1
|
Li Q, Ma C, Jing Y, Liu X. Multifunctional Nanofibrous Hollow Microspheres for Enhanced Periodontal Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402335. [PMID: 38757666 PMCID: PMC11267322 DOI: 10.1002/advs.202402335] [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: 03/04/2024] [Revised: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Destructive periodontitis destroys alveolar bone and eventually leads to tooth loss. While guided bone regeneration, which is based on creating a physical barrier to hinder the infiltration of epithelial and connective tissues into defect sites, has been widely used for alveolar bone regeneration, its outcomes remain variable. In this work, a multifunctional nanofibrous hollow microsphere (NFHMS) is developed for enhanced alveolar bone regeneration. The NFHMS is first prepared via combining a double emulsification and a thermally induced phase separation process. Next, E7, a short peptide with high specific affinity to bone marrow-derived stem cells (BMSCs), is conjugated onto the surface of NFHMS. After that, bone forming peptide (BFP), a short peptide derived from bone morphology protein 7 is loaded in calcium phosphate (CaP) nanoparticles, which are further encapsulated in the hollow space of the NFHMS-E7 to form NFHMS-E7-CaP/BFP. The NFHMS-E7-CaP/BFP selectively promoted the adhesion of BMSCs and expelled the adhesion of fibroblasts and epithelial cells. In addition, the BFP is sustainedly released from the NFHMS-E7-CaP/BFP to enhance the osteogenesis of BMSCs. A rat challenging fenestration defect model showed that the NFHMS-E7-CaP/BFP significantly enhanced alveolar bone tissue regeneration. This work provides a novel bioengineering approach for guided bone regeneration.
Collapse
Affiliation(s)
- Qian Li
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Chemical and Biomedical Engineering DepartmentUniversity of MissouriColumbiaMO65211USA
| | - Chi Ma
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Center of Excellence in HipScottish Rite for ChildrenDallasTX75219USA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical CenterDallasTX75390USA
| | - Yan Jing
- Department of OrthodonticsTexas A&M University School of DentistryDallasTX75246USA
| | - Xiaohua Liu
- Department of Biomedical SciencesTexas A&M University School of DentistryDallasTX75246USA
- Chemical and Biomedical Engineering DepartmentUniversity of MissouriColumbiaMO65211USA
| |
Collapse
|
2
|
Zhou Z, Wang J, Jiang C, Xu K, Xu T, Yu X, Fang J, Yang Y, Dai X. Advances in Hydrogels for Meniscus Tissue Engineering: A Focus on Biomaterials, Crosslinking, Therapeutic Additives. Gels 2024; 10:114. [PMID: 38391445 PMCID: PMC10887778 DOI: 10.3390/gels10020114] [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: 01/08/2024] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Meniscus tissue engineering (MTE) has emerged as a promising strategy for meniscus repair and regeneration. As versatile platforms, hydrogels have gained significant attention in this field, as they possess tunable properties that allow them to mimic native extracellular matrices and provide a suitable microenvironment. Additionally, hydrogels can be minimally invasively injected and can be adjusted to match the shape of the implant site. They can conveniently and effectively deliver bioactive additives and demonstrate good compatibility with other functional materials. These inherent qualities have made hydrogel a promising candidate for therapeutic approaches in meniscus repair and regeneration. This article provides a comprehensive review of the advancements made in the research on hydrogel application for meniscus tissue engineering. Firstly, the biomaterials and crosslinking strategies used in the formation of hydrogels are summarized and analyzed. Subsequently, the role of therapeutic additives, including cells, growth factors, and other active products, in facilitating meniscus repair and regeneration is thoroughly discussed. Furthermore, we summarize the key issues for designing hydrogels used in MTE. Finally, we conclude with the current challenges encountered by hydrogel applications and suggest potential solutions for addressing these challenges in the field of MTE. We hope this review provides a resource for researchers and practitioners interested in this field, thereby facilitating the exploration of new design possibilities.
Collapse
Affiliation(s)
- Zhuxing Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jiajie Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Chaoqian Jiang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Kaiwang Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Tengjing Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Xinning Yu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Jinghua Fang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| | - Yanyu Yang
- School of Materials and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xuesong Dai
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou 310000, China
| |
Collapse
|
3
|
Hu Z, Rong X, Liu X. E7-Conjugated Bio-Inspired Microspheres as a Biological Barrier for Guided Tissue Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58136-58150. [PMID: 38063848 PMCID: PMC10862379 DOI: 10.1021/acsami.3c12213] [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: 08/16/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/22/2023]
Abstract
Guided tissue regeneration (GTR), which is based on creating a physical barrier to prevent the downgrowth of epithelial and connective tissues into the defect site, has been widely used in clinical practice for periodontal regeneration for many years. However, its outcomes remain variable due to highly specific indications, the demand for proficient surgical skills, and frequent occurrence of complications. In this study, we developed a new GTR biomaterial that acts as a biological barrier for epithelial cells and fibroblasts while also serving as a scaffold for bone marrow-derived mesenchymal stem cells (BMSCs) and periodontal ligament stem cells (PDLSCs). This innovative GTR biomaterial is bioinspired injectable microspheres that are self-assembled from nanofibers, and their surfaces are conjugated with E7, a short peptide that selectively promotes BMSC and PDLSC adhesion but inhibits the attachment and spreading of epithelial cells and gingival fibroblasts. The selective affinity afforded by E7 on the surfaces of the nanofibrous microspheres facilitated the colonization of BMSCs in the periodontal defect, thereby substantially improving functional periodontal regeneration, as evidenced by enhanced new bone formation, reduced root exposure, and diminished attachment loss. The remarkable superiority of the bioinspired microspheres over conventional GTR materials in promoting periodontal regeneration underscores the potential of this innovative approach to enhance the efficacy of functional periodontal tissue regeneration.
