151
|
Sun L, Wang Y, Zhang S, Yang H, Mao Y. 3D bioprinted liver tissue and disease models: Current advances and future perspectives. BIOMATERIALS ADVANCES 2023; 152:213499. [PMID: 37295133 DOI: 10.1016/j.bioadv.2023.213499] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/23/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
Three-dimensional (3D) bioprinting is a promising technology for fabricating complex tissue constructs with biomimetic biological functions and stable mechanical properties. In this review, the characteristics of different bioprinting technologies and materials are compared, and development in strategies for bioprinting normal and diseased hepatic tissue are summarized. In particular, features of bioprinting and other bio-fabrication strategies, such as organoids and spheroids are compared to demonstrate the strengths and weaknesses of 3D printing technology. Directions and suggestions, such as vascularization and primary human hepatocyte culture, are provided for the future development of 3D bioprinting.
Collapse
Affiliation(s)
- Lejia Sun
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Dongcheng, Beijing, 100730, China; Department of General Surgery, The First affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yinhan Wang
- Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences & PUMC, Dongcheng, Beijing 100730, China
| | - Shuquan Zhang
- Peking Union Medical College (PUMC), Chinese Academy of Medical Sciences & PUMC, Dongcheng, Beijing 100730, China
| | - Huayu Yang
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Dongcheng, Beijing, 100730, China.
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Dongcheng, Beijing, 100730, China.
| |
Collapse
|
152
|
Zou S, Ye J, Wei Y, Xu J. Characterization of 3D-Bioprinted In Vitro Lung Cancer Models Using RNA-Sequencing Techniques. Bioengineering (Basel) 2023; 10:667. [PMID: 37370598 DOI: 10.3390/bioengineering10060667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/21/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVE To construct an in vitro lung cancer model using 3D bioprinting and evaluate the feasibility of the model. Transcriptome sequencing was used to compare the differential genes and functions of 2D and 3D lung cancer cells. METHODS 1. A549 cells were mixed with sodium alginate/gelatine/fibrinogen as 3D-printed biological ink to construct a hydrogel scaffold for the in vitro model of lung cancer; 2. A hydrogel scaffold was printed using a extrusion 3D bioprinter; 3. The printed lung cancer model was evaluated in vitro; and 4. A549 cells cultured in 2D and 3D tumour models in vitro were collected, and RNA-seq conducted bioinformatics analysis. RESULTS 1. The in vitro lung cancer model printed using 3D-bioprinting technology was a porous microstructure model, suitable for the survival of A549 cells. Compared with the 2D cell-line model, the 3D model is closer to the fundamental human growth environment; 2. There was no significant difference in cell survival rate between the 2D and 3D groups; 3. In the cell proliferation rate measurement, it was found that the cells in the 2D group had a speedy growth rate in the first five days, but after five days, the growth rate slowed down. Cell proliferation showed a declining process after the ninth day of cell culture. However, cells in the 3D group showed a slow growth process at the beginning, and the growth rate reached a peak on the 12th day. Then, the growth rate showed a downward trend; and 4. RNA-seq compared A549 cells from 2D and 3D lung cancer models. A total of 3112 genes were differentially expressed, including 1189 up-regulated and 1923 down-regulated genes, with p-value ≤ 0.05 and |Log2Ratio| ≥ 1 as screening conditions. After functional enrichment analysis of differential genes, these differential genes affect the biological regulation of A549 cells, thus promoting lung cancer progression. CONCLUSION This study uses 3D-bioprinting technology to construct a tumour model of lung cancer that can grow sustainably in vitro. Three-dimensional bioprinting may provide a new research platform for studying the lung cancer TME mechanism and anticancer drug screening.
Collapse
Affiliation(s)
- Sheng Zou
- The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China
| | - Jiayue Ye
- The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China
| | - Yiping Wei
- The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China
| | - Jianjun Xu
- The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China
| |
Collapse
|
153
|
Abstract
PURPOSE OF REVIEW Bioengineering of functional cardiac tissue composed of primary cardiomyocytes has great potential for myocardial regeneration and in vitro tissue modeling. 3D bioprinting was developed to create cardiac tissue in hydrogels that can mimic the structural, physiological, and functional features of native myocardium. Through a detailed review of the 3D printing technologies and bioink materials used in the creation of a heart tissue, this article discusses the potential of engineered heart tissues in biomedical applications. RECENT FINDINGS In this review, we discussed the recent progress in 3D bioprinting strategies for cardiac tissue engineering, including bioink and 3D bioprinting methods as well as examples of engineered cardiac tissue such as in vitro cardiac models and vascular channels. 3D printing is a powerful tool for creating in vitro cardiac tissues that are structurally and functionally similar to real tissues. The use of human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) enables the generation of patient-specific tissues. These tissues have the potential to be used for regenerative therapies, disease modeling, and drug testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Yi Xiang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Min Tang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Shaochen Chen
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of Bioengineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| |
Collapse
|
154
|
Qiu M, Li C, Cai Z, Li C, Yang K, Tulufu N, Chen B, Cheng L, Zhuang C, Liu Z, Qi J, Cui W, Deng L. 3D Biomimetic Calcified Cartilaginous Callus that Induces Type H Vessels Formation and Osteoclastogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207089. [PMID: 36999832 PMCID: PMC10238192 DOI: 10.1002/advs.202207089] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Indexed: 06/04/2023]
Abstract
The formation of a calcified cartilaginous callus (CACC) is crucial during bone repair. CACC can stimulate the invasion of type H vessels into the callus to couple angiogenesis and osteogenesis, induce osteoclastogenesis to resorb the calcified matrix, and promote osteoclast secretion of factors to enhance osteogenesis, ultimately achieving the replacement of cartilage with bone. In this study, a porous polycaprolactone/hydroxyapatite-iminodiacetic acid-deferoxamine (PCL/HA-SF-DFO) 3D biomimetic CACC is developed using 3D printing. The porous structure can mimic the pores formed by the matrix metalloproteinase degradation of the cartilaginous matrix, HA-containing PCL can mimic the calcified cartilaginous matrix, and SF anchors DFO onto HA for the slow release of DFO. The in vitro results show that the scaffold significantly enhances angiogenesis, promotes osteoclastogenesis and resorption by osteoclasts, and enhances the osteogenic differentiation of bone marrow stromal stem cells by promoting collagen triple helix repeat-containing 1 expression by osteoclasts. The in vivo results show that the scaffold significantly promotes type H vessels formation and the expression of coupling factors to promote osteogenesis, ultimately enhancing the regeneration of large-segment bone defects in rats and preventing dislodging of the internal fixation screw. In conclusion, the scaffold inspired by biological bone repair processes effectively promotes bone regeneration.
Collapse
Affiliation(s)
- Minglong Qiu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Changwei Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zhengwei Cai
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Cuidi Li
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Kai Yang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Nijiati Tulufu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Bo Chen
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Liang Cheng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Chengyu Zhuang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zhihong Liu
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Jin Qi
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Lianfu Deng
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| |
Collapse
|
155
|
Huang WH, Ding SL, Zhao XY, Li K, Guo HT, Zhang MZ, Gu Q. Collagen for neural tissue engineering: Materials, strategies, and challenges. Mater Today Bio 2023; 20:100639. [PMID: 37197743 PMCID: PMC10183670 DOI: 10.1016/j.mtbio.2023.100639] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023] Open
Abstract
Neural tissue engineering (NTE) has made remarkable strides in recent years and holds great promise for treating several devastating neurological disorders. Selecting optimal scaffolding material is crucial for NET design strategies that enable neural and non-neural cell differentiation and axonal growth. Collagen is extensively employed in NTE applications due to the inherent resistance of the nervous system against regeneration, functionalized with neurotrophic factors, antagonists of neural growth inhibitors, and other neural growth-promoting agents. Recent advancements in integrating collagen with manufacturing strategies, such as scaffolding, electrospinning, and 3D bioprinting, provide localized trophic support, guide cell alignment, and protect neural cells from immune activity. This review categorises and analyses collagen-based processing techniques investigated for neural-specific applications, highlighting their strengths and weaknesses in repair, regeneration, and recovery. We also evaluate the potential prospects and challenges of using collagen-based biomaterials in NTE. Overall, this review offers a comprehensive and systematic framework for the rational evaluation and applications of collagen in NTE.
Collapse
Affiliation(s)
- Wen-Hui Huang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Sheng-Long Ding
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Kai Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
| | - Hai-Tao Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
| | - Ming-Zhu Zhang
- Department of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, PR China
- Corresponding author.
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Chaoyang District, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Chaoyang District, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Huairou District, Beijing, 101499, PR China
- Corresponding author. Institute of Zoology, Chinese Academy of Sciences, No. 5 of Courtyard 1, Beichen West Road, Chaoyang District, Beijing 100101, PR China.
| |
Collapse
|
156
|
Badhe RV, Chatterjee A, Bijukumar D, Mathew MT. Current advancements in bio-ink technology for cartilage and bone tissue engineering. Bone 2023; 171:116746. [PMID: 36965655 PMCID: PMC10559728 DOI: 10.1016/j.bone.2023.116746] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/05/2023] [Accepted: 03/20/2023] [Indexed: 03/27/2023]
Abstract
In tissue engineering, the fate of a particular organ/tissue regeneration and repair mainly depends on three pillars - 3D architecture, cells used, and stimulus provided. 3D cell supportive structure development is one of the crucial pillars necessary for defining organ/tissue geometry and shape. In recent years, the advancements in 3D bio-printing (additive manufacturing) made it possible to develop very precise 3D architectures with the help of industrial software like Computer-Aided Design (CAD). The main requirement for the 3D printing process is the bio-ink, which can act as a source for cell support, proliferation, drug (growth factors, stimulators) delivery, and organ/tissue shape. The selection of the bio-ink depends upon the type of 3D tissue of interest. Printing tissues like bone and cartilage is always challenging because it is difficult to find printable biomaterial that can act as bio-ink and mimic the strength of the natural bone and cartilage tissues. This review describes different biomaterials used to develop bio-inks with different processing variables and cell-seeding densities for bone and cartilage 3D printing applications. The review also discusses the advantages, limitations, and cell bio-ink compatibility in each biomaterial section. The emphasis is given to bio-inks reported for 3D printing cartilage and bone and their applications in orthopedics and orthodontists. The critical/important performance and the architectural morphology requirements of desired bone and cartilage bio-inks were compiled in summary.
Collapse
Affiliation(s)
- Ravindra V Badhe
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA; Pharmaceutical Chemistry Department, Marathwada Mitramandal's College of Pharmacy, Thergaon, Pune, Maharashtra, India
| | - Abhinav Chatterjee
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | - Divya Bijukumar
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | - Mathew T Mathew
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA.
| |
Collapse
|
157
|
Ding Z, Tang N, Huang J, Cao X, Wu S. Global hotspots and emerging trends in 3D bioprinting research. Front Bioeng Biotechnol 2023; 11:1169893. [PMID: 37304138 PMCID: PMC10248473 DOI: 10.3389/fbioe.2023.1169893] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/08/2023] [Indexed: 06/13/2023] Open
Abstract
Three-dimensional (3D) bioprinting is an advanced tissue engineering technique that has received a lot of interest in the past years. We aimed to highlight the characteristics of articles on 3D bioprinting, especially in terms of research hotspots and focus. Publications related to 3D bioprinting from 2007 to 2022 were acquired from the Web of Science Core Collection database. We have used VOSviewer, CiteSpace, and R-bibliometrix to perform various analyses on 3,327 published articles. The number of annual publications is increasing globally, a trend expected to continue. The United States and China were the most productive countries with the closest cooperation and the most research and development investment funds in this field. Harvard Medical School and Tsinghua University are the top-ranked institutions in the United States and China, respectively. Dr. Anthony Atala and Dr. Ali Khademhosseini, the most productive researchers in 3D bioprinting, may provide cooperation opportunities for interested researchers. Tissue Engineering Part A contributed the largest publication number, while Frontiers in Bioengineering and Biotechnology was the most attractive journal with the most potential. As for the keywords in 3D bioprinting, Bio-ink, Hydrogels (especially GelMA and Gelatin), Scaffold (especially decellularized extracellular matrix), extrusion-based bioprinting, tissue engineering, and in vitro models (organoids particularly) are research hotspots analyzed in the current study. Specifically, the research topics "new bio-ink investigation," "modification of extrusion-based bioprinting for cell viability and vascularization," "application of 3D bioprinting in organoids and in vitro model" and "research in personalized and regenerative medicine" were predicted to be hotspots for future research.
