1
|
Miller J, Perrier Q, Rengaraj A, Bowlby J, Byers L, Peveri E, Jeong W, Ritchey T, Gambelli AM, Rossi A, Calafiore R, Tomei A, Orlando G, Asthana A. State of the Art of Bioengineering Approaches in Beta-Cell Replacement. CURRENT TRANSPLANTATION REPORTS 2025; 12:17. [PMID: 40342868 PMCID: PMC12055624 DOI: 10.1007/s40472-025-00470-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Purpose of the Review Despite recent advancements in technology for the treatment of type 1 diabetes (T1D), exogenous insulin delivery through automated devices remains the gold standard for treatment. This review will explore progress made in pancreatic islet bioengineering within the field of beta-cell replacement for T1D treatment. Recent Findings First, we will focus on the use of decellularized extracellular matrices (dECM) as a platform for pancreatic organoid development. These matrices preserve microarchitecture and essential biochemical signals for cell differentiation, offering a promising alternative to synthetic matrices. Second, advancements in 3D bioprinting for creating complex organ structures like pancreatic islets will be discussed. This technology allows for increased precision and customization of cellular models, crucial for replicating native pancreatic islet functionality. Finally, this review will explore the use of stem cell-derived organoids to generate insulin-producing islet-like cells. While these organoids face challenges such as functional immaturity and poor vascularization, they represent a significant advancement for disease modeling, drug screening, and autologous islet transplantation. Summary These innovative approaches promise to revolutionize T1D treatment by overcoming the limitations of traditional therapies based on human pancreatic islets.
Collapse
Affiliation(s)
- Jake Miller
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Quentin Perrier
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
- Univ. Grenoble Alpes, Department of Pharmacy, Grenoble Alpes University Hospital, Grenoble, France
| | - Arunkumar Rengaraj
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Joshua Bowlby
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Lori Byers
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Emma Peveri
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Wonwoo Jeong
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | - Thomas Ritchey
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
| | | | - Arianna Rossi
- Department of Engineering, University of Perugia, Perugia, Italy
| | | | - Alice Tomei
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL USA
| | - Giuseppe Orlando
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| | - Amish Asthana
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC USA
- Department of Surgery, Atrium Health Wake Forest Baptist, Winston-Salem, NC USA
| |
Collapse
|
2
|
Liu L, Ding Z, Huang Y, Zou J. Magnesium Ion/Gallic Acid MOF-Laden Multifunctional Acellular Matrix Hydrogels for Diabetic Wound Healing. ACS APPLIED BIO MATERIALS 2025. [PMID: 40317115 DOI: 10.1021/acsabm.4c01979] [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: 05/07/2025]
Abstract
The main objective for diabetic wound treatment is the design of a functional dressing that scavenges free radicals, alleviates inflammation, and is antibacterial while promoting neovascularization. Herein, a multifunctional acellular matrix hydrogel was prepared with the antimicrobial peptide jelleine-1 and a magnesium ion/gallic acid metal framework to exhibit antioxidant, anti-inflammatory, and proangiogenesis effects in diabetic wounds. The prepared hydrogel termed Gel-J-MOF efficiently released gallic acid in the acidic microenvironment of the diabetic wound, scavenged excess free radicals in vitro, and effectively reduced the levels of inflammation by regulating M2 macrophage polarization in vivo. The antimicrobial peptide jelleine-1 in the composite hydrogel effectively inhibited S. aureus and E. coli in vitro, promoting a suitable microenvironment for wound healing. In the later stage of wound healing, the composite hydrogel stimulated angiogenesis, accelerating the re-epithelialization and collagen deposition in the wound. In conclusion, this multifunctional composite hydrogel provides a regulated microenvironment for treating diabetic wounds and, therefore, has significant potential application promise in the treatment of chronic diabetic wounds.
