Home

Content of REVIEW in our journal

        Published in last 1 year |  In last 2 years |  In last 3 years |  All
    Please wait a minute...
    For Selected: Toggle Thumbnails
    Cellular modulation by the mechanical cues from biomaterials for tissue engineering
    Qiang Wei, Shenghao Wang, Feng Han, Huan Wang, Weidong Zhang, Qifan Yu, Changjiang Liu, Luguang Ding, Jiayuan Wang, Lili Yu, Caihong Zhu, Bin Li
    Biomaterials Translational    2021, 2 (4): 323-342.   DOI: 10.12336/biomatertransl.2021.04.001
    Abstract946)   HTML91)    PDF(pc) (77197KB)(1523)       Save

    Mechanical cues from the extracellular matrix (ECM) microenvironment are known to be significant in modulating the fate of stem cells to guide developmental processes and maintain bodily homeostasis. Tissue engineering has provided a promising approach to the repair or regeneration of damaged tissues. Scaffolds are fundamental in cell-based regenerative therapies. Developing artificial ECM that mimics the mechanical properties of native ECM would greatly help to guide cell functions and thus promote tissue regeneration. In this review, we introduce various mechanical cues provided by the ECM including elasticity, viscoelasticity, topography, and external stimuli, and their effects on cell behaviours. Meanwhile, we discuss the underlying principles and strategies to develop natural or synthetic biomaterials with different mechanical properties for cellular modulation, and explore the mechanism by which the mechanical cues from biomaterials regulate cell function toward tissue regeneration. We also discuss the challenges in multimodal mechanical modulation of cell behaviours and the interplay between mechanical cues and other microenvironmental factors.

    Table and Figures | Reference | Related Articles | Metrics
    History, progress and future challenges of artificial blood vessels: a narrative review
    Ke Hu, Yuxuan Li, Zunxiang Ke, Hongjun Yang, Chanjun Lu, Yiqing Li, Yi Guo, Weici Wang
    Biomaterials Translational    2022, 3 (1): 81-98.   DOI: 10.12336/biomatertransl.2022.01.008
    Abstract783)   HTML95)    PDF(pc) (2405KB)(1472)       Save

    Cardiovascular disease serves as the leading cause of death worldwide, with stenosis, occlusion, or severe dysfunction of blood vessels being its pathophysiological mechanism. Vascular replacement is the preferred surgical option for treating obstructed vascular structures. Due to the limited availability of healthy autologous vessels as well as the incidence of postoperative complications, there is an increasing demand for artificial blood vessels. From synthetic to natural, or a mixture of these components, numerous materials have been used to prepare artificial vascular grafts. Although synthetic grafts are more appropriate for use in medium to large-diameter vessels, they fail when replacing small-diameter vessels. Tissue-engineered vascular grafts are very likely to be an ideal alternative to autologous grafts in small-diameter vessels and are worthy of further investigation. However, a multitude of problems remain that must be resolved before they can be used in biomedical applications. Accordingly, this review attempts to describe these problems and provide a discussion of the generation of artificial blood vessels. In addition, we deliberate on current state-of-the-art technologies for creating artificial blood vessels, including advances in materials, fabrication techniques, various methods of surface modification, as well as preclinical and clinical applications. Furthermore, the evaluation of grafts both in vivo and in vitro, mechanical properties, challenges, and directions for further research are also discussed.

    Table and Figures | Reference | Related Articles | Metrics
    Proper animal experimental designs for preclinical research of biomaterials for intervertebral disc regeneration
    Yizhong Peng, Xiangcheng Qing, Hongyang Shu, Shuo Tian, Wenbo Yang, Songfeng Chen, Hui Lin, Xiao Lv, Lei Zhao, Xi Chen, Feifei Pu, Donghua Huang, Xu Cao, Zengwu Shao
    Biomaterials Translational    2021, 2 (2): 91-142.   DOI: 10.12336/biomatertransl.2021.02.003
    Abstract733)   HTML68)    PDF(pc) (3328KB)(2215)       Save

    Low back pain is a vital musculoskeletal disease that impairs life quality, leads to disability and imposes heavy economic burden on the society, while it is greatly attributed to intervertebral disc degeneration (IDD). However, the existing treatments, such as medicines, chiropractic adjustments and surgery, cannot achieve ideal disc regeneration. Therefore, advanced bioactive therapies are implemented, including stem cells delivery, bioreagents administration, and implantation of biomaterials etc. Among these researches, few reported unsatisfying regenerative outcomes. However, these advanced therapies have barely achieved successful clinical translation. The main reason for the inconsistency between satisfying preclinical results and poor clinical translation may largely rely on the animal models that cannot actually simulate the human disc degeneration. The inappropriate animal model also leads to difficulties in comparing the efficacies among biomaterials in different reaches. Therefore, animal models that better simulate the clinical charateristics of human IDD should be acknowledged. In addition, in vivo regenerative outcomes should be carefully evaluated to obtain robust results. Nevertheless, many researches neglect certain critical characteristics, such as adhesive properties for biomaterials blocking annulus fibrosus defects and hyperalgesia that is closely related to the clinical manifestations, e.g., low back pain. Herein, in this review, we summarized the animal models established for IDD, and highlighted the proper models and parameters that may result in acknowledged IDD models. Then, we discussed the existing biomaterials for disc regeneration and the characteristics that should be considered for regenerating different parts of discs. Finally, well-established assays and parameters for in vivo disc regeneration are explored.

    Table and Figures | Reference | Related Articles | Metrics
    Biodegradable magnesium alloys for orthopaedic applications
    Yu Lu, Subodh Deshmukh, Ian Jones, Yu-Lung Chiu
    Biomaterials Translational    2021, 2 (3): 214-235.   DOI: 10.12336/biomatertransl.2021.03.005
    Abstract659)   HTML68)    PDF(pc) (65304KB)(1384)       Save

    There is increasing interest in the development of bone repair materials for biomedical applications. Magnesium (Mg)-based alloys have a natural ability to biodegrade because they corrode in aqueous media; they are thus promising materials for orthopaedic device applications in that the need for a secondary surgical operation to remove the implant can be eliminated. Notably, Mg has superior biocompatibility because Mg is found in the human body in abundance. Moreover, Mg alloys have a low elastic modulus, close to that of natural bone, which limits stress shielding. However, there are still some challenges for Mg-based fracture fixation. The degradation of Mg alloys in biological fluids can be too rapid, resulting in a loss of mechanical integrity before complete healing of the bone fracture. In order to achieve an appropriate combination of bio-corrosion and mechanical performance, the microstructure needs to be tailored properly by appropriate alloy design, as well as the use of strengthening processes and manufacturing techniques. This review covers the evolution, current strategies and future perspectives of Mg-based orthopaedic implants.