Collapse
Affiliation(s)
- Zhiyu Ding
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ning Tang
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Junjie Huang
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xu Cao
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Song Wu
- Department of Orthopaedics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
158
|
Lewandrowski KU, Elfar JC, Li ZM, Burkhardt BW, Lorio MP, Winkler PA, Oertel JM, Telfeian AE, Dowling Á, Vargas RAA, Ramina R, Abraham I, Assefi M, Yang H, Zhang X, Ramírez León JF, Fiorelli RKA, Pereira MG, de Carvalho PST, Defino H, Moyano J, Lim KT, Kim HS, Montemurro N, Yeung A, Novellino P. The Changing Environment in Postgraduate Education in Orthopedic Surgery and Neurosurgery and Its Impact on Technology-Driven Targeted Interventional and Surgical Pain Management: Perspectives from Europe, Latin America, Asia, and The United States. J Pers Med 2023; 13:852. [PMID: 37241022 PMCID: PMC10221956 DOI: 10.3390/jpm13050852] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Personalized care models are dominating modern medicine. These models are rooted in teaching future physicians the skill set to keep up with innovation. In orthopedic surgery and neurosurgery, education is increasingly influenced by augmented reality, simulation, navigation, robotics, and in some cases, artificial intelligence. The postpandemic learning environment has also changed, emphasizing online learning and skill- and competency-based teaching models incorporating clinical and bench-top research. Attempts to improve work-life balance and minimize physician burnout have led to work-hour restrictions in postgraduate training programs. These restrictions have made it particularly challenging for orthopedic and neurosurgery residents to acquire the knowledge and skill set to meet the requirements for certification. The fast-paced flow of information and the rapid implementation of innovation require higher efficiencies in the modern postgraduate training environment. However, what is taught typically lags several years behind. Examples include minimally invasive tissue-sparing techniques through tubular small-bladed retractor systems, robotic and navigation, endoscopic, patient-specific implants made possible by advances in imaging technology and 3D printing, and regenerative strategies. Currently, the traditional roles of mentee and mentor are being redefined. The future orthopedic surgeons and neurosurgeons involved in personalized surgical pain management will need to be versed in several disciplines ranging from bioengineering, basic research, computer, social and health sciences, clinical study, trial design, public health policy development, and economic accountability. Solutions to the fast-paced innovation cycle in orthopedic surgery and neurosurgery include adaptive learning skills to seize opportunities for innovation with execution and implementation by facilitating translational research and clinical program development across traditional boundaries between clinical and nonclinical specialties. Preparing the future generation of surgeons to have the aptitude to keep up with the rapid technological advances is challenging for postgraduate residency programs and accreditation agencies. However, implementing clinical protocol change when the entrepreneur-investigator surgeon substantiates it with high-grade clinical evidence is at the heart of personalized surgical pain management.
Collapse
Affiliation(s)
- Kai-Uwe Lewandrowski
- Center For Advanced Spine Care of Southern Arizona, 4787 E Camp Lowell Drive, Tucson, AZ 85719, USA
- Department of Orthopaedics, Fundación Universitaria Sanitas, Bogotá 111321, Colombia
| | - John C. Elfar
- Department of Orthopaedic Surgery, College of Medicine—Tucson Campus, Health Sciences Innovation Building (HSIB), University of Arizona, 1501 N. Campbell Avenue, Tower 4, 8th Floor, Suite 8401, Tucson, AZ 85721, USA;
| | - Zong-Ming Li
- Departments of Orthopaedic Surgery and Biomedical Engineering, College of Medicine—Tucson Campus, Health Sciences Innovation Building (HSIB), University of Arizona, 1501 N. Campbell Avenue, Tower 4, 8th Floor, Suite 8401, Tucson, AZ 85721, USA;
| | - Benedikt W. Burkhardt
- Wirbelsäulenzentrum/Spine Center—WSC, Hirslanden Klinik Zurich, Witellikerstrasse 40, 8032 Zurich, Switzerland;
| | - Morgan P. Lorio
- Advanced Orthopaedics, 499 E. Central Pkwy, Ste. 130, Altamonte Springs, FL 32701, USA;
| | - Peter A. Winkler
- Department of Neurosurgery, Charite Universitaetsmedizin Berlin, 13353 Berlin, Germany;
| | - Joachim M. Oertel
- Klinik für Neurochirurgie, Universitätsdes Saarlandes, Kirrberger Straße 100, 66421 Homburg, Germany;
| | - Albert E. Telfeian
- Department of Neurosurgery, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, USA;
| | - Álvaro Dowling
- Orthopaedic Surgery, University of São Paulo, Brazilian Spine Society (SBC), Ribeirão Preto 14071-550, Brazil; (Á.D.); (H.D.)
| | - Roth A. A. Vargas
- Department of Neurosurgery, Foundation Hospital Centro Médico Campinas, Campinas 13083-210, Brazil;
| | - Ricardo Ramina
- Neurological Institute of Curitiba, Curitiba 80230-030, Brazil;
| | - Ivo Abraham
- Clinical Translational Sciences, University of Arizona, Roy P. Drachman Hall, Rm. B306H, Tucson, AZ 85721, USA;
| | - Marjan Assefi
- Department of Biology, Nano-Biology, University of North Carolina, Greensboro, NC 27413, USA;
| | - Huilin Yang
- Orthopaedic Department, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou 215031, China;
| | - Xifeng Zhang
- Department of Orthopaedics, First Medical Center, PLA General Hospital, Beijing 100853, China;
| | - Jorge Felipe Ramírez León
- Minimally Invasive Spine Center Bogotá D.C. Colombia, Reina Sofía Clinic Bogotá D.C. Colombia, Department of Orthopaedics Fundación Universitaria Sanitas, Bogotá 0819, Colombia;
| | - Rossano Kepler Alvim Fiorelli
- Department of General and Specialized Surgery, Gaffrée e Guinle University Hospital, Federal University of the State of Rio de Janeiro (UNIRIO), Rio de Janeiro 20270-004, Brazil;
| | - Mauricio G. Pereira
- Faculty of Medecine, University of Brasilia, Federal District, Brasilia 70919-900, Brazil;
| | | | - Helton Defino
- Orthopaedic Surgery, University of São Paulo, Brazilian Spine Society (SBC), Ribeirão Preto 14071-550, Brazil; (Á.D.); (H.D.)
| | - Jaime Moyano
- La Sociedad Iberolatinoamericana De Columna (SILACO), and the Spine Committee of the Ecuadorian Society of Orthopaedics and Traumatology (Comité de Columna de la Sociedad Ecuatoriana de Ortopedia y Traumatología), Quito 170521, Ecuador;
| | - Kang Taek Lim
- Good Doctor Teun Teun Spine Hospital, Anyang 14041, Republic of Korea;
| | - Hyeun-Sung Kim
- Department of Neurosurgery, Nanoori Hospital, Seoul 06048, Republic of Korea;
| | - Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliero Universitaria Pisana, University of Pisa, 56124 Pisa, Italy;
| | - Anthony Yeung
- Desert Institute for Spine Care, Phoenix, AZ 85020, USA;
| | - Pietro Novellino
- Guinle and State Institute of Diabetes and Endocrinology, Rio de Janeiro 20270-004, Brazil;
| |
Collapse
|
159
|
Li X, Xu M, Geng Z, Liu Y. Functional hydrogels for the repair and regeneration of tissue defects. Front Bioeng Biotechnol 2023; 11:1190171. [PMID: 37260829 PMCID: PMC10227617 DOI: 10.3389/fbioe.2023.1190171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023] Open
Abstract
Tissue defects can be accompanied by functional impairments that affect the health and quality of life of patients. Hydrogels are three-dimensional (3D) hydrophilic polymer networks that can be used as bionic functional tissues to fill or repair damaged tissue as a promising therapeutic strategy in the field of tissue engineering and regenerative medicine. This paper summarises and discusses four outstanding advantages of hydrogels and their applications and advances in the repair and regeneration of tissue defects. First, hydrogels have physicochemical properties similar to the extracellular matrix of natural tissues, providing a good microenvironment for cell proliferation, migration and differentiation. Second, hydrogels have excellent shape adaptation and tissue adhesion properties, allowing them to be applied to a wide range of irregularly shaped tissue defects and to adhere well to the defect for sustained and efficient repair function. Third, the hydrogel is an intelligent delivery system capable of releasing therapeutic agents on demand. Hydrogels are capable of delivering therapeutic reagents and releasing therapeutic substances with temporal and spatial precision depending on the site and state of the defect. Fourth, hydrogels are self-healing and can maintain their integrity when damaged. We then describe the application and research progress of functional hydrogels in the repair and regeneration of defects in bone, cartilage, skin, muscle and nerve tissues. Finally, we discuss the challenges faced by hydrogels in the field of tissue regeneration and provide an outlook on their future trends.
Collapse
|
160
|
Chen J, Cao Y, Jia O, Wang X, Luo Y, Cheuk YC, Zhu T, Zhu D, Zhang Y, Wang J. Monomethyl fumarate prevents alloimmune rejection in mouse heart transplantation by inducing tolerogenic dendritic cells. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37184280 DOI: 10.3724/abbs.2023088] [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: 05/16/2023] Open
Abstract
Dendritic cells (DCs) are important targets for eliciting allograft rejection after transplantation. Previous studies have demonstrated that metabolic reprogramming of DCs can transform their immune functions and induce their differentiation into tolerogenic DCs. In this study, we aim to investigate the protective effects and mechanisms of monomethyl fumarate (MMF), a bioactive metabolite of fumaric acid esters, in a mouse model of allogeneic heart transplantation. Bone marrow-derived DCs are harvested and treated with MMF to determine the impact of MMF on the phenotype and immunosuppressive function of DCs by flow cytometry and T-cell proliferation assays. RNA sequencing and Seahorse analyses are performed for mature DCs and MMF-treated DCs (MMF-DCs) to investigate the underlying mechanism. Our results show that MMF prolongs the survival time of heart grafts and inhibits the activation of DCs in vivo. MMF-DCs exhibit a tolerogenic phenotype and function in vitro. RNA sequencing and Seahorse analyses reveal that MMF activates the Nrf2 pathway and mediates metabolic reprogramming. Additionally, MMF-DC infusion prolongs cardiac allograft survival, induces regulatory T cells, and inhibits T-cell activation. MMF prevents allograft rejection in mouse heart transplantation by inducing tolerogenic DCs.
