1
|
González-Lara H, Parra-Pacheco B, Rico-García E, Aguirre-Becerra H, Feregrino-Pérez AA, García-Trejo JF. Black Soldier Fly Culture as a Source of Chitin and Chitosan for Its Potential Use in Concrete: An Overview. Polymers (Basel) 2025; 17:717. [PMID: 40292573 PMCID: PMC11945844 DOI: 10.3390/polym17060717] [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: 02/08/2025] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
Chitin is one of the most abundant biopolymers in nature and is found mainly in the exoskeletons of crustaceans and insects, in the cell walls of fungi, and in some species of mollusks. Chitosan is a derivative of chitin; it is much more accessible and has a broader range of applications, including improving the quality of materials such as films, plastics, and concrete. The rheological properties of chitin and chitosan refer to their behavior against deformation and flow and their ability to resist structural changes under mechanical stress conditions. These properties are fundamental in applications where the aim is to control the texture, viscosity, and handling of these biopolymers. Three types of methods for the extraction of chitin and chitosan can be classified: the first is the chemical method, which presents high yields but uses reagents that generate toxic residues; the second is the biological method, which takes advantage of chemical reactions of microorganisms but in some cases has low yields compared to chemical extraction; and the third is the enzymatic method, which uses reagents with a low production of toxic residues. However, low extraction yields are also reported. One of the primary sources of chitin and chitosan is the residue of shellfish and crustaceans. However, a new source of obtaining these compounds is the black soldier fly, which has the same yields of biopolymers as shellfish. In addition, this is a residue of the black soldier fly larvae culture, where protein, oil, and biofertilizers are generated by the bioconversion of organic waste. This work proposes the black soldier fly as an alternative source for extracting chitin and chitosan, using organic methodologies that do not generate toxic residues and have high yields. Including these biopolymers in concrete elaboration could have positive results in terms of flexibility, compressive strength, and workability.
Collapse
Affiliation(s)
- Hugo González-Lara
- División de Investigación y Posgrado, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Carretera a Chichimequillas Km. 1 s/n, Amazcala, El Marqués 76265, Querétaro, Mexico; (H.G.-L.); (B.P.-P.)
| | - Benito Parra-Pacheco
- División de Investigación y Posgrado, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Carretera a Chichimequillas Km. 1 s/n, Amazcala, El Marqués 76265, Querétaro, Mexico; (H.G.-L.); (B.P.-P.)
| | - Enrique Rico-García
- Cuerpo Académico de Bioingeniería Básica y Aplicada, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Las Campanas, Santiago de Querétaro 76010, Querétaro, Mexico; (E.R.-G.); (H.A.-B.); (A.A.F.-P.)
| | - Humberto Aguirre-Becerra
- Cuerpo Académico de Bioingeniería Básica y Aplicada, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Las Campanas, Santiago de Querétaro 76010, Querétaro, Mexico; (E.R.-G.); (H.A.-B.); (A.A.F.-P.)
| | - Ana Angélica Feregrino-Pérez
- Cuerpo Académico de Bioingeniería Básica y Aplicada, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Las Campanas, Santiago de Querétaro 76010, Querétaro, Mexico; (E.R.-G.); (H.A.-B.); (A.A.F.-P.)
| | - Juan Fernando García-Trejo
- Cuerpo Académico de Bioingeniería Básica y Aplicada, Facultad de Ingeniería, Universidad Autónoma de Querétaro, Cerro de las Campanas s/n, Las Campanas, Santiago de Querétaro 76010, Querétaro, Mexico; (E.R.-G.); (H.A.-B.); (A.A.F.-P.)
| |
Collapse
|
2
|
Pisani S, Evangelista A, Chesi L, Croce S, Avanzini MA, Dorati R, Genta I, Benazzo M, Comoli P, Conti B. Nanofibrous Scaffolds' Ability to Induce Mesenchymal Stem Cell Differentiation for Soft Tissue Regenerative Applications. Pharmaceuticals (Basel) 2025; 18:239. [PMID: 40006052 PMCID: PMC11859969 DOI: 10.3390/ph18020239] [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: 01/06/2025] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Mesenchymal stem cells (MSCs) have gained recognition as a highly versatile and promising cell source for repopulating bioengineered scaffolds due to their inherent capacity to differentiate into multiple cell types. However, MSC implantation techniques have often yielded inconsistent clinical results, underscoring the need for advanced approaches to enhance their therapeutic efficacy. Recent developments in three-dimensional (3D) bioengineered scaffolds have provided a significant breakthrough by closely mimicking the in vivo environment, addressing the limitations of traditional two-dimensional (2D) cell cultures. Among these, nanofibrous scaffolds have proven particularly effective, offering an optimal 3D framework, growth-permissive substrates, and the delivery of trophic factors crucial for MSC survival and regeneration. Furthermore, the selection of appropriate biomaterials can amplify the paracrine effects of MSCs, promoting both proliferation and targeted differentiation. The synergistic combination of MSCs with nanofibrous scaffolds has demonstrated remarkable potential in achieving repair, regeneration, and tissue-specific differentiation with enhanced safety and efficacy, paving the way for routine clinical applications. In this review, we examine the most recent studies (2013-2023) that explore the combined use of MSCs and nanofibrous scaffolds for differentiation into cardiogenic, epithelial, myogenic, tendon, and vascular cell lineages. Using PubMed, we identified and analyzed 275 relevant articles based on the search terms "Nanofibers", "Electrospinning", "Mesenchymal stem cells", and "Differentiation". This review highlights the critical advancements in the use of nanofibrous scaffolds as a platform for MSC differentiation and tissue regeneration. By summarizing key findings from the last decade, it provides valuable insights for researchers and clinicians aiming to optimize scaffold design, MSC integration, and translational applications. These insights could significantly influence future research directions and the development of more effective regenerative therapies.
Collapse
Affiliation(s)
- Silvia Pisani
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Aleksandra Evangelista
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Luca Chesi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Stefania Croce
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Maria Antonietta Avanzini
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| | - Marco Benazzo
- Otorhinolaryngology Unit, Department of Surgical Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Patrizia Comoli
- Department of Clinical, Surgical, Diagnostic & Pediatric Sciences, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (S.C.); (M.A.A.); (P.C.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.D.); (I.G.); (B.C.)
| |
Collapse
|
3
|
Tamo AK, Djouonkep LDW, Selabi NBS. 3D Printing of Polysaccharide-Based Hydrogel Scaffolds for Tissue Engineering Applications: A Review. Int J Biol Macromol 2024; 270:132123. [PMID: 38761909 DOI: 10.1016/j.ijbiomac.2024.132123] [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: 12/05/2023] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/20/2024]
Abstract
In tissue engineering, 3D printing represents a versatile technology employing inks to construct three-dimensional living structures, mimicking natural biological systems. This technology efficiently translates digital blueprints into highly reproducible 3D objects. Recent advances have expanded 3D printing applications, allowing for the fabrication of diverse anatomical components, including engineered functional tissues and organs. The development of printable inks, which incorporate macromolecules, enzymes, cells, and growth factors, is advancing with the aim of restoring damaged tissues and organs. Polysaccharides, recognized for their intrinsic resemblance to components of the extracellular matrix have garnered significant attention in the field of tissue engineering. This review explores diverse 3D printing techniques, outlining distinctive features that should characterize scaffolds used as ideal matrices in tissue engineering. A detailed investigation into the properties and roles of polysaccharides in tissue engineering is highlighted. The review also culminates in a profound exploration of 3D polysaccharide-based hydrogel applications, focusing on recent breakthroughs in regenerating different tissues such as skin, bone, cartilage, heart, nerve, vasculature, and skeletal muscle. It further addresses challenges and prospective directions in 3D printing hydrogels based on polysaccharides, paving the way for innovative research to fabricate functional tissues, enhancing patient care, and improving quality of life.
