
11
|
Xue J, Pisignano D, Xia Y. Maneuvering the Migration and Differentiation of Stem Cells with Electrospun Nanofibers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2000735. [PMID: 32775158 PMCID: PMC7404157 DOI: 10.1002/advs.202000735] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/30/2020] [Indexed: 05/21/2023]
Abstract
Electrospun nanofibers have been extensively explored as a class of scaffolding materials for tissue regeneration, because of their unique capability to mimic some features and functions of the extracellular matrix, including the fibrous morphology and mechanical properties, and to a certain extent the chemical/biological cues. This work reviews recent progress in applying electrospun nanofibers to direct the migration of stem cells and control their differentiation into specific phenotypes. First, the physicochemical properties that make electrospun nanofibers well-suited as a supporting material to expand stem cells by controlling their migration and differentiation are introduced. Then various systems are analyzed in conjunction with mesenchymal, neuronal, and embryonic stem cells, as well as induced pluripotent stem cells. Finally, some perspectives on the challenges and future opportunities in combining electrospun nanofibers with stem cells are offered to address clinical issues.
Collapse
Affiliation(s)
- Jiajia Xue
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
| | - Dario Pisignano
- Dipartimento di FisicaUniversità di PisaLargo B. Pontecorvo 3PisaI‐56127Italy
- NESTIstituto Nanoscienze‐CNRPiazza S. Silvestro 12PisaI‐56127Italy
| | - Younan Xia
- The Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- School of Chemistry and BiochemistrySchool of Chemical and Biomolecular EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
12
|
Nanodecoration of electrospun polymeric fibers with nanostructured silver coatings by ionized jet deposition for antibacterial tissues. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 113:110998. [PMID: 32487406 DOI: 10.1016/j.msec.2020.110998] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/27/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022]
Abstract
Silver-based nanomaterials are used as antibacterial agents in a number of applications, including wound dressing, where electrospun materials can effectively promote wound healing and tissue regeneration thanks to their biomimicry, flexibility and breathability. Incorporation of such nanomaterials in electrospun nonwovens is highly challenging if aiming at maximizing stability and antibacterial efficacy and minimizing silver detachment, without neglecting process straightforwardness and scalability. In this work nanostructured silver coatings were deposited by Ionized Jet Deposition (IJD) on Polylactic acid, a medical grade polyester-urethane and Polyamide 6,6 nanofibers. The resulting materials were thoroughly characterized to gain an in-depth view of coating morphology and substrate resistance to the low-temperature deposition process used. Morphology of silver coatings with well-cohesive grains having dimensions from a few tens to a few hundreds of nanometers was analyzed by SEM, TEM and AFM. TGA, DSC, FTIR and GPC showed that the polymers well withstand the deposition process with negligible effects on their properties, the only exception being the polylactic acid that resulted more susceptible to degradation. Finally, the efficacy against S. aureus and E. coli bacterial strains was demonstrated, indicating that electrospun fibers decorated with nanostructured silver by IJD represent a breakthrough solution in the field of antibacterial devices.
Collapse
|
13
|
Bloise N, Rountree I, Polucha C, Montagna G, Visai L, Coulombe KLK, Munarin F. Engineering Immunomodulatory Biomaterials for Regenerating the Infarcted Myocardium. Front Bioeng Biotechnol 2020; 8:292. [PMID: 32318563 PMCID: PMC7154131 DOI: 10.3389/fbioe.2020.00292] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Coronary artery disease is a severe ischemic condition characterized by the reduction of blood flow in the arteries of the heart that results in the dysfunction and death of cardiac tissue. Despite research over several decades on how to reduce long-term complications and promote angiogenesis in the infarct, the medical field has yet to define effective treatments for inducing revascularization in the ischemic tissue. With this work, we have developed functional biomaterials for the controlled release of immunomodulatory cytokines to direct immune cell fate for controlling wound healing in the ischemic myocardium. The reparative effects of colony-stimulating factor (CSF-1), and anti-inflammatory interleukins 4/6/13 (IL4/6/13) have been evaluated in vitro and in a predictive in vivo model of ischemia (the skin flap model) to optimize a new immunomodulatory biomaterial that we use for treating infarcted rat hearts. Alginate hydrogels have been produced by internal gelation with calcium carbonate (CaCO3) as carriers for the immunomodulatory cues, and their stability, degradation, rheological properties and release kinetics have been evaluated in vitro. CD14 positive human peripheral blood monocytes treated with the immunomodulatory biomaterials show polarization into pro-healing macrophage phenotypes. Unloaded and CSF-1/IL4 loaded alginate gel formulations have been implanted in skin flap ischemic wounds to test the safety and efficacy of the delivery system in vivo. Faster wound healing is observed with the new therapeutic treatment, compared to the wounds treated with the unloaded controls at day 14. The optimized therapy has been evaluated in a rat model of myocardial infarct (ischemia/reperfusion). Macrophage polarization toward healing phenotypes and global cardiac function measured with echocardiography and immunohistochemistry at 4 and 15 days demonstrate the therapeutic potential of the proposed immunomodulatory treatment in a clinically relevant infarct model.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, ICS Maugeri, IRCCS, Pavia, Italy