Collapse
Affiliation(s)
- Zhiai Hu
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
| | - Xin Rong
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
| | - Xiaohua Liu
- Department
of Biomedical Sciences, Texas A&M University
School of Dentistry, Dallas, Texas 75246, United States
- Chemical
and Biomedical Engineering Department, University
of Missouri, Columbia, Missouri 65211, United States
| |
Collapse
|
4
|
Li Q, Yu H, Zhao F, Cao C, Wu T, Fan Y, Ao Y, Hu X. 3D Printing of Microenvironment-Specific Bioinspired and Exosome-Reinforced Hydrogel Scaffolds for Efficient Cartilage and Subchondral Bone Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303650. [PMID: 37424038 PMCID: PMC10502685 DOI: 10.1002/advs.202303650] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Indexed: 07/11/2023]
Abstract
In clinical practice, repairing osteochondral defects presents a challenge due to the varying biological properties of articular cartilages and subchondral bones. Thus, elucidating how spatial microenvironment-specific biomimetic scaffolds can be used to simultaneously regenerate osteochondral tissue is an important research topic. Herein, a novel bioinspired double-network hydrogel scaffold produced via 3D printing with tissue-specific decellularized extracellular matrix (dECM) and human adipose mesenchymal stem cell (MSC)-derived exosomes is described. The bionic hydrogel scaffolds promote rat bone marrow MSC attachment, spread, migration, proliferation, and chondrogenic and osteogenic differentiation in vitro, as determined based on the sustained release of bioactive exosomes. Furthermore, the 3D-printed microenvironment-specific heterogeneous bilayer scaffolds efficiently accelerate the simultaneous regeneration of cartilage and subchondral bone tissues in a rat preclinical model. In conclusion, 3D dECM-based microenvironment-specific biomimetics encapsulated with bioactive exosomes can serve as a novel cell-free recipe for stem cell therapy when treating injured or degenerative joints. This strategy provides a promising platform for complex zonal tissue regeneration whilst holding attractive clinical translation potential.
Collapse
Affiliation(s)
- Qi Li
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
- Center of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730China
| | - Huilei Yu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Fengyuan Zhao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Chenxi Cao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Tong Wu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Yifei Fan
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Yingfang Ao
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| | - Xiaoqing Hu
- Department of Sports MedicineInstitute of Sports Medicine of Peking UniversityBeijing Key Laboratory of Sports InjuriesPeking University Third HospitalBeijing100191China
| |
Collapse
|
5
|
Mohammadalizadeh Z, Bahremandi-Toloue E, Karbasi S. Recent advances in modification strategies of pre- and post-electrospinning of nanofiber scaffolds in tissue engineering. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2022.105202] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
6
|
Li H, Liao Z, Yang Z, Gao C, Fu L, Li P, Zhao T, Cao F, Chen W, Yuan Z, Sui X, Liu S, Guo Q. 3D Printed Poly(ε-Caprolactone)/Meniscus Extracellular Matrix Composite Scaffold Functionalized With Kartogenin-Releasing PLGA Microspheres for Meniscus Tissue Engineering. Front Bioeng Biotechnol 2021; 9:662381. [PMID: 33996783 PMCID: PMC8119888 DOI: 10.3389/fbioe.2021.662381] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/22/2021] [Indexed: 01/04/2023] Open
Abstract
Meniscus tissue engineering (MTE) aims to fabricate ideal scaffolds to stimulate the microenvironment for recreating the damaged meniscal tissue. Indeed, favorable mechanical properties, suitable biocompatibility, and inherent chondrogenic capability are crucial in MTE. In this study, we present a composite scaffold by 3D printing a poly(ε-caprolactone) (PCL) scaffold as backbone, followed by injection with the meniscus extracellular matrix (MECM), and modification with kartogenin (KGN)-loaded poly(lactic-co-glycolic) acid (PLGA) microsphere (μS), which serves as a drug delivery system. Therefore, we propose a plan to improve meniscus regeneration via KGN released from the 3D porous PCL/MECM scaffold. The final results showed that the hydrophilicity and bioactivity of the resulting PCL/MECM scaffold were remarkably enhanced. In vitro synovium-derived mesenchymal stem cells (SMSCs) experiments suggested that introducing MECM components helped cell adhesion and proliferation and maintained promising ability to induce cell migration. Moreover, KGN-incorporating PLGA microspheres, which were loaded on scaffolds, showed a prolonged release profile and improved the chondrogenic differentiation of SMSCs during the 14-day culture. Particularly, the PCL/MECM-KGN μS seeded by SMSCs showed the highest secretion of total collagen and aggrecan. More importantly, the synergistic effect of the MECM and sustained release of KGN can endow the PCL/MECM-KGN μS scaffolds with not only excellent cell affinity and cell vitality preservation but also chondrogenic activity. Thus, the PCL/MECM-KGN μS scaffolds are expected to have good application prospects in the field of MTE.