Collapse
Affiliation(s)
- Arnaud Kamdem Tamo
- Institute of Microsystems Engineering IMTEK, University of Freiburg, 79110 Freiburg, Germany; Freiburg Center for Interactive Materials and Bioinspired Technologies FIT, University of Freiburg, 79110 Freiburg, Germany; Freiburg Materials Research Center FMF, University of Freiburg, 79104 Freiburg, Germany; Ingénierie des Matériaux Polymères (IMP), Université Claude Bernard Lyon 1, INSA de Lyon, Université Jean Monnet, CNRS, UMR 5223, 69622 Villeurbanne CEDEX, France.
| | - Lesly Dasilva Wandji Djouonkep
- College of Petroleum Engineering, Yangtze University, Wuhan 430100, China; Key Laboratory of Drilling and Production Engineering for Oil and Gas, Wuhan 430100, China
| | - Naomie Beolle Songwe Selabi
- Institute of Advanced Materials and Nanotechnology, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
4
|
Jiang Z, Yu J, Zhou H, Feng J, Xu Z, Wan M, Zhang W, He Y, Jia C, Shao S, Guo H, Liu B. Research hotspots and emerging trends of mesenchymal stem cells in cardiovascular diseases: a bibliometric-based visual analysis. Front Cardiovasc Med 2024; 11:1394453. [PMID: 38873270 PMCID: PMC11169657 DOI: 10.3389/fcvm.2024.1394453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Background Mesenchymal stem cells (MSCs) have important research value and broad application prospects in cardiovascular diseases (CVDs). However, few bibliometric analyses on MSCs in cardiovascular diseases are available. This study aims to provide a thorough review of the cooperation and influence of countries, institutions, authors, and journals in the field of MSCs in cardiovascular diseases, with the provision of discoveries in the latest progress, evolution paths, frontier research hotspots, and future research trends in the regarding field. Methods The articles related to MSCs in cardiovascular diseases were retrieved from the Web of Science. The bibliometric study was performed by CiteSpace and VOSviewer, and the knowledge map was generated based on data obtained from retrieved articles. Results In our study, a total of 4,852 publications launched before August 31, 2023 were accessed through the Web of Science Core Collection (WoSCC) database via our searching strategy. Significant fluctuations in global publications were observed in the field of MSCs in CVDs. China emerged as the nation with the largest number of publications, yet a shortage of high-quality articles was noted. The interplay among countries, institutions, journals and authors is visually represented in the enclosed figures. Importantly, current research trends and hotspots are elucidated. Cluster analysis on references has highlighted the considerable interest in exosomes, extracellular vesicles, and microvesicles. Besides, keywords analysis revealed a strong emphasis on myocardial infarction, therapy, and transplantation. Treatment methods-related keywords were prominent, while keywords associated with extracellular vesicles gathered significant attention from the long-term perspective. Conclusion MSCs in CVDs have become a topic of active research interest, showcasing its latent value and potential. By summarizing the latest progress, identifying the research hotspots, and discussing the future trends in the advancement of MSCs in CVDs, we aim to offer valuable insights for considering research prospects.
Collapse
Affiliation(s)
- Zhihang Jiang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajing Yu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Houle Zhou
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiaming Feng
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zehui Xu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Melisandre Wan
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weiwei Zhang
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuqing He
- Department of Preventive Medicine, College of Public Health, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyao Jia
- Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Shuijin Shao
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haidong Guo
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baonian Liu
- Department of Anatomy, School of Chinese Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Tekinay SH, Tekinay AB. Stem Cells and Nanofibers for Skin Regeneration and Wound Healing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1470:19-30. [PMID: 38904750 DOI: 10.1007/5584_2024_814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Stem cells have been attractive targets for tissue regeneration due to their inherent ability to differentiate into various specialized cell types; however, efforts for stem cell transplantation for the treatment of degenerated tissues have been hampered by the propensity of some stem cell types to form teratomas and the lessened viability of stem cells after transplantation. These disadvantages can be prevented using tailored extracellular matrix-like materials that can be used as an aid for the transplantation of stem cells. Nanomaterials, in particular nanofibers, have great potential to be used as extracellular matrix-like materials for this purpose. In this article, we will review the use of stem cells and nanofibers for skin regeneration and wound healing.
Collapse
Affiliation(s)
- Sarah H Tekinay
- Yildirim Beyazit University, School of Medicine, Ankara, Türkiye
| | | |
Collapse
|
6
|
Zhang W, Zhang J, Cui Y, Zhao Y, Lei X. Stem Cells and Exosome Applications for Cutaneous Wound Healing: From Ground to Microgravity Environment. Stem Cell Rev Rep 2023; 19:2094-2108. [PMID: 37458936 DOI: 10.1007/s12015-023-10571-9] [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] [Accepted: 05/27/2023] [Indexed: 10/17/2023]
Abstract
The increasing number of astronauts entering microgravity environments for long-term space missions has resulted in serious health problems, including accidental injury and trauma. Skin, as the largest organ and outermost layer of the human body, has the ability to self-renew and withstand a variety of harmful biological and environmental influences. Recent spaceflight experiments and simulated studies have begun to concern the effects of microgravity on the growth of skin cells and the process of cutaneous wound healing. However, the mechanisms of the adverse effects of microgravity on skin cells and potential intervention measures are still limited. Stem cells and their exosomes provide unique opportunities for the cutaneous wound healing as they have been used to improve skin repair. This review discusses the effects of microgravity on wound healing, from cell morphological changes to molecular level alterations. Furthermore, the current research on wound healing treatment utilizing stem cells and their exosomes on the ground is summarized. Finally, this review proposes promising therapeutic strategies using stem cells or exosomes for wound healing in the microgravity environment. Graphical Abstract.
Collapse
Affiliation(s)
- Wenya Zhang
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jinhua Zhang
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China
- The College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, 100044, China
| | - Yan Cui
- Department of General Surgery, Strategic Support Force Medical Center, Chaoyang District, Beijing, 100101, China
| | - Yongxiang Zhao
- Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, National Center for International Research of Bio-targeting Theranostics, Guangxi Medical University, Nanning, 530021, China.