    Table and Figures | Reference | Related Articles | Metrics
    Additive manufactured polyether-ether-ketone implants for orthopaedic applications: a narrative review
    Changning Sun, Jianfeng Kang, Chuncheng Yang, Jibao Zheng, Yanwen Su, Enchun Dong, Yingjie Liu, Siqi Yao, Changquan Shi, Huanhao Pang, Jiankang He, Ling Wang, Chaozong Liu, Jianhua Peng, Liang Liu, Yong Jiang, Dichen Li
    Biomaterials Translational    2022, 3 (2): 116-133.   DOI: 10.12336/biomatertransl.2022.02.001
    Abstract635)   HTML45)    PDF(pc) (5186KB)(1071)       Save

    Polyether-ether-ketone (PEEK) is believed to be the next-generation biomedical material for orthopaedic implants that may replace metal materials because of its good biocompatibility, appropriate mechanical properties and radiolucency. Currently, some PEEK implants have been used successfully for many years. However, there is no customised PEEK orthopaedic implant made by additive manufacturing licensed for the market, although clinical trials have been increasingly reported. In this review article, design criteria, including geometric matching, functional restoration, strength safety, early fixation, long-term stability and manufacturing capability, are summarised, focusing on the clinical requirements. An integrated framework of design and manufacturing processes to create customised PEEK implants is presented, and several typical clinical applications such as cranioplasty patches, rib prostheses, mandibular prostheses, scapula prostheses and femoral prostheses are described. The main technical challenge faced by PEEK orthopaedic implants lies in the poor bonding with bone and soft tissue due to its biological inertness, which may be solved by adding bioactive fillers and manufacturing porous architecture. The lack of technical standards is also one of the major factors preventing additive-manufactured customised PEEK orthopaedic implants from clinical translation, and it is good to see that the abundance of standards in the field of additive-manufactured medical devices is helping them enter the clinical market.

    Table and Figures | Reference | Related Articles | Metrics
    Magnesium-based materials in orthopaedics: material properties and animal models
    Xirui Jing, Qiuyue Ding, Qinxue Wu, Weijie Su, Keda Yu, Yanlin Su, Bing Ye, Qing Gao, Tingfang Sun, Xiaodong Guo
    Biomaterials Translational    2021, 2 (3): 197-213.   DOI: 10.12336/biomatertransl.2021.03.004
    Abstract489)   HTML60)    PDF(pc) (49069KB)(964)       Save

    As a new generation of medical metal materials, degradable magnesium-based materials have excellent mechanical properties and osteogenic promoting ability, making them promising materials for the treatment of refractory bone diseases. Animal models can be used to understand and evaluate the performance of materials in complex physiological environments, providing relevant data for preclinical evaluation of implants and laying the foundation for subsequent clinical studies. To date, many researchers have studied the biocompatibility, degradability and osteogenesis of magnesium-based materials, but there is a lack of review regarding the effects of magnesium-based materials in vivo. In view of the growing interest in these materials, this review briefly describes the properties of magnesium-based materials and focuses on the safety and efficacy of magnesium-based materials in vivo. Various animal models including rats, rabbits, dogs and pigs are covered to better understand and evaluate the progress and future of magnesium-based materials. This literature analysis reveals that the magnesium-based materials have good biocompatibility and osteogenic activity, thus causing no adverse reaction around the implants in vivo, and that they exhibit a beneficial effect in the process of bone repair. In addition, the degradation rate in vivo can also be improved by means of alloying and coating. These encouraging results show a promising future for the use of magnesium-based materials in musculoskeletal disorders.

    Table and Figures | Reference | Related Articles | Metrics
    Three-dimensional bio-printing of decellularized extracellular matrix-based bio-inks for cartilage regeneration: a systematic review
    Melika Sahranavard, Soulmaz Sarkari, SeyedehMina Safavi, Farnaz Ghorbani
    Biomaterials Translational    2022, 3 (2): 105-115.   DOI: 10.12336/biomatertransl.2022.02.004
    Abstract468)   HTML43)    PDF(pc) (1305KB)(816)       Save

    Cartilage injuries are common problems that increase with the population aging. Cartilage is an avascular tissue with a relatively low level of cellular mitotic activity, which makes it impossible to heal spontaneously. To compensate for this problem, three-dimensional bio-printing has attracted a great deal of attention in cartilage tissue engineering. This emerging technology aims to create three-dimensional functional scaffolds by accurately depositing layer-by-layer bio-inks composed of biomaterial and cells. As a novel bio-ink, a decellularized extracellular matrix can serve as an appropriate substrate that contains all the necessary biological cues for cellular interactions. Here, this review is intended to provide an overview of decellularized extracellular matrix-based bio-inks and their properties, sources, and preparation process. Following this, decellularized extracellular matrix-based bio-inks for cartilage tissue engineering are discussed, emphasizing cell behavior and in-vivo applications. Afterward, the current challenges and future outlook will be discussed to determine the conclusing remarks.

    Table and Figures | Reference | Related Articles | Metrics
    Update on the research and development of magnesium-based biodegradable implants and their clinical translation in orthopaedics
    Ying Luo, Jue Wang, Michael Tim Yun Ong, Patrick Shu-hang Yung, Jiali Wang, Ling Qin
    Biomaterials Translational    2021, 2 (3): 188-196.   DOI: 10.12336/biomatertransl.2021.03.003
    Abstract466)   HTML56)    PDF(pc) (375858KB)(1207)       Save

    Biodegradable magnesium (Mg) or its alloys are desirable materials for development into new-generation internal fixation devices or implants with high biocompatibility, adequate mechanical modulus, and osteopromotive properties, which may overcome some of the drawbacks of the existing permanent orthopaedic implants with regard to stress-shielding of bone and beam-hardening effects on radiographic images. This review summarises the current research status of Mg-based orthopaedic implants in animals and clinical trials. First, detailed information of animal studies including bone fracture repair and anterior cruciate ligament reconstruction with the use of Mg-based orthopaedic devices is introduced. Second, the repair mechanisms of the Mg-based orthopaedic implants are also reviewed. Afterwards, reports of recent clinical cases treated using Mg-based implants in orthopaedics are summarised. Finally, the challenges and the strategies of the use of Mg-based orthopaedic implants are discussed. Taken together, the collected efforts in basic research, translational work, and clinical applications of Mg-based orthopaedic implants over the last decades greatly contribute to the development of a new generation of biodegradable metals used for the design of innovative implants for better treatment of orthopaedic conditions in patients with challenging skeletal disorders or injuries.

    Table and Figures | Reference | Related Articles | Metrics
    Research and development strategy for biodegradable magnesium-based vascular stents: a review
    Jialin Niu, Hua Huang, Jia Pei, Zhaohui Jin, Shaokang Guan, Guangyin Yuan
    Biomaterials Translational    2021, 2 (3): 236-247.   DOI: 10.12336/biomatertransl.2021.03.06
    Abstract416)   HTML50)    PDF(pc) (36878KB)(819)       Save

    Magnesium alloys are an ideal material for biodegradable vascular stents, which can be completely absorbed in the human body, and have good biosafety and mechanical properties. However, the rapid corrosion rate and excessive localized corrosion, as well as challenges in the preparation and processing of microtubes for stents, are restricting the clinical application of magnesium-based vascular stents. In the present work we will give an overview of the recent progresses on biodegradable magnesium based vascular stents including magnesium alloy design, high-precision microtubes processing, stent shape optimisation and functional coating preparation. In particular, the Triune Principle in biodegradable magnesium alloy design is proposed based on our research experience, which requires three key aspects to be considered when designing new biodegradable magnesium alloys for vascular stents application, i.e. biocompatibility and biosafety, mechanical properties, and biodegradation. This review hopes to inspire the future studies on the design and development of biodegradable magnesium alloy-based vascular stents.