Collapse
Affiliation(s)
- Hao Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiyao Liao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhen Yang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Cangjian Gao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Liwei Fu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Pinxue Li
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Tianyuan Zhao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Fuyang Cao
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Chen
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Zhiguo Yuan
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
| | - Quanyi Guo
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
7
|
Wong KU, Zhang A, Akhavan B, Bilek MM, Yeo GC. Biomimetic Culture Strategies for the Clinical Expansion of Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2021. [PMID: 33599471 DOI: 10.1021/acsbiomaterials.0c01538] [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: 11/30/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) typically require significant ex vivo expansion to achieve the high cell numbers required for research and clinical applications. However, conventional MSC culture on planar (2D) plastic surfaces has been shown to induce MSC senescence and decrease cell functionality over long-term proliferation, and usually, it has a high labor requirement, a high usage of reagents, and therefore, a high cost. In this Review, we describe current MSC-based therapeutic strategies and outline the important factors that need to be considered when developing next-generation cell expansion platforms. To retain the functional value of expanded MSCs, ex vivo culture systems should ideally recapitulate the components of the native stem cell microenvironment, which include soluble cues, resident cells, and the extracellular matrix substrate. We review the interplay between these stem cell niche components and their biological roles in governing MSC phenotype and functionality. We discuss current biomimetic strategies of incorporating biochemical and biophysical cues in MSC culture platforms to grow clinically relevant cell numbers while preserving cell potency and stemness. This Review summarizes the current state of MSC expansion technologies and the challenges that still need to be overcome for MSC clinical applications to be feasible and sustainable.
Collapse
Affiliation(s)
- Kuan Un Wong
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anyu Zhang
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Behnam Akhavan
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Marcela M Bilek
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
8
|
Kim HD, Park J, Amirthalingam S, Jayakumar R, Hwang NS. Bioinspired inorganic nanoparticles and vascular factor microenvironment directed neo-bone formation. Biomater Sci 2021; 8:2627-2637. [PMID: 32242197 DOI: 10.1039/d0bm00041h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Various strategies have been explored to stimulate new bone formation. These strategies include using angiogenic stimulants in combination with inorganic biomaterials. Neovascularization during the neo-bone formation provides nutrients along with bone-forming minerals. Therefore, it is crucial to design a bone stimulating microenvironment composed of both pro-angiogenic and osteogenic factors. In this respect, human vascular endothelial growth factor (hVEGF) has been shown to promote blood vessel formation and bone formation. Furthermore, in recent years, whitlockite (WH), a novel phase of magnesium-containing calcium phosphate derivatives that exist in our bone tissue, has been synthesized and applied in bone tissue engineering. In this study, our aim is to explore the potential use of hVEGF and WH for bone tissue engineering. Our study demonstrated that hVEGF and a WH microenvironment synergistically stimulated osteogenic commitment of mesenchymal stem cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Hwan D Kim
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea.
| | - Jungha Park
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea.
| | - Sivashanmugam Amirthalingam
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - R Jayakumar
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, the Institute of Chemical Processes, Seoul National University, Seoul, 151-742, Republic of Korea. and Interdisciplinary Program in Bioengineering, Seoul National University, 151-742, Seoul, Republic of Korea and The BioMax/N-Bio Institute of Seoul National University, Seoul, 151-742, Republic of Korea
| |
Collapse
|
9
|
Chu W, Prodromou R, Day KN, Schneible JD, Bacon KB, Bowen JD, Kilgore RE, Catella CM, Moore BD, Mabe MD, Alashoor K, Xu Y, Xiao Y, Menegatti S. Peptides and pseudopeptide ligands: a powerful toolbox for the affinity purification of current and next-generation biotherapeutics. J Chromatogr A 2020; 1635:461632. [PMID: 33333349 DOI: 10.1016/j.chroma.2020.461632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Following the consolidation of therapeutic proteins in the fight against cancer, autoimmune, and neurodegenerative diseases, recent advancements in biochemistry and biotechnology have introduced a host of next-generation biotherapeutics, such as CRISPR-Cas nucleases, stem and car-T cells, and viral vectors for gene therapy. With these drugs entering the clinical pipeline, a new challenge lies ahead: how to manufacture large quantities of high-purity biotherapeutics that meet the growing demand by clinics and biotech companies worldwide. The protein ligands employed by the industry are inadequate to confront this challenge: while featuring high binding affinity and selectivity, these ligands require laborious engineering and expensive manufacturing, are prone to biochemical degradation, and pose safety concerns related to their bacterial origin. Peptides and pseudopeptides make excellent candidates to form a new cohort of ligands for the purification of next-generation biotherapeutics. Peptide-based ligands feature excellent target biorecognition, low or no toxicity and immunogenicity, and can be manufactured affordably at large scale. This work presents a comprehensive and systematic review of the literature on peptide-based ligands and their use in the affinity purification of established and upcoming biological drugs. A comparative analysis is first presented on peptide engineering principles, the development of ligands targeting different biomolecular targets, and the promises and challenges connected to the industrial implementation of peptide ligands. The reviewed literature is organized in (i) conventional (α-)peptides targeting antibodies and other therapeutic proteins, gene therapy products, and therapeutic cells; (ii) cyclic peptides and pseudo-peptides for protein purification and capture of viral and bacterial pathogens; and (iii) the forefront of peptide mimetics, such as β-/γ-peptides, peptoids, foldamers, and stimuli-responsive peptides for advanced processing of biologics.