| | - Xiaohua Lei
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Rybachuk O, Savytska N, Pinet É, Yaminsky Y, Medvediev V. Heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow derived stromal cells in vitroand in vivo. Biomed Mater 2023; 18. [PMID: 36542861 DOI: 10.1088/1748-605x/acadc3] [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: 06/27/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Synthetic hydrogels composed of polymer pore frames are commonly used in medicine, from pharmacologically targeted drug delivery to the creation of bioengineering constructions used in implantation surgery. Among various possible materials, the most common are poly-[N(2-hydroxypropyl)methacrylamide] (pHPMA) derivatives. One of the pHPMA derivatives is biocompatible hydrogel, NeuroGel. Upon contact with nervous tissue, the NeuroGel's structure can support the chemical and physiological conditions of the tissue necessary for the growth of native cells. Owing to the different pore diameters in the hydrogel, not only macromolecules, but also cells can migrate. This study evaluated the differentiation of bone marrow stromal cells (BMSCs) into neurons, as well as the effectiveness of using this biofabricated system in spinal cord injuryin vivo. The hydrogel was populated with BMSCs by injection or rehydration. After cultivation, these fragments (hydrogel + BMSCs) were implanted into the injured rat spinal cord. Fragments were immunostained before implantation and seven months after implantation. During cultivation with the hydrogel, both variants (injection/rehydration) of the BMSCs culture retained their viability and demonstrated a significant number of Ki-67-positive cells, indicating the preservation of their proliferative activity. In hydrogel fragments, BMSCs also maintained their viability during the period of cocultivation and were Ki-67-positive, but in significantly fewer numbers than in the cell culture. In addition, in fragments of hydrogel with grafted BMSCs, both by the injection or rehydration versions, we observed a significant number up to 57%-63.5% of NeuN-positive cells. These results suggest that the heterogeneous pHPMA hydrogel promotes neuronal differentiation of bone marrow-derived stromal cells. Furthermore, these data demonstrate the possible use of NeuroGel implants with grafted BMSCs for implantation into damaged areas of the spinal cord, with subsequent nerve fiber germination, nerve cell regeneration, and damaged segment restoration.
Collapse
Affiliation(s)
- Oksana Rybachuk
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,Institute of Genetic and Regenerative Medicine, M. D. Strazhesko National Scientific Center of Cardiology, Clinical and Regenerative Medicine, NAMS of Ukraine, Kyiv, Ukraine
| | - Natalia Savytska
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,German Center for Neurodegenerative Diseases, Tübingen, Germany
| | | | - Yurii Yaminsky
- State Institution 'Romodanov Neurosurgery Institute, NAMS of Ukraine', Kyiv, Ukraine
| | - Volodymyr Medvediev
- Bogomoletz Institute of Physiology NAS of Ukraine, Kyiv, Ukraine.,Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
8
|
Payushina OV, Tsomartova DA, Chereshneva YV, Ivanova MY, Lomanovskaya TA, Pavlova MS, Kuznetsov SL. Experimental Transplantation of Mesenchymal Stromal Cells as an Approach to Studying Their Differentiation In Vivo (Review). BIOL BULL+ 2022. [DOI: 10.1134/s1062359022060127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
9
|
Nanofiber Carriers of Therapeutic Load: Current Trends. Int J Mol Sci 2022; 23:ijms23158581. [PMID: 35955712 PMCID: PMC9368923 DOI: 10.3390/ijms23158581] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/10/2022] Open
Abstract
The fast advancement in nanotechnology has prompted the improvement of numerous methods for the creation of various nanoscale composites of which nanofibers have gotten extensive consideration. Nanofibers are polymeric/composite fibers which have a nanoscale diameter. They vary in porous structure and have an extensive area. Material choice is of crucial importance for the assembly of nanofibers and their function as efficient drug and biomedicine carriers. A broad scope of active pharmaceutical ingredients can be incorporated within the nanofibers or bound to their surface. The ability to deliver small molecular drugs such as antibiotics or anticancer medications, proteins, peptides, cells, DNA and RNAs has led to the biomedical application in disease therapy and tissue engineering. Although nanofibers have shown incredible potential for drug and biomedicine applications, there are still difficulties which should be resolved before they can be utilized in clinical practice. This review intends to give an outline of the recent advances in nanofibers, contemplating the preparation methods, the therapeutic loading and release and the various therapeutic applications.
Collapse
|
10
|
Bai L, Liu L, Esquivel M, Tardy BL, Huan S, Niu X, Liu S, Yang G, Fan Y, Rojas OJ. Nanochitin: Chemistry, Structure, Assembly, and Applications. Chem Rev 2022; 122:11604-11674. [PMID: 35653785 PMCID: PMC9284562 DOI: 10.1021/acs.chemrev.2c00125] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chitin, a fascinating biopolymer found in living organisms, fulfills current demands of availability, sustainability, biocompatibility, biodegradability, functionality, and renewability. A feature of chitin is its ability to structure into hierarchical assemblies, spanning the nano- and macroscales, imparting toughness and resistance (chemical, biological, among others) to multicomponent materials as well as adding adaptability, tunability, and versatility. Retaining the inherent structural characteristics of chitin and its colloidal features in dispersed media has been central to its use, considering it as a building block for the construction of emerging materials. Top-down chitin designs have been reported and differentiate from the traditional molecular-level, bottom-up synthesis and assembly for material development. Such topics are the focus of this Review, which also covers the origins and biological characteristics of chitin and their influence on the morphological and physical-chemical properties. We discuss recent achievements in the isolation, deconstruction, and fractionation of chitin nanostructures of varying axial aspects (nanofibrils and nanorods) along with methods for their modification and assembly into functional materials. We highlight the role of nanochitin in its native architecture and as a component of materials subjected to multiscale interactions, leading to highly dynamic and functional structures. We introduce the most recent advances in the applications of nanochitin-derived materials and industrialization efforts, following green manufacturing principles. Finally, we offer a critical perspective about the adoption of nanochitin in the context of advanced, sustainable materials.
Collapse
Affiliation(s)
- Long Bai
- Key
Laboratory of Bio-based Material Science & Technology (Ministry
of Education), Northeast Forestry University, Harbin 150040, P.R. China
- Bioproducts
Institute, Department of Chemical & Biological Engineering, Department
of Chemistry, and Department of Wood Science, 2360 East Mall, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Liang Liu
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Key Lab of Biomass-Based Green Fuel and Chemicals,
College of Chemical Engineering, Nanjing
Forestry University, 159 Longpan Road, Nanjing 210037, P.R. China
| | - Marianelly Esquivel
- Polymer
Research Laboratory, Department of Chemistry, National University of Costa Rica, Heredia 3000, Costa Rica
| | - Blaise L. Tardy
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076 Aalto, Finland
- Department
of Chemical Engineering, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Siqi Huan
- Key
Laboratory of Bio-based Material Science & Technology (Ministry
of Education), Northeast Forestry University, Harbin 150040, P.R. China
- Bioproducts
Institute, Department of Chemical & Biological Engineering, Department
of Chemistry, and Department of Wood Science, 2360 East Mall, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Xun Niu
- Bioproducts
Institute, Department of Chemical & Biological Engineering, Department
of Chemistry, and Department of Wood Science, 2360 East Mall, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Shouxin Liu
- Key
Laboratory of Bio-based Material Science & Technology (Ministry
of Education), Northeast Forestry University, Harbin 150040, P.R. China
| | - Guihua Yang
- State
Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of
Sciences, Jinan 250353, China
| | - Yimin Fan
- Jiangsu
Co-Innovation Center of Efficient Processing and Utilization of Forest
Resources, Jiangsu Key Lab of Biomass-Based Green Fuel and Chemicals,
College of Chemical Engineering, Nanjing
Forestry University, 159 Longpan Road, Nanjing 210037, P.R. China
| | - Orlando J. Rojas
- Bioproducts
Institute, Department of Chemical & Biological Engineering, Department
of Chemistry, and Department of Wood Science, 2360 East Mall, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Department
of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076 Aalto, Finland
| |
Collapse
|
11
|
Heras KL, Igartua M, Santos-Vizcaino E, Hernandez RM. Cell-based dressings: A journey through chronic wound management. BIOMATERIALS ADVANCES 2022; 135:212738. [PMID: 35929212 DOI: 10.1016/j.bioadv.2022.212738] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 06/15/2023]
Abstract
The field of regenerative medicine has undergone a paradigm shift in recent decades thanks to the emergence of novel therapies based on the use of living organisms. The development of cell-based strategies has become a trend for the treatment of different conditions and pathologies. In this sense, the need for more adequate, biomimetic and well-planned treatments for chronic wounds has found different and innovative strategies, based on the combination of cells with dressings, which seek to revolutionize the wound healing management. Therefore, the objective of this review is to analyze the current state and the latest advances in the research of cell-based dressings for chronic wounds, ranging from traditional and "second generation" bioengineered living skin equivalents to mesenchymal stem cell dressings; the latter include biopolymeric porous scaffolds, electrospun nanofiber meshes, hydrogels and 3D printed bio-printed dressings. Finally, this review updates the completed and ongoing clinical trials in this field and encourages researchers to rethink these new approaches, manufacturing processes and mechanisms of action, as well as their administration strategies and timings.