Collapse
Affiliation(s)
- Juntao Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yirui Cao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Ouyang Jia
- Nursing Department of Huashan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Xuanchuan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yongsheng Luo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Dong Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
- Department of Urology, Zhongshan Hospital, Fudan University (Xiamen branch), Xiamen 361015, China
| | - Yi Zhang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
- Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jina Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| |
Collapse
|
161
|
Jiang F, Xu XW, Chen FQ, Weng HF, Chen J, Ru Y, Xiao Q, Xiao AF. Extraction, Modification and Biomedical Application of Agarose Hydrogels: A Review. Mar Drugs 2023; 21:md21050299. [PMID: 37233493 DOI: 10.3390/md21050299] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/27/2023] Open
Abstract
Numerous compounds present in the ocean are contributing to the development of the biomedical field. Agarose, a polysaccharide derived from marine red algae, plays a vital role in biomedical applications because of its reversible temperature-sensitive gelling behavior, excellent mechanical properties, and high biological activity. Natural agarose hydrogel has a single structural composition that prevents it from adapting to complex biological environments. Therefore, agarose can be developed into different forms through physical, biological, and chemical modifications, enabling it to perform optimally in different environments. Agarose biomaterials are being increasingly used for isolation, purification, drug delivery, and tissue engineering, but most are still far from clinical approval. This review classifies and discusses the preparation, modification, and biomedical applications of agarose, focusing on its applications in isolation and purification, wound dressings, drug delivery, tissue engineering, and 3D printing. In addition, it attempts to address the opportunities and challenges associated with the future development of agarose-based biomaterials in the biomedical field. It should help to rationalize the selection of the most suitable functionalized agarose hydrogels for specific applications in the biomedical industry.
Collapse
Affiliation(s)
- Feng Jiang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- National R&D Center for Red Alga Processing Technology, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
| | - Xin-Wei Xu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Fu-Quan Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- National R&D Center for Red Alga Processing Technology, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| | - Hui-Fen Weng
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Jun Chen
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- National R&D Center for Red Alga Processing Technology, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| | - Yi Ru
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Qiong Xiao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- National R&D Center for Red Alga Processing Technology, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| | - An-Feng Xiao
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen 361021, China
- National R&D Center for Red Alga Processing Technology, Xiamen 361021, China
- Fujian Provincial Engineering Technology Research Center of Marine Functional Food, Xiamen 361021, China
- Xiamen Key Laboratory of Marine Functional Food, Xiamen 361021, China
| |
Collapse
|
162
|
Abstract
Bioprinting as an extension of 3D printing offers capabilities for printing tissues and organs for application in biomedical engineering. Conducting bioprinting in space, where the gravity is zero, can enable new frontiers in tissue engineering. Fabrication of soft tissues, which usually collapse under their own weight, can be accelerated in microgravity conditions as the external forces are eliminated. Furthermore, human colonization in space can be supported by providing critical needs of life and ecosystems by 3D bioprinting without relying on cargos from Earth, e.g., by development and long-term employment of living engineered filters (such as sea sponges-known as critical for initiating and maintaining an ecosystem). This review covers bioprinting methods in microgravity along with providing an analysis on the process of shipping bioprinters to space and presenting a perspective on the prospects of zero-gravity bioprinting.
Collapse
Affiliation(s)
- Misagh Rezapour Sarabi
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, U.K
| | - Savas Tasoglu
- Mechanical Engineering Department, School of Engineering, Koç University, Istanbul, Turkey 34450
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, Stuttgart, Germany 70569
- Koç University Translational Medicine Research Center (KUTTAM), Koç University, Istanbul, Turkey 34450
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Istanbul, Turkey 34450
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Istanbul, Turkey 34684
| |
Collapse
|
163
|
Sun H, Yang H, Mao Y. Personalized treatment for hepatocellular carcinoma in the era of targeted medicine and bioengineering. Front Pharmacol 2023; 14:1150151. [PMID: 37214451 PMCID: PMC10198383 DOI: 10.3389/fphar.2023.1150151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 04/24/2023] [Indexed: 05/24/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global health burden, causing approximately 8.3 million deaths each year, and it is the third leading cause of cancer-related death worldwide, with a relative 5-year survival rate of around 18%. Due to the advanced stage of diagnosis in most patients, systemic treatment based on targeted therapy has become the only feasible option. Genomic studies have established a profile of molecular alterations in hepatocellular carcinoma with potentially actionable mutations, but these mutations have yet to be translated into clinical practice. The first targeted drug approved for systemic treatment of patients with advanced hepatocellular carcinoma was Sorafenib, which was a milestone. Subsequent clinical trials have identified multiple tyrosine kinase inhibitors, such as Lenvatinib, Cabozantinib, and Regorafenib, for the treatment of hepatocellular carcinoma, with survival benefits for the patient. Ongoing systemic therapy studies and trials include various immune-based combination therapies, with some early results showing promise and potential for new therapy plans. Systemic therapy for hepatocellular carcinoma is complicated by the significant heterogeneity of the disease and its propensity for developing drug resistance. Therefore, it is essential to choose a better, individualized treatment plan to benefit patients. Preclinical models capable of preserving in vivo tumor characteristics are urgently needed to circumvent heterogeneity and overcome drug resistance. In this review, we summarize current approaches to targeted therapy for HCC patients and the establishment of several patient-derived preclinical models of hepatocellular carcinoma. We also discuss the challenges and opportunities of targeted therapy for hepatocellular carcinoma and how to achieve personalized treatment with the continuous development of targeted therapies and bioengineering technologies.
Collapse
Affiliation(s)
| | - Huayu Yang
- *Correspondence: Huayu Yang, ; Yilei Mao,
| | - Yilei Mao
- *Correspondence: Huayu Yang, ; Yilei Mao,
| |
Collapse
|
164
|
Liu Z, Luo Z, Yu H, Zhao E, Guo J, Mou P, Chen A, Chen J, Zhou Z, Zeng W. Near-infrared light-controlled kartogenin delivery of multifunctional Prussian blue nanocomposites for cartilage defect repair. NANOSCALE 2023; 15:9076-9093. [PMID: 37129436 DOI: 10.1039/d3nr00205e] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Articular cartilage injury repair remains a challenge for clinicians and researchers. Mesenchymal stem cells (MSCs) have multiple differentiation potentials and can be induced to differentiate into the chondrogenic lineage for cartilage defect repair; however, the insufficient capacity of chondrogenic differentiation and excess reactive oxygen species (ROS)-mediated oxidative stress, which always lead to differentiation into hypertrophic chondrocytes, still need to be resolved. Accordingly, kartogenin (KGN), which can promote chondrogenic differentiation of MSCs, has shown promise in promoting infected cartilage repair. However, realizing controllable release to prolong its action time and avoid hypertrophic differentiation is critical. We herein developed a mesoporous Prussian blue nanoparticle (mPB)-based near-infrared (NIR) light-responsive controlled nanosystem. KGN was encapsulated in temperature-stimulated responsive phase change materials (PCMs), which were used as excellent gating materials (KGN-PCM@mPBs). In addition, the mPBs could efficiently scavenge ROS by their enzyme-like antioxidative activities. Our study demonstrates that the nanocomposites could efficiently promote chondrogenic differentiation and successfully inhibit the hypertrophic differentiation of MSCs. By intra-articular injection of KGN-PCM@mPBs and NIR-triggered precisely controlled release, satisfactory cartilage repair effects can be achieved in a rat chondral defect model. Thus, this constructed NIR-mediated KGN-PCM@mPB nanoplatform may represent an effective cartilage repair strategy with satisfactory biosafety in clinical applications.
Collapse
Affiliation(s)
- Zunhan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Zhenyu Luo
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Haoda Yu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Enze Zhao
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Junfeng Guo
- Department of Sports Medicine Center, State Key Laboratory of Trauma, Burn and Combined Injury, the First Affiliated Hospital of the Army Military Medical University, Chongqing, 400038, China
| | - Ping Mou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Anjing Chen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Jiali Chen
- West China School of Nursing, Sichuan University/Department of Orthopedics, West China Hospital, Sichuan University Chengdu, 610041, P.R. China
| | - Zongke Zhou
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| | - Weinan Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
165
|
Jajoo SS, Chaudhary SM, Patil K, Kunte S, Lakade L, Jagtap C. A Systematic Review on Polyester Scaffolds in Dental Three-dimensional Cell Printing: Transferring Art from the Laboratories to the Clinics. Int J Clin Pediatr Dent 2023; 16:494-498. [PMID: 37496946 PMCID: PMC10367294 DOI: 10.5005/jp-journals-10005-2609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023] Open
Abstract
Objective The purpose of this systematic review is to describe developments in three-dimensional (3D) cell printing in the formation of dental pulp tissue using polyester as a scaffold to revitalize the damaged dental pulp tissue. Materials and methods A literature search for all the data published in PubMed and Google Scholar from January 2000 to April 2022 was conducted. Articles with the keywords 3D cell printing, scaffolds, polyester, dental pulp, and dentistry were used. Inclusion criteria consisted of any publication in electronic or print media directly studying or commenting on the use of polyester scaffolds in 3D cell printing technology in the regeneration of dental pulp. A total of 528 articles were selected, of which 27 duplicates and 286 irrelevant articles were discarded. A total of 215 articles were finally included in the systematic review. Result and conclusion For dental pulp regeneration, several scaffolds have been discovered to be appealing. Polylactic acid (PLA), polyglycolic acid (PGA), and their copolymers are nontoxic and biocompatible synthetic polyesters that degrade by hydrolysis and have received Food and Drug Administration (FDA) approval for a variety of applications. This review paper is intended to spark new ideas for using a certain scaffold in a specific regenerative approach to produce the desired pulp-dentin complex.
Collapse
Affiliation(s)
- Sakshi S Jajoo
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Shweta M Chaudhary
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Krishna Patil
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Sanket Kunte
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Laxmi Lakade
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| | - Chetana Jagtap
- Department of Pedodontics, Dental College and Hospital, Bharati Vidyapeeth (Deemed to be University), Pune, Maharashtra, India
| |
Collapse
|
166
|
Kyser AJ, Masigol M, Mahmoud MY, Ryan M, Lewis WG, Lewis AL, Frieboes HB, Steinbach-Rankins JM. Fabrication and characterization of bioprints with Lactobacillus crispatus for vaginal application. J Control Release 2023; 357:545-560. [PMID: 37076014 PMCID: PMC10696519 DOI: 10.1016/j.jconrel.2023.04.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Bacterial vaginosis (BV) is characterized by low levels of lactobacilli and overgrowth of potential pathogens in the female genital tract. Current antibiotic treatments often fail to treat BV in a sustained manner, and > 50% of women experience recurrence within 6 months post-treatment. Recently, lactobacilli have shown promise for acting as probiotics by offering health benefits in BV. However, as with other active agents, probiotics often require intensive administration schedules incurring difficult user adherence. Three-dimensional (3D)-bioprinting enables fabrication of well-defined architectures with tunable release of active agents, including live mammalian cells, offering the potential for long-acting probiotic delivery. One promising bioink, gelatin alginate has been previously shown to provide structural stability, host compatibility, viable probiotic incorporation, and cellular nutrient diffusion. This study formulates and characterizes 3D-bioprinted Lactobacillus crispatus-containing gelatin alginate scaffolds for gynecologic applications. Different weight to volume (w/v) ratios of gelatin alginate were bioprinted to determine formulations with highest printing resolution, and different crosslinking reagents were evaluated for effect on scaffold integrity via mass loss and swelling measurements. Post-print viability, sustained-release, and vaginal keratinocyte cytotoxicity assays were conducted. A 10:2 (w/v) gelatin alginate formulation was selected based on line continuity and resolution, while degradation and swelling experiments demonstrated greatest structural stability with dual genipin and calcium crosslinking, showing minimal mass loss and swelling over 28 days. 3D-bioprinted L. crispatus-containing scaffolds demonstrated sustained release and proliferation of live bacteria over 28 days, without impacting viability of vaginal epithelial cells. This study provides in vitro evidence for 3D-bioprinted scaffolds as a novel strategy to sustain probiotic delivery with the ultimate goal of restoring vaginal lactobacilli following microbiological disturbances.