Collapse
Affiliation(s)
- Lutong Liu
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
- Beijing Allgens Medical Science and Technology Co., Ltd., Beijing 100176, China
| | - Zhaozhao Ding
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325000, China
| | - Yong Huang
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| | - Junwei Zou
- Department of General Surgery, The Second Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, China
| |
Collapse
|
3
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
4
|
Kim M, Cho S, Hwang DG, Shim IK, Kim SC, Jang J, Jang J. Bioprinting of bespoke islet-specific niches to promote maturation of stem cell-derived islets. Nat Commun 2025; 16:1430. [PMID: 39920133 PMCID: PMC11805982 DOI: 10.1038/s41467-025-56665-5] [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: 12/11/2023] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Pancreatic islets are densely packed cellular aggregates containing various hormonal cell types essential for blood glucose regulation. Interactions among these cells markedly affect the glucoregulatory functions of islets along with the surrounding niche and pancreatic tissue-specific geometrical organization. However, stem cell (SC)-derived islets generated in vitro often lack the three-dimensional extracellular microenvironment and peri-vasculature, which leads to the immaturity of SC-derived islets, reducing their ability to detect glucose fluctuations and insulin release. Here, we bioengineer the in vivo-like pancreatic niches by optimizing the combination of pancreatic tissue-specific extracellular matrix and basement membrane proteins and utilizing bioprinting-based geometrical guidance to recreate the spatial pattern of islet peripheries. The bioprinted islet-specific niche promotes coordinated interactions between islets and vasculature, supporting structural and functional features resembling native islets. Our strategy not only improves SC-derived islet functionality but also offers significant potential for advancing research on islet development, maturation, and diabetic disease modeling, with future implications for translational applications.
Collapse
Affiliation(s)
- Myungji Kim
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Seungyeun Cho
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - In Kyong Shim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Song Cheol Kim
- Asan Institute for Life Science, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Republic of Korea
| | - Jiwon Jang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- Division of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Liu J, Song Q, Yin W, Li C, An N, Le Y, Wang Q, Feng Y, Hu Y, Wang Y. Bioactive scaffolds for tissue engineering: A review of decellularized extracellular matrix applications and innovations. EXPLORATION (BEIJING, CHINA) 2025; 5:20230078. [PMID: 40040827 PMCID: PMC11875452 DOI: 10.1002/exp.20230078] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/12/2024] [Indexed: 03/06/2025]
Abstract
Decellularized extracellular matrix (dECM) offers a three-dimensional, non-immunogenic scaffold, enriched with bioactive components, making it a suitable candidate for tissue regeneration. Although dECM-based scaffolds have been successfully implemented in preclinical and clinical settings within tissue engineering and regenerative medicine, the mechanisms of tissue remodeling and functional restoration are not fully understood. This review critically assesses the state-of-the-art in dECM scaffolds, including decellularization techniques for various tissues, quality control and cross-linking. It highlights the functional properties of dECM components and their latest applications in multiorgan tissue engineering and biomedicine. Additionally, the review addresses current challenges and limitations of decellularized scaffolds and offers perspectives on future directions in the field.
Collapse
Affiliation(s)
- Juan Liu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Qingru Song
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Wenzhen Yin
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Chen Li
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- College of Chemistry and Life SciencesBeijing University of TechnologyBeijingChina
| | - Ni An
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Yinpeng Le
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Institute of Smart Biomedical MaterialsSchool of Materials Science and EngineeringZhejiang Sci‐Tech UniversityHangzhouPeople's Republic of China
| | - Qi Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yutian Feng
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Yuelei Hu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yunfang Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| |
Collapse
|
6
|
Kim J, Shim IK, Lee YN, Kim M, Hwang DG, Kim J, Jo Y, Chae S, Kim J, Kim SC, Cho DW, Jang J. Engineering pore-enriched and pre-vascularized volumetric constructs for enhanced blood glucose regulation in type 1 diabetes therapy. Biofabrication 2024; 17:015034. [PMID: 39622163 DOI: 10.1088/1758-5090/ad998e] [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: 05/21/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Managing type 1 diabetes mellitus (T1DM) presents significant challenges because of the complexity of replicating the microenvironment of pancreatic islets and ensuring the long-term viability and function of transplanted insulin-producing cells (IPCs). This study developed a functional approach that utilizes 3D bioprinting technology to create pore-enriched and pre-vascularized tissue constructs incorporating a pancreatic tissue-derived decellularized extracellular matrix and human-induced pluripotent stem cells (hiPSCs) aimed at enhancing blood glucose regulation in T1DM. We designed a volumetric 3D pancreatic tissue construct that supported the engraftment, survival, and insulin-producing functionality of hiPSC-derived IPCs. The construct's porosity was optimized to enhance IPC delivery efficiency. Additionally, human umbilical vein endothelial cells co-cultured with IPCs in a patterned structure facilitated pre-vascularization, improving construct integration with host tissues and accelerating revascularization post-transplantation. Our results demonstrate high cell viability and sustained insulin production in diabetic rodent models, indicating the constructs' effectiveness in regulating blood glucose levels over an extended period. The findings of this study not only underscore the potential of 3D bioprinting for creating functional tissue constructs for T1DM treatment but also offer efficient cell delivery techniques applicable to other areas of regenerative medicine.