Collapse
Affiliation(s)
- Kevin Las Heras
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
12
|
Mahmood A, Patel D, Hickson B, DesRochers J, Hu X. Recent Progress in Biopolymer-Based Hydrogel Materials for Biomedical Applications. Int J Mol Sci 2022; 23:1415. [PMID: 35163339 PMCID: PMC8836285 DOI: 10.3390/ijms23031415] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/22/2022] [Accepted: 01/23/2022] [Indexed: 12/23/2022] Open
Abstract
Hydrogels from biopolymers are readily synthesized, can possess various characteristics for different applications, and have been widely used in biomedicine to help with patient treatments and outcomes. Polysaccharides, polypeptides, and nucleic acids can be produced into hydrogels, each for unique purposes depending on their qualities. Examples of polypeptide hydrogels include collagen, gelatin, and elastin, and polysaccharide hydrogels include alginate, cellulose, and glycosaminoglycan. Many different theories have been formulated to research hydrogels, which include Flory-Rehner theory, Rubber Elasticity Theory, and the calculation of porosity and pore size. All these theories take into consideration enthalpy, entropy, and other thermodynamic variables so that the structure and pore sizes of hydrogels can be formulated. Hydrogels can be fabricated in a straightforward process using a homogeneous mixture of different chemicals, depending on the intended purpose of the gel. Different types of hydrogels exist which include pH-sensitive gels, thermogels, electro-sensitive gels, and light-sensitive gels and each has its unique biomedical applications including structural capabilities, regenerative repair, or drug delivery. Major biopolymer-based hydrogels used for cell delivery include encapsulated skeletal muscle cells, osteochondral muscle cells, and stem cells being delivered to desired locations for tissue regeneration. Some examples of hydrogels used for drug and biomolecule delivery include insulin encapsulated hydrogels and hydrogels that encompass cancer drugs for desired controlled release. This review summarizes these newly developed biopolymer-based hydrogel materials that have been mainly made since 2015 and have shown to work and present more avenues for advanced medical applications.
Collapse
Affiliation(s)
- Ayaz Mahmood
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA;
| | - Dev Patel
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (D.P.); (B.H.); (J.D.)
| | - Brandon Hickson
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (D.P.); (B.H.); (J.D.)
| | - John DesRochers
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (D.P.); (B.H.); (J.D.)
| | - Xiao Hu
- Department of Physics and Astronomy, Rowan University, Glassboro, NJ 08028, USA;
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (D.P.); (B.H.); (J.D.)
- Department of Molecular and Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
13
|
Liu Y, Liu Y, Wu M, Zou R, Mao S, Cong P, Hou M, Jin H, Zhao Y, Bao Y. Adipose-derived mesenchymal stem cell-loaded β-chitin nanofiber hydrogel promote wound healing in rats. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 33:12. [PMID: 35050422 PMCID: PMC8776676 DOI: 10.1007/s10856-021-06630-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 11/28/2021] [Indexed: 05/14/2023]
Abstract
Because of stem cells are limited by the low efficiency of their cell homing and survival in vivo, cell delivery systems and scaffolds have attracted a great deal of attention for stem cells' successful clinical practice. β-chitin nanofibers (β-ChNF) were prepared from squid pens in this study. Fourier transform infrared spectroscopy, X-ray diffraction and scanning electron microscopy proved that β-ChNFs with the diameter of 5 to 10 nm were prepared. β-ChNF dispersion became gelled upon the addition of cell culture medium. Cell culture experiments showed that β-ChNFs exhibited negligible cytotoxicity towards ADSCs and L929 cells, and it was found that more exosomes were secreted by the globular ADSCs grown in the β-ChNF hydrogel. The vivo experiments of rats showed that the ADSCs-loaded β-ChNF hydrogel could directly cover the wound surface and significantly accelerate the wound healing and promote the generation of epithelization, granulation tissue and collagen. In addition, the ADSCs-loaded β-ChNF hydrogel clearly regulated the expressions of VEGFR, α-SMA, collagen I and collagen III. Finally, we showed that ADSCs-loaded β-ChNF hydrogel activated the TGFβ/smad signaling. The neutralization of TGFβ markedly reduced Smad phosphorylation and the expressions of TIMP1, VEGFR and α-SMA. Taken together, these findings suggest that ADSCs-loaded β-ChNF hydrogel promises for treating wounds that are challenge to heal via conventional methods. Graphical abstract.
Collapse
Affiliation(s)
- Ying Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Yunen Liu
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Mi Wu
- Jihua Laboratory, Foshan, 528200, China
| | - Rufei Zou
- Jihua Laboratory, Foshan, 528200, China
| | - Shun Mao
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Peifang Cong
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Mingxiao Hou
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China
| | - Hongxu Jin
- Emergency Medicine Department of General Hospital of Northern Theater Command, Laboratory of Rescue Center of Severe Wound and Trauma PLA, Shenyang, 110016, China.
| | - Yan Zhao
- Jihua Laboratory, Foshan, 528200, China.
| | - Yongli Bao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
14
|
Lv X, Wang L, Zou X, Huang S. Umbilical Cord Mesenchymal Stem Cell Therapy for Regenerative Treatment of Rheumatoid Arthritis: Opportunities and Challenges. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3927-3936. [PMID: 34584402 PMCID: PMC8462093 DOI: 10.2147/dddt.s323107] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/26/2021] [Indexed: 12/25/2022]
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease of unknown etiology with a high rate of disability. Traditional treatments for RA remain a challenging issue. For example, nonsteroidal anti-inflammatory drugs (NSAIDs) have no therapeutic effects on joint destruction, and the prominent side effects include gastrointestinal symptoms. RA is characterized by recurrence and bone attrition. Therefore, regenerative medicine and the use of umbilical cord mesenchymal stem cell (UC-MSC) therapies have recently emerged as potential options. UC-MSCs are multifunctional stem cells that are present in neonatal umbilical cord tissue and can differentiate into many kinds of cells, which have broad clinical application prospects in the tissue engineering of bone, cartilage, muscle, tendon, ligament, nerve, liver, endothelium, and myocardium. Moreover, UC-MSCs have advantages, such as convenient collection of materials and no ethical disputes; thus, these cells have attracted increasing attention from researchers. However, there are few clinical studies regarding UC-MSC therapy for RA. In this paper, we will review traditional drugs for RA treatment and then focus on UC-MSC therapy for RA, including preclinical and clinical UC-MSC applications for RA patients in the context of regenerative medicine. Finally, we will summarize the challenges and perspectives of UC-MSCs as a potential therapeutic strategy for RA. This review will help to design and discover more potent and efficacious treatments for RA patients and aid in advancing this class of cell therapy.