Collapse
Affiliation(s)
- Anthony J Kyser
- Department of Bioengineering, University of Louisville Speed School of Engineering, Louisville, KY 40202, USA.
| | - Mohammadali Masigol
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, USA.
| | - Mohamed Y Mahmoud
- Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Toxicology and Forensic Medicine, Faculty of Veterinary Medicine, Cairo University, Egypt.
| | - Mark Ryan
- Department of Bioengineering, University of Louisville Speed School of Engineering, Louisville, KY 40202, USA.
| | - Warren G Lewis
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA; Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA.
| | - Amanda L Lewis
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA; Glycobiology Research and Training Center, University of California San Diego, La Jolla, CA, USA.
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville Speed School of Engineering, Louisville, KY 40202, USA; Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; UofL Health - Brown Cancer Center, University of Louisville, KY 40202, USA.
| | - Jill M Steinbach-Rankins
- Department of Bioengineering, University of Louisville Speed School of Engineering, Louisville, KY 40202, USA; Center for Predictive Medicine, University of Louisville, Louisville, KY 40202, USA; Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY 40202, USA; Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA.
| |
Collapse
|
167
|
Choi KY, Ajiteru O, Hong H, Suh YJ, Sultan T, Lee H, Lee JS, Lee YJ, Lee OJ, Kim SH, Park CH. A digital light processing 3D-printed artificial skin model and full-thickness wound models using silk fibroin bioink. Acta Biomater 2023; 164:159-174. [PMID: 37121370 DOI: 10.1016/j.actbio.2023.04.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 03/31/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
A three-dimensional (3D) artificial skin model offers diverse platforms for skin transplantation, disease mechanisms, and biomaterial testing for skin tissue. However, implementing physiological complexes such as the neurovascular system with living cells in this stratified structure is extremely difficult. In this study, full-thickness skin models were fabricated from methacrylated silk fibroin (Silk-GMA) and gelatin (Gel-GMA) seeded with keratinocytes, fibroblasts, and vascular endothelial cells representing the epidermis and dermis layers through a digital light processing (DLP) 3D printer. Printability, mechanical properties, and cell viability of the skin hydrogels fabricated with different concentrations of Silk-GMA and Gel-GMA were analyzed to find the optimal concentrations for the 3D printing of the artificial skin model. After the skin model was DLP-3D printed using Gel-GMA 15% + Silk-GMA 5% bioink, cultured, and air-lifted for four weeks, well-proliferated keratinocytes and fibroblasts were observed in histological analysis, and increased expressions of Cytokeratin 13, Phalloidin, and CD31 were noted in immunofluorescence staining. Furthermore, full-thickness skin wound models were 3D-printed to evaluate the wound-healing capabilities of the skin hydrogel. When the epidermal growth factor (EGF) was applied, enhanced wound healing in the epidermis and dermis layer with the proliferation of keratinocytes and fibroblasts was observed. Also, the semi-quantitative reverse transcription-polymerase chain reaction revealed increased expression of Cytokeratin 13, fibroblast growth factor, and CD31 in the EGF-treated group relative to the control group. The DLP 3D-printed artificial skin model was mechanically stable and biocompatible for more than four weeks, demonstrating the potential for application in skin tissue engineering. STATEMENT OF SIGNIFICANCE: A full-thickness artificial skin model was 3D-printed in this study with a digital light processing technique using silk fibroin and gelatin, which mimics the structural and cellular compositions of the human skin. The 3D-printed skin hydrogel ensured the viability of the cells in the skin layers that proliferated well after air-lifting cultivation, shown in the histological analysis and immunofluorescence stainings. Furthermore, full-thickness skin wound models were 3D-printed to evaluate the wound healing capabilities of the skin hydrogel, which demonstrated enhanced wound healing in the epidermis and dermis layer with the application of epidermal growth factor on the wound compared to the control. The bioengineered hydrogel expands the applicability of artificial skin models for skin substitutes, wound models, and drug testing.
Collapse
Affiliation(s)
- Kyu Young Choi
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University Kangnam Sacred Heart Hospital, Seoul 07441, Republic of Korea
| | - Olatunji Ajiteru
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Heesun Hong
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ye Ji Suh
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Tipu Sultan
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Hanna Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ji Seung Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Young Jin Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Ok Joo Lee
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Soon Hee Kim
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea
| | - Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon-do, 24252, Republic of Korea; Department of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, 24252, Republic of Korea.
| |
Collapse
|
168
|
Kundak H, Bilisik K. Development of Three-Dimensional (3D) Biodegradable Polyglycolic Acid Fiber (PGA) Preforms for Scaffold Applications: Experimental Patterning and Fiber Volume Fraction-Porosity Modeling Study. Polymers (Basel) 2023; 15:polym15092083. [PMID: 37177227 PMCID: PMC10181393 DOI: 10.3390/polym15092083] [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: 02/25/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Three-dimensional (3D) biodegradable polyglycolic acid fiber (PGA) preforms were developed as temporary scaffolds for three-dimensional tissue regeneration applications. Three-dimensional biodegradable polyglycolic acid fiber (PGA) preforms including various degrees of interlaced structures called 3D plain, semi-interlaced, and orthogonal woven preforms were designed. Analytical relations and finite element model-based software (TexGen) on fiber volume fraction and porosity fraction were proposed to predict scaffolds' stiffness and strength properties considering micromechanics relations. It was revealed that yarn-to-yarn space, density, and angles of all 3D PGA fiber preforms were heterogeneous and demonstrated direction-dependent features (anisotropy). Total fiber volume fractions (Vfp) and porosity fraction (Vtpr) predicted by analytic and numerical modelling of all 3D scaffolds showed some deviations compared to the measured values. This was because yarn cross-sections in the scaffolds were changed from ideal circular yarn (fiber TOW) geometry to high-order ellipse (lenticular) due to inter-fiber pressure generated under a tensile-based macrostress environment during preform formation. Z-yarn modulus (Ez-yarn) and strength (σz-yarn) were probably critical values due to strong stiffness and strength in the through-the-thickness direction where hydrogel modulus and strengths were negligibly small. Morphology of the scaffold showed that PGA fiber sets in the preform were locally distorted, and they appeared as inconsistent and inhomogeneous continuous fiber forms. Additionally, various porosity shapes in the preform based on the virtual model featured complex shapes from nearly trapezoidal beams to partial or concave rectangular beams and ellipsoid rectangular cylinders. It was concluded that 3D polyglycolic acid fiber preforms could be a temporary supportive substrate for 3D tissue regeneration because cells in the scaffold's thickness can grow via through-the-thickness fiber (z-yarn), including various possible mechanobiology mechanisms.
Collapse
Affiliation(s)
- Hikmet Kundak
- Nano/Micro Fiber Preform Design and Composite Laboratory, Department of Textile Engineering, Faculty of Engineering, Erciyes University, Talas 38039, Kayseri, Turkey
| | - Kadir Bilisik
- Nano/Micro Fiber Preform Design and Composite Laboratory, Department of Textile Engineering, Faculty of Engineering, Erciyes University, Talas 38039, Kayseri, Turkey
- Nanotechnology Application and Research Centre (ERNAM), Erciyes University, Talas 38039, Kayseri, Turkey
| |
Collapse
|
169
|
Maresca JA, DeMel DC, Wagner GA, Haase C, Geibel JP. Three-Dimensional Bioprinting Applications for Bone Tissue Engineering. Cells 2023; 12:cells12091230. [PMID: 37174630 PMCID: PMC10177443 DOI: 10.3390/cells12091230] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/11/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
The skeletal system is a key support structure within the body. Bones have unique abilities to grow and regenerate after injury. Some injuries or degeneration of the tissues cannot rebound and must be repaired by the implantation of foreign objects following injury or disease. This process is invasive and does not always improve the quality of life of the patient. New techniques have arisen that can improve bone replacement or repair. 3D bioprinting employs a printer capable of printing biological materials in multiple directions. 3D bioprinting potentially requires multiple steps and additional support structures, which may include the use of hydrogels for scaffolding. In this review, we discuss normal bone physiology and pathophysiology and how bioprinting can be adapted to further the field of bone tissue engineering.
Collapse
Affiliation(s)
- Jamie A Maresca
- The John B. Pierce Laboratory, University of New Haven, New Haven, CT 06519, USA
| | - Derek C DeMel
- Yale School of Engineering & Applied Science, Yale University, New Haven, CT 06519, USA
| | - Grayson A Wagner
- Yale School of Engineering & Applied Science, Yale University, New Haven, CT 06519, USA
| | - Colin Haase
- The John B. Pierce Laboratory, University of New Haven, New Haven, CT 06519, USA
| | - John P Geibel
- The John B. Pierce Laboratory, University of New Haven, New Haven, CT 06519, USA
- Yale School of Engineering & Applied Science, Yale University, New Haven, CT 06519, USA
- Department of Surgery, School of Medicine, Yale University, New Haven, CT 06519, USA
| |
Collapse
|
170
|
Ming Z, Tang X, Liu J, Ruan B. Advancements in Research on Constructing Physiological and Pathological Liver Models and Their Applications Utilizing Bioprinting Technology. Molecules 2023; 28:molecules28093683. [PMID: 37175094 PMCID: PMC10180184 DOI: 10.3390/molecules28093683] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
In recent decades, significant progress has been made in liver tissue engineering through the use of 3D bioprinting technology. This technology offers the ability to create personalized biological structures with precise geometric design capabilities. The complex and multifaceted nature of liver diseases underscores the need for advanced technologies to accurately mimic the physiological and mechanical characteristics, as well as organ-level functions, of liver tissue in vitro. Bioprinting stands out as a superior option over traditional two-dimensional cell culture models and animal models due to its stronger biomimetic advantages. Through the use of bioprinting, it is possible to create liver tissue with a level of structural and functional complexity that more closely resembles the real organ, allowing for more accurate disease modeling and drug testing. As a result, it is a promising tool for restoring and replacing damaged tissue and organs in the field of liver tissue engineering and drug research. This article aims to present a comprehensive overview of the progress made in liver tissue engineering using bioprinting technology to provide valuable insights for researchers. The paper provides a detailed account of the history of liver tissue engineering, highlights the current 3D bioprinting methods and bioinks that are widely used, and accentuates the importance of existing in vitro liver tissue models based on 3D bioprinting and their biomedical applications. Additionally, the article explores the challenges faced by 3D bioprinting and predicts future trends in the field. The progress of 3D bioprinting technology is poised to bring new approaches to printing liver tissue in vitro, while offering powerful tools for drug development, testing, liver disease modeling, transplantation, and regeneration, which hold great academic and practical significance.
Collapse
Affiliation(s)
- Zibei Ming
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Xinyu Tang
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Jing Liu
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| | - Banfeng Ruan
- School of Biology, Food and Environment, Hefei University, Hefei 230601, China
| |
Collapse
|
171
|
Yunsheng D, Hui X, Jie W, Tingting Y, Naiqi K, Jiaxing H, Wei C, Yufei L, Qiang Y, Shufang W. Sustained release silicon from 3D bioprinting scaffold using silk/gelatin inks to promote osteogenesis. Int J Biol Macromol 2023; 234:123659. [PMID: 36796557 DOI: 10.1016/j.ijbiomac.2023.123659] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/20/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Repairing extensive bone defects that cannot self-heal has been a clinical challenge. The construction of scaffolds with osteogenic activity through tissue engineering can provide an effective strategy for bone regeneration. This study utilized gelatin, silk fibroin, and Si3N4 as scaffold materials to prepare silicon-functionalized biomacromolecules composite scaffolds using three-dimensional printing (3DP) technology. This system delivered positive outcomes when Si3N4 levels were 1 % (1SNS). The results showed that the scaffold had a porous reticular structure with a pore size of 600-700 μm. The Si3N4 nanoparticles were distributed uniformly in the scaffold. The scaffold could release Si ions for up to 28 days. In vitro experiments showed that the scaffold had good cytocompatibility, promoting the osteogenic differentiation of mesenchymal stem cells (MSCs). In vivo experiments on bone defects in rats showed that the 1SNS group facilitated bone regeneration. Therefore, the composite scaffold system showed potential for application in bone tissue engineering.