| | - Isobel Rountree
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Collin Polucha
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Giulia Montagna
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Electrical, Computer and Biomedical Engineering, Centre for Health Technologies (CHT), University of Pavia, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine, Center for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, ICS Maugeri, IRCCS, Pavia, Italy
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| | - Fabiola Munarin
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI, United States
| |
Collapse
|
14
|
Bloise N, Massironi A, Della Pina C, Alongi J, Siciliani S, Manfredi A, Biggiogera M, Rossi M, Ferruti P, Ranucci E, Visai L. Extra-Small Gold Nanospheres Decorated With a Thiol Functionalized Biodegradable and Biocompatible Linear Polyamidoamine as Nanovectors of Anticancer Molecules. Front Bioeng Biotechnol 2020; 8:132. [PMID: 32195232 PMCID: PMC7065572 DOI: 10.3389/fbioe.2020.00132] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/10/2020] [Indexed: 12/29/2022] Open
Abstract
Gold nanoparticles are elective candidate for cancer therapy. Current efforts are devoted to developing innovative methods for their synthesis. Besides, understanding their interaction with cells have become increasingly important for their clinical application. This work aims to describe a simple approach for the synthesis of extra-small gold nanoparticles for breast cancer therapy. In brief, a biocompatible and biodegradable polyamidoamine (named AGMA1-SH), bearing 20%, on a molar basis, thiol-functionalized repeat units, is employed to stabilize and coat extra-small gold nanospheres of different sizes (2.5, 3.5, and 5 nm in gold core), and to generate a nanoplatform for the link with Trastuzumab monoclonal antibody for HER2-positive breast cancer targeting. Dynamic light scattering, transmission electron microscopy, ultraviolet visible spectroscopy, X-ray powder diffraction, circular dichroism, protein quantification assays are used for the characterization. The targeting properties of the nanosystems are explored to achieve enhanced and selective uptake of AGMA1-SH-gold nanoparticles by in vitro studies against HER-2 overexpressing cells, SKBR-3 and compared to HER-2 low expressing cells, MCF-7, and normal fibroblast cell line, NIH-3T3. In vitro physicochemical characterization demonstrates that gold nanoparticles modified with AGMA1-SH are more stable in aqueous solution than the unmodified ones. Additionally, the greater gold nanoparticles size (5-nm) is associated with a higher stability and conjugation efficiency with Trastuzumab, which retains its folding and anticancer activity after the conjugation. In particular, the larger Trastuzumab functionalized nanoparticles displays the highest efficacy (via the pro-apoptotic protein increase, anti-apoptotic components decrease, survival-proliferation pathways downregulation) and internalization (via the activation of the classical clathrin-mediated endocytosis) in HER-2 overexpressing SKBR-3 cells, without eliciting significant effects on the other cell lines. The use of biocompatible AGMA1-SH for producing covalently stabilized gold nanoparticles to achieve selective targeting, cytotoxicity and uptake is completely novel, offering an important advancement for developing new anticancer conjugated-gold nanoparticles.
Collapse
Affiliation(s)
- Nora Bloise
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Pavia, Italy
| | - Alessio Massironi
- Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM PISA, Pisa, Italy
| | - Cristina Della Pina
- Dipartimento di Chimica, Università degli Studi di Milano e CNR-ISTM, Milan, Italy
| | - Jenny Alongi
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Stella Siciliani
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Amedea Manfredi
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | - Marco Biggiogera
- Department of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - Michele Rossi
- Dipartimento di Chimica, Università degli Studi di Milano e CNR-ISTM, Milan, Italy
| | - Paolo Ferruti
- Dipartimento di Chimica, Università degli Studi di Milano, Milan, Italy
| | | | - Livia Visai
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Pavia, Italy
| |
Collapse
|
15
|
Martelli G, Bloise N, Merlettini A, Bruni G, Visai L, Focarete ML, Giacomini D. Combining Biologically Active β-Lactams Integrin Agonists with Poly(l-lactic acid) Nanofibers: Enhancement of Human Mesenchymal Stem Cell Adhesion. Biomacromolecules 2020; 21:1157-1170. [PMID: 32011862 PMCID: PMC7997109 DOI: 10.1021/acs.biomac.9b01550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Regulating
stem cell adhesion and growth onto functionalized biomaterial
scaffolds is an important issue in the field of tissue engineering
and regenerative medicine. In this study, new electrospun scaffolds
of poly(l-lactic acid) (PLLA), as bioresorbable polymer,
and β-lactam compounds agonists of selected integrins, as functional
components with cell adhesive properties, are designed. The new β-lactam-PLLA
scaffolds contribute significantly in guiding protein translation
involved in human bone marrow mesenchymal stem cells (hBM-MSC) adhesion
and integrin gene expression. Scanning electron microscopy, confocal
laser scanning microscopy, and Western Blot analyses reveal that GM18-PLLA
shows the best results, promoting cell adhesion by significantly driving
changes in focal adhesion proteins distribution (β1 integrin and vinculin) and activation (pFAK), with a notable increase
of GM18-targets subunits integrin gene expression, α4 and β1. These novel functionalized submicrometric
fibrous scaffolds demonstrate, for the first time, the powerful combination
of selective β-lactams agonists of integrins with biomimetic
scaffolds, suggesting a designed rule that could be suitably applied
to tissue repair and regeneration.