Collapse
Affiliation(s)
- Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kevin N Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kaitlyn B Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Carly M Catella
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Matthew D Mabe
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kawthar Alashoor
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14642
| | - Yiman Xu
- College of Material Science and Engineering, Donghua University, 201620 Shanghai, People's Republic of China
| | - Yuanxin Xiao
- College of Textile, Donghua University, Songjiang District, Shanghai, 201620, People's Republic of China
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606.
| |
Collapse
|
10
|
Li Z, Wu N, Cheng J, Sun M, Yang P, Zhao F, Zhang J, Duan X, Fu X, Zhang J, Hu X, Chen H, Ao Y. Biomechanically, structurally and functionally meticulously tailored polycaprolactone/silk fibroin scaffold for meniscus regeneration. Am J Cancer Res 2020; 10:5090-5106. [PMID: 32308770 PMCID: PMC7163455 DOI: 10.7150/thno.44270] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/24/2020] [Indexed: 12/23/2022] Open
Abstract
Meniscus deficiency, the most common and refractory disease in human knee joints, often progresses to osteoarthritis (OA) due to abnormal biomechanical distribution and articular cartilage abrasion. However, due to its anisotropic spatial architecture, complex biomechanical microenvironment, and limited vascularity, meniscus repair remains a challenge for clinicians and researchers worldwide. In this study, we developed a 3D printing-based biomimetic and composite tissue-engineered meniscus scaffold consisting of polycaprolactone (PCL)/silk fibroin (SF) with extraordinary biomechanical properties and biocompatibility. We hypothesized that the meticulously tailored composite scaffold could enhance meniscus regeneration and cartilage protection. Methods: The physical property of the scaffold was characterized by scanning electron microscopy (SEM) observation, degradation test, frictional force of interface assessment, biomechanical testing, and fourier transform infrared (FTIR) spectroscopy analysis. To verify the biocompatibility of the scaffold, the viability, morphology, proliferation, differentiation, and extracellular matrix (ECM) production of synovium-derived mesenchymal stem cell (SMSC) on the scaffolds were assessed by LIVE/DEAD staining, alamarBlue assay, ELISA analysis, and qRT-PCR. The recruitment ability of SMSC was tested by dual labeling with CD29 and CD90 by confocal microscope at 1 week after implantation. The functionalized hybrid scaffold was then implanted into the meniscus defects on rabbit knee joint for meniscus regeneration, comparing with the Blank group (no scaffold) and PS group. The regenerated meniscus tissue was evaluated by histological and immunohistochemistry staining, and biomechanical test. Macroscopic and histological scoring was performed to assess the outcome of meniscus regeneration and cartilage protection in vivo. Results: The combination of SF and PCL could greatly balance the biomechanical properties and degradation rate to match the native meniscus. SF sponge, characterized by fine elasticity and low interfacial shear force, enhanced energy absorption capacity of the meniscus and improved chondroprotection. The SMSC-specific affinity peptide (LTHPRWP; L7) was conjugated to the scaffold to further increase the recruitment and retention of endogenous SMSCs. This meticulously tailored scaffold displayed superior biomechanics, structure, and function, creating a favorable microenvironment for SMSC proliferation, differentiation, and extracellular matrix (ECM) production. After 24 weeks of implantation, the histological assessment, biochemical contents, and biomechanical properties demonstrated that the polycaprolactone/silk fibroin-L7 (PS-L7) group was close to the native meniscus group, showing significantly better cartilage protection than the PS group. Conclusion: This tissue engineering scaffold could greatly strengthen meniscus regeneration and chondroprotection. Compared with traditional cell-based therapies, the meniscus tissue engineering approach with advantages of one-step operation and reduced cost has a promising potential for future clinical and translational studies.