Collapse
Affiliation(s)
- Dong Yunsheng
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Xiao Hui
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Wang Jie
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Yang Tingting
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Kang Naiqi
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Huang Jiaxing
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Cui Wei
- Qingdao Alticera Advanced Materials Co., Ltd, 266299 Shan Dong, China
| | - Liu Yufei
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China
| | - Yang Qiang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, 300211 Tianjin, China.
| | - Wang Shufang
- State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, The College of Life Science, Nankai University, 300071 Tianjin, China.
| |
Collapse
|
172
|
Yu Q, Wang Q, Zhang L, Deng W, Cao X, Wang Z, Sun X, Yu J, Xu X. The applications of 3D printing in wound healing: the external delivery of stem cells and antibiosis. Adv Drug Deliv Rev 2023; 197:114823. [PMID: 37068658 DOI: 10.1016/j.addr.2023.114823] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
As the global number of chronic wound patients rises, the financial burden and social pressure on patients increase daily. Stem cells have emerged as promising tissue engineering seed cells due to their enriched sources, multidirectional differentiation ability, and high proliferation rate. However, delivering them in vitro for the treatment of skin injury is still challenging. In addition, bacteria from the wound site and the environment can significantly impact wound healing. In the last decade, 3D bioprinting has dramatically enriched cell delivery systems. The produced scaffolds by this technique can be precisely localized within cells and perform antibacterial actions. In this review, we summarized the 3D bioprinting-based external delivery of stem cells and their antibiosis to improve wound healing.
Collapse
Affiliation(s)
- Qingtong Yu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Qilong Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Linzhi Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Xia Cao
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Zhe Wang
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China
| | - Ximing Xu
- School of Pharmacy, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
173
|
Hao L, Zhao S, Hao S, He Y, Feng M, Zhou K, He Y, Yang J, Mao H, Gu Z. Functionalized gelatin-alginate based bioink with enhanced manufacturability and biomimicry for accelerating wound healing. Int J Biol Macromol 2023; 240:124364. [PMID: 37044319 DOI: 10.1016/j.ijbiomac.2023.124364] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/10/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
Three-dimensional (3D) bioprinting is a promising technique to construct heterogeneous architectures that mimic cell microenvironment. However, the current bioinks for 3D bioprinting usually show some limitations, such as low printing accuracy, unsatisfactory mechanical properties and compromised cytocompatibility. Herein, a novel bioink comprising hydroxyphenyl propionic acid-conjugated gelatin and tyramine-modified alginate is developed for printing 3D constructs. The bioink takes advantage of an ionic/covalent intertwined network that combines covalent bonds formed by photo-mediated redox reaction and ionic bonds formed by chelate effect. Benefiting from the thermosensitivity of gelatin and the double-crosslinking mechanism, the developed bioink shows controllable rheological behaviors, enhanced mechanical behavior, improved printing accuracy and structure stability. Moreover, the printed cell-laden hydrogels exhibit a homogeneous cell distribution and considerable cell survival because the pre-crosslinking of the bioink prevents cellular sedimentation and the visible light crosslinking mechanism preserves cell viability. Further in vivo studies demonstrate that resulting cell-laden hydrogels are beneficial for the reduction of inflammation response and the promotion of collagen deposition and angiogenesis, thereby improving the quality of skin wound healing. This convenient and effective strategy is of great significance for accelerating the development of multifunctional bioinks and broadening the biomedical applications of 3D bioprinting.
Collapse
Affiliation(s)
- Lili Hao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Shijia Zhao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Shiqi Hao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Yuxin He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Miao Feng
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Ke Zhou
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China; NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing 210000, China
| | - Jiquan Yang
- Jiangsu Key Lab of 3D Printing Equipment and Manufacturing, Nanjing Normal University, Nanjing 210046, China
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China; NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing 210000, China.
| | - Zhongwei Gu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing 211816, China; NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing 210000, China
| |
Collapse
|
174
|
Guo M, Li T, Zhang WC, Duan Q, Dong XZ, Liu J, Jin F, Zheng ML. Wetting of Cell Aggregates on Microdisk Topography Structures Achieved by Maskless Optical Projection Lithography. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2300311. [PMID: 37026658 DOI: 10.1002/smll.202300311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/08/2023] [Indexed: 06/19/2023]
Abstract
Cell aggregates as a 3D culture model can effectively mimic the physiological processes such as embryonic development, immune response, and tissue renewal in vivo. Researches show that the topography of biomaterials plays an important role in regulating cell proliferation, adhesion, and differentiation. It is of great significance to understand how cell aggregates respond to surface topography. Herein, microdisk array structures with the optimized size are used to investigate the wetting of cell aggregates. Cell aggregates exhibit complete wetting with distinct wetting velocities on the microdisk array structures of different diameters. The wetting velocity of cell aggregates reaches a maximum of 293 µm h-1 on microdisk structures with a diameter of 2 µm and is a minimum of 247 µm h-1 on microdisk structures of 20 µm diameter, which suggests that the cell-substrates adhesion energy on the latter is smaller. Actin stress fibers, focal adhesions (FAs), and cell morphology are analyzed to reveal the mechanisms of variation of wetting velocity. Furthermore, it is demonstrated that cell aggregates adopt climb and detour wetting modes on small and large-sized microdisk structures, respectively. This work reveals the response of cell aggregates to micro-scale topography, providing guidance for better understanding of tissue infiltration.
Collapse
Affiliation(s)
- Min Guo
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, P. R. China
| | - Teng Li
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, P. R. China
| | - Wei-Cai Zhang
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, P. R. China
| | - Qi Duan
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Yanqihu Campus, Beijing, 101407, P. R. China
| | - Xian-Zi Dong
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
| | - Jie Liu
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
| | - Feng Jin
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
| | - Mei-Ling Zheng
- Laboratory of Organic NanoPhotonics and CAS Key Laboratory of Bio-Inspired Materials and Interfacial Science, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, No. 29, Zhongguancun East Road, Beijing, 100190, P. R. China
| |
Collapse
|
175
|
Lee HY, Lee JW. Current Status and Future Outlook of Additive Manufacturing Technologies for the Reconstruction of the Trachea. J Funct Biomater 2023; 14:jfb14040196. [PMID: 37103286 PMCID: PMC10141199 DOI: 10.3390/jfb14040196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023] Open
Abstract
Tracheal stenosis and defects occur congenitally and in patients who have undergone tracheal intubation and tracheostomy due to long-term intensive care. Such issues may also be observed during tracheal removal during malignant head and neck tumor resection. However, to date, no treatment method has been identified that can simultaneously restore the appearance of the tracheal skeleton while maintaining respiratory function in patients with tracheal defects. Therefore, there is an urgent need to develop a method that can maintain tracheal function while simultaneously reconstructing the skeletal structure of the trachea. Under such circumstances, the advent of additive manufacturing technology that can create customized structures using patient medical image data provides new possibilities for tracheal reconstruction surgery. In this study, the three-dimensional (3D) printing and bioprinting technologies used in tracheal reconstruction are summarized, and various research results related to the reconstruction of mucous membranes, cartilage, blood vessels, and muscle tissue, which are tissues required for tracheal reconstruction, are classified. The prospects for 3D-printed tracheas in clinical studies are also described. This review serves as a guide for the development of artificial tracheas and clinical trials using 3D printing and bioprinting.
Collapse
Affiliation(s)
- Hwa-Yong Lee
- Division of Science Education, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
176
|
Li T, Ma Z, Zhang Y, Yang Z, Li W, Lu D, Liu Y, Qiang L, Wang T, Ren Y, Wang W, He H, Zhou X, Mao Y, Zhu J, Wang J, Chen X, Dai K. Regeneration of Humeral Head Using a 3D Bioprinted Anisotropic Scaffold with Dual Modulation of Endochondral Ossification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205059. [PMID: 36755334 PMCID: PMC10131811 DOI: 10.1002/advs.202205059] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Indexed: 06/18/2023]
Abstract
Tissue engineering is theoretically thought to be a promising method for the reconstruction of biological joints, and thus, offers a potential treatment alternative for advanced osteoarthritis. However, to date, no significant progress is made in the regeneration of large biological joints. In the current study, a biomimetic scaffold for rabbit humeral head regeneration consisting of heterogeneous porous architecture, various bioinks, and different hard supporting materials in the cartilage and bone regions is designed and fabricated in one step using 3D bioprinting technology. Furthermore, orchestrated dynamic mechanical stimulus combined with different biochemical cues (parathyroid hormone [PTH] and chemical component hydroxyapatite [HA] in the outer and inner region, respectively) are used for dual regulation of endochondral ossification. Specifically, dynamic mechanical stimulus combined with growth factor PTH in the outer region inhibits endochondral ossification and results in cartilage regeneration, whereas dynamic mechanical stimulus combined with HA in the inner region promotes endochondral ossification and results in efficient subchondral bone regeneration. The strategy established in this study with the dual modulation of endochondral ossification for 3D bioprinted anisotropic scaffolds represents a versatile and scalable approach for repairing large joints.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Zhengjiang Ma
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Yuxin Zhang
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Zezheng Yang
- Department of OrthopedicsThe Fifth People's Hospital of ShanghaiFudan UniversityMinhang DistrictShanghai200240P. R. China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Dezhi Lu
- School of MedicineShanghai UniversityJing An DistrictShanghai200444China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Lei Qiang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Ya Ren
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Wenhao Wang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Hongtao He
- The Third Ward of Department of OrthopedicsThe Second Hospital of Dalian Medical UniversityNo. 467, Zhongshan Road, Shahekou DistrictDalianLiaoning Province116000P. R. China
| | - Xiaojun Zhou
- College of Biological Science and Medical EngineeringState Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsDonghua UniversityShanghai201620P. R. China
| | - Yuanqing Mao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Junfeng Zhu
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Xiaodong Chen
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| |
Collapse
|
177
|
Li ZA, Sant S, Cho SK, Goodman SB, Bunnell BA, Tuan RS, Gold MS, Lin H. Synovial joint-on-a-chip for modeling arthritis: progress, pitfalls, and potential. Trends Biotechnol 2023; 41:511-527. [PMID: 35995600 PMCID: PMC9938846 DOI: 10.1016/j.tibtech.2022.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 12/30/2022]
Abstract
Disorders of the synovial joint, such as osteoarthritis (OA) and rheumatoid arthritis (RA), afflict a substantial proportion of the global population. However, current clinical management has not been focused on fully restoring the native function of joints. Organ-on-chip (OoC), also called a microphysiological system, which typically accommodates multiple human cell-derived tissues/organs under physiological culture conditions, is an emerging platform that potentially overcomes the limitations of current models in developing therapeutics. Herein, we review major steps in the generation of OoCs for studying arthritis, discuss the challenges faced when these novel platforms enter the next phase of development and application, and present the potential for OoC technology to investigate the pathogenesis of joint diseases and the development of efficacious therapies.