    Table and Figures | Reference | Related Articles | Metrics
    Biofunctional magnesium coating of implant materials by physical vapour deposition
    Qingchuan Wang, Weidan Wang, Yanfang Li, Weirong Li, Lili Tan, Ke Yang
    Biomaterials Translational    2021, 2 (3): 248-256.   DOI: 10.12336/biomatertransl.2021.03.007
    Abstract404)   HTML56)    PDF(pc) (304378KB)(1195)       Save

    The lack of bioactivity of conventional medical materials leads to low osseointegration ability that may result in the occurrence of aseptic loosening in the clinic. To achieve high osseointegration, surface modifications with multiple biofunctions including degradability, osteogenesis, angiogenesis and antibacterial properties are required. However, the functions of conventional bioactive coatings are limited. Thus novel biofunctional magnesium (Mg) coatings are believed to be promising candidates for surface modification of implant materials for use in bone tissue repair. By physical vapour deposition, many previous researchers have deposited Mg coatings with high purity and granular microstructure on titanium alloys, polyetheretherketone, steels, Mg alloys and silicon. It was found that the Mg coatings with high-purity could considerably control the degradation rate in the initial stage of Mg alloy implantation, which is the most important problem for the application of Mg alloy implants. In addition, Mg coating on titanium (Ti) implant materials has been extensively studied both in vitro and in vivo, and the results indicated that their corrosion behaviour and biocompatibility are promising. Mg coatings continuously release Mg ions during the degradation process, and the alkaline environment caused by Mg degradation has obvious antibacterial effects. Meanwhile, the Mg coating has beneficial effects on osteogenesis and osseointegration, and increases the new bone-regenerating ability. Mg coatings also exhibit favourable osteogenic and angiogenic properties in vitro and increased long-term bone formation and early vascularization in vivo. Inhibitory effects of Mg coatings on osteoclasts have also been proven, which play a great role in osteoporotic patients. In addition, in order to obtain more biofunctions, other alloying elements such as copper have been added to the Mg coatings. Thus, Mg-coated Ti acquired biofunctions including degradability, osteogenesis, angiogenesis and antibacterial properties. These novel multi-functional Mg coatings are expected to significantly enhance the long-term safety of bone implants for the benefit of patients. This paper gives a brief review of studies of the microstructure, degradation behaviours and biofunctions of Mg coatings, and directions for future research are also proposed.

    Table and Figures | Reference | Related Articles | Metrics
    In silico modelling of the corrosion of biodegradable magnesium-based biomaterials: modelling approaches, validation and future perspectives
    Aditya Joshi, George Dias, Mark P. Staiger
    Biomaterials Translational    2021, 2 (3): 257-271.   DOI: 10.12336/biomatertransl.2021.03.008
    Abstract399)   HTML44)    PDF(pc) (34906KB)(804)       Save

    Metallic biomedical implants based on magnesium, zinc and iron alloys have emerged as bioresorbable alternatives to permanent orthopaedic implants over the last two decades. The corrosion rate of biodegradable metals plays a critical role in controlling the compatibility and functionality of the device in vivo. The broader adoption of biodegradable metals in orthopaedic applications depends on developing in vitro methods that accurately predict the biodegradation behaviour in vivo. However, the physiological environment is a highly complex corrosion environment to replicate in the laboratory, making the in vitro-to-in vivo translation of results very challenging. Accordingly, the results from in vitro corrosion tests fail to provide a complete schema of the biodegradation behaviour of the metal in vivo. In silico approach based on computer simulations aim to bridge the observed differences between experiments performed in vitro and vivo. A critical review of the state-of-the-art of computational modelling techniques for predicting the corrosion behaviour of magnesium alloy as a biodegradable metal is presented.

    Table and Figures | Reference | Related Articles | Metrics
    Advances and perspective on the translational medicine of biodegradable metals
    Hongtao Yang, Wenjiao Lin, Yufeng Zheng
    Biomaterials Translational    2021, 2 (3): 177-187.   DOI: 10.12336/biomatertransl.2021.03.002
    Abstract365)   HTML78)    PDF(pc) (63002KB)(772)       Save

    Biodegradable metals, designed to be safely degraded and absorbed by the body after fulfil the intended functions, are of particular interest in the 21st century. The marriage of advanced biodegradable metals with clinical needs have yield unprecedented possibility. Magnesium, iron, and zinc-based materials constitute the main components of temporary, implantable metallic medical devices. A burgeoning number of studies on biodegradable metals have driven the clinical translation of biodegradable metallic devices in the fields of cardiology and orthopaedics over the last decade. Their ability to degrade as well as their beneficial biological functions elicited during degradation endow this type of material with the potential to shift the paradigm in the treatment of musculoskeletal and cardiovascular diseases. This review provides an insight into the degradation mechanism of these metallic devices in specific application sites and introduces state-of-the-art translational research in the field of biodegradable metals, as well as highlighting some challenges for materials design strategies in the context of mechanical and biological compatibility.

    Table and Figures | Reference | Related Articles | Metrics
    Experimental and computational models for tissue-engineered heart valves: a narrative review
    Ge Yan, Yuqi Liu, Minghui Xie, Jiawei Shi, Weihua Qiao, Nianguo Dong
    Biomaterials Translational    2021, 2 (4): 361-375.   DOI: 10.12336/biomatertransl.2021.04.009
    Abstract324)   HTML37)    PDF(pc) (71332KB)(571)       Save

    Valvular heart disease is currently a common problem which causes high morbidity and mortality worldwide. Prosthetic valve replacements are widely needed to correct narrowing or backflow through the valvular orifice. Compared to mechanical valves and biological valves, tissue-engineered heart valves can be an ideal substitute because they have a low risk of thromboembolism and calcification, and the potential for remodelling, regeneration, and growth. In order to test the performance of these heart valves, various animal models and other models are needed to optimise the structure and function of tissue-engineered heart valves, which may provide a potential mechanism responsible for substantial enhancement in tissue-engineered heart valves. Choosing the appropriate model for evaluating the performance of the tissue-engineered valve is important, as different models have their own advantages and disadvantages. In this review, we summarise the current state-of-the-art animal models, bioreactors, and computational simulation models with the aim of creating more strategies for better development of tissue-engineered heart valves. This review provides an overview of major factors that influence the selection and design of a model for tissue-engineered heart valve. Continued efforts in improving and testing models for valve regeneration remain crucial in basic science and translational researches. Future research should focus on finding the right animal model and developing better in vitro testing systems for tissue-engineered heart valve.

    Table and Figures | Reference | Related Articles | Metrics
    Biological approaches to the repair and regeneration of the rotator cuff tendon-bone enthesis: a literature review
    Ahlam A. Abdalla, Catherine J. Pendegrass
    Biomaterials Translational    2023, 4 (2): 85-103.   DOI: 10.12336/biomatertransl.2023.02.004
    Abstract318)   HTML34)    PDF(pc) (834KB)(408)       Save

    Entheses are highly specialised organs connecting ligaments and tendons to bones, facilitating force transmission, and providing mechanical strengths to absorb forces encountered. Two types of entheses, fibrocartilaginous and fibrous, exist in interfaces. The gradual fibrocartilaginous type is in rotator cuff tendons and is more frequently injured due to the poor healing capacity that leads to loss of the original structural and biomechanical properties and is attributed to the high prevalence of retears. Fluctuating methodologies and outcomes of biological approaches are challenges to overcome for them to be routinely used in clinics. Therefore, stratifying the existing literature according to different categories (chronicity, extent of tear, and studied population) would effectively guide repair approaches. This literature review supports tissue engineering approaches to promote rotator cuff enthesis healing employing cells, growth factors, and scaffolds period. Outcomes suggest its promising role in animal studies as well as some clinical trials and that combination therapies are more beneficial than individualized ones. It then highlights the importance of tailoring interventions according to the tear extent, chronicity, and the population being treated. Contributing factors such as loading, deficiencies, and lifestyle habits should also be taken into consideration. Optimum results can be achieved if biological, mechanical, and environmental factors are approached. It is challenging to determine whether variations are due to the interventions themselves, the animal models, loading regimen, materials, or tear mechanisms. Future research should focus on tailoring interventions for different categories to formulate protocols, which would best guide regenerative medicine decision making.