Collapse
Affiliation(s)
- Jaewook Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In Kyong Shim
- Department of Biomedical Engineering, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yu Na Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jihwan Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | | | - Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Wang X, Zhang D, Singh YP, Yeo M, Deng G, Lai J, Chen F, Ozbolat IT, Yu Y. Progress in Organ Bioprinting for Regenerative Medicine. ENGINEERING 2024; 42:121-142. [DOI: 10.1016/j.eng.2024.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
8
|
Yao X, Gong Z, Yin W, Li H, Douroumis D, Huang L, Li H. Islet cell spheroids produced by a thermally sensitive scaffold: a new diabetes treatment. J Nanobiotechnology 2024; 22:657. [PMID: 39456025 PMCID: PMC11515210 DOI: 10.1186/s12951-024-02891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The primary issues in treating type 1 diabetes mellitus (T1DM) through the transplantation of healthy islets or islet β-cells are graft rejection and a lack of available donors. Currently, the majority of approaches use cell encapsulation technology and transplant replacement cells that can release insulin to address transplant rejection and donor shortages. However, existing encapsulation materials merely serve as carriers for islet cell growth. A new treatment approach for T1DM could be developed by creating a smart responsive material that encourages the formation of islet cell spheroids to replicate their 3D connections in vivo and controls the release of insulin aggregates. In this study, we used microfluidics to create thermally sensitive porous scaffolds made of poly(N-isopropyl acrylamide)/graphene oxide (PNIPAM/GO). The material was carefully shrunk under near-infrared light, enriched with mouse insulinoma pancreatic β cells (β-TC-6 cells), encapsulated, and cultivated to form 3D cell spheroids. The controlled contraction of the thermally responsive porous scaffold regulated insulin release from the spheroids, demonstrated using the glucose-stimulated insulin release assay (GSIS), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence assay. Eventually, implantation of the spheroids into C57BL/6 N diabetic mice enhanced the therapeutic effect, potentially offering a novel approach to the management of T1DM.
Collapse
Affiliation(s)
- Xueting Yao
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, P. R. China
| | - Zehua Gong
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hanbing Li
- Department of Pharmaceutical Sciences, Institute of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Dennis Douroumis
- Centre for Research Innovation, CRI, University of Greenwich, Kent, ME4 4TB, UK
| | - Lijiang Huang
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
| | - Huaqiong Li
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China.
| |
Collapse
|
9
|
Lian L, Xie M, Luo Z, Zhang Z, Maharjan S, Mu X, Garciamendez-Mijares CE, Kuang X, Sahoo JK, Tang G, Li G, Wang D, Guo J, González FZ, Abril Manjarrez Rivera V, Cai L, Mei X, Kaplan DL, Zhang YS. Rapid Volumetric Bioprinting of Decellularized Extracellular Matrix Bioinks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304846. [PMID: 38252896 PMCID: PMC11260906 DOI: 10.1002/adma.202304846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/28/2023] [Indexed: 01/24/2024]
Abstract
Decellularized extracellular matrix (dECM)-based hydrogels are widely applied to additive biomanufacturing strategies for relevant applications. The extracellular matrix components and growth factors of dECM play crucial roles in cell adhesion, growth, and differentiation. However, the generally poor mechanical properties and printability have remained as major limitations for dECM-based materials. In this study, heart-derived dECM (h-dECM) and meniscus-derived dECM (Ms-dECM) bioinks in their pristine, unmodified state supplemented with the photoinitiator system of tris(2,2-bipyridyl) dichlororuthenium(II) hexahydrate and sodium persulfate, demonstrate cytocompatibility with volumetric bioprinting processes. This recently developed bioprinting modality illuminates a dynamically evolving light pattern into a rotating volume of the bioink, and thus decouples the requirement of mechanical strengths of bioprinted hydrogel constructs with printability, allowing for the fabrication of sophisticated shapes and architectures with low-concentration dECM materials that set within tens of seconds. As exemplary applications, cardiac tissues are volumetrically bioprinted using the cardiomyocyte-laden h-dECM bioink showing favorable cell proliferation, expansion, spreading, biomarker expressions, and synchronized contractions; whereas the volumetrically bioprinted Ms-dECM meniscus structures embedded with human mesenchymal stem cells present appropriate chondrogenic differentiation outcomes. This study supplies expanded bioink libraries for volumetric bioprinting and broadens utilities of dECM toward tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Liming Lian
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Maobin Xie
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zeyu Luo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Zhenrui Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Sushila Maharjan
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Xuan Mu
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Carlos Ezio Garciamendez-Mijares
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Xiao Kuang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jugal Kishore Sahoo
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Guosheng Tang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Gang Li
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Di Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Jie Guo
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Federico Zertuche González
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Victoria Abril Manjarrez Rivera
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Ling Cai
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA
| |
Collapse
|
10
|
Wan H, Xiang J, Mao G, Pan S, Li B, Lu Y. Recent Advances in the Application of 3D-Printing Bioinks Based on Decellularized Extracellular Matrix in Tissue Engineering. ACS OMEGA 2024; 9:24219-24235. [PMID: 38882108 PMCID: PMC11170705 DOI: 10.1021/acsomega.4c02847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/10/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024]
Abstract
In recent years, 3D bioprinting with various types of bioinks has been widely used in tissue engineering to fabricate human tissues and organs with appropriate biological functions. Decellularized extracellular matrix (dECM) is an excellent bioink candidate because it is enriched with a variety of bioactive proteins and bioactive factors and can provide a suitable environment for tissue repair or tissue regeneration while reducing the likelihood of severe immune rejection. In this Review, we systematically review recent advances in 3D bioprinting and decellularization technologies and comprehensively detail the latest research and applications of dECM as a bioink for tissue engineering in various systems, with the aim of providing a reference for researchers in tissue engineering to better understand the properties of dECM bioinks.
