1
|
Li S, Huang C, Liu H, Wang Z, Han X, Chen Z, Huang J, Wang Z. Optimization of cellular microenvironment of custom 3D printed brain-mimetic hydrogel and its application in drug neurotoxicity evaluation. BIOMATERIALS ADVANCES 2025; 172:214257. [PMID: 40023084 DOI: 10.1016/j.bioadv.2025.214257] [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: 09/02/2024] [Revised: 02/11/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
In this study, a customized biomimetic brain-mimetic hydrogel model was developed using Digital Light Processing (DLP) 3D printing technology. This model aims to optimize the cellular microenvironment for the growth of neural stem cells and to evaluate the neurotoxicity of various drugs effectively. By precisely controlling the material composition, printing process, and structural design, the pore structure, water absorption, mechanical properties, and formability of the hydrogel were optimized, creating a more biomimetic microenvironment for the survival and proliferation of neural stem cells. Our developed composite hydrogel (GelMA-HAMA-SilMA-Gelatin) achieved a relatively low elastic modulus, excellent water absorption, good biocompatibility, and superior printability by adjusting printing parameters. This makes it better suited to approximate the optimal microenvironment for neural stem cell growth, enhancing cellular proliferation and vitality. Furthermore, our research demonstrated that the 3D hollow brain-mimetic hydrogel model, which features macronutrient channels and microporous networks, significantly improved the survival and proliferation of neural stem cells. Using this model, the neurotoxicity of acrylamide and oxaliplatin was evaluated, confirming the model's effectiveness in evaluating drug neurotoxicity and its ability to demonstrate cellular sensitivity to drug dosages. Compared to conventional two-dimensional adherent cell culture models, our three-dimensional brain-mimetic model can more accurately simulate the complex in vivo environment, offering new perspectives for drug screening and neurotoxicity evaluation. This study not only demonstrates the application potential of 3D printing technology in optimizing cellular microenvironments and drug screening but also emphasizes the importance of considering the cellular microenvironment during drug development. Ultimately, it provides new strategies and tools for evaluating drug safety.
Collapse
Affiliation(s)
- Shuying Li
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Chuanzhen Huang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China.
| | - Hanlian Liu
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China.
| | - Zhichao Wang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Xu Han
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhuang Chen
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Jun Huang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-Efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhen Wang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China
| |
Collapse
|
2
|
Cao L, Wang H, Guo W, Shen H, Hong FF, Xu G. Fabrication of sustainable, antibacterial, and water-stable paper-based materials from kapok fiber, carboxymethyl cellulose and cationic starch for disposable hygiene products. Int J Biol Macromol 2025; 314:144112. [PMID: 40354862 DOI: 10.1016/j.ijbiomac.2025.144112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Natural paper-based materials are desirable candidate materials for disposable hygiene products due to their environmental sustainability, cost-effectiveness, and biodegradability. However, their practical application is often hindered by poor water stability and limited functional properties. In this study, we developed a wet-laid web formation and hot-pressing technique to produce porous, layered paper-based materials with high porosity, flexibility, water stability, and antibacterial properties. These materials were created using naturally derived components, including kapok fiber, carboxymethyl cellulose (CMC), and cationic starch (CS). The synergistic interaction between CMC and CS significantly enhances the mechanical properties and water stability, achieving a 146.09 % improvement compared to materials without CMC/CS. The resulting paper-based materials also exhibit water stability for up to 30 days. Kapok fibers contribute excellent antimicrobial properties, with >95 % inhibition of both Escherichia coli and Staphylococcus aureus. Furthermore, the materials are biodegradable in soil, completely degrading after 60 days. This study provides novel insights into the valorization of kapok fiber and presents a sustainable approach to producing high-performance paper-based materials for disposable hygiene products applications.
Collapse
Affiliation(s)
- Liyao Cao
- College of Fine Arts and Design, Wenzhou University, Wenzhou 325035, China
| | - Hongchang Wang
- Shanghai Composite Materials Technology Co., Ltd, Shanghai, 201112, China; Shanghai Aerospace Resin based Composite Engineering Technology & Research Center, Shanghai, 201112, China
| | - Weina Guo
- College of Textiles, Donghua University, Shanghai 201620, China
| | - Hua Shen
- College of Textiles, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Science and Technology Ministry of Education, Donghua University, Shanghai 201620, China
| | - Feng F Hong
- Scientific Research Base for Bacterial Nanofiber Manufacturing and Composite Technology, College of Biological Science and Medical Engineering, Donghua University, North Ren Min Road 2999, Shanghai 201620, China.
| | - Guangbiao Xu
- College of Textiles, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Science and Technology Ministry of Education, Donghua University, Shanghai 201620, China.
| |
Collapse
|
3
|
Dmitrenko M, Mikhailovskaya O, Dubovenko R, Mazur A, Kuzminova A, Prikhodko I, Semenov K, Su R, Penkova A. Nanofiltration Membranes from Poly(sodium-p-styrenesulfonate)/Polyethylenimine Polyelectrolyte Complex Modified with Carbon Nanoparticles for Enhanced Water Treatment. Polymers (Basel) 2025; 17:1306. [PMID: 40430602 PMCID: PMC12114722 DOI: 10.3390/polym17101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Industrial wastewater poses a significant environmental challenge due to its harmful effects. The development of sustainable membrane processes for water treatment and the environmentally friendly production of polymer membranes is one of the major challenges of our time. An alternative approach is to prepare polyelectrolyte complex (PEC) membranes using the aqueous phase separation (APS) method without the use of toxic solvents. In this work, PEC nanofiltration membranes of poly(sodium-p-styrenesulfonate) (PSS)/polyethylenimine (PEI) modified with carbon nanoparticles (graphene oxide, polyhydroxylated fullerene (HF), multi-walled carbon nanotubes) were developed for enhanced water treatment from anionic food dyes and heavy metal ions. The effect of varying the PSS/PEI monomer ratio, carbon nanoparticles, the content of the optimal HF modifier, and the cross-linking agent on the membrane properties was studied in detail. The changes in the structure and physicochemical properties of the PEC-based membranes were investigated using spectroscopic, microscopic, thermogravimetric analysis methods, and contact angle measurements. The PSS and PEI interactions during PEC formation and the effect of PEI protonation on membrane properties were investigated using computational methods. The optimal cross-linked PEC/HF(1%) (1:1.75 PSS/PEI) membrane had more than 2 times higher permeability compared to the pristine PEC membrane, with dye and heavy metal ion rejection of 99.99 and >97%, respectively.
Collapse
Affiliation(s)
- Mariia Dmitrenko
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Olga Mikhailovskaya
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Roman Dubovenko
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Anton Mazur
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Anna Kuzminova
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Igor Prikhodko
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| | - Konstantin Semenov
- Pavlov First Saint Petersburg State Medical University, L’va Tolstogo ulitsa 6–8, St. Petersburg 197022, Russia;
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China;
| | - Anastasia Penkova
- St. Petersburg State University, 7/9 Universitetskaya nab., St. Petersburg 199034, Russia; (O.M.); (R.D.); (A.M.); (A.K.); (I.P.)
| |
Collapse
|
4
|
Ng XJ, Esser TU, Trossmann VT, Rudisch C, Fiedler M, Roshanbinfar K, Lamberger Z, Stahlhut P, Lang G, Scheibel T, Engel FB. Enhancing Form Stability: Shrink-Resistant Hydrogels Made of Interpenetrating Networks of Recombinant Spider Silk and Collagen-I. Adv Healthc Mater 2025; 14:e2500311. [PMID: 40143764 PMCID: PMC12057611 DOI: 10.1002/adhm.202500311] [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: 01/18/2025] [Revised: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Tissue engineering enables the production of tissues and organ-like structures as models for drug testing and mechanistical studies or functional replacements for injured tissues. Available cytocompatible materials are limited in number, suffer from insufficient mechanical properties, and cells interacting with them often cause construct shrinkage. As shape is important for function, identifying cytocompatible, shrink-resistant materials are a major aim. Here, it is shown that hydrogels made of interpenetrating networks of collagen-I and recombinant spider silk protein eADF4(C16)-RGD nanofibrils exhibit synergistic and tunable mechanical properties. Composite hydrogels allow cell adhesion and spreading and are resistant to shrinkage mediated by fibroblasts, C2C12 myoblasts, and human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. Myoblasts differentiate and fuse into myotubes, and hiPSC-cardiomyocytes can be cultured long-term, show spontaneous contractions, and remain drug responsive. Collectively, a novel composite material is developed to overcome the challenge of post-fabrication matrix shrinkage conferring high shape fidelity suitable for tissue engineering.
Collapse
Affiliation(s)
- Xuen J. Ng
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Tilman U. Esser
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Vanessa T. Trossmann
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Christoph Rudisch
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Maren Fiedler
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Zan Lamberger
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Gregor Lang
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Thomas Scheibel
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
- Bayreuth Center for Colloids and Interfaces (BZKG)University of Bayreuth95447BayreuthGermany
- Bavarian Polymer Institute (BPI)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Molecular Biosciences (BZMB)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Material Science (BayMAT)University of Bayreuth95447BayreuthGermany
- Faculty of MedicineUniversity of Würzburg97080WürzburgGermany
| | - Felix B. Engel
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| |
Collapse
|
5
|
Saosamniang P, Matsumura K, Okajima MK, Kaneko T. Directing mineralization of ZnO nanoparticles in cyanobacterial liquid crystalline polysaccharides for cancer therapies. Int J Biol Macromol 2025; 304:140716. [PMID: 39920940 DOI: 10.1016/j.ijbiomac.2025.140716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Effective cancer therapy faces significant challenges, including non-selective toxicity, limited structural stability, inconsistent nanoparticle (NP) morphology, and instability under varying biological conditions. These issues hindering targeted delivery and therapeutic efficacy. Previous approaches using polysaccharide-based nanomaterials have shown promise; however, problems such as inconsistent NP sizes and shapes, poor mechanical stability, and limited pH resilience restrict their clinical potential. This study hypothesized that sacran, a cyanobacterial liquid crystalline (LC) polysaccharide, can stabilize ZnO NPs, allowing for controlled mineralization, enhanced stability, and selective cytotoxicity. We developed ZnO nanocomposite xerogels in an LC sacran matrix, yielding block-like ZnO NPs (25-70 nm) with high surface-area-to-volume ratios that improve cellular uptake in tumor environments. Incorporating these NPs into chemically crosslinked sacran matrices resulted in a 3-fold increase in mechanical strength and a 10-fold improvement in swelling capacity compared to physically crosslinked systems. Additionally, the sacran-ZnO nanocomposites demonstrated robust stability under various pH conditions, indicating their resilience in diverse biological environments. Cytotoxicity assays revealed that higher concentrations of ZnO NP selectively increased toxicity toward human lung cancer cells (A549), with less impact on human dermal fibroblasts (HDFa). Moreover, HDFa successfully attached to and proliferated on the smooth surfaces of the xerogels, emphasizing their compatibility with normal cells. This highlights the potential of sacran-ZnO nanocomposite xerogels as cancer-selective therapeutic materials, offering stability and effectiveness even under varying biological conditions, while addressing key challenges associated with earlier NP-based therapies.
Collapse
Affiliation(s)
- Pruetsakorn Saosamniang
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kazuaki Matsumura
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Maiko K Okajima
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Tatsuo Kaneko
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
6
|
Resmi R, Parvathy J, Anjali S, Amrita N, Jyothi A, Harikrishnan VS, John A, Joseph R. Platelet-Rich Plasma Loaded Alginate-Based Injectable Hydrogel for Meniscal Tear Repair: In Vivo Evaluation in Lapine Model. J Biomed Mater Res B Appl Biomater 2025; 113:e35541. [PMID: 39891920 DOI: 10.1002/jbm.b.35541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/28/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Platelet-rich plasma (PRP) has been employed for orthopedic applications for decades due to the abundance of bioactive cues/growth factors that ameliorate the proliferation and migration of relevant cells involved in tissue repair/regeneration. In this work, PRP was incorporated into injectable compositions of alginate-based hydrogel and evaluated in vitro and in vivo. In vitro tests revealed that PRP addition promoted cell adhesion, cell proliferation, and distribution of seeded fibrochondrocytes on the hydrogel. Further, the DNA quantification and sGAG estimation confirmed the production of fibrocartilage-specific extracellular matrix, predominantly type 1 collagen and sGAG. For in vivo evaluation, tears were created surgically in the rabbit menisci and were filled with injectable hydrogel. Sham and hydrogel without PRP were used as controls. Histopathological evaluation after 3 months of implantation revealed that the healing was partial for sham control, but complete for hydrogel without PRP. The hydrogel served as the scaffold for fibrocartilage tissue regeneration. On the other hand, PRP-incorporated hydrogel showed good healing with low signs of inflammation as evidenced by histology and biochemical content. The healing was complete, and the nature of the regenerated tissues was very close to native tissue indicating that alginate-based hydrogel is a promising candidate for meniscal tissue repair.