Collapse
|
11
|
Wang G, Man Z, Xin H, Li Y, Wu C, Sun S. Enhanced adhesion and proliferation of bone marrow mesenchymal stem cells on β‑tricalcium phosphate modified by an affinity peptide. Mol Med Rep 2018; 19:375-381. [PMID: 30431109 PMCID: PMC6297790 DOI: 10.3892/mmr.2018.9655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/09/2018] [Indexed: 11/09/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are often used in orthopedic tissue engineering, and bone marrow-derived mesenchymal stem cells (BMSCs) are currently considered the gold standard. One of the most important issues in MSC-based tissue engineering therapy is the low number of MSCs in pathological tissues. Achieving efficient recruitment of MSCs to defective or damaged tissues in vivo has been a difficult hurdle. The aim of the present study was to construct a biomaterial that can effectively recruit BMSCs to facilitate the repair of pathological tissues. So functional β-tricalcium phosphate (β-TCP) was synthesized using the BMSC affinity peptide DPIYALSWSGMA (DPI) adsorbed onto β-TCP through an adsorption/freeze-drying strategy. C57BL/6 mouse-derived BMSCs were seeded onto the DPI peptide-modified β-TCP (β-TCP-DPI); in vitro experiments demonstrated that β-TCP-DPI enhanced BMSC adhesion and proliferation compared with unmodified β-TCP. Results from the present study indicated that functional β-TCP may be used as an ideal scaffold in tissue engineering and in regenerative medicine.
Collapse
Affiliation(s)
- Guozong Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Hua Xin
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 222000, P.R. China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Changshun Wu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
12
|
Wang G, Man Z, Zhang N, Xin H, Li Y, Sun T, Sun S. Biopanning of mouse bone marrow mesenchymal stem cell affinity for cyclic peptides. Mol Med Rep 2018; 19:407-413. [PMID: 30431079 PMCID: PMC6297745 DOI: 10.3892/mmr.2018.9626] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 10/16/2018] [Indexed: 12/13/2022] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a refractory disease present worldwide. In the development of therapies for this disease, mesenchymal stem cells (MSC) are a promising candidate cell source in tissue engineering (TE) and regenerative medicine. MSCs harvested from bone marrow (BM) are the gold standard. A significant barrier for BMMSC-based therapies is the inability and decreased number of BMMSCs in the tissues of interest. The ability to recruit BMMSCs efficiently to defective or injured sites in tissues or organs, for example the necrotic area of the femoral head in vivo, has been a major concern. In the present study, a peptide sequence (CDNVAQSVC), termed D7, was identified through phage display technology using C57BL/6 mouse BMMSCs. Subsequent analysis suggested that the identified loop-constrained heptapeptide exhibited a high specific affinity for mouse BMMSCs. Due to this specific affinity for BMMSCs, the present study provides a selective method to improve MSC-based TE strategies for the treatment of ONFH.
Collapse
Affiliation(s)
- Guozong Wang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Zhentao Man
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Nianping Zhang
- The Teaching and Research Section of Surgery, The First Clinical College of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Hua Xin
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 222000, P.R. China
| | - Yi Li
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Tiantong Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
13
|
Xie X, Zhu J, Hu X, Dai L, Fu X, Zhang J, Duan X, Ao Y. A co-culture system of rat synovial stem cells and meniscus cells promotes cell proliferation and differentiation as compared to mono-culture. Sci Rep 2018; 8:7693. [PMID: 29769537 PMCID: PMC5955983 DOI: 10.1038/s41598-018-25709-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 03/09/2018] [Indexed: 12/25/2022] Open
Abstract
A meniscus tear often happens during active sports. It needs to be repaired or replaced surgically to avoid further damage to the articular cartilage. To address the shortage of autologous meniscal cells, we designed a co-culture system of synovial stem cells (SMSCs) and meniscal cells (MCs) to produce a large cell number and to maintain characteristics of MCs. Different ratios of SMSCs and MCs at 3:1, 1:1, and 1:3 were tested. Mono-culture of SMSCs or MCs served as control groups. Proliferation and differentiation abilities were compared. The expression of extracellular matrix (ECM) genes in MCs was assessed using an ECM array to reveal the mechanism at the gene level. The co-culture system of SMSCs/MCs at the ratio of 1:3 showed better results than the control groups or those at other ratios. This co-culture system may be a promising strategy for meniscus repair with tissue engineering.