Collapse
Affiliation(s)
- Zhong Alan Li
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sung Kwon Cho
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Stuart B Goodman
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rocky S Tuan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
178
|
Hofmann E, Fink J, Pignet AL, Schwarz A, Schellnegger M, Nischwitz SP, Holzer-Geissler JCJ, Kamolz LP, Kotzbeck P. Human In Vitro Skin Models for Wound Healing and Wound Healing Disorders. Biomedicines 2023; 11:biomedicines11041056. [PMID: 37189674 DOI: 10.3390/biomedicines11041056] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
Skin wound healing is essential to health and survival. Consequently, high amounts of research effort have been put into investigating the cellular and molecular components involved in the wound healing process. The use of animal experiments has contributed greatly to the knowledge of wound healing, skin diseases, and the exploration of treatment options. However, in addition to ethical concerns, anatomical and physiological inter-species differences often influence the translatability of animal-based studies. Human in vitro skin models, which include essential cellular and structural components for wound healing analyses, would improve the translatability of results and reduce animal experiments during the preclinical evaluation of novel therapy approaches. In this review, we summarize in vitro approaches, which are used to study wound healing as well as wound healing-pathologies such as chronic wounds, keloids, and hypertrophic scars in a human setting.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna-Lisa Pignet
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Marlies Schellnegger
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Sebastian P Nischwitz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Judith C J Holzer-Geissler
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
179
|
Derman ID, Yeo M, Castaneda DC, Callender M, Horvath M, Mo Z, Xiong R, Fleming E, Chen P, Peeples ME, Palucka K, Oh J, Ozbolat IT. High-Throughput Bioprinting of the Nasal Epithelium using Patient-derived Nasal Epithelial Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534723. [PMID: 37034627 PMCID: PMC10081172 DOI: 10.1101/2023.03.29.534723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Human nasal epithelial cells (hNECs) are an essential cell source for the reconstruction of the respiratory pseudostratified columnar epithelium composed of multiple cell types in the context of infection studies and disease modeling. Hitherto, manual seeding has been the dominant method for creating nasal epithelial tissue models. However, the manual approach is slow, low-throughput and has limitations in terms of achieving the intricate 3D structure of the natural nasal epithelium in a uniform manner. 3D Bioprinting has been utilized to reconstruct various epithelial tissue models, such as cutaneous, intestinal, alveolar, and bronchial epithelium, but there has been no attempt to use of 3D bioprinting technologies for reconstruction of the nasal epithelium. In this study, for the first time, we demonstrate the reconstruction of the nasal epithelium with the use of primary hNECs deposited on Transwell inserts via droplet-based bioprinting (DBB), which enabled high-throughput fabrication of the nasal epithelium in Transwell inserts of 24-well plates. DBB of nasal progenitor cells ranging from one-tenth to one-half of the cell seeding density employed during the conventional cell seeding approach enabled a high degree of differentiation with the presence of cilia and tight-junctions over a 4-week air-liquid interface culture. Single cell RNA sequencing of these cultures identified five major epithelial cells populations, including basal, suprabasal, goblet, club, and ciliated cells. These cultures recapitulated the pseudostratified columnar epithelial architecture present in the native nasal epithelium and were permissive to respiratory virus infection. These results denote the potential of 3D bioprinting for high-throughput fabrication of nasal epithelial tissue models not only for infection studies but also for other purposes such as disease modeling, immunological studies, and drug screening.
Collapse
|
180
|
Luo Y, Liu Y, Wang B, Tu X. CHIR99021-Treated Osteocytes with Wnt Activation in 3D-Printed Module Form an Osteogenic Microenvironment for Enhanced Osteogenesis and Vasculogenesis. Int J Mol Sci 2023; 24:ijms24066008. [PMID: 36983081 PMCID: PMC10052982 DOI: 10.3390/ijms24066008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Finding a bone implant that has high bioactivity that can safely drive stem cell differentiation and simulate a real in vivo microenvironment is a challenge for bone tissue engineering. Osteocytes significantly regulate bone cell fate, and Wnt-activated osteocytes can reversely regulate bone formation by regulating bone anabolism, which may improve the biological activity of bone implants. To achieve a safe application, we used the Wnt agonist CHIR99021 (C91) to treat MLO-Y4 for 24 h, in a co-culture with ST2 for 3 days after withdrawal. We found that the expression of Runx2 and Osx increased, promoted osteogenic differentiation, and inhibited adipogenic differentiation in the ST2 cells, and these effects were eliminated by the triptonide. Therefore, we hypothesized that C91-treated osteocytes form an osteogenic microenvironment (COOME). Subsequently, we constructed a bio-instructive 3D printing system to verify the function of COOME in 3D modules that mimic the in vivo environment. Within PCI3D, COOME increased the survival and proliferation rates to as high as 92% after 7 days and promoted ST2 cell differentiation and mineralization. Simultaneously, we found that the COOME-conditioned medium also had the same effects. Therefore, COOME promotes ST2 cell osteogenic differentiation both directly and indirectly. It also promotes HUVEC migration and tube formation, which can be explained by the high expression of Vegf. Altogether, these results indicate that COOME, combined with our independently developed 3D printing system, can overcome the poor cell survival and bioactivity of orthopedic implants and provide a new method for clinical bone defect repair.
Collapse
Affiliation(s)
- Yisheng Luo
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yangxi Liu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Bo Wang
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xiaolin Tu
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
181
|
Lu Y, Cheng D, Niu B, Wang X, Wu X, Wang A. Properties of Poly (Lactic-co-Glycolic Acid) and Progress of Poly (Lactic-co-Glycolic Acid)-Based Biodegradable Materials in Biomedical Research. Pharmaceuticals (Basel) 2023; 16:ph16030454. [PMID: 36986553 PMCID: PMC10058621 DOI: 10.3390/ph16030454] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
In recent years, biodegradable polymers have gained the attention of many researchers for their promising applications, especially in drug delivery, due to their good biocompatibility and designable degradation time. Poly (lactic-co-glycolic acid) (PLGA) is a biodegradable functional polymer made from the polymerization of lactic acid (LA) and glycolic acid (GA) and is widely used in pharmaceuticals and medical engineering materials because of its biocompatibility, non-toxicity, and good plasticity. The aim of this review is to illustrate the progress of research on PLGA in biomedical applications, as well as its shortcomings, to provide some assistance for its future research development.
Collapse
Affiliation(s)
- Yue Lu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Dongfang Cheng
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Baohua Niu
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Xiuzhi Wang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xiaxia Wu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Aiping Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai 264005, China
- Correspondence:
| |
Collapse
|
182
|
Liao W, Duan X, Xie F, Zheng D, Yang P, Wang X, Hu Z. 3D-bioprinted double-crosslinked angiogenic alginate/chondroitin sulfate patch for diabetic wound healing. Int J Biol Macromol 2023; 236:123952. [PMID: 36894059 DOI: 10.1016/j.ijbiomac.2023.123952] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/09/2023]
Abstract
Improving chronic wound healing remains a challenge in the clinical practice. In this study, we developed double-crosslinked angiogenic 3D-bioprinted patches for diabetic wound healing by the photocovalent crosslinking of vascular endothelial growth factor (VEGF) using ultraviolet (UV) irradiation. 3D printing technology can precisely customize the structure and composition of patches to meet different clinical requirements. The biological polysaccharide alginate and chondroitin sulfate methacryloyl were used as biomaterials to construct the biological patch, which could be crosslinked using calcium ion crosslinking and photocrosslinking, thereby improving its mechanical properties. More importantly, acrylylated VEGF could be easily and rapidly photocrosslinked under UV irradiation, which simplified the step of chemically coupling growth factors and prolonged VEGF release time. These characteristics suggest that 3D-bioprinted double-crosslinked angiogenic patches are ideal candidates for diabetic wound healing and other tissue engineering applications.
Collapse
Affiliation(s)
- Weifang Liao
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Nanchang, Jiangxi, China
| | - Xunxin Duan
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Nanchang, Jiangxi, China
| | - Fusheng Xie
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Nanchang, Jiangxi, China
| | - Dongxi Zheng
- School of Mechanical and Intelligent Manufacturing, Jiujiang University, Jiujiang, Jiangxi, China
| | - Pu Yang
- Department of Plastic and Aesthetic (Burn) Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiangguo Wang
- Animal Science and Technology College, Beijing University of Agriculture, Beijing, China.
| | - Zhijian Hu
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China; Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Nanchang, Jiangxi, China.
| |
Collapse
|
183
|
Hofmann E, Schwarz A, Fink J, Kamolz LP, Kotzbeck P. Modelling the Complexity of Human Skin In Vitro. Biomedicines 2023; 11:biomedicines11030794. [PMID: 36979772 PMCID: PMC10045055 DOI: 10.3390/biomedicines11030794] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023] Open
Abstract
The skin serves as an important barrier protecting the body from physical, chemical and pathogenic hazards as well as regulating the bi-directional transport of water, ions and nutrients. In order to improve the knowledge on skin structure and function as well as on skin diseases, animal experiments are often employed, but anatomical as well as physiological interspecies differences may result in poor translatability of animal-based data to the clinical situation. In vitro models, such as human reconstructed epidermis or full skin equivalents, are valuable alternatives to animal experiments. Enormous advances have been achieved in establishing skin models of increasing complexity in the past. In this review, human skin structures are described as well as the fast evolving technologies developed to reconstruct the complexity of human skin structures in vitro.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
184
|
Liu H, Gong Y, Zhang K, Ke S, Wang Y, Wang J, Wang H. Recent Advances in Decellularized Matrix-Derived Materials for Bioink and 3D Bioprinting. Gels 2023; 9:gels9030195. [PMID: 36975644 PMCID: PMC10048399 DOI: 10.3390/gels9030195] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
As an emerging 3D printing technology, 3D bioprinting has shown great potential in tissue engineering and regenerative medicine. Decellularized extracellular matrices (dECM) have recently made significant research strides and have been used to create unique tissue-specific bioink that can mimic biomimetic microenvironments. Combining dECMs with 3D bioprinting may provide a new strategy to prepare biomimetic hydrogels for bioinks and hold the potential to construct tissue analogs in vitro, similar to native tissues. Currently, the dECM has been proven to be one of the fastest growing bioactive printing materials and plays an essential role in cell-based 3D bioprinting. This review introduces the methods of preparing and identifying dECMs and the characteristic requirements of bioink for use in 3D bioprinting. The most recent advances in dECM-derived bioactive printing materials are then thoroughly reviewed by examining their application in the bioprinting of different tissues, such as bone, cartilage, muscle, the heart, the nervous system, and other tissues. Finally, the potential of bioactive printing materials generated from dECM is discussed.