Collapse
Affiliation(s)
- Rajalekshmi Resmi
- Division of Polymeric Medical Devices, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Jayasree Parvathy
- Corporate R&D Center, HLL Life Care Limited, Thiruvananthapuram, Kerala, India
| | - Sudha Anjali
- Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Natarajan Amrita
- Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Arun Jyothi
- Department of Orthopaedics, Sree Uthradom Thirunal (SUT) Hospital, Thiruvananthapuram, Kerala, India
| | - V S Harikrishnan
- Division of Laboratory Animal Science, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| | - Annie John
- Department of Biochemistry, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Roy Joseph
- Division of Polymeric Medical Devices, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
7
|
Brunel LG, Long CM, Christakopoulos F, Cai B, Johansson PK, Singhal D, Enejder A, Myung D, Heilshorn SC. Interpenetrating networks of fibrillar and amorphous collagen promote cell spreading and hydrogel stability. Acta Biomater 2025; 193:128-142. [PMID: 39798635 PMCID: PMC11908676 DOI: 10.1016/j.actbio.2025.01.009] [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: 09/06/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Hydrogels composed of collagen, the most abundant protein in the human body, are widely used as scaffolds for tissue engineering due to their ability to support cellular activity. However, collagen hydrogels with encapsulated cells often experience bulk contraction due to cell-generated forces, and conventional strategies to mitigate this undesired deformation often compromise either the fibrillar microstructure or cytocompatibility of the collagen. To support the spreading of encapsulated cells while preserving the structural integrity of the gels, we present an interpenetrating network (IPN) of two distinct collagen networks with different crosslinking mechanisms and microstructures. First, a physically self-assembled collagen network preserves the fibrillar microstructure and enables the spreading of encapsulated human corneal mesenchymal stromal cells. Second, an amorphous collagen network covalently crosslinked with bioorthogonal chemistry fills the voids between fibrils and stabilizes the gel against cell-induced contraction. This collagen IPN balances the biofunctionality of natural collagen with the stability of covalently crosslinked, engineered polymers. Taken together, these data represent a new avenue for maintaining both the fiber-induced spreading of cells and the structural integrity of collagen hydrogels by leveraging an IPN of fibrillar and amorphous collagen networks. STATEMENT OF SIGNIFICANCE: Collagen hydrogels are widely used as scaffolds for tissue engineering due to their support of cellular activity. However, collagen hydrogels often undergo undesired changes in size and shape due to cell-generated forces, and conventional strategies to mitigate this deformation typically compromise either the fibrillar microstructure or cytocompatibility of the collagen. In this study, we introduce an innovative interpenetrating network (IPN) that combines physically self-assembled, fibrillar collagen-ideal for promoting cell adhesion and spreading-with covalently crosslinked, amorphous collagen-ideal for enhancing bulk hydrogel stability. Our IPN design maintains the native fibrillar structure of collagen while significantly improving resistance against cell-induced contraction, providing a promising solution to enhance the performance and reliability of collagen hydrogels for tissue engineering applications.
Collapse
Affiliation(s)
- Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Chris M Long
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Fotis Christakopoulos
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Patrik K Johansson
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Diya Singhal
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - David Myung
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA; Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA; VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Krishna VS, Subashini V, Hariharan A, Chidambaram D, Raaju A, Gopichandran N, Nanthanalaxmi MP, Lekhavadhani S, Shanmugavadivu A, Selvamurugan N. Role of crosslinkers in advancing chitosan-based biocomposite scaffolds for bone tissue engineering: A comprehensive review. Int J Biol Macromol 2024; 283:137625. [PMID: 39547606 DOI: 10.1016/j.ijbiomac.2024.137625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Bone tissue engineering (BTE) aims to repair and regenerate damaged bone tissue by combining cells, scaffolds, and signaling molecules. Various macromolecules, including natural polymers like chitosan (CS), collagen, hyaluronic acid, and alginate, as well as synthetic polymers such as polyethylene glycol and polylactic acid, are used in scaffold fabrication. Among these, CS holds significant potential in BTE due to its biocompatibility, biodegradability, and other features. The inherent mechanical weaknesses of CS-based scaffolds require the implementation of crosslinking strategies to improve their stability and overall performance. Physical crosslinkers like ultra-violet irradiation and freeze-thaw cycles are biocompatible but offer limited mechanical strength. Chemical crosslinkers like glutaraldehyde significantly improve mechanical strength, but they may induce cytotoxicity. We briefly outline here the critical role of physical and chemical crosslinkers in improving the physicochemical properties, mechanical strength, biocompatibility, and biological functions of CS-based scaffolds, including effective bone regeneration. The influence of crosslinking on the CS-based scaffolds' bioactivity, including the controlled release of bioactive molecules, is also discussed. A thorough understanding of crosslinker chemistry and application in CS-based scaffolds is essential for advancing bone regeneration therapies.
Collapse
Affiliation(s)
- Venkatasubramanian Sai Krishna
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Velan Subashini
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Adithya Hariharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Deekshaa Chidambaram
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Adityaa Raaju
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nikthesh Gopichandran
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Muthuvaira Prasath Nanthanalaxmi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Sundaravadhanan Lekhavadhani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
9
|
Kim JR, Cho YS, Park JH, Kim TH. Poly(HEMA-co-MMA) Hydrogel Scaffold for Tissue Engineering with Controllable Morphology and Mechanical Properties Through Self-Assembly. Polymers (Basel) 2024; 16:3014. [PMID: 39518224 PMCID: PMC11548049 DOI: 10.3390/polym16213014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Poly(2-hydroxyethyl methacrylate) (PHEMA) has been widely used in medical materials for several decades. However, the poor mechanical properties of this material have limited its application in the field of tissue engineering. The purpose of this study was to fabricate a scaffold with suitable mechanical properties and in vitro cell responses for soft tissue by using poly(HEMA-co-MMA) with various concentration ratios of hydroxyethyl methacrylate (HEMA) and methyl methacrylate (MMA). To customize the concentration ratio of HEMA and MMA, the characteristics of the fabricated scaffold with various concentration ratios were investigated through structural morphology, FT-IR, mechanical property, and contact angle analyses. Moreover, in vitro cell responses were observed according to the various concentration ratios of HEMA and MMA. Consequently, various morphologies and pore sizes were observed by changing the HEMA and MMA ratio. The mechanical properties and contact angle of the fabricated scaffolds were measured according to the HEMA and MMA concentration ratio. The results were as follows: compressive maximum stress: 254.24-932.42 KPa; tensile maximum stress: 4.37-30.64 KPa; compressive modulus: 16.14-38.80 KPa; tensile modulus: 0.5-2 KPa; and contact angle: 36.89-74.74°. In terms of the in vitro cell response, the suitable cell adhesion and proliferation of human dermal fibroblast (HDF) cells were observed in the whole scaffold. Therefore, a synthetic hydrogel scaffold with enhanced mechanical properties and suitable fibroblast cell responses could be easily fabricated for use with soft tissue using a specific HEMA and MMA concentration ratio.
Collapse
Affiliation(s)
- Ja-Rok Kim
- R&D Center, TE BioS, Co., Ltd., 194-41, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju-si 28160, Republic of Korea; (J.-R.K.); (J.-H.P.)
| | - Yong Sang Cho
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), 80 Cheombok-ro, Dong-gu, Daegu 41061, Republic of Korea;
| | - Jae-Hong Park
- R&D Center, TE BioS, Co., Ltd., 194-41, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju-si 28160, Republic of Korea; (J.-R.K.); (J.-H.P.)
| | - Tae-Hyun Kim
- R&D Center, TE BioS, Co., Ltd., 194-41, Osongsaengmyeong 1-ro, Heungdeok-gu, Cheongju-si 28160, Republic of Korea; (J.-R.K.); (J.-H.P.)
| |
Collapse
|
10
|
Benkhira I, Zermane F, Cheknane B, Trache D, Brosse N, Paolone A, Chader H, Sobhi W. Preparation and characterization of amidated pectin-gelatin-oxidized tannic acid hydrogel films supplemented with in-situ reduced silver nanoparticles for wound-dressing applications. Int J Biol Macromol 2024; 277:134158. [PMID: 39059528 DOI: 10.1016/j.ijbiomac.2024.134158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 07/13/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Wound dressings play a crucial role in protecting injured tissues and promoting the healing process. Traditional fabrication of antibacterial wound dressings can be complex and may involve toxic components. In this study, we developed an innovative hydrogel film (AP:GE@OTA/Ag) composed of amidated pectin (AP), gelatin (GE), oxidized tannic acid (OTA) at varying concentrations, and in-situ reduced silver nanoparticles (AgNPs). FTIR and XRD analyses confirmed that crosslinking occurs via interactions between OTA quinone groups and free amino groups in AP and GE. TEM imaging demonstrated the well-dispersed AgNPs with an average particle size of 58.64 nm, while the TG measurements indicated the enhancement of the thermal stability compared to AP:GE films. The AP:GE@OTA/Ag films exhibited superior fluid uptake ability (90.96 % at 2 h), water retention capacity (91.69 % at 2 h), and water vapor transmission rate (1903.29 g/m2/day), alongside improved tensile strength (38 MPa). Additionally, these films showed excellent cytocompatibility and sustained potent antimicrobial activity against S. aureus and E. coli with low AgNPs loadings of 1.02 ± 0.13 μg/cm2. NIT-1 mouse insulinoma cells demonstrated robust proliferation when cultured with the prepared dressings. These films significantly accelerated wound repair in a skin excision model, indicating their potential clinical applications for wound healing.
Collapse
Affiliation(s)
- Ilyas Benkhira
- Laboratoire Chimie Physique Des Interfaces Des Matériaux Appliqués à l'Environnement, Département de Génie Des Procédés, Université Saad Dahlab Blida 1, 09000 Blida, Algeria.
| | - Faiza Zermane
- Laboratoire Chimie Physique Des Interfaces Des Matériaux Appliqués à l'Environnement, Département de Génie Des Procédés, Université Saad Dahlab Blida 1, 09000 Blida, Algeria
| | - Benamar Cheknane
- Laboratoire Chimie Physique Des Interfaces Des Matériaux Appliqués à l'Environnement, Département de Génie Des Procédés, Université Saad Dahlab Blida 1, 09000 Blida, Algeria
| | - Djalal Trache
- Energetic Materials Laboratory (EMLab), Teaching and Research Unit of Energetic Processes, Polytechnic Military School, BP 17, Bordj El-Bahri, 16046 Algiers, Algeria
| | - Nicolas Brosse
- LERMAB, Faculty of Science and Technology, University of Lorraine, Vandoeuvre-Les-Nancy, 54506, France
| | - Annalisa Paolone
- Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche, Piazzale A. Moro 5, I-00185 Rome, Italy
| | - Henni Chader
- Department of Pharmacy, Faculty of Medicine, University of Algiers 1, Algiers 16001, Algeria
| | - Widad Sobhi
- Research Center of Biotechnology (CRBt), Constantine 25000, Algeria
| |
Collapse
|
11
|
McLeod KA, Di Gregorio M, Tinney D, Carmichael J, Zuanazzi D, Siqueira WL, Rizkalla A, Hamilton DW. Galectin-3/Gelatin Electrospun Scaffolds Modulate Collagen Synthesis in Skin Healing but Do Not Improve Wound Closure Kinetics. Bioengineering (Basel) 2024; 11:960. [PMID: 39451336 PMCID: PMC11504234 DOI: 10.3390/bioengineering11100960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic wounds remain trapped in a pro-inflammatory state, with strategies targeted at inducing re-epithelialization and the proliferative phase of healing desirable. As a member of the lectin family, galectin-3 is implicated in the regulation of macrophage phenotype and epithelial migration. We investigated if local delivery of galectin-3 enhanced skin healing in a full-thickness excisional C57BL/6 mouse model. An electrospun gelatin scaffold loaded with galectin-3 was developed and compared to topical delivery of galectin-3. Electrospun gelatin/galectin-3 scaffolds had an average fiber diameter of 200 nm, with 83% scaffold porosity approximately and an average pore diameter of 1.15 μm. The developed scaffolds supported dermal fibroblast adhesion, matrix deposition, and proliferation in vitro. In vivo treatment of 6 mm full-thickness excisional wounds with gelatin/galectin-3 scaffolds did not influence wound closure, re-epithelialization, or macrophage phenotypes, but increased collagen synthesis. In comparison, topical delivery of galectin-3 [6.7 µg/mL] significantly increased arginase-I cell density at day 7 versus untreated and gelatin/galectin-3 scaffolds (p < 0.05). A preliminary assessment of increasing the concentration of topical galectin-3 demonstrated that at day 7, galectin-3 [12.5 µg/mL] significantly increased both epithelial migration and collagen content in a concentration-dependent manner. In conclusion, local delivery of galectin 3 shows potential efficacy in modulating skin healing in a concentration-dependent manner.
Collapse
Affiliation(s)
- Karrington A. McLeod
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Madeleine Di Gregorio
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
| | - Dylan Tinney
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - Justin Carmichael
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
| | - David Zuanazzi
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
| | - Walter L. Siqueira
- Biochemistry Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada; (D.Z.); (W.L.S.)
- College of Dentistry, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amin Rizkalla
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Douglas W. Hamilton
- Faculty of Engineering, School of Biomedical Engineering, University of Western Ontario, London, ON N6A 3K7, Canada (A.R.)
- Department of Anatomy and Cell Biology, University of Western Ontario, London, ON N6A 5C1, Canada; (D.T.); (J.C.)
- School of Dentistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
12
|
Lee YJ, Ajiteru O, Lee JS, Lee OJ, Choi KY, Kim SH, Park CH. Highly conductive, stretchable, and biocompatible graphene oxide biocomposite hydrogel for advanced tissue engineering. Biofabrication 2024; 16:045032. [PMID: 39116889 DOI: 10.1088/1758-5090/ad6cf7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 08/08/2024] [Indexed: 08/10/2024]
Abstract
The importance of hydrogels in tissue engineering cannot be overemphasized due to their resemblance to the native extracellular matrix. However, natural hydrogels with satisfactory biocompatibility exhibit poor mechanical behavior, which hampers their application in stress-bearing soft tissue engineering. Here, we describe the fabrication of a double methacrylated gelatin bioink covalently linked to graphene oxide (GO) via a zero-length crosslinker, digitally light-processed (DLP) printable into 3D complex structures with high fidelity. The resultant natural hydrogel (GelGOMA) exhibits a conductivity of 15.0 S m-1as a result of the delocalization of theπ-orbital from the covalently linked GO. Furthermore, the hydrogel shows a compressive strength of 1.6 MPa, and a 2.0 mm thick GelGOMA can withstand a 1.0 kg ms-1momentum. The printability and mechanical strengths of GelGOMAs were demonstrated by printing a fish heart with a functional fluid pumping mechanism and tricuspid valves. Its biocompatibility, electroconductivity, and physiological relevance enhanced the proliferation and differentiation of myoblasts and neuroblasts and the contraction of human-induced pluripotent stem cell-derived cardiomyocytes. GelGOMA demonstrates the potential for the tissue engineering of functional hearts and wearable electronic devices.