    Table and Figures | Reference | Related Articles | Metrics
    Endogenous repair theory enriches construction strategies for orthopaedic biomaterials: a narrative review
    Yizhong Peng, Jinye Li, Hui Lin, Shuo Tian, Sheng Liu, Feifei Pu, Lei Zhao, Kaige Ma, Xiangcheng Qing, Zengwu Shao
    Biomaterials Translational    2021, 2 (4): 343-360.   DOI: 10.12336/biomatertransl.2021.04.008
    Abstract311)   HTML27)    PDF(pc) (28393KB)(757)       Save

    The development of tissue engineering has led to new strategies for mitigating clinical problems; however, the design of the tissue engineering materials remains a challenge. The limited sources and inadequate function, potential risk of microbial or pathogen contamination, and high cost of cell expansion impair the efficacy and limit the application of exogenous cells in tissue engineering. However, endogenous cells in native tissues have been reported to be capable of spontaneous repair of the damaged tissue. These cells exhibit remarkable plasticity, and thus can differentiate or be reprogrammed to alter their phenotype and function after stimulation. After a comprehensive review, we found that the plasticity of these cells plays a major role in establishing the cell source in the mechanism involved in tissue regeneration. Tissue engineering materials that focus on assisting and promoting the natural self-repair function of endogenous cells may break through the limitations of exogenous seed cells and further expand the applications of tissue engineering materials in tissue repair. This review discusses the effects of endogenous cells, especially stem cells, on injured tissue repairing, and highlights the potential utilisation of endogenous repair in orthopaedic biomaterial constructions for bone, cartilage, and intervertebral disc regeneration.

    Table and Figures | Reference | Related Articles | Metrics
    New perspective of skeletal stem cells
    Guixin Yuan, Zan Li, Xixi Lin, Na Li, Ren Xu
    Biomaterials Translational    2022, 3 (4): 280-294.   DOI: 10.12336/biomatertransl.2022.04.007
    Abstract293)   HTML26)    PDF(pc) (7147KB)(505)       Save

    Tissue–resident stem cells are a group of stem cells distinguished by their capacity for self–renewal and multilineage differentiation capability with tissue specificity. Among these tissue–resident stem cells, skeletal stem cells (SSCs) were discovered in the growth plate region through a combination of cell surface markers and lineage tracing series. With the process of unravelling the anatomical variation of SSCs, researchers were also keen to investigate the developmental diversity outside the long bones, including in the sutures, craniofacial sites, and spinal regions. Recently, fluorescence–activated cell sorting, lineage tracing, and single–cell sequencing have been used to map lineage trajectories by studying SSCs with different spatiotemporal distributions. The SSC niche also plays a pivotal role in regulating SSC fate, such as cell–cell interactions mediated by multiple signalling pathways. This review focuses on discussing the spatial and temporal distribution of SSCs, and broadening our understanding of the diversity and plasticity of SSCs by summarizing the progress of research into SSCs in recent years.

    Table and Figures | Reference | Related Articles | Metrics
    Manufacturing artificial bone allografts: a perspective
    Emma Steijvers, Armaan Ghei, Zhidao Xia
    Biomaterials Translational    2022, 3 (1): 65-80.   DOI: 10.12336/biomatertransl.2022.01.007
    Abstract291)   HTML30)    PDF(pc) (1261KB)(808)       Save

    Bone grafts have traditionally come from four sources: the patients’ own tissue (autograft), tissue from a living or cadaveric human donor (allograft), animal donors (xenograft) and synthetic artificial biomaterials (ceramics, cement, polymers, and metal). However, all of these have advantages and drawbacks. The most commercially successful bone grafts so far are allografts, which hold 57% of the current bone graft market; however, disease transmission and scarcity are still significant drawbacks limiting their use. Tissue-engineered grafts have great potential, in which human stem cells and synthetical biomaterials are combined to produce bone-like tissue in vitro, but this is yet to be approved for widespread clinical practice. It is hypothesised that artificial bone allografts can be mass-manufactured to replace conventional bone allografts through refined bone tissue engineering prior to decellularisation. This review article aims to review current literature on (1) conventional bone allograft preparation; (2) bone tissue engineering including the use of synthetic biomaterials as bone graft substitute scaffolds, combined with osteogenic stem cells in vitro; (3) potential artificial allograft manufacturing processes, including mass production of engineered bone tissue, osteogenic enhancement, decellularisation, sterilisation and safety assurance for regulatory approval. From these assessments, a practical route map for mass production of artificial allografts for clinical use is proposed.

    Table and Figures | Reference | Related Articles | Metrics
    Bioactive peptides for anticancer therapies
    Yehao Zhang, Cong Wang, Wenhui Zhang, Xinming Li
    Biomaterials Translational    2023, 4 (1): 5-17.   DOI: 10.12336/biomatertransl.2023.01.003
    Abstract290)   HTML30)    PDF(pc) (858KB)(811)       Save

    Cancer is a serious concern in public health worldwide. Numerous modalities including surgery, radiotherapy, and chemotherapy, have been used for cancer therapies in clinic. Despite progress in anticancer therapies, the usage of these methods for cancer treatment is often related to deleterious side effects and multidrug resistance of conventional anticancer drugs, which have prompted the development of novel therapeutic methods. Anticancer peptides (ACPs), derived from naturally occurring and modified peptides, have received great attention in these years and emerge as novel therapeutic and diagnostic candidates for cancer therapies, because of several advantages over the current treatment modalities. In this review, the classification and properties of ACPs, the mode of action and mechanism of membrane disruption, as well as the natural sources of bioactive peptides with anticancer activities were summarised. Because of their high efficacy for inducing cancer cell death, certain ACPs have been developed to work as drugs and vaccines, evaluated in varied phases of clinical trials. We expect that this summary could facilitate the understanding and design of ACPs with increased specificity and toxicity towards malignant cells and with reduced side effects to normal cells.

    Table and Figures | Reference | Related Articles | Metrics
    Human pluripotent stem cells: tools for regenerative medicine
    Peter W. Andrews
    Biomaterials Translational    2021, 2 (4): 294-300.   DOI: 10.12336/biomatertransl.2021.04.004
    Abstract290)   HTML34)    PDF(pc) (247KB)(528)       Save

    Human embryonic stem cells and induced pluripotent stem cells, together denoted as pluripotent stem cells have opened up unprecedented opportunities for developments in human healthcare over the past 20 years. Although much about the properties and behaviour of these cells required to underpin their applications has been discovered over this time, a number of issues remain. This brief review considers the history of these developments and some of the underlying biology, pointing out some of the problems still to be resolved, particularly in relation to their genetic stability and possible malignancy.