Collapse
Affiliation(s)
- Xiaolan Lv
- Department of Laboratory Medicine, Liuzhou Maternity and Child Healthcare Hospital, Liu Zhou, Guang Xi, People's Republic of China
| | - Liming Wang
- Shaanxi Jiuzhou Biomedical Science and Technology Group, Xi'an, Shaan Xi, People's Republic of China
| | - XiaoRong Zou
- Department of Hematology, 986 Hospital of Fourth Military Medical University, Xi'an, Shaan Xi, People's Republic of China
| | - Shigao Huang
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| |
Collapse
|
15
|
Fan F, Saha S, Hanjaya-Putra D. Biomimetic Hydrogels to Promote Wound Healing. Front Bioeng Biotechnol 2021; 9:718377. [PMID: 34616718 PMCID: PMC8488380 DOI: 10.3389/fbioe.2021.718377] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/13/2021] [Indexed: 01/13/2023] Open
Abstract
Wound healing is a common physiological process which consists of a sequence of molecular and cellular events that occur following the onset of a tissue lesion in order to reconstitute barrier between body and external environment. The inherent properties of hydrogels allow the damaged tissue to heal by supporting a hydrated environment which has long been explored in wound management to aid in autolytic debridement. However, chronic non-healing wounds require added therapeutic features that can be achieved by incorporation of biomolecules and supporting cells to promote faster and better healing outcomes. In recent decades, numerous hydrogels have been developed and modified to match the time scale for distinct stages of wound healing. This review will discuss the effects of various types of hydrogels on wound pathophysiology, as well as the ideal characteristics of hydrogels for wound healing, crosslinking mechanism, fabrication techniques and design considerations of hydrogel engineering. Finally, several challenges related to adopting hydrogels to promote wound healing and future perspectives are discussed.
Collapse
Affiliation(s)
- Fei Fan
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Sanjoy Saha
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
| | - Donny Hanjaya-Putra
- Bioengineering Graduate Program, Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN, United States
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN, United States
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, United States
| |
Collapse
|
16
|
Gu C, Feng J, Waqas A, Deng Y, Zhang Y, Chen W, Long J, Huang S, Chen L. Technological Advances of 3D Scaffold-Based Stem Cell/Exosome Therapy in Tissues and Organs. Front Cell Dev Biol 2021; 9:709204. [PMID: 34568322 PMCID: PMC8458970 DOI: 10.3389/fcell.2021.709204] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, biomaterial scaffolds have been widely applied in the field of tissue engineering and regenerative medicine. Due to different production methods, unique types of three-dimensional (3D) scaffolds can be fabricated to meet the structural characteristics of tissues and organs, and provide suitable 3D microenvironments. The therapeutic effects of stem cell (SC) therapy in tissues and organs are considerable and have attracted the attention of academic researchers worldwide. However, due to the limitations and challenges of SC therapy, exosome therapy can be used for basic research and clinical translation. The review briefly introduces the materials (nature or polymer), shapes (hydrogels, particles and porous solids) and fabrication methods (crosslinking or bioprinting) of 3D scaffolds, and describes the recent progress in SC/exosome therapy with 3D scaffolds over the past 5 years (2016-2020). Normal SC/exosome therapy can improve the structure and function of diseased and damaged tissues and organs. In addition, 3D scaffold-based SC/exosome therapy can significantly improve the structure and function cardiac and neural tissues for the treatment of various refractory diseases. Besides, exosome therapy has the same therapeutic effects as SC therapy but without the disadvantages. Hence, 3D scaffold therapy provides an alternative strategy for treatment of refractory and incurable diseases and has entered a transformation period from basic research into clinical translation as a viable therapeutic option in the future.
Collapse
Affiliation(s)
- Chenyang Gu
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia Feng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- School of Medicine, Southeast University, Nanjing, China
| | - Ahmed Waqas
- School of Medicine, Southeast University, Nanjing, China
| | - Yushu Deng
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yifan Zhang
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wanghao Chen
- Department of Neurosurgery, Ninth People Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Long
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Shiying Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lukui Chen
- Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Chen L, Carlton M, Chen X, Kaur N, Ryan H, Parker TJ, Lin Z, Xiao Y, Zhou Y. Effect of fibronectin, FGF-2, and BMP4 in the stemness maintenance of BMSCs and the metabolic and proteomic cues involved. Stem Cell Res Ther 2021; 12:165. [PMID: 33676544 PMCID: PMC7936451 DOI: 10.1186/s13287-021-02227-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/14/2021] [Indexed: 02/08/2023] Open
Abstract
Background Growing evidence suggests that the pluripotent state of mesenchymal stem cells (MSCs) relies on specific local microenvironmental cues such as adhesion molecules and growth factors. Fibronectin (FN), fibroblast growth factor 2 (FGF2), and bone morphogenetic protein 4 (BMP4) are the key players in the regulation of stemness and lineage commitment of MSCs. Therefore, this study was designed to investigate the pluripotency and multilineage differentiation of bone marrow-derived MSCs (BMSCs) with the introduction of FN, FGF-2, and BMP4 and to identify the metabolic and proteomic cues involved in stemness maintenance. Methods To elucidate the stemness of BMSCs when treated with FN, FGF-2, and BMP4, the pluripotency markers of OCT4, SOX2, and c-MYC in BMSCs were monitored by real-time PCR and/or western blot. The nuclear translocation of OCT4, SOX2, and c-MYC was investigated by immunofluorescence staining. Multilineage differentiation of the treated BMSCs was determined by relevant differentiation markers. To identify the molecular signatures of BMSC stemness, gas chromatography-mass spectrometry (GC-MS), liquid chromatography-tandem mass spectrometry (LC-MS/MS), and bioinformatics analysis were utilized to determine the metabolite and protein profiles associated with stem cell maintenance. Results Our results demonstrated that the expression of stemness markers decreased with BMSC passaging, and the manipulation of the microenvironment with fibronectin and growth factors (FGF2 and BMP4) can significantly improve BMSC stemness. Of note, we revealed 7 differentially expressed metabolites, the target genes of these metabolites may have important implications in the maintenance of BMSCs through their effects on metabolic activity, energy production, and potentially protein production. We also identified 21 differentially abundant proteins, which involved in multiple pathways, including metabolic, autophagy-related, and signaling pathways regulating the pluripotency of stem cells. Additionally, bioinformatics analysis comfirned the correlation between metabolic and proteomic profiling, suggesting that the importance of metabolism and proteome networks and their reciprocal communication in the preservation of stemness. Conclusions These results indicate that the culture environment supplemented with the culture cocktail (FN, FGF2, and BMP4) plays an essential role in shaping the pluripotent state of BMSCs. Both the metabolism and proteome networks are involved in this process and the modulation of cell-fate decision making. All these findings may contribute to the application of MSCs for regenerative medicine. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02227-7.