Collapse
Affiliation(s)
- Young Jin Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
- CURE 3D, Department of Cardiac Surgery, University Hospital Düsseldorf, Düsseldorf, Nordrhein-Westfalen 40225, Germany
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Kyu Young Choi
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Kangnam, Seoul 07441, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute (NBRM), Hallym University College of Medicine, Chuncheon, Gangwon-do 24252, Republic of Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Republic of Korea
| |
Collapse
|
13
|
Fakhri N, Khalili A, Sachlos T, Rezai P. Fabrication of Porous Collagen Scaffolds Containing Embedded Channels with Collagen Membrane Linings. MICROMACHINES 2024; 15:1031. [PMID: 39203682 PMCID: PMC11356104 DOI: 10.3390/mi15081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024]
Abstract
Tissues and organs contain an extracellular matrix (ECM). In the case of blood vessels, endothelium cells are anchored to a specialized basement membrane (BM) embedded inside the interstitial matrix (IM). We introduce a multi-structural collagen-based scaffold with embedded microchannels that mimics in vivo structures within vessels. Our scaffold consists of two parts, each containing two collagen layers, i.e., a 3D porous collagen layer analogous to IM lined with a thin 2D collagen film resembling the BM. Enclosed microchannels were fabricated using contact microprinting. Microchannel test structures with different sizes ranging from 300 to 800 µm were examined for their fabrication reproducibility. The heights and perimeters of the fabricated microchannels were ~20% less than their corresponding values in the replication PDMS mold; however, microchannel widths were significantly closer to their replica dimensions. The stiffness, permeability, and pore size properties of the 2D and 3D collagen layers were measured. The permeability of the 2D collagen film was negligible, making it suitable for mimicking the BM of large blood vessels. A leakage test at various volumetric flow rates applied to the microchannels showed no discharge, thereby verifying the reliability of the proposed integrated 2D/3D collagen parts and the contact printing method used for bonding them in the scaffold. In the future, multi-cell culturing will be performed within the 3D porous collagen and against the 2D membrane inside the microchannel, hence preparing this scaffold for studying a variety of blood vessel-tissue interfaces. Also, thicker collagen scaffold tissues will be fabricated by stacking several layers of the proposed scaffold.
Collapse
Affiliation(s)
| | | | - Terry Sachlos
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
14
|
El Moujahed S, Errachidi F, Morosanu AM, Abou Oualid H, Avramescu SM, Dragoi Cudalbeanu M, Ouazzani Chahdi F, Kandri Rodi Y, Dinica RM. Sustainable Collagen Film Preparation with Tannins Extracted from Moroccan Pomegranate Byproduct Varieties: Thermal, Structural, and Nanoscaled Studies. ACS OMEGA 2024; 9:27428-27437. [PMID: 38947794 PMCID: PMC11209680 DOI: 10.1021/acsomega.4c02321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/20/2024] [Accepted: 05/23/2024] [Indexed: 07/02/2024]
Abstract
Recently, obtaining collagen films using a cross-linking technique has been a successful strategy. The current investigation used six cross-linker extracts (CE) from six different pomegranate varieties' byproducts to make and characterize collagen-tannin films using acid-soluble collagen (SC). The polymeric film has a yellow hue after CE incorporation. Fourier transform infrared spectroscopy assessed the impact of CE and its successful interaction within the matrix. The shifts verify different interactions between extracts and collagen functional groups, where they likely form new hydrogen bonds, retaining their helix structure without damaging the matrix. Scanning electron microscopy was used to analyze the morphology and fiber size. The average diameter of the fibers was found to be about 3.64 μm. Thermal behaviors (denaturation and degradation) were investigated by thermogravimetric analysis. The weight losses of cross-linked films increased by around 20% compared to non-cross-linked ones. This phenomenon was explained by the absence of telopeptide sections in the collagen helical structure, typically reinforced by lysine and hydroxylysine covalent linkages. Nanoscaled observations were also accomplished using transmission electron microscopy (TEM) on SC and SC-CE. The TEM analysis confirmed the CE polymerization degree effect on the cross-linking density via the overlap sequences, ranging up to 32.38 ± 2.37 nm on the fibril. The prepared biodegradable collagen-tannin film showed higher cross-linking density, which is expected to improve the biomaterial applications of collagen films while exploiting the underrated pomegranate byproducts.
Collapse
Affiliation(s)
- Sara El Moujahed
- Laboratory
of Applied Organic Chemistry, Faculty of Sciences and Technologies, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Faouzi Errachidi
- Laboratory
of Functional Ecology and Engineering Environment, Faculty of Sciences
and Technologies, Sidi Mohamed Ben Abdellah
University, Fez 30050, Morocco
| | - Ana-Maria Morosanu
- Institute
of Biology Bucharest, Romanian Academy, Bucharest 060031, Romania
| | | | - Sorin Marius Avramescu
- Department
of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, Bucharest 050663, Romania
| | - Mihaela Dragoi Cudalbeanu
- Faculty of
Land Reclamation and Environmental Engineering, University of Agronomic Sciences and Veterinary Medicine of Bucharest, Bucharest 011464, Romania
| | - Fouad Ouazzani Chahdi
- Laboratory
of Applied Organic Chemistry, Faculty of Sciences and Technologies, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Youssef Kandri Rodi
- Laboratory
of Applied Organic Chemistry, Faculty of Sciences and Technologies, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Rodica-Mihaela Dinica
- Laboratory
of Organic Chemistry, Faculty of Sciences and Environment, Dunarea de Jos University of Galati, Galati 800008, Romania
| |
Collapse
|
15
|
Abraham S, Gupta P, Govarthanan K, Rao S, Santra TS. Direction-oriented fiber guiding with a tunable tri-layer-3D scaffold for periodontal regeneration. RSC Adv 2024; 14:19806-19822. [PMID: 38899033 PMCID: PMC11186324 DOI: 10.1039/d4ra01459f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Multilayered scaffolds mimicking mechanical and biological host tissue architectures are the current prerequisites for successful tissue regeneration. We propose our tunable tri-layered scaffold, designed to represent the native periodontium for potential regenerative applications. The fused deposition modeling platform is used to fabricate the novel movable three-layered polylactic acid scaffold mimicking in vivo cementum, periodontal ligament, and alveolar bone layers. The scaffold is further provided with multiple angulated fibers, offering directional guidance and facilitating the anchorage dependence on cell adhesion. Additionally, surface modifications of the scaffold were made by incorporating coatings like collagen and different concentrations of gelatin methacryloyl to enrich the cell adhesion and proliferation. The surface characterization of our designed scaffolds was performed using tribological studies, atomic force microscopy, contact angle measurement, scanning electron microscopy, and micro-computed tomography. Furthermore, the material characterization of this scaffold was investigated by attenuated total reflectance-Fourier transformed infrared spectroscopy. The scaffold's mechanical characterization, such as strength and compression modulus, was demonstrated by compression testing. The L929 mouse fibroblast cells and MG63 human osteosarcoma cells have been cultured on the scaffold. The scaffold's superior biocompatibility was evaluated using fluorescence dye with fluorescence microscopy, scanning electron microscopy, in vitro wound healing assay, MTT assay, and flow cytometry. The mineralization capability of the scaffolds was also studied. In conclusion, our study demonstrated the construction of a multilayered movable scaffold, which is highly biocompatible and most suitable for various downstream applications such as periodontium and in situ tissue regeneration of complex, multilayered tissues.
Collapse
Affiliation(s)
- Sarin Abraham
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Pallavi Gupta
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Kavitha Govarthanan
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem) Bengaluru Karnataka 560065 India
| | - Suresh Rao
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| | - Tuhin Subhra Santra
- Department of Engineering Design, Indian Institute of Technology Madras Chennai 600036 India
| |
Collapse
|
16
|
Byun H, Han Y, Kim E, Jun I, Lee J, Jeong H, Huh SJ, Joo J, Shin SR, Shin H. Cell-homing and immunomodulatory composite hydrogels for effective wound healing with neovascularization. Bioact Mater 2024; 36:185-202. [PMID: 38463552 PMCID: PMC10924181 DOI: 10.1016/j.bioactmat.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/23/2024] [Indexed: 03/12/2024] Open
Abstract
Wound healing in cases of excessive inflammation poses a significant challenge due to compromised neovascularization. Here, we propose a multi-functional composite hydrogel engineered to overcome such conditions through recruitment and activation of macrophages with adapted degradation of the hydrogel. The composite hydrogel (G-TSrP) is created by combining gelatin methacryloyl (GelMA) and nanoparticles (TSrP) composed of tannic acid (TA) and Sr2+. These nanoparticles are prepared using a one-step mineralization process assisted by metal-phenolic network formation. G-TSrP exhibits the ability to eliminate reactive oxygen species and direct polarization of macrophages toward M2 phenotype. It has been observed that the liberation of TA and Sr2+ from G-TSrP actively facilitate the recruitment and up-regulation of the expression of extracellular matrix remodeling genes of macrophages, and thereby, coordinate in vivo adapted degradation of the G-TSrP. Most significantly, G-TSrP accelerates angiogenesis despite the TA's inhibitory properties, which are counteracted by the released Sr2+. Moreover, G-TSrP enhances wound closure under inflammation and promotes normal tissue formation with strong vessel growth. Genetic analysis confirms macrophage-mediated wound healing by the composite hydrogel. Collectively, these findings pave the way for the development of biomaterials that promote wound healing by creating regenerative environment.
Collapse
Affiliation(s)
- Hayeon Byun
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, USA
| | - Yujin Han
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Eunhyung Kim
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Indong Jun
- Environmental Safety Group, Korea Institute of Science & Technology Europe (KIST-EUROPE), Saarbrücken 66123, Germany
| | - Jinkyu Lee
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Hyewoo Jeong
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Seung Jae Huh
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Cambridge, MA 02139, USA
| | - Heungsoo Shin
- Department of Bioengineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
- Institute of Nano Science and Technology, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| |
Collapse
|
17
|
Ziverec A, Bax D, Cameron R, Best S, Pasdeloup M, Courtial EJ, Mallein-Gerin F, Malcor JD. The diazirine-mediated photo-crosslinking of collagen improves biomaterial mechanical properties and cellular interactions. Acta Biomater 2024; 180:230-243. [PMID: 38574880 DOI: 10.1016/j.actbio.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/22/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024]
Abstract
In tissue engineering, crosslinking with carbodiimides such as EDC is omnipresent to improve the mechanical properties of biomaterials. However, in collagen biomaterials, EDC reacts with glutamate or aspartate residues, inactivating the binding sites for cellular receptors and rendering collagen inert to many cell types. In this work, we have developed a crosslinking method that ameliorates the rigidity, stability, and degradation rate of collagen biomaterials, whilst retaining key interactions between cells and the native collagen sequence. Our approach relies on the UV-triggered reaction of diazirine groups grafted on lysines, leaving critical amino acid residues intact. Notably, GxxGER recognition motifs for collagen-binding integrins, ablated by EDC crosslinking, were left unreacted, enabling cell attachment, spreading, and colonization on films and porous scaffolds. In addition, our procedure conserves the architecture of biomaterials, improves their resistance to collagenase and cellular contraction, and yields material stiffness akin to that obtained with EDC. Importantly, diazirine-crosslinked collagen can host mesenchymal stem cells, highlighting its strong potential as a substrate for tissue repair. We have therefore established a new crosslinking strategy to modulate the mechanical features of collagen porous scaffolds without altering its biological properties, thereby offering an advantageous alternative to carbodiimide treatment. STATEMENT OF SIGNIFICANCE: This article describes an approach to improve the mechanical properties of collagen porous scaffolds, without impacting collagen's natural interactions with cells. This is significant because collagen crosslinking is overwhelmingly performed using carbodiimides, which results in a critical loss of cellular affinity. By contrast, our method leaves key cellular binding sites in the collagen sequence intact, enabling cell-biomaterial interactions. It relies on the fast, UV-triggered reaction of diazirine with collagen, and does not produce toxic by-products. It also supports the culture of mesenchymal stem cells, a pivotal cell type in a wide range of tissue repair applications. Overall, our approach offers an attractive option for the crosslinking of collagen, a prominent material in the growing field of tissue engineering.
Collapse
Affiliation(s)
- Audrey Ziverec
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Daniel Bax
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Rd, Cambridge CB3 0FS, United Kingdom
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Edwin-Joffrey Courtial
- 3dFAB, Univ Lyon, Université Lyon1, CNRS, INSA, CPE-Lyon, ICBMS, UMR 5246, 43, Bd du 11 novembre 1918, 69622 Villeurbanne, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France
| | - Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, 69367 Lyon Cedex 07, France.
| |
Collapse
|
18
|
Sionkowska A, Kulka-Kamińska K, Brudzyńska P, Lewandowska K, Piwowarski Ł. The Influence of Various Crosslinking Conditions of EDC/NHS on the Properties of Fish Collagen Film. Mar Drugs 2024; 22:194. [PMID: 38786585 PMCID: PMC11123180 DOI: 10.3390/md22050194] [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: 03/26/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
The process of crosslinking improves the physicochemical properties of biopolymer-based composites, making them valuable for biomedical applications. EDC/NHS-crosslinked collagen materials have a significant potential for tissue engineering applications, due to their enhanced properties and biocompatibility. Chemical crosslinking of samples can be carried out in several ways, which is crucial and has a direct effect on the final properties of the obtained material. In this study, the effect of crosslinking conditions on the properties of collagen films using EDC and NHS was investigated. Studies included FTIR spectroscopy, AFM, swelling and degradation tests, mechanical testing and contact angle measurements. Evaluation of prepared collagen films indicated that both crosslinking agents and crosslinking conditions influenced film properties. Notable alternations were observed in the infrared spectrum of the sample, to which EDC was added directly to the fish collagen solution. The same sample indicated the lowest Young modulus, tensile strength and breaking force parameters and the highest elongation at break. All samples reached the maximum swelling degree two hours after immersion in PBS solution; however, the immersion-crosslinked samples exhibited a significantly lower degree of swelling and were highly durable. The highest roughness was observed for the collagen film crosslinked with EDC, whereas the lowest was observed for the specimen crosslinked with EDC with NHS addition. The crosslinking agents increased the surface roughness of the collagen film, except for the sample modified with the addition of EDC and NHS mixture. All films were characterized by hydrophilic character. The films' modification resulted in a decrease in their hydrophilicity and wettability. Our research allows for a comparison of proposed EDC/NHS crosslinking conditions and their influence on the physicochemical properties of fish collagen thin films. EDC and NHS are promising crosslinking agents for the modification of fish collagen used in biomedical applications.