Collapse
Affiliation(s)
- Huaying Liu
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing 100091, China
| | - Yuxuan Gong
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing 100091, China
| | - Kaihui Zhang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing 100091, China
- College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Shen Ke
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing 100091, China
| | - Yue Wang
- National Institutes for Food and Drug Control, Beijing 102629, China
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
- Correspondence: (J.W.); (H.W.)
| | - Haibin Wang
- College of Life Sciences and Bioengineering, School of Physical Science and Engineering, Beijing Jiaotong University, Beijing 100091, China
- Correspondence: (J.W.); (H.W.)
| |
Collapse
|
185
|
Dong Q, Su X, Li X, Zhou H, Jian H, Bai S, Yin J, You Q. In vitro construction of lung cancer organoids by 3D bioprinting for drug evaluation. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2023.131288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2023]
|
186
|
Wu CA, Zhu Y, Venkatesh A, Stark CJ, Lee SH, Woo YJ. Optimization of Freeform Reversible Embedding of Suspended Hydrogel Microspheres for Substantially Improved Three-Dimensional Bioprinting Capabilities. Tissue Eng Part C Methods 2023; 29:85-94. [PMID: 36719778 PMCID: PMC10024587 DOI: 10.1089/ten.tec.2022.0214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/23/2023] [Indexed: 02/01/2023] Open
Abstract
Three-dimensional (3D) bioprinting demonstrates technology that is capable of producing structures comparable to native tissues in the human body. The freeform reversible embedding of suspended hydrogels (FRESH) technique involves hydrogel-based bio-inks printed within a thermo-reversible support bath to provide mechanical strength to the printed construct. Smaller and more uniform microsphere sizes of FRESH were reported to aid in enhancing printing resolution and construct accuracy. Therefore, we sought to optimize the FRESH generation protocol, particularly by varying stir speed and stir duration, in hopes to further improve microsphere size and uniformity. We observed optimal conditions at a stir speed of 600 rpm and stir duration for 20 h that generated the smallest microspheres with the best uniformity. Comparison of using the optimized FRESH to the commercial FRESH LifeSupport to bioprint single filament and geometrical constructs revealed reduced single filament diameters and higher angular precision in the optimized FRESH bio-printed constructs compared with those printed in the commercial FRESH. Overall, our refinement of the FRESH manufacturing protocol represents an important step toward enhancing 3D bioprinting resolution and construct fidelity. Improving such technologies allows for the fabrication of highly accurate constructs with anatomical properties similar to native counterparts. Such work has significant implications in the field of tissue engineering for producing accurate human organ model systems. Impact statement Freeform reversible embedding of suspended hydrogels (FRESH) is a method of sacrificial three-dimensional (3D) bioprinting that offers support to reinforce bio-ink extrusion during printing. During FRESH generation, the stir speed and stir duration of the mixture can significantly impact FRESH microsphere characteristics. In this study, we optimized FRESH microspheres to significantly improve resolution and accuracy in bioprinting. This advancement in FRESH-based 3D bioprinting technologies allows for the fabrication of highly accurate constructs with anatomical properties similar to native counterparts and has significant implications in the field of tissue engineering and translational medicine.
Collapse
Affiliation(s)
- Catherine A. Wu
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| | - Akshay Venkatesh
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
| | - Charles J. Stark
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
| | - Seung Hyun Lee
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
| | - Y. Joseph Woo
- Department of Cardiothoracic Surgery and Stanford University, Stanford, California, USA
- Department of Bioengineering, Stanford University, Stanford, California, USA
| |
Collapse
|
187
|
Mirdamadi ES, Khosrowpour Z, Jafari D, Gholipourmalekabadi M, Solati-Hashjin M. 3D-printed PLA/Gel hybrid in liver tissue engineering: Effects of architecture on biological functions. Biotechnol Bioeng 2023; 120:836-851. [PMID: 36479982 DOI: 10.1002/bit.28301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/28/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
The liver is one of the vital organs in the body, and the gold standard of treatment for liver function impairment is liver transplantation, which poses many challenges. The specific three-dimensional (3D) structure of liver, which significantly impacts the growth and function of its cells, has made biofabrication with the 3D printing of scaffolds suitable for this approach. In this study, to investigate the effect of scaffold geometry on the performance of HepG2 cells, poly-lactic acid (PLA) polymer was used as the input of the fused deposition modeling (FDM) 3D-printing machine. Samples with simple square and bioinspired hexagonal cross-sectional designs were printed. One percent and 2% of gelatin coating were applied to the 3D printed PLA to improve the wettability and surface properties of the scaffold. Scanning electron microscopy pictures were used to analyze the structural properties of PLA-Gel hybrid scaffolds, energy dispersive spectroscopy to investigate the presence of gelatin, water contact angle measurement for wettability, and weight loss for degradation. In vitro tests were performed by culturing HepG2 cells on the scaffold to evaluate the cell adhesion, viability, cytotoxicity, and specific liver functions. Then, high-precision scaffolds were printed and the presence of gelatin was detected. Also, the effect of geometry on cell function was confirmed in viability, adhesion, and functional tests. The albumin and urea production of the Hexagonal PLA scaffold was about 1.22 ± 0.02-fold higher than the square design in 3 days. This study will hopefully advance our understanding of liver tissue engineering toward a promising perspective for liver regeneration.
Collapse
Affiliation(s)
- Elnaz Sadat Mirdamadi
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.,Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, USA
| | - Zahra Khosrowpour
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Davod Jafari
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Solati-Hashjin
- BioFabrication Lab (BFL), Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
188
|
Wang J, Qin X, Xia S, Liu S, Ren H. Orthotopic implantable liver decellularized scaffold for acute liver failure. ENGINEERED REGENERATION 2023; 4:12-19. [DOI: 10.1016/j.engreg.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023] Open
|
189
|
Shopova D, Yaneva A, Bakova D, Mihaylova A, Kasnakova P, Hristozova M, Sbirkov Y, Sarafian V, Semerdzhieva M. (Bio)printing in Personalized Medicine—Opportunities and Potential Benefits. Bioengineering (Basel) 2023; 10:bioengineering10030287. [PMID: 36978678 PMCID: PMC10045778 DOI: 10.3390/bioengineering10030287] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
The global development of technologies now enters areas related to human health, with a transition from conventional to personalized medicine that is based to a significant extent on (bio)printing. The goal of this article is to review some of the published scientific literature and to highlight the importance and potential benefits of using 3D (bio)printing techniques in contemporary personalized medicine and also to offer future perspectives in this research field. The article is prepared according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Web of Science, PubMed, Scopus, Google Scholar, and ScienceDirect databases were used in the literature search. Six authors independently performed the search, study selection, and data extraction. This review focuses on 3D bio(printing) in personalized medicine and provides a classification of 3D bio(printing) benefits in several categories: overcoming the shortage of organs for transplantation, elimination of problems due to the difference between sexes in organ transplantation, reducing the cases of rejection of transplanted organs, enhancing the survival of patients with transplantation, drug research and development, elimination of genetic/congenital defects in tissues and organs, and surgery planning and medical training for young doctors. In particular, we highlight the benefits of each 3D bio(printing) applications included along with the associated scientific reports from recent literature. In addition, we present an overview of some of the challenges that need to be overcome in the applications of 3D bioprinting in personalized medicine. The reviewed articles lead to the conclusion that bioprinting may be adopted as a revolution in the development of personalized, medicine and it has a huge potential in the near future to become a gold standard in future healthcare in the world.
Collapse
Affiliation(s)
- Dobromira Shopova
- Department of Prosthetic Dentistry, Faculty of Dental Medicine, Medical University, 4000 Plovdiv, Bulgaria
- Correspondence: ; Tel.: +359-887417078
| | - Antoniya Yaneva
- Department of Medical Informatics, Biostatistics and eLearning, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| | - Desislava Bakova
- Department of Healthcare Management, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| | - Anna Mihaylova
- Department of Healthcare Management, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| | - Petya Kasnakova
- Department of Healthcare Management, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| | - Maria Hristozova
- Department of Healthcare Management, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| | - Yordan Sbirkov
- Department of Medical Biology, Medical University, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University, 4000 Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Medical University, 4000 Plovdiv, Bulgaria
- Research Institute, Medical University, 4000 Plovdiv, Bulgaria
| | - Mariya Semerdzhieva
- Department of Healthcare Management, Faculty of Public Health, Medical University, 4000 Plovdiv, Bulgaria
| |
Collapse
|
190
|
Jin Q, Yu C, Xu L, Zhang G, Ju J, Hou R. Combined light-cured and sacrificial hydrogels for fabrication of small-diameter bionic vessels by 3D bioprinting. Technol Health Care 2023:THC220393. [PMID: 36872804 DOI: 10.3233/thc-220393] [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: 03/06/2023]
Abstract
BACKGROUND Bionic grafts can replace autologous tissue through tissue engineering in cases of cardiovascular disease. However, small-diameter vessel grafts remain challenging to precellularize. OBJECTIVE Bionic small-diameter vessels with endothelial and smooth muscle cells (SMCs) manufactured with a novel approach. METHODS A 1-mm-diameter bionic blood vessel was constructed by combining light-cured hydrogel gelatin-methacryloyl (GelMA) with sacrificial hydrogel Pluronic F127. Mechanical properties of GelMA (Young's modulus and tensile stress) were tested. Cell viability and proliferation were detected using Live/dead staining and CCK-8 assays, respectively. The histology and function of the vessels were observed using hematoxylin and eosin and immunofluorescence staining. RESULTS GelMA and Pluronic were printed together using extrusion. The temporary Pluronic support was removed by cooling during GelMA crosslinking, yielding a hollow tubular construct. A bionic bilayer vascular structure was fabricated by loading SMCs into the GelMA bioink, followed by perfusion with endothelial cells. In the structure, both cell types maintained good cell viability. The vessel showed good histological morphology and function. CONCLUSION Using light-cured and sacrificial hydrogels, we formed a small ca bionic vessel with a small caliber containing SMCs and endothelial cells, demonstrating an innovative approach for construction of bionic vascular tissues.
Collapse
Affiliation(s)
- Qianheng Jin
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China.,Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chenghao Yu
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China.,Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Lei Xu
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China
| | - Guangliang Zhang
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China
| | - Jihui Ju
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China
| | - Ruixing Hou
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.,Suzhou Ruihua Orthopedic Hospital, Suzhou, Jiangsu, China
| |
Collapse
|
191
|
Abstract
Heart disease is a significant burden on global health care systems and is a leading cause of death each year. To improve our understanding of heart disease, high quality disease models are needed. These will facilitate the discovery and development of new treatments for heart disease. Traditionally, researchers have relied on 2D monolayer systems or animal models of heart disease to elucidate pathophysiology and drug responses. Heart-on-a-chip (HOC) technology is an emerging field where cardiomyocytes among other cell types in the heart can be used to generate functional, beating cardiac microtissues that recapitulate many features of the human heart. HOC models are showing great promise as disease modeling platforms and are poised to serve as important tools in the drug development pipeline. By leveraging advances in human pluripotent stem cell-derived cardiomyocyte biology and microfabrication technology, diseased HOCs are highly tuneable and can be generated via different approaches such as: using cells with defined genetic backgrounds (patient-derived cells), adding small molecules, modifying the cells' environment, altering cell ratio/composition of microtissues, among others. HOCs have been used to faithfully model aspects of arrhythmia, fibrosis, infection, cardiomyopathies, and ischemia, to name a few. In this review, we highlight recent advances in disease modeling using HOC systems, describing instances where these models outperformed other models in terms of reproducing disease phenotypes and/or led to drug development.