Collapse
Affiliation(s)
- Lingling Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology & Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China
| | - Morgan Carlton
- Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Xiaodan Chen
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology & Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China
| | - Navdeep Kaur
- Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Hollie Ryan
- Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Tony J Parker
- Faculty of Health, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia
| | - Zhengmei Lin
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology & Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China.
| | - Yin Xiao
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology & Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, Guangdong, China. .,Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia.
| | - Yinghong Zhou
- Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, Queensland, 4000, Australia.
| |
Collapse
|
18
|
Applications of Mesenchymal Stem Cells in Skin Regeneration and Rejuvenation. Int J Mol Sci 2021; 22:ijms22052410. [PMID: 33673711 PMCID: PMC7957487 DOI: 10.3390/ijms22052410] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells derived from adult stem cells. Primary MSCs can be obtained from diverse sources, including bone marrow, adipose tissue, and umbilical cord blood. Recently, MSCs have been recognized as therapeutic agents for skin regeneration and rejuvenation. The skin can be damaged by wounds, caused by cutting or breaking of the tissue, and burns. Moreover, skin aging is a process that occurs naturally but can be worsened by environmental pollution, exposure to ultraviolet radiation, alcohol consumption, tobacco use, and undernourishment. MSCs have healing capacities that can be applied in damaged and aged skin. In skin regeneration, MSCs increase cell proliferation and neovascularization, and decrease inflammation in skin injury lesions. In skin rejuvenation, MSCs lead to production of collagen and elastic fibers, inhibition of metalloproteinase activation, and promote protection from ultraviolet radiation-induced senescence. In this review, we focus on how MSCs and MSC-derived molecules improve diseased and aged skin. Additionally, we emphasize that induced pluripotent stem cell (iPSC)-derived MSCs are potentially advanced MSCs, which are suitable for cell therapy.
Collapse
|
19
|
Zou J, Du J, Tu H, Chen H, Cong K, Bi Z, Sun J. Resveratrol benefits the lineage commitment of bone marrow mesenchymal stem cells into osteoblasts via miR-320c by targeting Runx2. J Tissue Eng Regen Med 2021; 15:347-360. [PMID: 33481337 DOI: 10.1002/term.3176] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 01/09/2021] [Indexed: 11/07/2022]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are a potential source of osteoblasts and have been widely used in clinical therapies due to their pluripotency. Recent publications have found that resveratrol (RSVL) played a crucial role in the proliferation and differentiation of BMSCs; however, the underlying molecular mechanism of RSVL-induced BMSCs osteogenic differentiation needs to be fully elucidated. The objective of this study was to explore functions of miRNAs in the RSVL-treated BMSCs and its effects on the differentiation potentials of BMSCs. The findings demonstrated that RSVL enhanced the osteogenesis and suppressed the adipogenesis of BMSCs in a dose-dependent manner. Besides, a novel regulatory axis containing miR-320c, and its target Runx2 was found during the differentiation process of BMSCs under RSVL treatment. Increase of miR-320c reduced the osteogenic potential of BMSCs, while knockdown of miR-320c played a positive role in the osteogenesis of BMSCs. In contrast, overexpression of miR-320c accelerated the adipogenic differentiation, while knockdown of miR-320c restrained the adipogenic differentiation of BMSCs. The results confirmed that Runx2 might be the direct target of miR-320c in RSVL-promoted osteogenic differentiation of BMSCs. This study revealed that RSVL might be used for the treatment of bone loss related diseases and miR-320c could be regarded as a novel and potential target to regulate the biological functions of BMSCs.
Collapse
Affiliation(s)
- Jilong Zou
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jianyang Du
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hualei Tu
- Department of Burn, The Fifth Hospital in Harbin, Harbin, China
| | - Hongjun Chen
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kai Cong
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenggang Bi
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiabing Sun
- Department of Orthopaedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
Bhattacharyya A, Janarthanan G, Noh I. Nano-biomaterials for designing functional bioinks towards complex tissue and organ regeneration in 3D bioprinting. ADDITIVE MANUFACTURING 2021; 37:101639. [DOI: 10.1016/j.addma.2020.101639] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
21
|
Huang J, Frauenlob M, Shibata Y, Wang L, Nakajima T, Nonoyama T, Tsuda M, Tanaka S, Kurokawa T, Gong JP. Chitin-Based Double-Network Hydrogel as Potential Superficial Soft-Tissue-Repairing Materials. Biomacromolecules 2020; 21:4220-4230. [PMID: 32936628 DOI: 10.1021/acs.biomac.0c01003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chitin is a biopolymer, which has been proven to be a biomedical material candidate, yet the weak mechanical properties seriously limit their potentials. In this work, a chitin-based double-network (DN) hydrogel has been designed as a potential superficial repairing material. The hydrogel was synthesized through a double-network (DN) strategy composing hybrid regenerated chitin nanofiber (RCN)-poly (ethylene glycol diglycidyl ether) (PEGDE) as the first network and polyacrylamide (PAAm) as the second network. The hybrid RCN-PEGDE/PAAm DN hydrogel was strong and tough, possessing Young's modulus (elasticity) E 0.097 ± 0.020 MPa, fracture stress σf 0.449 ± 0.025 MPa, and work of fracture Wf 5.75 ± 0.35 MJ·m-3. The obtained DN hydrogel was strong enough for surgical requirements in the usage of soft tissue scaffolds. In addition, chitin endowed the DN hydrogel with good bacterial resistance and accelerated fibroblast proliferation, which increased the NIH3T3 cell number by nearly five times within 3 days. Subcutaneous implantation studies showed that the DN hydrogel did not induce inflammation after 4 weeks, suggesting a good biosafety in vivo. These results indicated that the hybrid RCN-PEGDE/PAAm DN hydrogel had great prospect as a rapid soft-tissue-repairing material.
Collapse
Affiliation(s)
- Junchao Huang
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Martin Frauenlob
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Yuki Shibata
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Lei Wang
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan
| | - Tasuku Nakajima
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan
| | - Takayuki Nonoyama
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan
| | - Masumi Tsuda
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan
| | - Shinya Tanaka
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan
| | - Takayuki Kurokawa
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan
| | - Jian Ping Gong
- Laboratory of Soft and Wet Matter, Faculty of Advanced Life Science, Hokkaido University, Sapporo 060-0810, Japan.,Global Station for Soft Matter, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo 001-0021, Japan.,Institute for Chemical Reaction Design and Discovery (WPI-ICReDD), Hokkaido University, Sapporo 001-0021, Japan
| |
Collapse
|
22
|
Liubaviciute A, Ivaskiene T, Biziuleviciene G. Modulated mesenchymal stromal cells improve skin wound healing. Biologicals 2020; 67:1-8. [DOI: 10.1016/j.biologicals.2020.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/26/2020] [Accepted: 08/04/2020] [Indexed: 12/20/2022] Open
|
23
|
Goetzke R, Keijdener H, Franzen J, Ostrowska A, Nüchtern S, Mela P, Wagner W. Differentiation of Induced Pluripotent Stem Cells towards Mesenchymal Stromal Cells is Hampered by Culture in 3D Hydrogels. Sci Rep 2019; 9:15578. [PMID: 31666572 PMCID: PMC6821810 DOI: 10.1038/s41598-019-51911-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 10/10/2019] [Indexed: 01/08/2023] Open
Abstract
Directed differentiation of induced pluripotent stem cells (iPSCs) towards specific lineages remains a major challenge in regenerative medicine, while there is a growing perception that this process can be influenced by the three-dimensional environment. In this study, we investigated whether iPSCs can differentiate towards mesenchymal stromal cells (MSCs) when embedded into fibrin hydrogels to enable a one-step differentiation procedure within a scaffold. Differentiation of iPSCs on tissue culture plastic or on top of fibrin hydrogels resulted in a typical MSC-like phenotype. In contrast, iPSCs embedded into fibrin gel gave rise to much smaller cells with heterogeneous growth patterns, absence of fibronectin, faint expression of CD73 and CD105, and reduced differentiation potential towards osteogenic and adipogenic lineage. Transcriptomic analysis demonstrated that characteristic genes for MSCs and extracellular matrix were upregulated on flat substrates, whereas genes of neural development were upregulated in 3D culture. Furthermore, the 3D culture had major effects on DNA methylation profiles, particularly within genes for neuronal and cardiovascular development, while there was no evidence for epigenetic maturation towards MSCs. Taken together, iPSCs could be differentiated towards MSCs on tissue culture plastic or on a flat fibrin hydrogel. In contrast, the differentiation process was heterogeneous and not directed towards MSCs when iPSCs were embedded into the hydrogel.