Collapse
Affiliation(s)
- Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland; (K.K.-K.); (P.B.); (K.L.)
| | - Karolina Kulka-Kamińska
- Department of Biomaterials and Cosmetic Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland; (K.K.-K.); (P.B.); (K.L.)
| | - Patrycja Brudzyńska
- Department of Biomaterials and Cosmetic Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland; (K.K.-K.); (P.B.); (K.L.)
| | - Katarzyna Lewandowska
- Department of Biomaterials and Cosmetic Chemistry, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100 Torun, Poland; (K.K.-K.); (P.B.); (K.L.)
| | - Łukasz Piwowarski
- SanColl Sp. z o. o., Juliusza Słowackiego 24, 35-060 Rzeszów, Poland;
| |
Collapse
|
19
|
Carnes ME, Gonyea CR, Coburn JM, Pins GD. A biomimetic approach to modulating the sustained release of fibroblast growth factor 2 from fibrin microthread scaffolds. EXPLORATION OF BIOMAT-X 2024; 1:58-83. [PMID: 39070763 PMCID: PMC11274095 DOI: 10.37349/ebmx.2024.00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2024]
Abstract
Aim The pleiotropic effect of fibroblast growth factor 2 (FGF2) on promoting myogenesis, angiogenesis, and innervation makes it an ideal growth factor for treating volumetric muscle loss (VML) injuries. While an initial delivery of FGF2 has demonstrated enhanced regenerative potential, the sustained delivery of FGF2 from scaffolds with robust structural properties as well as biophysical and biochemical signaling cues has yet to be explored for treating VML. The goal of this study is to develop an instructive fibrin microthread scaffold with intrinsic topographic alignment cues as well as regenerative signaling cues and a physiologically relevant, sustained release of FGF2 to direct myogenesis and ultimately enhance functional muscle regeneration. Methods Heparin was passively adsorbed or carbodiimide-conjugated to microthreads, creating a biomimetic binding strategy, mimicking FGF2 sequestration in the extracellular matrix (ECM). It was also evaluated whether FGF2 incorporated into fibrin microthreads would yield sustained release. It was hypothesized that heparin-conjugated and co-incorporated (co-inc) fibrin microthreads would facilitate sustained release of FGF2 from the scaffold and enhance in vitro myoblast proliferation and outgrowth. Results Toluidine blue staining and Fourier transform infrared spectroscopy confirmed that carbodiimide-conjugated heparin bound to fibrin microthreads in a dose-dependent manner. Release kinetics revealed that heparin-conjugated fibrin microthreads exhibited sustained release of FGF2 over a period of one week. An in vitro assay demonstrated that FGF2 released from microthreads remained bioactive, stimulating myoblast proliferation over four days. Finally, a cellular outgrowth assay suggests that FGF2 promotes increased outgrowth onto microthreads. Conclusions It was anticipated that the combined effects of fibrin microthread structural properties, topographic alignment cues, and FGF2 release profiles will facilitate the fabrication of a biomimetic scaffold that enhances the regeneration of functional muscle tissue for the treatment of VML injuries.
Collapse
Affiliation(s)
- Meagan E. Carnes
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Cailin R. Gonyea
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - Jeannine M. Coburn
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - George D. Pins
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| |
Collapse
|
20
|
Varga V, Smeller L, Várdai R, Kocsis B, Zsoldos I, Cruciani S, Pala R, Hornyák I. Water-Insoluble, Thermostable, Crosslinked Gelatin Matrix for Soft Tissue Implant Development. Int J Mol Sci 2024; 25:4336. [PMID: 38673921 PMCID: PMC11050114 DOI: 10.3390/ijms25084336] [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: 02/26/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
In this present study, the material science background of crosslinked gelatin (GEL) was investigated. The aim was to assess the optimal reaction parameters for the production of a water-insoluble crosslinked gelatin matrix suitable for heat sterilization. Matrices were subjected to enzymatic degradation assessments, and their ability to withstand heat sterilization was evaluated. The impact of different crosslinkers on matrix properties was analyzed. It was found that matrices crosslinked with butanediol diglycidyl ether (BDDE) and poly(ethylene glycol) diglycidyl ether (PEGDE) were resistant to enzymatic degradation and heat sterilization. Additionally, at 1 v/v % crosslinker concentration, the crosslinked weight was lower than the starting weight, suggesting simultaneous degradation and crosslinking. The crosslinked weight and swelling ratio were optimal in the case of the matrices that were crosslinked with 3% and 5% v/v BDDE and PEGDE. FTIR analysis confirmed crosslinking, and the reduction of free primary amino groups indicated effective crosslinking even at a 1% v/v crosslinker concentration. Moreover, stress-strain and compression characteristics of the 5% v/v BDDE crosslinked matrix were comparable to native gelatin. Based on material science measurements, the crosslinked matrices may be promising candidates for scaffold development, including properties such as resistance to enzymatic degradation and heat sterilization.
Collapse
Affiliation(s)
- Viktória Varga
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
- Department of Materials Science and Technology, University of Győr, 9026 Győr, Hungary; (B.K.); (I.Z.)
| | - László Smeller
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary;
| | - Róbert Várdai
- Laboratory of Plastics and Rubber Technology, Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, 1111 Budapest, Hungary;
- Research Centre for Natural Sciences, Institute of Materials and Environmental Chemistry, 1111 Budapest, Hungary
| | - Bence Kocsis
- Department of Materials Science and Technology, University of Győr, 9026 Győr, Hungary; (B.K.); (I.Z.)
| | - Ibolya Zsoldos
- Department of Materials Science and Technology, University of Győr, 9026 Győr, Hungary; (B.K.); (I.Z.)
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (R.P.)
| | - Renzo Pala
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy; (S.C.); (R.P.)
| | - István Hornyák
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary;
- Department of Materials Science and Technology, University of Győr, 9026 Győr, Hungary; (B.K.); (I.Z.)
| |
Collapse
|
21
|
Köksal B, Kartal RB, Günay US, Durmaz H, Yildiz AA, Yildiz ÜH. Fabrication of gelatin-polyester based biocomposite scaffold via one-step functionalization of melt electrowritten polymer blends in aqueous phase. Int J Biol Macromol 2024; 265:130938. [PMID: 38493814 DOI: 10.1016/j.ijbiomac.2024.130938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The rapid manufacturing of biocomposite scaffold made of saturated-Poly(ε-caprolactone) (PCL) and unsaturated Polyester (PE) blends with gelatin and modified gelatin (NCO-Gel) is demonstrated. Polyester blend-based scaffold are fabricated with and without applying potential in the melt electrowriting system. Notably, the applied potential induces phase separation between PCL and PE and drives the formation of PE rich spots at the interface of electrowritten fibers. The objective of the current study is to control the phase separation between saturated and unsaturated polyesters occurring in the melt electro-writing process and utilization of this phenomenon to improve efficiency of biofunctionalization at the interface of scaffold via Aza-Michael addition reaction. Electron-deficient triple bonds of PE spots on the fibers exhibit good potential for the biofunctionalization via the aza-Michael addition reaction. PE spots are found to be pronounced in which blend compositions are PCL-PE as 90:10 and 75:25 %. The biofunctionalization of scaffold is monitored through CN bond formation appeared at 400 eV via X-ray photoelectron spectroscopy (XPS) and XPS chemical mapping. The described biofunctionalization methodology suggest avoiding use of multi-step chemical modification on additive manufacturing products and thereby rapid prototyping of functional polymer blend based scaffolds with enhanced biocompatibility and preserved mechanical properties. Additionally one-step additive manufacturing method eliminates side effects of toxic solvents and long modification steps during scaffold fabrication.
Collapse
Affiliation(s)
- Büşra Köksal
- Department of Chemistry, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey
| | | | - Ufuk Saim Günay
- Department of Chemistry, Istanbul Technical University, 34469 Maslak, İstanbul, Turkey
| | - Hakan Durmaz
- Department of Chemistry, Istanbul Technical University, 34469 Maslak, İstanbul, Turkey
| | - Ahu Arslan Yildiz
- Department of Bioengineering, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey
| | - Ümit Hakan Yildiz
- Department of Chemistry, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey; Department of Polymer Science and Engineering, Izmir Institute of Technology, 35430 Urla, İzmir, Turkey.
| |
Collapse
|
22
|
King NC, McGuire KR, Bejar-Chapa M, Hoftiezer YAJ, Randolph MA, Winograd JM. Photochemical Tissue Bonding of Amnion Allograft Membranes for Peripheral Nerve Repair: A Biomechanical Analysis. J Reconstr Microsurg 2024; 40:232-238. [PMID: 37696294 DOI: 10.1055/s-0043-1772670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
BACKGROUND Photochemical tissue bonding (PTB) is a technique for peripheral nerve repair in which a collagenous membrane is bonded around approximated nerve ends. Studies using PTB with cryopreserved human amnion have shown promising results in a rat sciatic nerve transection model including a more rapid and complete return of function, larger axon size, and thicker myelination than suture repair. Commercial collagen membranes, such as dehydrated amnion allograft, are readily available, offer ease of storage, and have no risk of disease transmission or tissue rejection. However, the biomechanical properties of these membranes using PTB are currently unknown in comparison to PTB of cryopreserved human amnion and suture neurorrhaphy. METHODS Rat sciatic nerves (n = 10 per group) were transected and repaired using either suture neurorrhaphy or PTB with one of the following membranes: cryopreserved human amnion, monolayer human amnion allograft (crosslinked and noncrosslinked), trilayer human amnion/chorion allograft (crosslinked and noncrosslinked), or swine submucosa. Repaired nerves were subjected to mechanical testing. RESULTS During ultimate stress testing, the repair groups that withstood the greatest strain increases were suture neurorrhaphy (69 ± 14%), PTB with crosslinked trilayer amnion (52 ± 10%), and PTB with cryopreserved human amnion (46 ± 20%), although the differences between these groups were not statistically significant. Neurorrhaphy repairs had a maximum load (0.98 ± 0.30 N) significantly greater than all other repair groups except for noncrosslinked trilayer amnion (0.51 ± 0.27 N). During fatigue testing, all samples repaired with suture, or PTBs with either crosslinked or noncrosslinked trilayer amnion were able to withstand strain increases of at least 50%. CONCLUSION PTB repairs with commercial noncrosslinked amnion allograft membranes can withstand physiological strain and have comparable performance to repairs with human amnion, which has demonstrated efficacy in vivo. These results indicate the need for further testing of these membranes using in vivo animal model repairs.
Collapse
Affiliation(s)
- Nicholas C King
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kalyn R McGuire
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria Bejar-Chapa
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yannick A J Hoftiezer
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Mark A Randolph
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jonathan M Winograd
- Peripheral Nerve Research Laboratory, Division of Plastic and Reconstructive Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Salvatore L, Russo F, Natali ML, Rajabimashhadi Z, Bagheri S, Mele C, Lionetto F, Sannino A, Gallo N. On the effect of pepsin incubation on type I collagen from horse tendon: Fine tuning of its physico-chemical and rheological properties. Int J Biol Macromol 2024; 256:128489. [PMID: 38043667 DOI: 10.1016/j.ijbiomac.2023.128489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/10/2023] [Accepted: 11/27/2023] [Indexed: 12/05/2023]
Abstract
Type I collagen is commonly recognized as the gold standard biomaterial for the manufacturing of medical devices for health-care related applications. In recent years, with the final aim of developing scaffolds with optimal bioactivity, even more studies focused on the influence of processing parameters on collagen properties, since processing can strongly affect the architecture of collagen at various length scales and, consequently, scaffolds macroscopic performances. The ability to finely tune scaffold properties in order to closely mimic the tissues' hierarchical features, preserving collagen's natural conformation, is actually of great interest. In this work, the effect of the pepsin-based extraction step on the material final properties was investigated. Thus, the physico-chemical properties of fibrillar type I collagens upon being extracted under various conditions were analyzed in depth. Correlations of collagen structure at the supramolecular scale with its microstructural properties were done, confirming the possibility of tuning rheological, viscoelastic and degradation properties of fibrillar type I collagen.