    Reference | Related Articles | Metrics
    Adipose-derived cells: building blocks of three-dimensional microphysiological systems
    Trivia P. Frazier, Katie Hamel, Xiying Wu, Emma Rogers, Haley Lassiter, Jordan Robinson, Omair Mohiuddin, Michael Henderson, Jeffrey M. Gimble
    Biomaterials Translational    2021, 2 (4): 301-306.   DOI: 10.12336/biomatertransl.2021.04.005
    Abstract288)   HTML26)    PDF(pc) (262KB)(690)       Save

    Microphysiological systems (MPS) created with human-derived cells and biomaterial scaffolds offer a potential in vitro alternative to in vivo animal models. The adoption of three-dimensional MPS models has economic, ethical, regulatory, and scientific implications for the fields of regenerative medicine, metabolism/obesity, oncology, and pharmaceutical drug discovery. Key opinion leaders acknowledge that MPS tools are uniquely positioned to aid in the objective to reduce, refine, and eventually replace animal experimentation while improving the accuracy of the finding’s clinical translation. Adipose tissue has proven to be an accessible and available source of human-derived stromal vascular fraction (SVF) cells, a heterogeneous population available at point of care, and adipose-derived stromal/stem cells, a relatively homogeneous population requiring plastic adherence and culture expansion of the SVF cells. The adipose-derived stromal/stem cells or SVF cells, in combination with human tissue or synthetic biomaterial scaffolds, can be maintained for extended culture periods as three-dimensional MPS models under angiogenic, stromal, adipogenic, or osteogenic conditions. This review highlights recent literature relating to the versatile use of adipose-derived cells as fundamental components of three-dimensional MPS models for discovery research and development. In this context, it compares the merits and limitations of the adipose-derived stromal/stem cells relative to SVF cell models and considers the likely directions that this emerging field of scientific discovery will take in the near future.

    Table and Figures | Reference | Related Articles | Metrics
    Advances and perspective on animal models and hydrogel biomaterials for diabetic wound healing
    Yiqiang Hu, Yuan Xiong, Ranyang Tao, Hang Xue, Lang Chen, Ze Lin, Adriana C. Panayi, Bobin Mi, Guohui Liu
    Biomaterials Translational    2022, 3 (3): 188-200.   DOI: 10.12336/biomatertransl.2022.03.003
    Abstract284)   HTML34)    PDF(pc) (2004KB)(669)       Save

    Diabetic wounds are a common complication in diabetes patients. Due to peripheral nerve damage and vascular dysfunction, diabetic wounds are prone to progress to local ulcers, wound gangrene and even to require amputation, bringing huge psychological and economic burdens to patients. However, the current treatment methods for diabetic wounds mainly include wound accessories, negative pressure drainage, skin grafting and surgery; there is still no ideal treatment to promote diabetic wound healing at present. Appropriate animal models can simulate the physiological mechanism of diabetic wounds, providing a basis for translational research in treating diabetic wound healing. Although there are no animal models that can fully mimic the pathophysiological mechanisms of diabetic wounds in humans, it is vital to explore animal simulation models used in basic research and preclinical studies of diabetic wounds. In addition, hydrogel materials are regarded as a promising treatment for diabetic wounds because of their good antimicrobial activity, biocompatibility, biodegradation and appropriate mechanical properties. Herein, we review and discuss the different animal models used to investigate the pathological mechanisms of diabetic wounds. We further discuss the promising future application of hydrogel biomaterials in diabetic wound healing.

    Table and Figures | Reference | Related Articles | Metrics
    Recent development of hydrogen sulfide-releasing biomaterials as novel therapies:a narrative review
    Jingyu Fan, Elizabeth Pung, Yuan Lin, Qian Wang
    Biomaterials Translational    2022, 3 (4): 250-263.   DOI: 10.12336/biomatertransl.2022.04.005
    Abstract281)   HTML26)    PDF(pc) (940KB)(825)       Save

    Hydrogen sulfide (H2S) has been reported as an endogenous gasotransmitter that contributes to the modulation of a myriad of biological signalling pathways, which includes maintaining homeostasis in living organisms at physiological concentrations, controlling protein sulfhydration and persulfidation for signalling processes, mediating neurodegeneration, and regulating inflammation and innate immunity, etc. As a result, researchers are actively exploring effective approaches to evaluate the properties and the distribution of H2S in vivo. Furthermore, the regulation of the physiological conditions of H2S in vivo introduces the opportunity to further study the molecular mechanisms by which H2S regulates cellular functions. In recent years, many H2S–releasing compounds and biomaterials that can deliver H2S to various body systems have been developed to provide sustained and stable H2S delivery. Additionally, various designs of these H2S–releasing biomaterials have been proposed to aid in the normal conduction of physiological processes, such as cardioprotection and wound healing, by modulating different signalling pathways and cell functionalities. Using biomaterials as a platform to control the delivery of H2S introduces the opportunity to fine tune the physiological concentration of H2S in vivo, a key to many therapeutic applications. In this review, we highlight recent research works concerning the development and application of H2S–releasing biomaterials with a special emphasis to different release triggering conditions in in vivo studies. We believe that the further exploration of the molecular mechanisms underlying H2S donors and their function when incorporated with various biomaterials will potentially help us understand the pathophysiological mechanisms of different diseases and assist the development of H2S–based therapies.

    Table and Figures | Reference | Related Articles | Metrics
    Osteoarthritis animal models for biomaterial-assisted osteochondral regeneration
    Yi Wang, Yangyang Chen, Yulong Wei
    Biomaterials Translational    2022, 3 (4): 264-279.   DOI: 10.12336/biomatertransl.2022.04.006
    Abstract281)   HTML19)    PDF(pc) (435KB)(604)       Save

    Clinical therapeutics for the regeneration of osteochondral defects (OCD) in the early stages of osteoarthritis remain an enormous challenge in orthopaedics. For in-depth studies of tissue engineering and regenerative medicine in terms of OCD treatment, the utility of an optimal OCD animal model is crucial for assessing the effects of implanted biomaterials on the repair of damaged osteochondral tissues. Currently, the most frequently used in vivo animal models for OCD regeneration include mice, rats, rabbits, dogs, pigs, goats, sheep, horses and nonhuman primates. However, there is no single “gold standard” animal model to accurately recapitulate human disease in all aspects, thus understanding the benefits and limitations of each animal model is critical for selecting the most suitable one. In this review, we aim to elaborate the complex pathological changes in osteoarthritic joints and to summarise the advantages and limitations of OCD animal models utilised for biomaterial testing along with the methodology of outcome assessment. Furthermore, we review the surgical procedures of OCD creation in different species, and the novel biomaterials that promote OCD regeneration. Above all, it provides a significant reference for selection of an appropriate animal model for use in preclinical in vivo studies of biomaterial-assisted osteochondral regeneration in osteoarthritic joints.

    Table and Figures | Reference | Related Articles | Metrics
    Mechanical environment for in vitro cartilage tissue engineering assisted by in silico models
    Rob Jess, Tao Ling, Yi Xiong, Chris J. Wright, Feihu Zhao
    Biomaterials Translational    2023, 4 (1): 18-26.   DOI: 10.12336/biomatertransl.2023.01.004
    Abstract237)   HTML19)    PDF(pc) (372KB)(336)       Save

    Mechanobiological study of chondrogenic cells and multipotent stem cells for articular cartilage tissue engineering (CTE) has been widely explored. The mechanical stimulation in terms of wall shear stress, hydrostatic pressure and mechanical strain has been applied in CTE in vitro. It has been found that the mechanical stimulation at a certain range can accelerate the chondrogenesis and articular cartilage tissue regeneration. This review explicitly focuses on the study of the influence of the mechanical environment on proliferation and extracellular matrix production of chondrocytes in vitro for CTE. The multidisciplinary approaches used in previous studies and the need for in silico methods to be used in parallel with in vitro methods are also discussed. The information from this review is expected to direct facial CTE research, in which mechanobiology has not been widely explored yet.