Collapse
Affiliation(s)
- Xing Xie
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Jingxian Zhu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Xiaoqing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Linghui Dai
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Xin Fu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Jiying Zhang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Xiaoning Duan
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China
| | - Yingfang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injury, Third Hospital of Peking University, Beijing, China.
| |
Collapse
|
14
|
Sánchez-Téllez DA, Téllez-Jurado L, Rodríguez-Lorenzo LM. Hydrogels for Cartilage Regeneration, from Polysaccharides to Hybrids. Polymers (Basel) 2017; 9:E671. [PMID: 30965974 PMCID: PMC6418920 DOI: 10.3390/polym9120671] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/24/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022] Open
Abstract
The aims of this paper are: (1) to review the current state of the art in the field of cartilage substitution and regeneration; (2) to examine the patented biomaterials being used in preclinical and clinical stages; (3) to explore the potential of polymeric hydrogels for these applications and the reasons that hinder their clinical success. The studies about hydrogels used as potential biomaterials selected for this review are divided into the two major trends in tissue engineering: (1) the use of cell-free biomaterials; and (2) the use of cell seeded biomaterials. Preparation techniques and resulting hydrogel properties are also reviewed. More recent proposals, based on the combination of different polymers and the hybridization process to improve the properties of these materials, are also reviewed. The combination of elements such as scaffolds (cellular solids), matrices (hydrogel-based), growth factors and mechanical stimuli is needed to optimize properties of the required materials in order to facilitate tissue formation, cartilage regeneration and final clinical application. Polymer combinations and hybrids are the most promising materials for this application. Hybrid scaffolds may maximize cell growth and local tissue integration by forming cartilage-like tissue with biomimetic features.
Collapse
Affiliation(s)
- Daniela Anahí Sánchez-Téllez
- Instituto Politécnico Nacional-ESIQIE, Depto. Ing. en Metalurgia y Materiales, UPALM-Zacatenco, Mexico City 07738, Mexico.
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain.
| | - Lucía Téllez-Jurado
- Instituto Politécnico Nacional-ESIQIE, Depto. Ing. en Metalurgia y Materiales, UPALM-Zacatenco, Mexico City 07738, Mexico.
| | - Luís María Rodríguez-Lorenzo
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine, Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Av. Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029 Madrid, Spain.
- Department Polymeric Nanomaterials and Biomaterials, ICTP-CSIC, Juan de la Cierva 3, 28006 Madrid, Spain.
| |
Collapse
|
15
|
Kim HD, Amirthalingam S, Kim SL, Lee SS, Rangasamy J, Hwang NS. Biomimetic Materials and Fabrication Approaches for Bone Tissue Engineering. Adv Healthc Mater 2017; 6. [PMID: 29171714 DOI: 10.1002/adhm.201700612] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/09/2017] [Indexed: 01/14/2023]
Abstract
Various strategies have been explored to overcome critically sized bone defects via bone tissue engineering approaches that incorporate biomimetic scaffolds. Biomimetic scaffolds may provide a novel platform for phenotypically stable tissue formation and stem cell differentiation. In recent years, osteoinductive and inorganic biomimetic scaffold materials have been optimized to offer an osteo-friendly microenvironment for the osteogenic commitment of stem cells. Furthermore, scaffold structures with a microarchitecture design similar to native bone tissue are necessary for successful bone tissue regeneration. For this reason, various methods for fabricating 3D porous structures have been developed. Innovative techniques, such as 3D printing methods, are currently being utilized for optimal host stem cell infiltration, vascularization, nutrient transfer, and stem cell differentiation. In this progress report, biomimetic materials and fabrication approaches that are currently being utilized for biomimetic scaffold design are reviewed.
Collapse
Affiliation(s)
- Hwan D. Kim
- School of Chemical and Biological Engineering; The Institute of Chemical Processes; Seoul National University; Seoul 151-742 Republic of Korea
| | | | - Seunghyun L. Kim
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Seunghun S. Lee
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
| | - Jayakumar Rangasamy
- Centre for Nanosciences and Molecular Medicine; Amrita University; Kochi 682041 India
| | - Nathaniel S. Hwang
- School of Chemical and Biological Engineering; The Institute of Chemical Processes; Seoul National University; Seoul 151-742 Republic of Korea
- Interdisciplinary Program in Bioengineering; Seoul National University; Seoul 151-742 Republic of Korea
- The BioMax Institute of Seoul National University; Seoul 151-742 Republic of Korea
| |
Collapse
|
16
|
Yun X, Wang Y, Li M, Jin Y, Han Y, Dong T. Application of permselective poly(ε-caprolactone) film for equilibrium-modified atmosphere packaging of strawberry in cold storage. J FOOD PROCESS PRES 2017. [DOI: 10.1111/jfpp.13247] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xueyan Yun
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
| | - Yu Wang
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
| | - Mengting Li
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
- Inner Mongolia Yili Industrial Group Co, Ltd Liquid Milk Business Unit, No. 8 Jinshan Road. Jinshan Development Zone; Hohhot Inner Mongolia 010110 China