Collapse
Affiliation(s)
- Luca Salvatore
- Typeone Biomaterials Srl, Via Europa 167, Calimera, 73021 Lecce, Italy.
| | - Francesca Russo
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | | | - Zahra Rajabimashhadi
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Sonia Bagheri
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Claudio Mele
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Francesca Lionetto
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Alessandro Sannino
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Nunzia Gallo
- Typeone Biomaterials Srl, Via Europa 167, Calimera, 73021 Lecce, Italy; Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| |
Collapse
|
24
|
Khan R, Haider S, Khan MUA, Haider A, Razak SIA, Hasan A, Khan R, Wahit MU. Fabrication of amine-functionalized and multi-layered PAN-(TiO 2)-gelatin nanofibrous wound dressing: In-vitro evaluation. Int J Biol Macromol 2023; 253:127169. [PMID: 37783243 DOI: 10.1016/j.ijbiomac.2023.127169] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/16/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023]
Abstract
The development of advanced multifunctional wound dressings remains a major challenge. Herein, a novel multilayer (ML) electrospun nanofibers (NFs) wound dressing based on diethylenetriamine (DETA) functionalized polyacrylonitrile (PAN), TiO2 nanoparticles (NPs) coating (Ct), and bioderived gelatin (Gel) was developed for potential applications in wound healing. The ML PAN-DETA-Ct-Gel membrane was developed by combining electrospinning, chemical functionalization, synthesis, and electrospray techniques, using a layer-by-layer method. The ML PAN-DETA-Ct-Gel membrane is comprised of an outer layer of PAN-DETA as a barrier to external microorganisms and structural support, an interlayer TiO2 NPs (Ct) as antibacterial function, and a contact layer (Gel) to improve biocompatibility and cell viability. The NFs membranes were characterized by scanning electron microscopy (SEM), surface profilometry, BET analysis, and water contact angle techniques to investigate their morphology, surface roughness, porosity, and wettability. The ML PAN-DETA-Ct-Gel wound dressing exhibited good surface roughness, porosity, and better wettability. Cell morphology, proliferation, and viability were determined using fibroblasts (3T3), and antibacterial assays were performed against six pathogens. The ML PAN-DETA-Ct-Gel NFs membrane showed good cell morphology, proliferation, viability, and antibacterial activity compared with other membranes. This new class of ML NFs membranes offers a multifunctional architecture with adequate biocompatibility, cell viability, and antibacterial activity.
Collapse
Affiliation(s)
- Rawaiz Khan
- Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia (UTM), 81310 UTM Skudai, Johor Bahru, Johor, Malaysia
| | - Sajjad Haider
- Department of Chemical Engineering, College of Engineering, King Saud University, Riyadh 11421, Saudi Arabia; King Salman Center for Disability Research, Riyadh 11614, Saudi Arabia.
| | - Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Adnan Haider
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Punjab, Pakistan
| | - Saiful Izwan Abd Razak
- Sports Innovation & Technology Centre, Institute of Human Centred Engineering, Universiti Teknologi Malaysia, 81300 Skudai, Johor, Malaysia; BioInspired Device and Tissue Engineering Research Group, School of Biomedical Engineering and Health Sciences, Faculty of Engineering, Universiti Teknologi Malaysia, 81300 Skudai, Johor, Malaysia.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Raees Khan
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Punjab, Pakistan
| | - Mat Uzir Wahit
- Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia (UTM), 81310 UTM Skudai, Johor Bahru, Johor, Malaysia; Center for Advanced Composite Materials (CACM), Universiti Teknologi Malaysia (UTM), 81310 UTM Skudai, Johor Bahru, Johor, Malaysia
| |
Collapse
|
25
|
Olguín Y, Selva M, Benavente D, Orellana N, Montenegro I, Madrid A, Jaramillo-Pinto D, Otero MC, Corrales TP, Acevedo CA. Effect of Electrical Stimulation on PC12 Cells Cultured in Different Hydrogels: Basis for the Development of Biomaterials in Peripheral Nerve Tissue Engineering. Pharmaceutics 2023; 15:2760. [PMID: 38140099 PMCID: PMC10747664 DOI: 10.3390/pharmaceutics15122760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/26/2023] [Accepted: 12/02/2023] [Indexed: 12/24/2023] Open
Abstract
Extensive damage to peripheral nerves is a health problem with few therapeutic alternatives. In this context, the development of tissue engineering seeks to obtain materials that can help recreate environments conducive to cellular development and functional repair of peripheral nerves. Different hydrogels have been studied and presented as alternatives for future treatments to emulate the morphological characteristics of nerves. Along with this, other research proposes the need to incorporate electrical stimuli into treatments as agents that promote cell growth and differentiation; however, no precedent correlates the simultaneous effects of the types of hydrogel and electrical stimuli. This research evaluates the neural differentiation of PC12 cells, relating the effect of collagen, alginate, GelMA, and PEGDA hydrogels with electrical stimulation modulated in four different ways. Our results show significant correlations for different cultivation conditions. Electrical stimuli significantly increase neural differentiation for specific experimental conditions dependent on electrical frequency, not voltage. These backgrounds allow new material treatment schemes to be formulated through electrical stimulation in peripheral nerve tissue engineering.
Collapse
Affiliation(s)
- Yusser Olguín
- Departamento de Química y Medio Ambiente, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile
- Centro Científico y Tecnológico de Valparaíso (CCTVal), Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (D.B.); (C.A.A.)
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (M.S.); (N.O.); (T.P.C.)
| | - Mónica Selva
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (M.S.); (N.O.); (T.P.C.)
| | - Diego Benavente
- Centro Científico y Tecnológico de Valparaíso (CCTVal), Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (D.B.); (C.A.A.)
| | - Nicole Orellana
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (M.S.); (N.O.); (T.P.C.)
| | - Ivan Montenegro
- Centro de Investigaciones Biomédicas, Escuela de Obstetricia, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Reñaca, Viña del Mar 2520000, Chile;
| | - Alejandro Madrid
- Laboratorio de Productos Naturales y Síntesis Orgánica (LPNSO), Departamento de Ciencias y Geografía, Facultad de Ciencias Naturales y Exactas, Universidad de Playa Ancha, Avda. Leopoldo Carvallo 270, Playa Ancha, Valparaíso 2390123, Chile;
| | - Diego Jaramillo-Pinto
- Departamento de Física, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile;
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad Técnica Federico Santa María, Valparaíso 2390123, Chile
| | - María Carolina Otero
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Republica 252, Santiago 8370071, Chile;
| | - Tomas P. Corrales
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (M.S.); (N.O.); (T.P.C.)
- Departamento de Física, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile;
- Millenium Nucleus in NanoBioPhysics (NNBP), Universidad Técnica Federico Santa María, Valparaíso 2390123, Chile
| | - Cristian A. Acevedo
- Centro Científico y Tecnológico de Valparaíso (CCTVal), Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (D.B.); (C.A.A.)
- Centro de Biotecnología, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile; (M.S.); (N.O.); (T.P.C.)
- Departamento de Física, Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2390123, Chile;
| |
Collapse
|
26
|
Lee C. Injectable glucose oxidase-immobilized gelatin hydrogel prevents tumor recurrence via oxidation therapy. Colloids Surf B Biointerfaces 2023; 232:113581. [PMID: 37857184 DOI: 10.1016/j.colsurfb.2023.113581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/21/2023]
Abstract
In clinical practice, surgery is the preferred treatment for breast cancer; however, the high recurrence rate due to residual tumors after surgery remains a major issue. Hydrogels can reduce the side effects of residual tumors and exert strong anticancer effects, thereby showing potential as therapeutic agents for suppressing tumor recurrence after surgery. Glucose oxidase (GOD)-immobilized gelatin hydrogels (GOD-gelatin hydrogel) were prepared by bioorthogonal click chemistry. Then, the anticancer effect, tumor recurrence inhibition, and biodegradability of the resulting hydrogels were evaluated through cell and animal experiments. GOD-gelatin hydrogel showed cytotoxicity and anticancer effect via H2O2 generation. Unlike free GOD, GOD-gelatin hydrogel remained in the surgical site after implant and continued to suppress tumor recurrence over time. The proposed GOD-gelatin hydrogel system can be easily implanted at the surgical site after tumor surgery, representing a novel treatment to suppress tumor recurrence without any systemic toxicity.
Collapse
Affiliation(s)
- Changkyu Lee
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, the Republic of Korea.
| |
Collapse
|
27
|
Miwa H, Antao OQ, Kelly‐Scumpia KM, Baghdasarian S, Mayer DP, Shang L, Sanchez GM, Archang MM, Scumpia PO, Weinstein JS, Di Carlo D. Improved Humoral Immunity and Protection against Influenza Virus Infection with a 3d Porous Biomaterial Vaccine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302248. [PMID: 37750461 PMCID: PMC10625058 DOI: 10.1002/advs.202302248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/03/2023] [Indexed: 09/27/2023]
Abstract
New vaccine platforms that activate humoral immunity and generate neutralizing antibodies are required to combat emerging pathogens, including influenza virus. A slurry of antigen-loaded hydrogel microparticles that anneal to form a porous scaffold with high surface area for antigen uptake by infiltrating immune cells as the biomaterial degrades is demonstrated to enhance humoral immunity. Antigen-loaded-microgels elicited a robust cellular humoral immune response, with increased CD4+ T follicular helper (Tfh) cells and prolonged germinal center (GC) B cells comparable to the commonly used adjuvant, aluminum hydroxide (Alum). Increasing the weight fraction of polymer material led to increased material stiffness and antigen-specific antibody titers superior to Alum. Vaccinating mice with inactivated influenza virus loaded into this more highly cross-linked formulation elicited a strong antibody response and provided protection against a high dose viral challenge. By tuning physical and chemical properties, adjuvanticity can be enhanced leading to humoral immunity and protection against a pathogen, leveraging two different types of antigenic material: individual protein antigen and inactivated virus. The flexibility of the platform may enable design of new vaccines to enhance innate and adaptive immune cell programming to generate and tune high affinity antibodies, a promising approach to generate long-lasting immunity.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Olivia Q. Antao
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Kindra M. Kelly‐Scumpia
- Division of CardiologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Sevana Baghdasarian
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Daniel P. Mayer
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Lily Shang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
| | - Gina M. Sanchez
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Maani M. Archang
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- MSTP ProgramDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
| | - Philip O. Scumpia
- Division of DermatologyDepartment of MedicineDavid Geffen School of MedicineUniversity of California Los AngelesLos AngelesCA90095USA
- Department of DermatologyVA Greater Los Angeles Healthcare SystemLos AngelesCA90073USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
| | - Jason S Weinstein
- Center for Immunity and InflammationRutgers New Jersey Medical SchoolNewarkNJ07103USA
| | - Dino Di Carlo
- Department of BioengineeringUniversity of California Los AngelesLos AngelesCA90095USA
- Jonsson Comprehensive Cancer CenterUniversity of California Los AngelesLos AngelesCA90095USA
- Department of Mechanical and Aerospace EngineeringUniversity of California Los AngelesLos AngelesCA90095USA
- California Nano Systems Institute (CNSI)University of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
28
|
Liu D, Meng Q, Hu J. Bacterial Nanocellulose Hydrogel: A Promising Alternative Material for the Fabrication of Engineered Vascular Grafts. Polymers (Basel) 2023; 15:3812. [PMID: 37765666 PMCID: PMC10534661 DOI: 10.3390/polym15183812] [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: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/29/2023] Open
Abstract
Blood vessels are crucial in the human body, providing essential nutrients to all tissues while facilitating waste removal. As the incidence of cardiovascular disease rises, the demand for efficient treatments increases concurrently. Currently, the predominant interventions for cardiovascular disease are autografts and allografts. Although effective, they present limitations including high costs and inconsistent success rates. Recently, synthetic vascular grafts, made from artificial materials, have emerged as promising alternatives to traditional methods. Among these materials, bacterial cellulose hydrogel exhibits significant potential for tissue engineering applications, particularly in developing nanoscale platforms that regulate cell behavior and promote tissue regeneration, attributed to its notable physicochemical and biocompatible properties. This study reviews recent progress in fabricating engineered vascular grafts using bacterial nanocellulose, demonstrating the efficacy of bacterial cellulose hydrogel as a biomaterial for synthetic vascular grafts, specifically for stimulating angiogenesis and neovascularization.
Collapse
Affiliation(s)
| | | | - Jinguang Hu
- Department of Chemical and Petroleum Engineering, University of Calgary, 2500 University Drive, Calgary, AB T2N 1N4, Canada; (D.L.); (Q.M.)
| |
Collapse
|
29
|
Dutta D, Graupner N, Müssig J, Brüggemann D. Assembly of Rolled-Up Collagen Constructs on Porous Alumina Textiles. ACS NANOSCIENCE AU 2023; 3:286-294. [PMID: 37601922 PMCID: PMC10436369 DOI: 10.1021/acsnanoscienceau.3c00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 08/22/2023]
Abstract
Developing new techniques to prepare free-standing tubular scaffolds has always been a challenge in the field of regenerative medicine. Here, we report a new and simple way to prepare free-standing collagen constructs with rolled-up architecture by self-assembling nanofibers on porous alumina (Al2O3) textiles modified with different silanes, carbon or gold. Following self-assembly and cross-linking with glutaraldehyde, collagen nanofibers spontaneously rolled up on the modified Al2O3 textiles and detached. The resulting collagen constructs had an inner diameter of approximately 2 to 4 mm in a rolled-up state and could be easily detached from the underlying textiles. Mechanical testing of wet collagen scaffolds following detachment yielded mean values of 3.5 ± 1.9 MPa for the tensile strength, 41.0 ± 20.8 MPa for the Young's modulus and 8.1 ± 3.7% for the elongation at break. No roll-up was observed on Al2O3 textiles without any modification, where collagen did not assemble into fibers, either. Blends of collagen and chitosan were also found to roll into fibrous constructs on silanized Al2O3 textiles, while fibrinogen nanofibers or blends of collagen and elastin did not yield such structures. Based on these differences, we hypothesize that textile surface charge and protein charge, in combination with the porous architecture of protein nanofibers and differences in mechanical strain, are key factors in inducing a scaffold roll-up. Further studies are required to develop the observed roll-up effect into a reproducible biofabrication process that can enable the controlled production of free-standing collagen-based tubes for soft tissue engineering.