    Table and Figures | Reference | Related Articles | Metrics
    Recent updates on the biological basis of heterogeneity in bone marrow stromal cells/skeletal stem cells
    Deepika Arora, Pamela Gehron Robey
    Biomaterials Translational    2022, 3 (1): 3-16.   DOI: 10.12336/biomatertransl.2022.01.002
    Abstract209)   HTML35)    PDF(pc) (2307KB)(854)       Save

    Based on studies over the last several decades, the self-renewing skeletal lineages derived from bone marrow stroma could be an ideal source for skeletal tissue engineering. However, the markers for osteogenic precursors; i.e., bone marrow-derived skeletal stem cells (SSCs), in association with other cells of the marrow stroma (bone marrow stromal cells, BMSCs) and their heterogeneous nature both in vivo and in vitro remain to be clarified. This review aims to highlight: i) the importance of distinguishing BMSCs/SSCs from other “mesenchymal stem/stromal cells”, and ii) factors that are responsible for their heterogeneity, and how these factors impact on the differentiation potential of SSCs towards bone. The prospective role of SSC enrichment, their expansion and its impact on SSC phenotype is explored. Emphasis has also been given to emerging single cell RNA sequencing approaches in scrutinizing the unique population of SSCs within the BMSC population, along with their committed progeny. Understanding the factors involved in heterogeneity may help researchers to improvise their strategies to isolate, characterize and adopt best culture practices and source identification to develop standard operating protocols for developing reproducible stem cells grafts. However, more scientific understanding of the molecular basis of heterogeneity is warranted that may be obtained from the robust high-throughput functional transcriptomics of single cells or clonal populations.

    Table and Figures | Reference | Related Articles | Metrics
    Oral stem cells, decoding and mapping the resident cells populations
    Xuechen Zhang, Ana Justo Caetano, Paul T. Sharpe, Ana Angelova Volponi
    Biomaterials Translational    2022, 3 (1): 24-30.   DOI: 10.12336/biomatertransl.2022.01.004
    Abstract206)   HTML21)    PDF(pc) (1244KB)(795)       Save

    The teeth and their supporting tissues provide an easily accessible source of oral stem cells. These different stem cell populations have been extensively studied for their properties, such as high plasticity and clonogenicity, expressing stem cell markers and potency for multilineage differentiation in vitro. Such cells with stem cell properties have been derived and characterised from the dental pulp tissue, the apical papilla region of roots in development, as well as the supporting tissue of periodontal ligament that anchors the tooth within the alveolar socket and the soft gingival tissue. Studying the dental pulp stem cell populations in a continuously growing mouse incisor model, as a traceable in vivo model, enables the researchers to study the properties, origin and behaviour of mesenchymal stem cells. On the other side, the oral mucosa with its remarkable scarless wound healing phenotype, offers a model to study a well-coordinated system of healing because of coordinated actions between epithelial, mesenchymal and immune cells populations. Although described as homogeneous cell populations following their in vitro expansion, the increasing application of approaches that allow tracing of individual cells over time, along with single-cell RNA-sequencing, reveal that different oral stem cells are indeed diverse populations and there is a highly organised map of cell populations according to their location in resident tissues, elucidating diverse stem cell niches within the oral tissues. This review covers the current knowledge of diverse oral stem cells, focusing on the new approaches in studying these cells. These approaches “decode” and “map” the resident cells populations of diverse oral tissues and contribute to a better understanding of the “stem cells niche architecture and interactions. Considering the high accessibility and simplicity in obtaining these diverse stem cells, the new findings offer potential in development of translational tissue engineering approaches and innovative therapeutic solutions.

    Table and Figures | Reference | Related Articles | Metrics
    Mesenchymal stem cell–derived extracellular vesicles: a possible therapeutic strategy for orthopaedic diseases: a narrative review
    Zhao–Lin Zeng, Hui Xie
    Biomaterials Translational    2022, 3 (3): 175-187.   DOI: 10.12336/biomatertransl.2022.03.002
    Abstract194)   HTML27)    PDF(pc) (2654KB)(839)       Save

    Accumulating evidence suggests that the therapeutic role of mesenchymal stem cells (MSCs) in bone diseases is closely related to paracrine–generated extracellular vesicles (EVs). MSC–derived EVs (MSC–EVs) carry proteins, nucleic acids, and lipids to the extracellular space and affect the bone microenvironment. They have similar biological functions to MSCs, such as the ability to repair organ and tissue damage. In addition, MSC–EVs also have the advantages of long half–life, low immunogenicity, attractive stability, ability to pass through the blood–brain barrier, and demonstrate excellent performance with potential practical applications in bone diseases. In this review, we summarise the current applications and mechanisms of MSC–EVs in osteoporosis, osteoarthritis, bone tumours, osteonecrosis of the femoral head, and fractures, as well as the development of MSC–EVs combined with materials science in the field of orthopaedics. Additionally, we explore the critical challenges involved in the clinical application of MSC–EVs in orthopaedic diseases.

    Table and Figures | Reference | Related Articles | Metrics
    Therapeutic potential of oncolytic viruses in the era of precision oncology
    Monchupa Kingsak, Thongpon Meethong, Jinnawat Jongkhumkrong, Li Cai, Qian Wang
    Biomaterials Translational    2023, 4 (2): 67-84.   DOI: 10.12336/biomatertransl.2023.02.003
    Abstract192)   HTML35)    PDF(pc) (1311KB)(228)       Save

    Oncolytic virus (OV) therapy has been shown to be an effective targeted cancer therapy treatment in recent years, providing an avenue of treatment that poses no damage to surrounding healthy tissues. Not only do OVs cause direct oncolysis, but they also amplify both innate and adaptive immune responses generating long-term anti-tumour immunity. Genetically engineered OVs have become the common promising strategy to enhance anti-tumour immunity, safety, and efficacy as well as targeted delivery. The studies of various OVs have been accomplished through phase I-III clinical trial studies. In addition, the uses of carrier platforms of organic materials such as polymer chains, liposomes, hydrogels, and cell carriers have played a vital role in the potentially targeted delivery of OVs. The mechanism, rational design, recent clinical trials, applications, and the development of targeted delivery platforms of OVs will be discussed in this review.

    Table and Figures | Reference | Related Articles | Metrics
    Mesenchymal stem cells and COVID-19: the process of discovery and of translation
    Arnold I. Caplan
    Biomaterials Translational    2021, 2 (4): 307-311.   DOI: 10.12336/biomatertransl.2021.04.006
    Abstract183)   HTML19)    PDF(pc) (238KB)(434)       Save

    Mesenchymal stem cells were developed as a cell-based therapeutic in the 1990’s. The translation of culture expanded mesenchymal stem cells from a basic science focus into a modern therapeutic has taken 30 years. The current state of the basic science information argues that mesenchymal stem cells may be curative for coronavirus disease 2019 (COVID-19). Indeed, early small-scale clinical trials have shown positive results. The issue raised is how to assemble the resources to get this cell-based therapy approved for clinical use. The technology is complex, the COVID-19 viral infections are life threatening, the cost is high, but human life is precious. What will it take to perfect this potentially curative technology?

    Reference | Related Articles | Metrics
    Fabrication, microstructure and properties of advanced ceramic–reinforced composites for dental implants: a review
    Mugilan Thanigachalam, Aezhisai Vallavi Muthusamy Subramanian
    Biomaterials Translational    2023, 4 (3): 151-165.   DOI: 10.12336/biomatertransl.2023.03.004
    Abstract170)   HTML16)    PDF(pc) (2244KB)(299)       Save

    The growing field of dental implant research and development has emerged to rectify the problems associated with human dental health issues. Bio–ceramics are widely used in the medical field, particularly in dental implants, ortho implants, and medical and surgical tools. Various materials have been used in those applications to overcome the limitations and problems associated with their performance and its impact on dental implants. In this article we review and describe the fabrication methods employed for ceramic composites, the microstructure analyses used to identify significant effects on fracture behaviour, and various methods of enhancing mechanical properties. Further, the collective data show that the sintering technique improves the density, hardness, fracture toughness, and flexural strength of alumina– and zirconia–based composites compared with other methods. Future research aspects and suggestions are discussed systematically.