| | - Ye Jin
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
| | - Yumei Han
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
| | - Tungalag Dong
- College of Food Science and Engineering; Inner Mongolia Agricultural University, 306 Zhaowuda Road; Hohhot Inner Mongolia 010018 China
| |
Collapse
|
17
|
Wang PY, Thissen H, Kingshott P. Modulation of human multipotent and pluripotent stem cells using surface nanotopographies and surface-immobilised bioactive signals: A review. Acta Biomater 2016; 45:31-59. [PMID: 27596488 DOI: 10.1016/j.actbio.2016.08.054] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/30/2016] [Accepted: 08/30/2016] [Indexed: 02/08/2023]
Abstract
The ability to control the interactions of stem cells with synthetic surfaces is proving to be effective and essential for the quality of passaged stem cells and ultimately the success of regenerative medicine. The stem cell niche is crucial for stem cell self-renewal and differentiation. Thus, mimicking the stem cell niche, and here in particular the extracellular matrix (ECM), in vitro is an important goal for the expansion of stem cells and their applications. Here, surface nanotopographies and surface-immobilised biosignals have been identified as major factors that control stem cell responses. The development of tailored surfaces having an optimum nanotopography and displaying suitable biosignals is proposed to be essential for future stem cell culture, cell therapy and regenerative medicine applications. While early research in the field has been restricted by the limited availability of micro- and nanofabrication techniques, new approaches involving the use of advanced fabrication and surface immobilisation methods are starting to emerge. In addition, new cell types such as induced pluripotent stem cells (iPSCs) have become available in the last decade, but have not been fully understood. This review summarises significant advances in the area and focuses on the approaches that are aimed at controlling the behavior of human stem cells including maintenance of their self-renewal ability and improvement of their lineage commitment using nanotopographies and biosignals. More specifically, we discuss developments in biointerface science that are an important driving force for new biomedical materials and advances in bioengineering aiming at improving stem cell culture protocols and 3D scaffolds for clinical applications. Cellular responses revolve around the interplay between the surface properties of the cell culture substrate and the biomolecular composition of the cell culture medium. Determination of the precise role played by each factor, as well as the synergistic effects amongst the factors, all of which influence stem cell responses is essential for future developments. This review provides an overview of the current state-of-the-art in the design of complex material surfaces aimed at being the next generation of tools tailored for applications in cell culture and regenerative medicine. STATEMENT OF SIGNIFICANCE This review focuses on the effect of surface nanotopographies and surface-bound biosignals on human stem cells. Recently, stem cell research attracts much attention especially the induced pluripotent stem cells (iPSCs) and direct lineage reprogramming. The fast advance of stem cell research benefits disease treatment and cell therapy. On the other hand, surface property of cell adhered materials has been demonstrated very important for in vitro cell culture and regenerative medicine. Modulation of cell behavior using surfaces is costeffective and more defined. Thus, we summarise the recent progress of modulation of human stem cells using surface science. We believe that this review will capture a broad audience interested in topographical and chemical patterning aimed at understanding complex cellular responses to biomaterials.
Collapse
|
18
|
Li M, Ke QF, Tao SC, Guo SC, Rui BY, Guo YP. Fabrication of hydroxyapatite/chitosan composite hydrogels loaded with exosomes derived from miR-126-3p overexpressed synovial mesenchymal stem cells for diabetic chronic wound healing. J Mater Chem B 2016; 4:6830-6841. [PMID: 32263577 DOI: 10.1039/c6tb01560c] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The exploration of an effective diabetic chronic wound healing process still remains a great challenge. Herein, we used gene overexpression technology to obtain synovial mesenchymal stem cells (SMSCs) and the miR-126-3p highly expressed SMSCs (SMSCs-126). The exosomes derived from miR-126-3p overexpressed SMSCs (SMSCs-126-Exos) with a particle size of 85 nm were encapsulated in hydroxyapatite/chitosan (HAP-CS) composite hydrogels (HAP-CS-SMSCs-126-Exos) as wound dressings. The SMSCs-126-Exos, CS and low-crystallinity HAP nanorods with a length of 200 nm and a diameter of 50 nm are uniformly dispersed within the whole composite hydrogel. The HAP-CS-SMSCs-126-Exos possess the controlled release property of SMSCs-126-Exos for at least 6 days. The released SMSCs-126-Exos nanoparticles stimulate the proliferation and migration of human dermal fibroblasts and human dermal microvascular endothelial cells (HMEC-1). At the same time, the migration and capillary-network formation of HMEC-1 are promoted through the activation of MAPK/ERK and PI3K/AKT. In vivo tests demonstrate that the HAP-CS-SMSCs-126-Exos successfully promote wound surface re-epithelialization, accelerate angiogenesis, and expedite collagen maturity due to the presence of HAP, CS and SMSCs-126-Exos. Therefore, the HAP-CS-SMSCs-126-Exos possess great potential application for diabetic chronic wound healing, and especially provide the possibility of using exosomes derived from modified cells as a new approach to bring wonderful functionality and controllability in future chronic wound therapy.