Collapse
Affiliation(s)
- Deepanjalee Dutta
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
| | - Nina Graupner
- The Biological Materials Group, Biomimetics, Faculty 5, HSB - City University of Applied Sciences Bremen, Neustadtswall 30, 28199 Bremen, Germany
| | - Jörg Müssig
- The Biological Materials Group, Biomimetics, Faculty 5, HSB - City University of Applied Sciences Bremen, Neustadtswall 30, 28199 Bremen, Germany
| | - Dorothea Brüggemann
- Institute for Biophysics, University of Bremen, Otto-Hahn-Allee 1, 28359 Bremen, Germany
- MAPEX Center for Materials and Processes, University of Bremen, 28359 Bremen, Germany
| |
Collapse
|
30
|
Roldán E, Reeves ND, Cooper G, Andrews K. Can we achieve biomimetic electrospun scaffolds with gelatin alone? Front Bioeng Biotechnol 2023; 11:1160760. [PMID: 37502104 PMCID: PMC10368888 DOI: 10.3389/fbioe.2023.1160760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023] Open
Abstract
Introduction: Gelatin is a natural polymer commonly used in biomedical applications in combination with other materials due to its high biocompatibility, biodegradability, and similarity to collagen, principal protein of the extracellular matrix (ECM). The aim of this study was to evaluate the suitability of gelatin as the sole material to manufacture tissue engineering scaffolds by electrospinning. Methods: Gelatin was electrospun in nine different concentrations onto a rotating collector and the resulting scaffold's mechanical properties, morphology and topography were assessed using mechanical testing, scanning electron microscopy and white light interferometry, respectively. After characterizing the scaffolds, the effects of the concentration of the solvents and crosslinking agent were statistically evaluated with multivariate analysis of variance and linear regressions. Results: Fiber diameter and inter-fiber separation increased significantly when the concentration of the solvents, acetic acid (HAc) and dimethyl sulfoxide (DMSO), increased. The roughness of the scaffolds decreased as the concentration of dimethyl sulfoxide increased. The mechanical properties were significantly affected by the DMSO concentration. Immersed crosslinked scaffolds did not degrade until day 28. The manufactured gelatin-based electrospun scaffolds presented comparable mechanical properties to many human tissues such as trabecular bone, gingiva, nasal periosteum, oesophagus and liver tissue. Discussion: This study revealed for the first time that biomimetic electrospun scaffolds with gelatin alone can be produced for a significant number of human tissues by appropriately setting up the levels of factors and their interactions. These findings also extend statistical relationships to a form that would be an excellent starting point for future research that could optimize factors and interactions using both traditional statistics and machine learning techniques to further develop specific human tissue.
Collapse
Affiliation(s)
- Elisa Roldán
- Department of Engineering, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Neil D. Reeves
- Research Centre for Musculoskeletal Science and Sports Medicine, Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| | - Glen Cooper
- School of Engineering, University of Manchester, Manchester, United Kingdom
| | - Kirstie Andrews
- Department of Engineering, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, United Kingdom
| |
Collapse
|
31
|
Sanapalli BKR, Yele V, Singh MK, Thumbooru SN, Parvathaneni M, Karri VVSR. Human beta defensin-2 loaded PLGA nanoparticles impregnated in collagen-chitosan composite scaffold for the management of diabetic wounds. Biomed Pharmacother 2023; 161:114540. [PMID: 36934557 DOI: 10.1016/j.biopha.2023.114540] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/26/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023] Open
Abstract
Diabetic wound (DW) is the most devastating complication resulting in significant mortality and morbidity in diabetic patients. The standard treatment of DW care fails to address the prerequisites of treating DW owing to its multifactorial pathophysiology. Henceforth, developing a single treatment strategy to handle all the loopholes may effectively manage DW. The objective of the current study was to formulate Human beta defensin-2 (HBD-2) loaded Poly (lactic-co-glycolic acid) (PLGA) nanoparticle impregnated in collagen/chitosan (COL-CS) composite scaffolds for the accelerated healing of DW. Upon investigation, the developed biodegradable crosslinked scaffold possesses low matrix degradation, optimum porosity, and sustained drug release than the non-crosslinked scaffold. In vitro studies revealed that the HBD-2 COL-CS scaffold was biocompatible and accelerated cell migration and angiogenesis. The HBD-2 COL-CS scaffold showed significant antimicrobial activity in S. aureus, E. coli, and P. aeruginosa. The in vivo studies revealed that the HBD-2 COL-CS treated group accelerated healing compared to those in COL-CS and control groups. The ELISA results indicated a significant decrease in MMP-9, TNF-α, MPO, NAG, and NO with an increase in IL-10 in HBD-2 COL-CS treated group. The accelerated healing in HBD-2 COL-CS treated group might be due to the synergistic effects of PLGA (collagen synthesis and deposition and positive angiogenic effect), HBD-2 (anti-inflammatory, antibacterial, positive angiogenic effect, cell proliferation, and migration), COL (established wound healer and stabilizer) and CS (antibacterial, controlled drug release).
Collapse
Affiliation(s)
- Bharat Kumar Reddy Sanapalli
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Vidyasrilekha Yele
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Mantosh Kumar Singh
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Shilpa N Thumbooru
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu 643001, India.
| | - Madhukiran Parvathaneni
- Department of Biotechnology, Harrisburg University of Science & Technology, 326 Market Street, Harrisburg, PA 17101, USA; Arni Medica, 4475 South Clinton Ave, Suite 230, South Plainfield, NJ 07080, USA; CRC Pharma LLC, 333 Littleton Road, Parsippany, NJ 07054, USA.
| | | |
Collapse
|
32
|
Rizwan A, Gulfam M, Jo SH, Seo JW, Ali I, Thang Vu T, Joo SB, Park SH, Taek Lim K. Gelatin-based NIR and reduction-responsive injectable hydrogels cross-linked through IEDDA click chemistry for drug delivery application. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.112019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
33
|
Barroca N, da Silva DM, Pinto SC, Sousa JPM, Verstappen K, Klymov A, Fernández-San-Argimiro FJ, Madarieta I, Murua O, Olalde B, Papadimitriou L, Karali K, Mylonaki K, Stratakis E, Ranella A, Marques PAAP. Interfacing reduced graphene oxide with an adipose-derived extracellular matrix as a regulating milieu for neural tissue engineering. BIOMATERIALS ADVANCES 2023; 148:213351. [PMID: 36842343 DOI: 10.1016/j.bioadv.2023.213351] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/31/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Enthralling evidence of the potential of graphene-based materials for neural tissue engineering is motivating the development of scaffolds using various structures related to graphene such as graphene oxide (GO) or its reduced form. Here, we investigated a strategy based on reduced graphene oxide (rGO) combined with a decellularized extracellular matrix from adipose tissue (adECM), which is still unexplored for neural repair and regeneration. Scaffolds containing up to 50 wt% rGO relative to adECM were prepared by thermally induced phase separation assisted by carbodiimide (EDC) crosslinking. Using partially reduced GO enables fine-tuning of the structural interaction between rGO and adECM. As the concentration of rGO increased, non-covalent bonding gradually prevailed over EDC-induced covalent conjugation with the adECM. Edge-to-edge aggregation of rGO favours adECM to act as a biomolecular physical crosslinker to rGO, leading to the softening of the scaffolds. The unique biochemistry of adECM allows neural stem cells to adhere and grow. Importantly, high rGO concentrations directly control cell fate by inducing the differentiation of both NE-4C cells and embryonic neural progenitor cells into neurons. Furthermore, primary astrocyte fate is also modulated as increasing rGO boosts the expression of reactivity markers while unaltering the expression of scar-forming ones.
Collapse
Affiliation(s)
- Nathalie Barroca
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| | - Daniela M da Silva
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Susana C Pinto
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Joana P M Sousa
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal
| | - Kest Verstappen
- Radboud University Nijmegen Medical Centre, Department of Regenerative Biomaterials, 6500HB Nijmegen, the Netherlands
| | - Alexey Klymov
- Radboud University Nijmegen Medical Centre, Department of Regenerative Biomaterials, 6500HB Nijmegen, the Netherlands
| | | | - Iratxe Madarieta
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Olatz Murua
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Beatriz Olalde
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Lina Papadimitriou
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Kanelina Karali
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Konstantina Mylonaki
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece
| | - Anthi Ranella
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH), Heraklion, 71003, Greece.
| | - Paula A A P Marques
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| |
Collapse
|
34
|
Liang Y, Zou Y, Wu S, Song D, Xu W, Zhu K. Preparation and properties of chitin/silk fibroin biocompatible composite fibers. JOURNAL OF BIOMATERIALS SCIENCE, POLYMER EDITION 2022; 34:860-874. [PMID: 36369874 DOI: 10.1080/09205063.2022.2147746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In the present world chitin is used enormously in various fields, such as biopharmaceuticals, medical and clinical bioproducts, food packaging, etc. However, its development has been curbed by the impaired performance and cumbersome dissolution process when chitin materials are dissolved and regenerated by physical or chemical methods. To further obtain the regenerated chitin fiber material with improved performance, silk fibroin was introduced into the chitin matrix material, and chitin/silk fibroin biocompatible composite fibers were obtained by formic acid/calcium chloride/ethanol ternary system and top-down wet spinning technology. The produced composite fibers outperformed previously reported chitin-silk composites in terms of the tensile strength (160 MPa) and failure strain (25%). The fibers also performed good cell compatibility and strong cellular affinity for non-toxicity. The cell viabilities of the fibers were about 20% greater than those of silk fiber after three days of co-culture with NIH-3T3. Furthermore, no hemolysis occurs in the presence of chitin/silk fibers, demonstrating their superior hemocompatibility. The fibers had a hemolysis index as low as 1%, which is far lower than the acceptable level of 5%. The material offers prospective opportunities for biomaterial applications in anticoagulation, absorbable surgical sutures, etc.
Collapse
Affiliation(s)
- Yaoting Liang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| | - Yongkang Zou
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Shuangquan Wu
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Dengpeng Song
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| | - Weilin Xu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| | - Kunkun Zhu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan 430200, China
| |
Collapse
|
35
|
Tian X, Zhao K, Teng A, Li Y, Wang W. A rethinking of collagen as tough biomaterials in meat packaging: assembly from native to synthetic. Crit Rev Food Sci Nutr 2022; 64:957-977. [PMID: 35997287 DOI: 10.1080/10408398.2022.2111401] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Due to the high moisture-associated typical rheology and the changeable and harsh processing conditions in the production process, packaging materials for meat products have higher requirements including a sufficient mechanical strength and proper ductility. Collagen, a highly conserved structural protein consisting of a triple helix of Gly-X-Y repeats, has been proved to be suitable packaging material for meat products. The treated animal digestive tract (i.e. the casing) is the perfect natural packaging material for wrapping meat into sausage. Its thin walls, strong toughness and impact resistance make it the oldest and best edible meat packaging. Collagen casing is another wisdom of meat packaging, which is made by collagen fibers from hide skin, presenting a rapid growth in casing market. To strengthen mechanical strength and barrier behaviors of collagen-based packaging materials, different physical, chemical, and biological cross-linking methods are springing up exuberantly, as well as a variety of reinforcement approaches including nanotechnology. In addition, the rapid development of biomimetic technology also provides a good research idea and means for the promotion of collagen's assembly and relevant mechanical properties. This review can offer some reference on fundamental theory and practical application of collagenous materials in meat products.
Collapse
Affiliation(s)
- Xiaojing Tian
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - KaiXuan Zhao
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Anguo Teng
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| | - Yu Li
- College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Wenhang Wang
- College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin, China
| |
Collapse
|
36
|
The Effect of the Topmost Layer and the Type of Bone Morphogenetic Protein-2 Immobilization on the Mesenchymal Stem Cell Response. Int J Mol Sci 2022; 23:ijms23169287. [PMID: 36012551 PMCID: PMC9408842 DOI: 10.3390/ijms23169287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/16/2022] Open
Abstract
Recombinant human bone morphogenetic protein-2 (rhBMP-2) plays a key role in the stem cell response, not only via its influence on osteogenesis, but also on cellular adhesion, migration, and proliferation. However, when applied clinically, its supra-physiological levels cause many adverse effects. Therefore, there is a need to concomitantly retain the biological activity of BMP-2 and reduce its doses. Currently, the most promising strategies involve site-specific and site-directed immobilization of rhBMP-2. This work investigated the covalent and electrostatic binding of rhBMP-2 to ultrathin-multilayers with chondroitin sulfate (CS) or diazoresin (DR) as the topmost layer. Angle-resolved X-ray photoelectron spectroscopy was used to study the exposed chemical groups. The rhBMP-2 binding efficiency and protein state were studied with time-of-flight secondary ion mass spectrometry. Quartz crystal microbalance, atomic force microscopy, and enzyme-linked immunosorbent assay were used to analyze protein–substrate interactions. The effect of the topmost layer was tested on initial cell adhesion and short-term osteogenesis marker expression. The results show the highest expression of selected osteomarkers in cells cultured on the DR-ended layer, while the cellular flattening was rather poor compared to the CS-ended system. rhBMP-2 adhesion was observed only on negatively charged layers. Cell flattening became more prominent in the presence of the protein, even though the osteogenic gene expression decreased.
Collapse
|
37
|
He X, Li W, Liu S, Li Y, Chen Y, Dan N, Dan W, Zhu M. Fabrication of high-strength, flexible, porous collagen-based scaffolds to promote tissue regeneration. Mater Today Bio 2022; 16:100376. [PMID: 35991626 PMCID: PMC9386468 DOI: 10.1016/j.mtbio.2022.100376] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/01/2022]
Abstract
Collagen-based scaffolds lack mechanical strength, flexibility, and tunable pore structure, affecting tissue repair outcomes and restricting their wide clinical application. Here, two kinds of scaffolds were prepared by a combination of vacuum homogenization, natural air drying, water soaking, lyophilization, and crosslinking. Compared with the scaffolds made of collagen molecules (Col-M), the scaffolds made of collagen aggregates (Col-A) exhibited higher mechanical strength (ultimate tensile strength: 1.38 ± 0.26 MPa vs 15.46 ± 1.55 MPa), stronger flexibility, advanced cell adhesion, survival, and proliferation. Subcutaneous implantation in rats showed that Col-A scaffolds promoted cell infiltration, macrophage polarization, and vascularization. Furthermore, the Col-A scaffolds inhibited abdominal bulges due to their adequate mechanical support, and they also promoted vascularized muscle regeneration in a rat abdominal hernia defect model. Our study provides a novel strategy for generating high-strength, flexible, porous collagen-based scaffolds, which can be applied to tissue repair with mechanical strength requirements. It broadens their application range in the field of regenerative medicine.