    Table and Figures | Reference | Related Articles | Metrics
    Animal models for testing biomaterials in periodontal regeneration
    Qiao Sun, Yicun Li, Ping Luo, Hong He
    Biomaterials Translational    2023, 4 (3): 142-150.   DOI: 10.12336/biomatertransl.2023.03.003
    Abstract167)   HTML30)    PDF(pc) (746KB)(272)       Save

    Periodontitis is a prevalent oral disease. It can cause tooth loss and has a significant impact on patients’ quality of life. While existing treatments can only slow the progression of periodontitis, they are unable to achieve complete regeneration and functional reconstruction of periodontal tissues. As a result, regenerative therapies based on biomaterials have become a focal point of research in the field of periodontology. Despite numerous studies reporting the superiority of new materials in periodontal regeneration, limited progress has been made in translating these findings into clinical practice. This may be due to the lack of appropriate animal models to simulate the tissue defects caused by human periodontitis. This review aims to provide an overview of established animal models for periodontal regeneration, examine their advantages and limitations, and outline the steps for model construction. The objective is to determine the most relevant animal models for periodontal regeneration based on the hypothesis and expected outcomes.

    Table and Figures | Reference | Related Articles | Metrics
    Advances in electrode interface materials and modification technologies for brain-computer interfaces
    Yunke Jiao, Miao Lei, Jianwei Zhu, Ronghang Chang, Xue Qu
    Biomaterials Translational    2023, 4 (4): 213-233.   DOI: 10.12336/biomatertransl.2023.04.003
    Abstract164)   HTML28)    PDF(pc) (4326KB)(329)       Save

    Recent advances in neuroelectrode interface materials and modification technologies are reviewed. Brain-computer interface is the new method of human-computer interaction, which not only can realise the exchange of information between the human brain and external devices, but also provides a brand-new means for the diagnosis and treatment of brain-related diseases. The neural electrode interface part of brain-computer interface is an important area for electrical, optical and chemical signal transmission between brain tissue system and external electronic devices, which determines the performance of brain-computer interface. In order to solve the problems of insufficient flexibility, insufficient signal recognition ability and insufficient biocompatibility of traditional rigid electrodes, researchers have carried out extensive studies on the neuroelectrode interface in terms of materials and modification techniques. This paper introduces the biological reactions that occur in neuroelectrodes after implantation into brain tissue and the decisive role of the electrode interface for electrode function. Following this, the latest research progress on neuroelectrode materials and interface materials is reviewed from the aspects of neuroelectrode materials and modification technologies, firstly taking materials as a clue, and then focusing on the preparation process of neuroelectrode coatings and the design scheme of functionalised structures.

    Table and Figures | Reference | Related Articles | Metrics
    Bioactive elements manipulate bone regeneration
    Long Bai, Peiran Song, Jiacan Su
    Biomaterials Translational    2023, 4 (4): 248-269.   DOI: 10.12336/biomatertransl.2023.04.005
    Abstract164)   HTML31)    PDF(pc) (3289KB)(393)       Save

    While bone tissue is known for its inherent regenerative abilities, various pathological conditions and trauma can disrupt its meticulously regulated processes of bone formation and resorption. Bone tissue engineering aims to replicate the extracellular matrix of bone tissue as well as the sophisticated biochemical mechanisms crucial for effective regeneration. Traditionally, the field has relied on external agents like growth factors and pharmaceuticals to modulate these processes. Although efficacious in certain scenarios, this strategy is compromised by limitations such as safety issues and the transient nature of the compound release and half-life. Conversely, bioactive elements such as zinc (Zn), magnesium (Mg) and silicon (Si), have garnered increasing interest for their therapeutic benefits, superior stability, and reduced biotic risks. Moreover, these elements are often incorporated into biomaterials that function as multifaceted bioactive components, facilitating bone regeneration via release on-demand. By elucidating the mechanistic roles and therapeutic efficacy of the bioactive elements, this review aims to establish bioactive elements as a robust and clinically viable strategy for advanced bone regeneration.

    Table and Figures | Reference | Related Articles | Metrics
    An update of nanotopographical surfaces in modulating stem cell fate: a narrative review
    Shuqin Cao, Quan Yuan
    Biomaterials Translational    2022, 3 (1): 55-64.   DOI: 10.12336/biomatertransl.2022.01.006
    Abstract163)   HTML16)    PDF(pc) (399KB)(434)       Save

    Stem cells have been one of the ideal sources for tissue regeneration owing to their capability of self-renewal and differentiation. In vivo, the extracellular microenvironment plays a vital role in modulating stem cell fate. When developing biomaterials for regenerative medicine, incorporating biochemical and biophysical cues to mimic extracellular matrix can enhance stem cell lineage differentiation. More specifically, modulating the stem cell fate can be achieved by controlling the nanotopographic features on synthetic surfaces. Optimization of nanotopographical features leads to desirable stem cell functions, which can maximize the effectiveness of regenerative treatment. In this review, nanotopographical surfaces, including static patterned surface, dynamic patterned surface, and roughness are summarized, and their fabrication, as well as the impact on stem cell behaviour, are discussed. Later, the recent progress of applying nanotopographical featured biomaterials for altering different types of stem cells is presented, which directs the design and fabrication of functional biomaterial. Last, the perspective in fundamental research and for clinical application in this field is discussed.

    Table and Figures | Reference | Related Articles | Metrics
    Mesenchymal stem cell differentiation and usage for biotechnology applications: tissue engineering and food manufacturing
    Dafna Benayahu
    Biomaterials Translational    2022, 3 (1): 17-23.   DOI: 10.12336/biomatertransl.2022.01.003
    Abstract147)   HTML18)    PDF(pc) (199KB)(503)       Save

    Recent advances in the field of stem cell research now enable their utilisation for biotechnology applications in regenerative medicine and food tech. The first use of stem cells as biomedical devices employed a combination of cells and scaffold to restore, improve, or replace damaged tissues and to grow new viable tissue for replacement organs. This approach has also been adopted to replace meat production in the food industry. Mesenchymal stem cells are the source material used to induce cells to differentiate into the desired lineage. These technologies require mass propagation and rely on supplying the regulatory factors that direct differentiation. Mesenchymal stem cells can differentiate into fibroblastic and skeletal cells; fibroblastic/chondrogenic/osteogenic/myogenic and adipogenic lineages. Each differentiation fate requires specific key molecular regulators and appropriate activation conditions. Stem cell commitment determination involves a concerted effort of coordinated activation and silencing of lineage-specific genes. Transcription factors which bind gene promoters and chromatin-remodelling proteins are key players in the control process of lineage commitment and differentiation from embryogenesis through adulthood. Consequently, a major research challenge is to characterise such molecular pathways that coordinate lineage-specific differentiation and function. Revealing the mechanisms of action and the main factors will provide the knowledge necessary to control activation and regulation to achieve a specific lineage. Growing cells on a scaffold is a support system that mimics natural tissue and transduces the appropriate signals of the tissue niche for appropriate cellular function. The outcome of such research will deepen the understanding of cell differentiation to promote and advance the biotech, allowing the cell expansion required for their usage in therapy or the development of food tech.