Collapse
Affiliation(s)
- Giulia Martelli
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Nora Bloise
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Andrea Merlettini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giovanna Bruni
- Department of Chemistry, Section of Physical Chemistry, University of Pavia, Viale Taramelli 16, 27100 Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Maria Letizia Focarete
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Daria Giacomini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
16
|
Near-Infrared, Light-Triggered, On-Demand Anti-inflammatories and Antibiotics Release by Graphene Oxide/Elecrospun PCL Patch for Wound Healing. C — JOURNAL OF CARBON RESEARCH 2019. [DOI: 10.3390/c5040063] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Very recently, significant attention has been focused on the adsorption and cell adhesion properties of graphene oxide (GO), because it is expected to allow high drug loading and controlled drug release, as well as the promotion of cell adhesion and proliferation. This is particularly interesting in the promotion of wound healing, where antibiotics and anti-inflammatories should be locally released for a prolonged time to allow fibroblast proliferation. Here, we designed an implantable patch consisting of poly(caprolactone) electrospun covered with GO, henceforth named GO–PCL, endowed with high ibuprofen (5.85 mg cm−2), ketoprofen (0.86 mg cm−2), and vancomycin (0.95 mg cm−2) loading, used as anti-inflammatory and antibiotic models respectively, and capable of responding to near infrared (NIR)-light stimuli in order to promptly release the payload on-demand beyond three days. Furthermore, we demonstrated the GO is able to promote fibroblast adhesion, a key characteristic to potentially provide wound healing in vivo.
Collapse
|
17
|
Abstract
Soft and hard tissue engineering has expanded the frontiers of oral/maxillofacial augmentation. Soft tissue grafting enhancements include improving flap prevascularization and using stem cells and other cells to create not only the graft, but also the vascularization and soft tissue scaffolding for the graft. Hard tissue grafts have been enhanced by osteoinductive factors, such as bone morphogenic proteins, that have allowed the elimination of harvesting autogenous bone and thus decrease the need for other surgical sites. Advancements in bone graft scaffolds have developed via seeding with stem cells and improvement of the silica/calcium/phosphate composite to improve graft characteristics and healing.
Collapse
Affiliation(s)
- Dolphus R Dawson
- Division of Periodontology, Department of Oral Health Practice, College of Dentistry, University of Kentucky, 800 Rose Street, D-444 Dental Sciences Building, Lexington, KY 40536-0297, USA.
| | - Ahmed El-Ghannam
- Department of Mechanical Engineering and Engineering Science, University of North Carolina at Charlotte, 9201 University City Boulevard, Charlotte, NC 28223-0001, USA
| | - Joseph E Van Sickels
- Division of Oral and Maxillofacial Surgery, College of Dentistry, University of Kentucky, 800 Rose Street, Lexington, KY 40536-0297, USA
| | - Noel Ye Naung
- Division of Oral and Maxillofacial Surgery, College of Dentistry, University of Kentucky, 800 Rose Street, Lexington, KY 40536-0297, USA
| |
Collapse
|
18
|
Ranucci E, Manfredi A. Polyamidoamines: Versatile Bioactive Polymers with Potential for Biotechnological Applications. CHEMISTRY AFRICA-A JOURNAL OF THE TUNISIAN CHEMICAL SOCIETY 2019. [DOI: 10.1007/s42250-019-00046-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
19
|
Bandiera A, Passamonti S, Dolci LS, Focarete ML. Composite of Elastin-Based Matrix and Electrospun Poly(L-Lactic Acid) Fibers: A Potential Smart Drug Delivery System. Front Bioeng Biotechnol 2018; 6:127. [PMID: 30258842 PMCID: PMC6143670 DOI: 10.3389/fbioe.2018.00127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/24/2018] [Indexed: 11/13/2022] Open
Abstract
Stimuli-responsive hydrogel matrices are inspiring manifold applications in controlled delivery of bioactive compounds. Elastin-derived polypeptides form hydrogel matrices that may release bioactive moieties as a function of local increase of active elastases, as it would occur in several processes like inflammation. In view of the development of a patch for healing wounds, recombinant elastin-based polypeptides were combined with a proteolysis-resistant scaffold, made of electrospun poly-L-lactic acid (PLLA) fibers. The results of this study demonstrated the compatibility of these two components. An efficient procedure to obtain a composite material retaining the main features of each component was established. The release of the elastin moiety was monitored by means of a simple protocol. Our data showed that electrospun PLLA can form a composite with fusion proteins bound to elastin-derived polypeptides. Therefore, our approach allows designing a therapeutic agent delivery platform to realize devices capable of responding and interacting with biological systems at the molecular level.