Collapse
|
38
|
Imani M, Mohajeri N, Rastegar M, Zarghami N. Synthesis and Characterization of N-rich Fluorescent Bio-dots as a Reporter in the Design of Dual-labeled FRET Probe for TaqMan PCR: a Feasibility Study. Biotechnol Appl Biochem 2022; 70:645-658. [PMID: 35900086 DOI: 10.1002/bab.2387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 07/12/2022] [Indexed: 11/07/2022]
Abstract
DNA-based analytical techniques have provided an advantageous sensing assay in the realm of biotechnology. Bio-inspired fluorescent nanodots are a novel type of biological staining agent with excellent optical properties widely used for cellular imaging and diagnostics. In the present research, we successfully synthesized bio-dots with excellent optical properties and high-quantum yield from DNA sodium salt through the hydrothermal method. We conjugated the bio-dots with 3' Eclipse® Dark Quencher (Eclipse) labeled single strand oligodeoxyribonucleotide according to carbodiimide chemistry, to design a fluorescence resonance energy transfer (FRET) probe. The results confirmed the prosperous synthesis and surface functionalization of the bio-dot. Analysis of size, zeta potential, and FTIR spectroscopy verified successful bioconjugation of the bio-dots with probes. UV-Visibility analysis and fluorescence intensity profile of the bio-dot and bio-dot@probes represented a concentration-dependent quenching of fluorescent signal of bio-dot by Eclipse after probe conjugation. The results demonstrated that TaqMan PCR was not feasible using the designed bio-dot@probes. Our results indicated that bio-dot can be used as an efficient fluorescent tag in the design of fluorescently labeled oligonucleotides with high biocompatibility and optical features. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mahsa Imani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasrin Mohajeri
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mojgan Rastegar
- Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
39
|
Origin of critical nature and stability enhancement in collagen matrix based biomaterials: Comprehensive modification technologies. Int J Biol Macromol 2022; 216:741-756. [PMID: 35908679 DOI: 10.1016/j.ijbiomac.2022.07.199] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/17/2022] [Accepted: 07/24/2022] [Indexed: 02/08/2023]
Abstract
Collagen is the most abundant protein in animals and one of the most important extracellular matrices that chronically plays an important role in biomaterials. However, the major concern about native collagen is the lack of its thermal stability and weak resistance to proteolytic degradation. Currently, a series of modification technologies have been explored for critical nature and stability enhancement in collagen matrix-based biomaterials, and prosperously large-scale progress has been achieved. The establishment of covalent bonds among collagen noumenon has been verified assuringly to have pregnant influences on its physicochemical properties and biological properties, enlightening to discuss the disparate modification technologies on specific effects on the multihierarchical structures and pivotal performances of collagen. In this review, various existing modification methods were classified from a new perspective, scilicet whether to introduce exogenous substances, to reveal the basic scientific theories of collagen modification. Understanding the role of modification technologies in the enhancement of collagen performance is crucial for developing novel collagen-based biomaterials. Moreover, the different modification effects caused by the interaction sites between the modifier and collagen, and the structure-activity relationship between the structure of the modifier and the properties of collagen were reviewed.
Collapse
|
40
|
Schirmer U, Ludolph J, Rothe H, Hauptmann N, Behrens C, Bittrich E, Schliephake H, Liefeith K. Tailored Polyelectrolyte Multilayer Systems by Variation of Polyelectrolyte Composition and EDC/NHS Cross-Linking: Physicochemical Characterization and In Vitro Evaluation. NANOMATERIALS 2022; 12:nano12122054. [PMID: 35745395 PMCID: PMC9228333 DOI: 10.3390/nano12122054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/09/2022] [Accepted: 06/12/2022] [Indexed: 12/17/2022]
Abstract
The layer-by-layer (LbL) self-assembly technique is an effective method to immobilize components of the extracellular matrix (ECM) such as collagen and heparin onto, e.g., implant surfaces/medical devices with the aim of forming polyelectrolyte multilayers (PEMs). Increasing evidence even suggests that cross-linking influences the physicochemical character of PEM films since mechanical cues inherent to the substrate may be as important as its chemical nature to influence the cellular behavior. In this study, for the first-time different collagen/heparin films have been prepared and cross-linked with EDC/NHS chemistry. Quartz crystal microbalance, zeta potential analyzer, diffuse reflectance Fourier transform infrared spectroscopy, atomic force microscopy and ellipsometry were used to characterize film growth, stiffness, and topography of different film systems. The analysis of all data proves a nearly linear film growth for all PEM systems, the efficacy of cross-linking and the corresponding changes in the film rigidity after cross-linking and an appropriate surface topography. Furthermore, preliminary cell culture experiments illustrated those cellular processes correlate roughly with the quantity of newly created covalent amide bonds. This allows a precise adjustment of the physicochemical properties of the selected film architecture regarding the desired application and target cells. It could be shown that collagen improves the biocompatibility of heparin containing PEMs and due to their ECM-analogue nature both molecules are ideal candidates intended to be used for any biomedical application with a certain preference to improve the performance of bone implants or bone augmentation strategies.
Collapse
Affiliation(s)
- Uwe Schirmer
- Institute for Bioprocessing and Analytical Measurement Techniques, 37308 Heiligenstadt, Germany; (U.S.); (J.L.); (H.R.); (N.H.)
| | - Johanna Ludolph
- Institute for Bioprocessing and Analytical Measurement Techniques, 37308 Heiligenstadt, Germany; (U.S.); (J.L.); (H.R.); (N.H.)
| | - Holger Rothe
- Institute for Bioprocessing and Analytical Measurement Techniques, 37308 Heiligenstadt, Germany; (U.S.); (J.L.); (H.R.); (N.H.)
| | - Nicole Hauptmann
- Institute for Bioprocessing and Analytical Measurement Techniques, 37308 Heiligenstadt, Germany; (U.S.); (J.L.); (H.R.); (N.H.)
| | - Christina Behrens
- Department of Oral and Maxillofacial Surgery, George-Augusta-University, 37075 Goettingen, Germany; (C.B.); (H.S.)
| | - Eva Bittrich
- Center Macromolecular Structure Analysis, Leibniz Institute of Polymer Research, 01005 Dresden, Germany;
| | - Henning Schliephake
- Department of Oral and Maxillofacial Surgery, George-Augusta-University, 37075 Goettingen, Germany; (C.B.); (H.S.)
| | - Klaus Liefeith
- Institute for Bioprocessing and Analytical Measurement Techniques, 37308 Heiligenstadt, Germany; (U.S.); (J.L.); (H.R.); (N.H.)
- Correspondence: ; Tel.:+49-3606-671500
| |
Collapse
|
41
|
Malcor JD, Mallein-Gerin F. Biomaterial functionalization with triple-helical peptides for tissue engineering. Acta Biomater 2022; 148:1-21. [PMID: 35675889 DOI: 10.1016/j.actbio.2022.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/09/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
In the growing field of tissue engineering, providing cells in biomaterials with the adequate biological cues represents an increasingly important challenge. Yet, biomaterials with excellent mechanical properties often are often biologically inert to many cell types. To address this issue, researchers resort to functionalization, i.e. the surface modification of a biomaterial with active molecules or substances. Functionalization notably aims to replicate the native cellular microenvironment provided by the extracellular matrix, and in particular by collagen, its major component. As our understanding of biological processes regulating cell behaviour increases, functionalization with biomolecules binding cell surface receptors constitutes a promising strategy. Amongst these, triple-helical peptides (THPs) that reproduce the architectural and biological properties of collagen are especially attractive. Indeed, THPs containing binding sites from the native collagen sequence have successfully been used to guide cell response by establishing cell-biomaterial interactions. Notably, the GFOGER motif recognising the collagen-binding integrins is extensively employed as a cell adhesive peptide. In biomaterials, THPs efficiently improved cell adhesion, differentiation and function on biomaterials designed for tissue repair (especially for bone, cartilage, tendon and heart), vascular graft fabrication, wound dressing, drug delivery or immunomodulation. This review describes the key characteristics of THPs, their effect on cells when combined to biomaterials and their strong potential as biomimetic tools for regenerative medicine. STATEMENT OF SIGNIFICANCE: This review article describes how triple-helical peptides constitute efficient tools to improve cell-biomaterial interactions in tissue engineering. Triple helical peptides are bioactive molecules that mimic the architectural and biological properties of collagen. They have been successfully used to specifically recognize cell-surface receptors and provide cells seeded on biomaterials with controlled biological cues. Functionalization with triple-helical peptides has enabled researchers to improve cell function for regenerative medicine applications, such as tissue repair. However, despite encouraging results, this approach remains limited and under-exploited, and most functionalization strategies reported in the literature rely on biomolecules that are unable to address collagen-binding receptors. This review will assist researchers in selecting the correct tools to functionalize biomaterials in efforts to guide cellular response.
Collapse
Affiliation(s)
- Jean-Daniel Malcor
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France.
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering, CNRS UMR 5305, University Claude Bernard-Lyon 1 and University of Lyon, 7 Passage du Vercors, Cedex 07, Lyon 69367, France
| |
Collapse
|
42
|
Paradiso F, Lenna S, Gazze SA, Garcia Parra J, Murphy K, Margarit L, Gonzalez D, Francis L, Taraballi F. Mechanomimetic 3D Scaffolds as a Humanized In Vitro Model for Ovarian Cancer. Cells 2022; 11:824. [PMID: 35269446 PMCID: PMC8909508 DOI: 10.3390/cells11050824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
The mechanical homeostasis of tissues can be altered in response to trauma or disease, such as cancer, resulting in altered mechanotransduction pathways that have been shown to impact tumor development, progression, and the efficacy of therapeutic approaches. Specifically, ovarian cancer progression is parallel to an increase in tissue stiffness and fibrosis. With in vivo models proving difficult to study, tying tissue mechanics to altered cellular and molecular properties necessitate advanced, tunable, in vitro 3D models able to mimic normal and tumor mechanic features. First, we characterized normal human ovary and high-grade serous (HGSC) ovarian cancer tissue stiffness to precisely mimic their mechanical features on collagen I-based sponge scaffolds, soft (NS) and stiff (MS), respectively. We utilized three ovarian cancer cell lines (OVCAR-3, Caov-3, and SKOV3) to evaluate changes in viability, morphology, proliferation, and sensitivity to doxorubicin and liposomal doxorubicin treatment in response to a mechanically different microenvironment. High substrate stiffness promoted the proliferation of Caov-3 and SKOV3 cells without changing their morphology, and upregulated mechanosensors YAP/TAZ only in SKOV3 cells. After 7 days in culture, both OVCAR3 and SKOV3 decreased the MS scaffold storage modulus (stiffness), suggesting a link between cell proliferation and the softening of the matrix. Finally, high matrix stiffness resulted in higher OVCAR-3 and SKOV3 cell cytotoxicity in response to doxorubicin. This study demonstrates the promise of biomimetic porous scaffolds for effective inclusion of mechanical parameters in 3D cancer modeling. Furthermore, this work establishes the use of porous scaffolds for studying ovarian cancer cells response to mechanical changes in the microenvironment and as a meaningful platform from which to investigate chemoresistance and drug response.
Collapse
Affiliation(s)
- Francesca Paradiso
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Stefania Lenna
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| | - S. Andrea Gazze
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Jezabel Garcia Parra
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Kate Murphy
- Department of Pathology, Singleton Hospital, Swansea Bay University Health Board, Swansea SA2 8QA, UK;
| | - Lavinia Margarit
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Deyarina Gonzalez
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Lewis Francis
- Reproductive Biology and Gynaecological Oncology Group, Swansea University Medical School, Singleton Park, Swansea SA2 8PP, UK; (S.A.G.); (J.G.P.); (L.M.); (D.G.); (L.F.)
| | - Francesca Taraballi
- Center for Musculoskeletal Regeneration, Houston Methodist Academic Institute, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX 77030, USA; (F.P.); (S.L.)
- Orthopedics and Sports Medicine, Houston Methodist Hospital, 6445 Main St., Houston, TX 77030, USA
| |
Collapse
|
43
|
Possible Treatment of Myocardial Infarct Based on Tissue Engineering Using a Cellularized Solid Collagen Scaffold Functionalized with Arg-Glyc-Asp (RGD) Peptide. Int J Mol Sci 2021; 22:ijms222212563. [PMID: 34830447 PMCID: PMC8620820 DOI: 10.3390/ijms222212563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/23/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, the clinical impact of cell therapy after a myocardial infarction (MI) is limited by low cell engraftment due to low cell retention, cell death in inflammatory and poor angiogenic infarcted areas, secondary migration. Cells interact with their microenvironment through integrin mechanoreceptors that control their survival/apoptosis/differentiation/migration and proliferation. The association of cells with a three-dimensional material may be a way to improve interactions with their integrins, and thus outcomes, especially if preparations are epicardially applied. In this review, we will focus on the rationale for using collagen as a polymer backbone for tissue engineering of a contractile tissue. Contractilities are reported for natural but not synthetic polymers and for naturals only for: collagen/gelatin/decellularized-tissue/fibrin/Matrigel™ and for different material states: hydrogels/gels/solids. To achieve a thick/long-term contractile tissue and for cell transfer, solid porous compliant scaffolds are superior to hydrogels or gels. Classical methods to produce solid scaffolds: electrospinning/freeze-drying/3D-printing/solvent-casting and methods to reinforce and/or maintain scaffold properties by reticulations are reported. We also highlight the possibility of improving integrin interaction between cells and their associated collagen by its functionalizing with the RGD-peptide. Using a contractile patch that can be applied epicardially may be a way of improving ventricular remodeling and limiting secondary cell migration.