Collapse
Affiliation(s)
- Min Li
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China.
| | | | | | | | | | | |
Collapse
|
19
|
Dadaci M, Karagülle N, Sönmez E, Dadaci Z, İşci ET, İnce B, Vargel İ, Pişkin E, Erk AY. Evaluation of the effectiveness of biodegradable electrospun caprolactoneand poly(lactic acid-ε-caprolactone) nerve conduits for peripheral nerveregenerations in a rat sciatic nerve defect model. Turk J Med Sci 2016; 46:539-48. [PMID: 27511522 DOI: 10.3906/sag-1412-110] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 07/02/2015] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND/AIM The aim of this study was to compare electrospun caprolactone (EC) and poly(lactic acid-ε-caprolactone) (PLCL) nerve conduits with nerve graft in a rat sciatic nerve defect model. MATERIALS AND METHODS A total of 32 male Wistar albino rats were divided into 4 groups, with 8 rats in each group. A nerve defect of 1 cm was constructed in the left sciatic nerve of the rats. These defects were left denuded in the sham group, and reconstructed with nerve grafts, PLCL, and EC nerve conduits in the other groups. After 3 months, nerve regenerations were evaluated macroscopically, microscopically, and electrophysiologically. The numbers of myelinated axons in the cross-sections of the nerves were compared between the groups. RESULTS Macroscopically, all nerve coaptations were intact and biodegradation was detected in nerve conduits. Electromyographic assessment and count of myelinated axons in the cross-sections of the nerves displayed the best regeneration in the nerve graft group (P < 0.001) and similar results were obtained in the PLCL and EC nerve conduit groups (P = 0.79). Light and electron microscopy studies demonstrated nerve regeneration in both nerve conduit groups. CONCLUSION EC nerve conduits and PLCL nerve conduits yielded similar results and may be alternatives to nerve grafts as they biodegrade.
Collapse
Affiliation(s)
- Mehmet Dadaci
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - Nimet Karagülle
- Department of Chemical Engineering, Faculty of Engineering, Mersin University, Mersin, Turkey
| | - Erhan Sönmez
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, İzmir Katip Çelebi University, İzmir, Turkey
| | - Zeynep Dadaci
- Department of Ophthalmology, Faculty of Medicine, Mevlana University, Konya, Turkey
| | - Evren Tevfik İşci
- Reconstructive and Aesthetic Surgery, Clinic of Plastic, Acıbadem Family Hospital, İstanbul, Turkey
| | - Bilsev İnce
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | - İbrahim Vargel
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Erhan Pişkin
- Department of Chemical Engineering, Faculty of Engineering, Hacettepe University, Ankara, Turkey
| | - Ali Yücel Erk
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
20
|
Rambhia KJ, Ma PX. Controlled drug release for tissue engineering. J Control Release 2015; 219:119-128. [PMID: 26325405 DOI: 10.1016/j.jconrel.2015.08.049] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 08/23/2015] [Accepted: 08/25/2015] [Indexed: 11/19/2022]
Abstract
Tissue engineering is often referred to as a three-pronged discipline, with each prong corresponding to 1) a 3D material matrix (scaffold), 2) drugs that act on molecular signaling, and 3) regenerative living cells. Herein we focus on reviewing advances in controlled release of drugs from tissue engineering platforms. This review addresses advances in hydrogels and porous scaffolds that are synthesized from natural materials and synthetic polymers for the purposes of controlled release in tissue engineering. We pay special attention to efforts to reduce the burst release effect and to provide sustained and long-term release. Finally, novel approaches to controlled release are described, including devices that allow for pulsatile and sequential delivery. In addition to recent advances, limitations of current approaches and areas of further research are discussed.
Collapse
Affiliation(s)
- Kunal J Rambhia
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
21
|
Makris EA, Gomoll AH, Malizos KN, Hu JC, Athanasiou KA. Repair and tissue engineering techniques for articular cartilage. Nat Rev Rheumatol 2015; 11:21-34. [PMID: 25247412 PMCID: PMC4629810 DOI: 10.1038/nrrheum.2014.157] [Citation(s) in RCA: 850] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Chondral and osteochondral lesions due to injury or other pathology commonly result in the development of osteoarthritis, eventually leading to progressive total joint destruction. Although current progress suggests that biologic agents can delay the advancement of deterioration, such drugs are incapable of promoting tissue restoration. The limited ability of articular cartilage to regenerate renders joint arthroplasty an unavoidable surgical intervention. This Review describes current, widely used clinical repair techniques for resurfacing articular cartilage defects; short-term and long-term clinical outcomes of these techniques are discussed. Also reviewed is a developmental pipeline of acellular and cellular regenerative products and techniques that could revolutionize joint care over the next decade by promoting the development of functional articular cartilage. Acellular products typically consist of collagen or hyaluronic-acid-based materials, whereas cellular techniques use either primary cells or stem cells, with or without scaffolds. Central to these efforts is the prominent role that tissue engineering has in translating biological technology into clinical products; therefore, concomitant regulatory processes are also discussed.
Collapse
Affiliation(s)
- Eleftherios A Makris
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Andreas H Gomoll
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Konstantinos N Malizos
- Department of Orthopaedic Surgery and Musculoskeletal Trauma, University of Thessaly, Biopolis, Larisa 41110, Greece
| | - Jerry C Hu
- Department of Biomedical Engineering, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kyriacos A Athanasiou
- Department of Orthopaedic Surgery, University of California Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|