Collapse
Affiliation(s)
- Roman Goetzke
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Hans Keijdener
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Julia Franzen
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Alina Ostrowska
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Selina Nüchtern
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Petra Mela
- Department of Biohybrid & Medical Textiles (BioTex), AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany. .,Medical Materials and Implants, Department of Mechanical Engineering and Munich School of BioEngineering, Technical University of Munich, Garching, Germany.
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, Stem Cell Biology and Cellular Engineering, RWTH Aachen University Medical School, Aachen, Germany. .,Institute for Biomedical Engineering - Cell Biology, RWTH Aachen University Medical School, Aachen, Germany.
| |
Collapse
|
24
|
Zhou X, Zhang D, Wang M, Zhang D, Xu Y. Three-Dimensional Printed Titanium Scaffolds Enhance Osteogenic Differentiation and New Bone Formation by Cultured Adipose Tissue-Derived Stem Cells Through the IGF-1R/AKT/Mammalian Target of Rapamycin Complex 1 (mTORC1) Pathway. Med Sci Monit 2019; 25:8043-8054. [PMID: 31655847 PMCID: PMC6833923 DOI: 10.12659/msm.918517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background This study aimed to investigate the effects of three-dimensional (3D) printed titanium (3DTi) scaffolds on osteogenic differentiation and new bone formation by 3D cultured adipose tissue-derived stem cells (ADSCs) in vitro, and the effects of bone regeneration in vivo using a full-thickness mandibular defect rat model, and the mechanisms involved. Material/Methods Alpha-beta titanium alloy (Ti6Al4V) 3DTi scaffolds were prepared with Cellmatrix hydrogel and 3D culture medium. ADSCs were impregnated into the 3DTi scaffolds. ADSC viability and proliferation were assessed using the cell counting kit-8 (CCK-8) assay, and alkaline phosphatase (ALP) levels were measured. Real-time polymerase chain reaction (RT-PCR) and Western blot were performed to assess the expression of osteogenesis-related mRNA for RUNX2, OPN, OCN, and IGF-1 genes and proteins. A rat model of full-thickness mandibular defect was evaluated with micro-computed tomography (microCT) scanning, and histochemistry with Alizarin red and von Giesen’s stain were used to evaluate osteogenesis. Results ADSC viability and proliferation were not affected by culture with 3DTi scaffolds. Expression of osteogenesis-related mRNA and proteins for RUNX2, OPN, OCN, and IGF-1, expression of ALP, and histochemical findings showed that the use of 3DTi scaffolds enhanced osteogenic differentiation and new bone formation by ADSCs, with upregulation of components of the IGF-1R/AKT/mTORC1 pathway. Conclusions The 3D culture of ADSCs with 3DTi scaffolds enhanced osteogenic differentiation and new bone formation through the IGF-1R/AKT/mTORC1 pathway. This improved method of osteointegration may have clinical application in the preparation of bone grafts before implantation for improved repair of mandibular bone defects.
Collapse
Affiliation(s)
- Xiaoyu Zhou
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Dongjie Zhang
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, China (mainland)
| | - Mengling Wang
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Ding Zhang
- Department of Stomatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China (mainland)
| | - Yisheng Xu
- Orthopedics Department, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
25
|
da Silva LP, Reis RL, Correlo VM, Marques AP. Hydrogel-Based Strategies to Advance Therapies for Chronic Skin Wounds. Annu Rev Biomed Eng 2019; 21:145-169. [DOI: 10.1146/annurev-bioeng-060418-052422] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chronic skin wounds are the leading cause of nontraumatic foot amputations worldwide and present a significant risk of morbidity and mortality due to the lack of efficient therapies. The intrinsic characteristics of hydrogels allow them to benefit cutaneous healing essentially by supporting a moist environment. This property has long been explored in wound management to aid in autolytic debridement. However, chronic wounds require additional therapeutic features that can be provided by a combination of hydrogels with biochemical mediators or cells, promoting faster and better healing. We survey hydrogel-based approaches with potential to improve the healing of chronic wounds by reviewing their effects as observed in preclinical models. Topics covered include strategies to ablate infection and resolve inflammation, the delivery of bioactive agents to accelerate healing, and tissue engineering approaches for skin regeneration. The article concludes by considering the relevance of treating chronic skin wounds using hydrogel-based strategies.
Collapse
Affiliation(s)
- Lucília P. da Silva
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Vitor M. Correlo
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Alexandra P. Marques
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| |
Collapse
|
26
|
Liu Y, Zhou S, Gao Y, Zhai Y. Electrospun nanofibers as a wound dressing for treating diabetic foot ulcer. Asian J Pharm Sci 2019; 14:130-143. [PMID: 32104445 PMCID: PMC7032134 DOI: 10.1016/j.ajps.2018.04.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 03/25/2018] [Accepted: 04/19/2018] [Indexed: 01/14/2023] Open
Abstract
Diabetes is one of the most prevalent diseases in the world with high-mortality and complex complications including diabetic foot ulcer (DFU). It has been reported that the difficulties in repairing the wound related to DFU has much relationship with the wound infection, change of inflammatory responses, lack of extracellular matrix (ECM), and the failure of angiogenesis. Following the development of medical materials and pharmaceutical technology, nanofibers has been developed by electrospinning with huge porosity, excellent humidity absorption, a better oxygen exchange rate, and some antibacterial activities. That is to say, as a potential material, nanofibers must be a wonderful candidate for the DFU treatment with so many benefits. Careful selection of polymers from natural resource and synthetic resource can widen the nanofibrous application. Popular methods applied for the nanofibrous fabrication consist of uniaxial electrospinning and coaxial electrospinning. Furthermore, nanofibers loading chemical, biochemical active pharmaceutical ingredient (API) or even stem cells can be wonderful dosage forms for the treatment of DFU. This review summarizes the present techniques applied in the fabrication of nanofibrous dressing (ND) that utilizes a variety of materials and active agents to offer a better health care for the patients suffering from DFU.