Collapse
|
44
|
Functionalization of Electrospun Polycaprolactone Scaffolds with Matrix-Binding Osteocyte-Derived Extracellular Vesicles Promotes Osteoblastic Differentiation and Mineralization. Ann Biomed Eng 2021; 49:3621-3635. [PMID: 34664147 PMCID: PMC8671272 DOI: 10.1007/s10439-021-02872-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Synthetic polymeric materials have demonstrated great promise for bone tissue engineering based on their compatibility with a wide array of scaffold-manufacturing techniques, but are limited in terms of the bioactivity when compared to naturally occurring materials. To enhance the regenerative properties of these materials, they are commonly functionalised with bioactive factors to guide growth within the developing tissue. Extracellular matrix vesicles (EVs) play an important role in facilitating endochondral ossification during long bone development and have recently emerged as important mediators of cell-cell communication coordinating bone regeneration, and thus represent an ideal target to enhance the regenerative properties of synthetic scaffolds. Therefore, in this paper we developed tools and protocols to enable the attachment of MLO-Y4 osteocyte-derived EVs onto electrospun polycaprolactone (PCL) scaffolds for bone repair. Initially, we optimize a method for the functionalization of PCL materials with collagen type-1 and fibronectin, inspired by the behaviour of matrix vesicles during endochondral ossification, and demonstrate that this is an effective method for the adhesion of EVs to the material surface. We then used this functionalization process to attach osteogenic EVs, collected from mechanically stimulated MLO-Y4 osteocytes, to collagen-coated electrospun PCL scaffolds. The EV-functionalized scaffold promoted osteogenic differentiation (measured by increased ALP activity) and mineralization of the matrix. In particular, EV-functionalised scaffolds exhibited significant increases in matrix mineralization particularly at earlier time points compared to uncoated and collagen-coated controls. This approach to matrix-based adhesion of EVs provides a mechanism for incorporating vesicle signalling into polyester scaffolds and demonstrates the potential of osteocyte derived EVs to enhance the rate of bone tissue regeneration.
Collapse
|
45
|
Kornmuller A, Flynn LE. Development and characterization of matrix-derived microcarriers from decellularized tissues using electrospraying techniques. J Biomed Mater Res A 2021; 110:559-575. [PMID: 34581474 DOI: 10.1002/jbm.a.37306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/27/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022]
Abstract
Stirred bioreactor systems integrating microcarriers represent a promising approach for therapeutic cell manufacturing. While a variety of microcarriers are commercially available, current options do not integrate the tissue-specific composition of the extracellular matrix (ECM), which can play critical roles in directing cell function. The current study sought to generate microcarriers comprised exclusively of ECM from multiple tissue sources. More specifically, porcine decellularized dermis, porcine decellularized myocardium, and human decellularized adipose tissue were digested with α-amylase to obtain ECM suspensions that could be electrosprayed into liquid nitrogen to generate 3D microcarriers that were stable over a range of ECM concentrations without the need for chemical crosslinking or other additives. Characterization studies confirmed that all three microcarrier types had similar soft and compliant mechanical properties and were of a similar size range, but that their composition varied depending on the native tissue source. In vivo testing in immunocompetent mice revealed that the microcarriers integrated into the host tissues, supporting the infiltration of host cells including macrophages and endothelial cells at 2 weeks post-implantation. In vitro cell culture studies validated that the novel microcarriers supported the attachment of tissue-specific stromal cell populations under dynamic culture conditions within spinner flasks, with a significant increase in live cell numbers observed over 1 week on the dermal- and adipose-derived microcarriers. Overall, the findings demonstrate the versatility of the electrospraying methods and support the further development of the microcarriers as cell culture and delivery platforms.
Collapse
Affiliation(s)
- Anna Kornmuller
- School of Biomedical Engineering, Amit Chakma Engineering Building, The University of Western Ontario, London, Ontario, Canada
| | - Lauren E Flynn
- School of Biomedical Engineering, Amit Chakma Engineering Building, The University of Western Ontario, London, Ontario, Canada.,Department of Chemical & Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
46
|
Zhuravleva IY, Karpova EV, Dokuchaeva AA, Kuznetsova EV, Vladimirov SV, Ksenofontov AL, Nichay NR. Bovine jugular vein conduit: What affects its elastomechanical properties and thermostability? J Biomed Mater Res A 2021; 110:394-408. [PMID: 34390309 DOI: 10.1002/jbm.a.37296] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
The aim of this study was to compare the mechanical properties and thermal stability of the venous wall depending on the treatment method used, and, accordingly, on those structural changes in the tissue that this treatment causes. Bovine jugular vein walls (BJVWs) cross-linked with glutaraldehyde (GA), ethylene glycol diglycidyl ether (DE), and Contegra commercial conduit were evaluated using uniaxial stretching [with and without pre-conditioning (PreC)], differential scanning calorimetry, amino acid analysis, and attenuated total reflection infrared spectroscopy. Fresh BJVW was used as a control. It was shown that failure stress in non-PreC GA-treated and DE-treated materials was lower than that in fresh and Contegra counterparts. Contegra samples were the stiffest among the tested materials. Cyclic preloading leads to distortion of the mechanical behavior of this material, which is heterogeneous in composition and structure. The denaturation temperatures (Td ) of all cross-linked BJVWs were higher than the Td of the fresh vein. The microstructures of the tested BJVWs did not exhibit any differences, but the cross-linking density and hydration of the DE-vein were the highest. GA-cross-linking or DE-cross-linking and isopropanol exposure (Contegra) changed the protein secondary structures of the tested materials in different ways. We hypothesized that the protein secondary structure and hydration degree are the main causes of differences in the mechanical properties and thermal stability of BJVW.
Collapse
Affiliation(s)
- Irina Yu Zhuravleva
- E. Meshalkin National Medical Research Center of the RF Ministry of Health, Novosibirsk, Russian Federation
| | - Elena V Karpova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry SB RAS, Novosibirsk, Russian Federation
| | - Anna A Dokuchaeva
- E. Meshalkin National Medical Research Center of the RF Ministry of Health, Novosibirsk, Russian Federation
| | - Elena V Kuznetsova
- E. Meshalkin National Medical Research Center of the RF Ministry of Health, Novosibirsk, Russian Federation
| | - Sergei V Vladimirov
- E. Meshalkin National Medical Research Center of the RF Ministry of Health, Novosibirsk, Russian Federation
| | - Alexander L Ksenofontov
- A. Belozersky Research Institute of Physico-Chemical Biology MSU, Moscow, Russian Federation
| | - Natalia R Nichay
- E. Meshalkin National Medical Research Center of the RF Ministry of Health, Novosibirsk, Russian Federation
| |
Collapse
|
47
|
Schussler O, Chachques JC, Alifano M, Lecarpentier Y. Key Roles of RGD-Recognizing Integrins During Cardiac Development, on Cardiac Cells, and After Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:179-203. [PMID: 34342855 DOI: 10.1007/s12265-021-10154-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Cardiac cells interact with the extracellular matrix (ECM) proteins through integrin mechanoreceptors that control many cellular events such as cell survival, apoptosis, differentiation, migration, and proliferation. Integrins play a crucial role in cardiac development as well as in cardiac fibrosis and hypertrophy. Integrins recognize oligopeptides present on ECM proteins and are involved in three main types of interaction, namely with collagen, laminin, and the oligopeptide RGD (Arg-Gly-Asp) present on vitronectin and fibronectin proteins. To date, the specific role of integrins recognizing the RGD has not been addressed. In this review, we examine their role during cardiac development, their role on cardiac cells, and their upregulation during pathological processes such as heart fibrosis and hypertrophy. We also examine their role in regenerative and angiogenic processes after myocardial infarction (MI) in the peri-infarct area. Specific targeting of these integrins may be a way of controlling some of these pathological events and thereby improving medical outcomes.
Collapse
Affiliation(s)
- Olivier Schussler
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.
| | - Juan C Chachques
- Department of Cardiac Surgery Pompidou Hospital, Laboratory of Biosurgical Research, Carpentier Foundation, University Paris Descartes, 75015, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, APHP Centre, University of Paris, Paris, France.,INSERM U1138 Team "Cancer, Immune Control, and Escape", Cordeliers Research Center, University of Paris, Paris, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien, Meaux, France
| |
Collapse
|
48
|
Graceffa V. Physical and mechanical cues affecting biomaterial-mediated plasmid DNA delivery: insights into non-viral delivery systems. J Genet Eng Biotechnol 2021; 19:90. [PMID: 34142237 PMCID: PMC8211807 DOI: 10.1186/s43141-021-00194-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Whilst traditional strategies to increase transfection efficiency of non-viral systems aimed at modifying the vector or the polyplexes/lipoplexes, biomaterial-mediated gene delivery has recently sparked increased interest. This review aims at discussing biomaterial properties and unravelling underlying mechanisms of action, for biomaterial-mediated gene delivery. DNA internalisation and cytoplasmic transport are initially discussed. DNA immobilisation, encapsulation and surface-mediated gene delivery (SMD), the role of extracellular matrix (ECM) and topographical cues, biomaterial stiffness and mechanical stimulation are finally outlined. MAIN TEXT Endocytic pathways and mechanisms to escape the lysosomal network are highly variable. They depend on cell and DNA complex types but can be diverted using appropriate biomaterials. 3D scaffolds are generally fabricated via DNA immobilisation or encapsulation. Degradation rate and interaction with the vector affect temporal patterns of DNA release and transgene expression. In SMD, DNA is instead coated on 2D surfaces. SMD allows the incorporation of topographical cues, which, by inducing cytoskeletal re-arrangements, modulate DNA endocytosis. Incorporation of ECM mimetics allows cell type-specific transfection, whereas in spite of discordances in terms of optimal loading regimens, it is recognised that mechanical loading facilitates gene transfection. Finally, stiffer 2D substrates enhance DNA internalisation, whereas in 3D scaffolds, the role of stiffness is still dubious. CONCLUSION Although it is recognised that biomaterials allow the creation of tailored non-viral gene delivery systems, there still are many outstanding questions. A better characterisation of endocytic pathways would allow the diversion of cell adhesion processes and cytoskeletal dynamics, in order to increase cellular transfection. Further research on optimal biomaterial mechanical properties, cell ligand density and loading regimens is limited by the fact that such parameters influence a plethora of other different processes (e.g. cellular adhesion, spreading, migration, infiltration, and proliferation, DNA diffusion and release) which may in turn modulate gene delivery. Only a better understanding of these processes may allow the creation of novel robust engineered systems, potentially opening up a whole new area of biomaterial-guided gene delivery for non-viral systems.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
- Department of Life Sciences, Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
| |
Collapse
|
49
|
Wu Z, Korntner SH, Mullen AM, Skoufos I, Tzora A, Zeugolis DI. In the quest of the optimal tissue source (porcine male and female articular, tracheal and auricular cartilage) for the development of collagen sponges for articular cartilage. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
50
|
Arifin MA, Mel M, Swan SY, Samsudin N, Hashim YZHY, Salleh HM. Optimization of ultraviolet/ozone (UVO 3) process conditions for the preparation of gelatin coated polystyrene (PS) microcarriers. Prep Biochem Biotechnol 2021; 52:181-196. [PMID: 34010098 DOI: 10.1080/10826068.2021.1923031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The aim of this study was to develop gelatin coated polystyrene (PS) microcarriers with good cell adhesion and proliferation properties. PS microspheres, prepared using oil-in water (o/w) solvent evaporation method, were loaded with oxygen containing functional groups using an ultraviolet/ozone (UVO3) system. Using water-soluble carbodiimide chemistry, gelatin was subsequently immobilized on UVO3 treated PS microspheres. The amount of immobilized gelatin was found to be directly proportional to the surface carboxyl (COOH) concentration on PS microspheres. Face Centered Central Composite Design (FCCD) was employed to optimize the process conditions of UVO3 treatment to maximize the surface COOH concentration on PS microspheres for allowing higher gelatin immobilization. Statistical results revealed that, the optimized process conditions were ozone flow rate of ∼64,603 ppm, exposure time of ∼60 minutes and sample amount of 5.05 g. Under these conditions, the surface COOH concentration on PS microspheres was ∼1,505 nmol/g with the corresponding amount of immobilized gelatin was ∼2,725 µg/g. Characterization analyses strongly suggest that the optimized UVO3 treatment and successive gelatin immobilization have successfully improved surface wettability and dispersion stability of PS microspheres. Moreover, gelatin coated PS microcarriers were also proven as able to support the growth of CHO-K1 cells in high cell density culture.
Collapse
Affiliation(s)
- Mohd Azmir Arifin
- Faculty of Chemical and Process Engineering Technology, Lebuhraya Tun Razak, Universiti Malaysia Pahang, Kuantan, Malaysia
| | - Maizirwan Mel
- Department of Biotechnology Engineering, International Islamic University Malaysia Kulliyyah of Engineering, Kuala Lumpur, Malaysia
| | - Sia Yiik Swan
- Faculty of Chemical and Process Engineering Technology, Lebuhraya Tun Razak, Universiti Malaysia Pahang, Kuantan, Malaysia
| | - Nurhusna Samsudin
- International Institute for Halal Research and Training, International Islamic University Malaysia, Kuala Lumpur, Malaysia
| | - Yumi Zuhanis Has-Yun Hashim
- International Institute for Halal Research and Training, International Islamic University Malaysia, Kuala Lumpur, Malaysia
| | - Hamzah Mohd Salleh
- International Institute for Halal Research and Training, International Islamic University Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|