    Reference | Related Articles | Metrics
    Organoid extracellular vesicle-based therapeutic strategies for bone therapy
    Han Liu, Jiacan Su
    Biomaterials Translational    2023, 4 (4): 199-212.   DOI: 10.12336/biomatertransl.2023.04.002
    Abstract147)   HTML27)    PDF(pc) (1653KB)(402)       Save

    With the rapid development of population ageing, bone-related diseases seriously affecting the life of the elderly. Over the past few years, organoids, cell clusters with specific functions and structures that are self-induced from stem cells after three-dimensional culture in vitro, have been widely used for bone therapy. Moreover, organoid extracellular vesicles (OEVs) have emerging as promising cell-free nanocarriers due to their vigoroso physiological effects, significant biological functions, stable loading capacity, and great biocompatibility. In this review, we first provide a comprehensive overview of biogenesis, internalisation, isolation, and characterisation of OEVs. We then comprehensively highlight the differences between OEVs and traditional EVs. Subsequently, we present the applications of natural OEVs in disease treatment. We also summarise the engineering modifications of OEVs, including engineering parental cells and engineering OEVs after isolation. Moreover, we provide an outlook on the potential of natural and engineered OEVs in bone-related diseases. Finally, we critically discuss the advantages and challenges of OEVs in the treatment of bone diseases. We believe that a comprehensive discussion of OEVs will provide more innovative and efficient solutions for complex bone diseases.

    Table and Figures | Reference | Related Articles | Metrics
    Research progress and clinical translation of three–dimensional printed porous tantalum in orthopaedics
    Jiawei Ying, Haiyu Yu, Liangliang Cheng, Junlei Li, Bin Wu, Liqun Song, Pinqiao Yi, Haiyao Wang, Lingpeng Liu, Dewei Zhao
    Biomaterials Translational    2023, 4 (3): 166-179.   DOI: 10.12336/biomatertransl.2023.03.005
    Abstract146)   HTML11)    PDF(pc) (1526KB)(285)       Save

    With continuous developments in additive manufacturing technology, tantalum (Ta) metal has been manufactured into orthopaedic implants with a variety of forms, properties and uses by three–dimensional printing. Based on extensive research in recent years, the design, processing and performance aspects of this new orthopaedic implant material have been greatly improved. Besides the bionic porous structure and mechanical characteristics that are similar to human bone tissue, porous tantalum is considered to be a viable bone repair material due to its outstanding corrosion resistance, biocompatibility, bone integration and bone conductivity. Numerous in vitro, in vivo, and clinical studies have been carried out in order to analyse the safety and efficacy of these implants in orthopaedic applications. This study reviews the most recent advances in manufacturing, characteristics and clinical application of porous tantalum materials.

    Table and Figures | Reference | Related Articles | Metrics
    Engineered microorganism–based delivery systems for targeted cancer therapy: a narrative review
    Xin Huang, Haoyu Guo, Lutong Wang, Zengwu Shao
    Biomaterials Translational    2022, 3 (3): 201-212.   DOI: 10.12336/biomatertransl.2022.03.004
    Abstract145)   HTML16)    PDF(pc) (1207KB)(343)       Save

    Microorganisms with innate and artificial advantages have been regarded as intelligent drug delivery systems for cancer therapy with the help of engineering technology. Although numerous studies have confirmed the promising prospects of microorganisms in cancer, several problems such as immunogenicity and toxicity should be addressed before further clinical applications. This review aims to investigate the development of engineered microorganism–based delivery systems for targeted cancer therapy. The main types of microorganisms such as bacteria, viruses, fungi, microalgae, and their components and characteristics are introduced in detail. Moreover, the engineering strategies and biomaterials design of microorganisms are further discussed. Most importantly, we discuss the innovative attempts and therapeutic effects of engineered microorganisms in cancer. Taken together, engineered microorganism–based delivery systems hold tremendous promise for biomedical applications in targeted cancer therapy.

    Table and Figures | Reference | Related Articles | Metrics
    The long and winding road: homeostatic and disordered haematopoietic microenvironmental niches: a narrative review
    Suzanne M. Watt
    Biomaterials Translational    2022, 3 (1): 31-54.   DOI: 10.12336/biomatertransl.2022.01.005
    Abstract114)   HTML17)    PDF(pc) (1774KB)(702)       Save

    Haematopoietic microenvironmental niches have been described as the ‘gatekeepers’ for the blood and immune systems. These niches change during ontogeny, with the bone marrow becoming the predominant site of haematopoiesis in post-natal life under steady state conditions. To determine the structure and function of different haematopoietic microenvironmental niches, it is essential to clearly define specific haematopoietic stem and progenitor cell subsets during ontogeny and to understand their temporal appearance and anatomical positioning. A variety of haematopoietic and non-haematopoietic cells contribute to haematopoietic stem and progenitor cell niches. The latter is reported to include endothelial cells and mesenchymal stromal cells (MSCs), skeletal stem cells and/or C-X-C motif chemokine ligand 12-abundant-reticular cell populations, which form crucial components of these microenvironments under homeostatic conditions. Dysregulation or deterioration of such cells contributes to significant clinical disorders and diseases worldwide and is associated with the ageing process. A critical appraisal of these issues and of the roles of MSC/C-X-C motif chemokine ligand 12-abundant-reticular cells and the more recently identified skeletal stem cell subsets in bone marrow haematopoietic niche function under homeostatic conditions and during ageing will form the basis of this research review. In the context of haematopoiesis, clinical translation will deal with lessons learned from the vast experience garnered from the development and use of MSC therapies to treat graft versus host disease in the context of allogeneic haematopoietic transplants, the recent application of these MSC therapies to treating emerging and severe coronavirus disease 2019 (COVID-19) infections, and, given that skeletal stem cell ageing is one proposed driver for haematopoietic ageing, the potential contributions of these stem cells to haematopoiesis in healthy bone marrow and the benefits and challenges of using this knowledge for rejuvenating the age-compromised bone marrow haematopoietic niches and restoring haematopoiesis.

    Table and Figures | Reference | Related Articles | Metrics
    Recent advances of medical polyhydroxyalkanoates in musculoskeletal system
    Chen-Hui Mi, Xin-Ya Qi, Yan-Wen Ding, Jing Zhou, Jin-Wei Dao, Dai-Xu Wei
    Biomaterials Translational    2023, 4 (4): 234-247.   DOI: 10.12336/biomatertransl.2023.04.004
    Abstract104)   HTML20)    PDF(pc) (2169KB)(333)       Save

    Infection and rejection in musculoskeletal trauma often pose challenges for natural healing, prompting the exploration of biomimetic organ and tissue transplantation as a common alternative solution. Polyhydroxyalkanoates (PHAs) are a large family of biopolyesters synthesised in microorganism, demonstrating excellent biocompatibility and controllable biodegradability for tissue remodelling and drug delivery. With different monomer-combination and polymer-types, multi-mechanical properties of PHAs making them have great application prospects in medical devices with stretching, compression, twist in long time, especially in musculoskeletal tissue engineering. This review systematically summarises the applications of PHAs in multiple tissues repair and drug release, encompassing areas such as bone, cartilage, joint, skin, tendons, ligament, cardiovascular tissue, and nervous tissue. It also discusses challenges encountered in their application, including high production costs, potential cytotoxicity, and uncontrollable particle size distribution. In conclusion, PHAs offer a compelling avenue for musculoskeletal system applications, striking a balance between biocompatibility and mechanical performance. However, addressing challenges in their production and application requires further research to unleash their full potential in tackling the complexities of musculoskeletal regeneration.

    Table and Figures | Reference | Related Articles | Metrics