Collapse
Affiliation(s)
- Omar Mourad
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada.,Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Ryan Yee
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada.,Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Mengyuan Li
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada.,Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada
| | - Sara S Nunes
- Toronto General Hospital Research Institute (O.M., R.Y., M.L., S.S.N.), University Health Network, Toronto, Canada.,Ajmera Transplant Center (S.S.N.), University Health Network, Toronto, Canada.,Institute of Biomedical Engineering (O.M., R.Y., M.L., S.S.N.), University of Toronto, Canada.,Department of Laboratory Medicine and Pathobiology (S.S.N.), University of Toronto, Canada.,Heart and Stroke/Richard Lewar Centre of Excellence (S.S.N.), University of Toronto, Canada
| |
Collapse
|
192
|
Yan Y, Ji Q, Fu R, Liu C, Yang J, Yin X, Li Q, Huang R. Biomaterials and tissue engineering strategies for posterior lamellar eyelid reconstruction: Replacement or regeneration? Bioeng Transl Med 2023. [DOI: 10.1002/btm2.10497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Affiliation(s)
- Yuxin Yan
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiumei Ji
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rao Fu
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Chuanqi Liu
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Jing Yang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Xiya Yin
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Plastic and Burn Surgery West China Hospital, Sichuan University Chengdu China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ru‐Lin Huang
- Department of Plastic and Reconstructive Surgery Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
193
|
Zhang C, Wang G, Lin H, Shang Y, Liu N, Zhen Y, An Y. Cartilage 3D bioprinting for rhinoplasty using adipose-derived stem cells as seed cells: Review and recent advances. Cell Prolif 2023; 56:e13417. [PMID: 36775884 PMCID: PMC10068946 DOI: 10.1111/cpr.13417] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/10/2023] [Accepted: 01/18/2023] [Indexed: 02/14/2023] Open
Abstract
Nasal deformities due to various causes affect the aesthetics and use of the nose, in which case rhinoplasty is necessary. However, the lack of cartilage for grafting has been a major problem and tissue engineering seems to be a promising solution. 3D bioprinting has become one of the most advanced tissue engineering methods. To construct ideal cartilage, bio-ink, seed cells, growth factors and other methods to promote chondrogenesis should be considered and weighed carefully. With continuous progress in the field, bio-ink choices are becoming increasingly abundant, from a single hydrogel to a combination of hydrogels with various characteristics, and more 3D bioprinting methods are also emerging. Adipose-derived stem cells (ADSCs) have become one of the most popular seed cells in cartilage 3D bioprinting, owing to their abundance, excellent proliferative potential, minimal morbidity during harvest and lack of ethical considerations limitations. In addition, the co-culture of ADSCs and chondrocytes is commonly used to achieve better chondrogenesis. To promote chondrogenic differentiation of ADSCs and construct ideal highly bionic tissue-engineered cartilage, researchers have used a variety of methods, including adding appropriate growth factors, applying biomechanical stimuli and reducing oxygen tension. According to the process and sequence of cartilage 3D bioprinting, this review summarizes and discusses the selection of hydrogel and seed cells (centered on ADSCs), the design of printing, and methods for inducing the chondrogenesis of ADSCs.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Guanhuier Wang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Hongying Lin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yujia Shang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Na Liu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China.,Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, China
| |
Collapse
|
194
|
Gao J, Li M, Cheng J, Liu X, Liu Z, Liu J, Tang P. 3D-Printed GelMA/PEGDA/F127DA Scaffolds for Bone Regeneration. J Funct Biomater 2023; 14:jfb14020096. [PMID: 36826895 PMCID: PMC9962173 DOI: 10.3390/jfb14020096] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
Tissue-engineered scaffolds are an effective method for the treatment of bone defects, and their structure and function are essential for bone regeneration. Digital light processing (DLP) printing technology has been widely used in bone tissue engineering (BTE) due to its high printing resolution and gentle printing process. As commonly used bioinks, synthetic polymers such as polyethylene glycol diacrylate (PEGDA) and Pluronic F127 diacrylate (F127DA) have satisfactory printability and mechanical properties but usually lack sufficient adhesion to cells and tissues. Here, a compound BTE scaffold based on PEGDA, F127DA, and gelatin methacrylate (GelMA) was successfully prepared using DLP printing technology. The scaffold not only facilitated the adhesion and proliferation of cells, but also effectively promoted the osteogenic differentiation of mesenchymal stem cells in an osteoinductive environment. Moreover, the bone tissue volume/total tissue volume (BV/TV) of the GelMA/PEGDA/F127DA (GPF) scaffold in vivo was 49.75 ± 8.50%, higher than the value of 37.10 ± 7.27% for the PEGDA/F127DA (PF) scaffold and 20.43 ± 2.08% for the blank group. Therefore, the GPF scaffold prepared using DLP printing technology provides a new approach to the treatment of bone defects.
Collapse
Affiliation(s)
- Jianpeng Gao
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Ming Li
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Junyao Cheng
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Xiao Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- Medical School of Chinese PLA, Beijing 100039, China
| | - Zhongyang Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
| | - Jianheng Liu
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Correspondence: (J.L.); (P.T.)
| | - Peifu Tang
- Department of Orthopaedics, Chinese PLA General Hospital, Beijing 100039, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing 100853, China
- Correspondence: (J.L.); (P.T.)
| |
Collapse
|
195
|
Chen L, Wei L, Su X, Qin L, Xu Z, Huang X, Chen H, Hu N. Preparation and Characterization of Biomimetic Functional Scaffold with Gradient Structure for Osteochondral Defect Repair. Bioengineering (Basel) 2023; 10:bioengineering10020213. [PMID: 36829707 PMCID: PMC9952804 DOI: 10.3390/bioengineering10020213] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
Osteochondral (OC) defects cannot adequately repair themselves due to their sophisticated layered structure and lack of blood supply in cartilage. Although therapeutic interventions are reaching an advanced stage, current clinical therapies to repair defects are in their infancy. Among the possible therapies, OC tissue engineering has shown considerable promise, and multiple approaches utilizing scaffolds, cells, and bioactive factors have been pursued. The most recent trend in OC tissue engineering has been to design gradient scaffolds using different materials and construction strategies (such as bi-layered, multi-layered, and continuous gradient structures) to mimic the physiological and mechanical properties of OC tissues while further enabling OC repair. This review focuses specifically on design and construction strategies for gradient scaffolds and their role in the successful engineering of OC tissues. The current dilemmas in the field of OC defect repair and the efforts of tissue engineering to address these challenges were reviewed. In addition, the advantages and limitations of the typical fabrication techniques for gradient scaffolds were discussed, with examples of recent studies summarizing the future prospects for integrated gradient scaffold construction. This updated and enlightening review could provide insights into our current understanding of gradient scaffolds in OC tissue engineering.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiao Huang
- Correspondence: (X.H.); (H.C.); (N.H.); Tel.: +86-023-89011202 (X.H. & H.C. & N.H.)
| | - Hong Chen
- Correspondence: (X.H.); (H.C.); (N.H.); Tel.: +86-023-89011202 (X.H. & H.C. & N.H.)
| | - Ning Hu
- Correspondence: (X.H.); (H.C.); (N.H.); Tel.: +86-023-89011202 (X.H. & H.C. & N.H.)
| |
Collapse
|
196
|
Mao X, Wang Z. Research Progress of Three-Dimensional Bioprinting Artificial Cardiac Tissue. Tissue Eng Regen Med 2023; 20:1-9. [PMID: 36401767 PMCID: PMC9852375 DOI: 10.1007/s13770-022-00495-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular disease is one of the main diseases that endanger human life and health, and heart failure often occurs when the cardiovascular disease develops to the end-stage. Heart transplantation is the most effective treatment. However, there has always been a shortage of living heart organs. With the development of regenerative medicine, researchers have turned to bioprinting technology that can build tissues and organs in vitro. A large number of relevant literature on three-dimensional (3D) bioprinted hearts were searched and screened in Google Scholar. 3D bioprinting technology can accurately print biomaterials containing living cells into 3D functional living tissues, providing a feasible solution to the shortage of transplantable organs. As one of the most important organs in the human body, the research on 3D bioprinting of the heart has currently become a hot topic. This paper briefly overviews 3D bioprinting technology and the progress in bioprinting cardiac tissue. It is believed that in the future, bio-printed hearts will become a reality, making a new way of providing artificial organs for heart transplantation.
Collapse
Affiliation(s)
- Xin Mao
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, People's Republic of China
| | - Zhehui Wang
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, People's Republic of China.
| |
Collapse
|
197
|
Wanigasekara J, Cullen PJ, Bourke P, Tiwari B, Curtin JF. Advances in 3D culture systems for therapeutic discovery and development in brain cancer. Drug Discov Today 2023; 28:103426. [PMID: 36332834 DOI: 10.1016/j.drudis.2022.103426] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
This review focuses on recent advances in 3D culture systems that promise more accurate therapeutic models of the glioblastoma multiforme (GBM) tumor microenvironment (TME), such as the unique anatomical, cellular, and molecular features evident in human GBM. The key components of a GBM TME are outlined, including microbiomes, vasculature, extracellular matrix (ECM), infiltrating parenchymal and peripheral immune cells and molecules, and chemical gradients. 3D culture systems are evaluated against 2D culture systems and in vivo animal models. The main 3D culture techniques available are compared, with an emphasis on identifying key gaps in knowledge for the development of suitable platforms to accurately model the intricate components of the GBM TME.
Collapse
Affiliation(s)
- Janith Wanigasekara
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Environmental Sustainability and Health Institute (ESHI), Technological University Dublin, Dublin, Ireland; Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland; FOCAS Research Institute, Technological University Dublin, Dublin, Ireland.
| | - Patrick J Cullen
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
| | - Paula Bourke
- School of Biosystems and Food Engineering, University College Dublin, Dublin, Ireland
| | - Brijesh Tiwari
- Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - James F Curtin
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Environmental Sustainability and Health Institute (ESHI), Technological University Dublin, Dublin, Ireland; FOCAS Research Institute, Technological University Dublin, Dublin, Ireland; Faculty of Engineering and Built Environment, Technological University Dublin, Dublin, Ireland.
| |
Collapse
|
198
|
Binelli MR, Kan A, Rozas LEA, Pisaturo G, Prakash N, Studart AR. Complex Living Materials Made by Light-Based Printing of Genetically Programmed Bacteria. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207483. [PMID: 36444840 DOI: 10.1002/adma.202207483] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/23/2022] [Indexed: 06/16/2023]
Abstract
Living materials with embedded microorganisms can genetically encode attractive sensing, self-repairing, and responsive functionalities for applications in medicine, robotics, and infrastructure. While the synthetic toolbox for genetically engineering bacteria continues to expand, technologies to shape bacteria-laden living materials into complex 3D geometries are still rather limited. Here, it is shown that bacteria-laden hydrogels can be shaped into living materials with unusual architectures and functionalities using readily available light-based printing techniques. Bioluminescent and melanin-producing bacteria are used to create complex materials with autonomous chemical-sensing capabilities by harnessing the metabolic activity of wild-type and engineered microorganisms. The shaping freedom offered by printing technologies and the rich biochemical diversity available in bacteria provides ample design space for the creation and exploration of complex living materials with programmable functionalities for a broad range of applications.
Collapse
Affiliation(s)
- Marco R Binelli
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| | - Anton Kan
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| | - Luis E A Rozas
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| | - Giovanni Pisaturo
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| | - Namita Prakash
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| | - André R Studart
- Complex Materials, Department of Materials, ETH Zürich, Vladimir-Prelog-Weg 5, Zürich, 8093, Switzerland
| |
Collapse
|
199
|
Cadamuro F, Nicotra F, Russo L. 3D printed tissue models: From hydrogels to biomedical applications. J Control Release 2023; 354:726-745. [PMID: 36682728 DOI: 10.1016/j.jconrel.2023.01.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 01/09/2023] [Accepted: 01/16/2023] [Indexed: 01/24/2023]
Abstract
The development of new advanced constructs resembling structural and functional properties of human organs and tissues requires a deep knowledge of the morphological and biochemical properties of the extracellular matrices (ECM), and the capacity to reproduce them. Manufacturing technologies like 3D printing and bioprinting represent valuable tools for this purpose. This review will describe how morphological and biochemical properties of ECM change in different tissues, organs, healthy and pathological states, and how ECM mimics with the required properties can be generated by 3D printing and bioprinting. The review describes and classifies the polymeric materials of natural and synthetic origin exploited to generate the hydrogels acting as "inks" in the 3D printing process, with particular emphasis on their functionalization allowing crosslinking and conjugation with signaling molecules to develop bio-responsive and bio-instructive ECM mimics.
Collapse
Affiliation(s)
- Francesca Cadamuro
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milano, Italy; CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland.
| |
Collapse
|
200
|
Li Z, Ruan C, Niu X. Collagen-based bioinks for regenerative medicine: Fabrication, application and prospective. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2023. [DOI: 10.1016/j.medntd.2023.100211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|