Collapse
Affiliation(s)
- Yan Liu
- Shenyang Pharmaceutical University, No.103, Shenyang 110016, China
| | - Shiya Zhou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yanlin Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinglei Zhai
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
27
|
Mohebbi S, Nezhad MN, Zarrintaj P, Jafari SH, Gholizadeh SS, Saeb MR, Mozafari M. Chitosan in Biomedical Engineering: A Critical Review. Curr Stem Cell Res Ther 2019; 14:93-116. [PMID: 30207244 DOI: 10.2174/1574888x13666180912142028] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/29/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022]
Abstract
Biomedical engineering seeks to enhance the quality of life by developing advanced materials and technologies. Chitosan-based biomaterials have attracted significant attention because of having unique chemical structures with desired biocompatibility and biodegradability, which play different roles in membranes, sponges and scaffolds, along with promising biological properties such as biocompatibility, biodegradability and non-toxicity. Therefore, chitosan derivatives have been widely used in a vast variety of uses, chiefly pharmaceuticals and biomedical engineering. It is attempted here to draw a comprehensive overview of chitosan emerging applications in medicine, tissue engineering, drug delivery, gene therapy, cancer therapy, ophthalmology, dentistry, bio-imaging, bio-sensing and diagnosis. The use of Stem Cells (SCs) has given an interesting feature to the use of chitosan so that regenerative medicine and therapeutic methods have benefited from chitosan-based platforms. Plenty of the most recent discussions with stimulating ideas in this field are covered that could hopefully serve as hints for more developed works in biomedical engineering.
Collapse
Affiliation(s)
- Shabnam Mohebbi
- Department of Chemical Engineering, Tabriz University, Tabriz, Iran
| | | | - Payam Zarrintaj
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed Hassan Jafari
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Saman Seyed Gholizadeh
- Department of Microbiology, College of Basic Science, Islamic Azad University, Shiraz Branch, Shiraz, Iran
| | - Mohammad Reza Saeb
- Departments of Resin and Additives, Institute for Color Science and Technology, P.O. Box 16765-654, Tehran, Iran
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran, Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Ling S, Chen W, Fan Y, Zheng K, Jin K, Yu H, Buehler MJ, Kaplan DL. Biopolymer nanofibrils: structure, modeling, preparation, and applications. Prog Polym Sci 2018; 85:1-56. [PMID: 31915410 PMCID: PMC6948189 DOI: 10.1016/j.progpolymsci.2018.06.004] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biopolymer nanofibrils exhibit exceptional mechanical properties with a unique combination of strength and toughness, while also presenting biological functions that interact with the surrounding environment. These features of biopolymer nanofibrils profit from their hierarchical structures that spun angstrom to hundreds of nanometer scales. To maintain these unique structural features and to directly utilize these natural supramolecular assemblies, a variety of new methods have been developed to produce biopolymer nanofibrils. In particular, cellulose nanofibrils (CNFs), chitin nanofibrils (ChNFs), silk nanofibrils (SNFs) and collagen nanofibrils (CoNFs), as the four most abundant biopolymer nanofibrils on earth, have been the focus of research in recent years due to their renewable features, wide availability, low-cost, biocompatibility, and biodegradability. A series of top-down and bottom-up strategies have been accessed to exfoliate and regenerate these nanofibrils for versatile advanced applications. In this review, we first summarize the structures of biopolymer nanofibrils in nature and outline their related computational models with the aim of disclosing fundamental structure-property relationships in biological materials. Then, we discuss the underlying methods used for the preparation of CNFs, ChNFs, SNF and CoNFs, and discuss emerging applications for these biopolymer nanofibrils.
Collapse
Affiliation(s)
- Shengjie Ling
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Wenshuai Chen
- Key Laboratory of Bio-based Material Science & Technology, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Yimin Fan
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Ke Zheng
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Kai Jin
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Haipeng Yu
- Key Laboratory of Bio-based Material Science & Technology, Ministry of Education, Northeast Forestry University, Harbin, China
| | - Markus J. Buehler
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| |
Collapse
|
29
|
3D Culture of Bone Marrow-Derived Mesenchymal Stem Cells (BMSCs) Could Improve Bone Regeneration in 3D-Printed Porous Ti6Al4V Scaffolds. Stem Cells Int 2018; 2018:2074021. [PMID: 30254680 PMCID: PMC6145055 DOI: 10.1155/2018/2074021] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 07/10/2018] [Accepted: 07/30/2018] [Indexed: 01/14/2023] Open
Abstract
Mandibular bone defect reconstruction is an urgent challenge due to the requirements for daily eating and facial aesthetics. Three-dimensional- (3D-) printed titanium (Ti) scaffolds could provide patient-specific implants for bone defects. Appropriate load-bearing properties are also required during bone reconstruction, which makes them potential candidates for mandibular bone defect reconstruction implants. However, in clinical practice, the insufficient osteogenesis of the scaffolds needs to be further improved. In this study, we first encapsulated bone marrow-derived mesenchymal stem cells (BMSCs) into Matrigel. Subsequently, the BMSC-containing Matrigels were infiltrated into porous Ti6Al4V scaffolds. The Matrigels in the scaffolds provided a 3D culture environment for the BMSCs, which was important for osteoblast differentiation and new bone formation. Our results showed that rats with a full thickness of critical mandibular defects treated with Matrigel-infiltrated Ti6Al4V scaffolds exhibited better new bone formation than rats with local BMSC injection or Matrigel-treated defects. Our data suggest that Matrigel is able to create a more favorable 3D microenvironment for BMSCs, and Matrigel containing infiltrated BMSCs may be a promising method for enhancing the bone formation properties of 3D-printed Ti6Al4V scaffolds. We suggest that this approach provides an opportunity to further improve the efficiency of stem cell therapy for the treatment of mandibular bone defects.
Collapse
|
30
|
Huang Y, Wang Y, Chen L, Zhang L. Facile construction of mechanically tough collagen fibers reinforced by chitin nanofibers as cell alignment templates. J Mater Chem B 2018; 6:918-929. [DOI: 10.1039/c7tb02945d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Reconstituted collagen fibers with excellent mechanical performance were successfully fabricated with sodium alginate as coagulate and chitin nanofibers as reinforcing filler and applied as a fibroblast alignment templated scaffold.
Collapse
Affiliation(s)
- Yao Huang
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Yixiang Wang
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Lingyun Chen
- Department of Agricultural
- Food and Nutritional Science
- University of Alberta
- Edmonton
- Canada
| | - Lina Zhang
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
- China
| |
Collapse
|
31
|
Datta S, Rameshbabu AP, Bankoti K, Maity PP, Das D, Pal S, Roy S, Sen R, Dhara S. Oleoyl-Chitosan-Based Nanofiber Mats Impregnated with Amniotic Membrane Derived Stem Cells for Accelerated Full-Thickness Excisional Wound Healing. ACS Biomater Sci Eng 2017; 3:1738-1749. [DOI: 10.1021/acsbiomaterials.7b00189] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | - Dipankar Das
- Department
of Applied Chemistry, Indian School of Mines, Dhanbad 826004, India
| | - Sagar Pal
- Department
of Applied Chemistry, Indian School of Mines, Dhanbad 826004, India
| | - Sabyasachi Roy
- Department
of Gynaecology, Midnapore Medical College, Paschim Medinipur 721101, India
| | | | | |
Collapse
|