Collapse
Affiliation(s)
| | | | - Luisa Stella Dolci
- Department of Chemistry "G. Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Maria Letizia Focarete
- Department of Chemistry "G. Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), Alma Mater Studiorum - Università di Bologna, Bologna, Italy.,Health Sciences and Technologies-Interdepartmental Center for Industrial Research, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| |
Collapse
|
20
|
Mauro N, Scialabba C, Pitarresi G, Giammona G. Enhanced adhesion and in situ photothermal ablation of cancer cells in surface-functionalized electrospun microfiber scaffold with graphene oxide. Int J Pharm 2017; 526:167-177. [PMID: 28442269 DOI: 10.1016/j.ijpharm.2017.04.045] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 04/13/2017] [Accepted: 04/18/2017] [Indexed: 01/01/2023]
Abstract
The physicochemical characteristics of a biomaterial surface highly affect the interaction with living cells. Recently, much attention has been focused on the adhesion properties of functional biomaterials toward cancer cells, since is expected to control metastatic spread of a tumor, which is related to good probability containing the progression of disease burden. Here, we designed an implantable poly(caprolactone)-based electrospun microfiber scaffold, henceforth PCLMF-GO, to simultaneously capture and kill cancer cells by tuning physicochemical features of the hybrid surface through nitrogen plasma activation and hetero-phase graphene oxide (GO) covalent functionalization. The surface immobilization of GO implies enhanced cell adhesion and proliferation, promoting the selective adhesion of cancer cells, even if allowing cancer associated fibroblast (CAFs) capture. We also display that the functionalization with GO, thanks to the high near-infrared (NIR) absorbance, enables the discrete photothermal eradication of the captured cancer cells in situ (≈98%).
Collapse
Affiliation(s)
- Nicolò Mauro
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche" (STEBICEF), University of Palermo, Via Archirafi, 32, 90123 Palermo, Italy.
| | - Cinzia Scialabba
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche" (STEBICEF), University of Palermo, Via Archirafi, 32, 90123 Palermo, Italy
| | - Giovanna Pitarresi
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche" (STEBICEF), University of Palermo, Via Archirafi, 32, 90123 Palermo, Italy
| | - Gaetano Giammona
- Laboratory of Biocompatible Polymers, Department of "Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche" (STEBICEF), University of Palermo, Via Archirafi, 32, 90123 Palermo, Italy; Mediterranean Center for Human Advanced Biotechnologies (Med-Chab), Viale delle Scienze Ed. 18, 90128 Palermo, Italy
| |
Collapse
|
21
|
Tomaselli S, Ramirez DOS, Carletto RA, Varesano A, Vineis C, Zanzoni S, Molinari H, Ragona L. Electrospun Lipid Binding Proteins Composite Nanofibers with Antibacterial Properties. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/13/2016] [Indexed: 01/24/2023]
Affiliation(s)
- Simona Tomaselli
- Istituto per lo Studio delle Macromolecole (ISMAC); CNR, via Corti 12 20133 Milano Italy
| | | | | | - Alessio Varesano
- Istituto per lo Studio delle Macromolecole (ISMAC); CNR, C.so G. Pella 16 13900 Biella Italy
| | - Claudia Vineis
- Istituto per lo Studio delle Macromolecole (ISMAC); CNR, C.so G. Pella 16 13900 Biella Italy
| | - Serena Zanzoni
- Dipartimento di Biotecnologie; Università degli Studi di Verona; Strada le Grazie 15 37134 Verona Italy
| | - Henriette Molinari
- Istituto per lo Studio delle Macromolecole (ISMAC); CNR, via Corti 12 20133 Milano Italy
| | - Laura Ragona
- Istituto per lo Studio delle Macromolecole (ISMAC); CNR, via Corti 12 20133 Milano Italy
| |
Collapse
|