Collapse
Affiliation(s)
- Haoxin Wan
- Department
of Thoracic Surgery, The First Affiliated
Hospital of Soochow University, Suzhou 215000, China
| | - Jian Xiang
- Affiliated
Hospital of Yangzhou University, Yangzhou 225000, China
| | - Guocai Mao
- Department
of Thoracic Surgery, The First Affiliated
Hospital of Soochow University, Suzhou 215000, China
| | - Shu Pan
- Department
of Thoracic Surgery, The First Affiliated
Hospital of Soochow University, Suzhou 215000, China
| | - Bing Li
- The
Second Affiliated Hospital of Soochow University, Suzhou 215000, China
| | - Yi Lu
- Clinical
Medical College, Yangzhou University, Yangzhou 225000, China
| |
Collapse
|
11
|
Sathisaran I. 3D printing and bioprinting in the battle against diabetes and its chronic complications. Front Bioeng Biotechnol 2024; 12:1363483. [PMID: 38863489 PMCID: PMC11165705 DOI: 10.3389/fbioe.2024.1363483] [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: 12/30/2023] [Accepted: 04/22/2024] [Indexed: 06/13/2024] Open
Abstract
Diabetes is a metabolic disorder characterized by high blood sugar. Uncontrolled blood glucose affects the circulatory system in an organism by intervening blood circulation. The high blood glucose can lead to macrovascular (large blood vessels) and microvascular (small blood vessels) complications. Due to this, the vital organs (notably brain, eyes, feet, heart, kidneys, lungs and nerves) get worsen in diabetic patients if not treated at the earliest. Therefore, acquiring treatment at an appropriate time is very important for managing diabetes and other complications that are caused due to diabetes. The root cause for the occurrence of various health complications in diabetic patients is the uncontrolled blood glucose levels. This review presents a consolidated account of the applications of various types of three-dimensional (3D) printing and bioprinting technologies in treating diabetes as well as the complications caused due to impaired blood glucose levels. Herein, the development of biosensors (for the diagnosis), oral drug formulations, transdermal drug carriers, orthotic insoles and scaffolds (for the treatment) are discussed. Next to this, the fabrication of 3D bioprinted organs and cell-seeded hydrogels (pancreas engineering for producing insulin and bone engineering for managing bone defects) are explained. As the final application, 3D bioprinting of diabetic disease models for high-throughput screening of ant-diabetic drugs are discussed. Lastly, the challenges and future perspective associated with the use of 3D printing and bioprinting technologies against diabetes and its related chronic complications have been put forward.
Collapse
Affiliation(s)
- Indumathi Sathisaran
- Department of Bioscience and Engineering, National Institute of Technology Calicut, Kozhikode, Kerala, India
| |
Collapse
|
12
|
Wang X, Wei P, Hu C, Zeng H, Fan Z. 3D printing of Rg3-loaded hydrogel scaffolds: anti-inflammatory and scar-formation related collagen inhibitory effects for scar-free wound healing. J Mater Chem B 2024; 12:4673-4685. [PMID: 38647236 DOI: 10.1039/d3tb02941g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
During the process of wound healing, the stimulation of inflammatory factors often leads to abnormal proliferation of blood vessels and collagen, ultimately resulting in scar formation. To address this challenge, we fabricate a novel dermal extracellular matrix (DECM) hydrogel scaffold loaded with ginsenoside Rg3 (Rg3) using 3D printing technology. Mesoporous silica nanoparticles (MSNs) are introduced into the system to encase the Rg3 to control its release rate and enhance its bioavailability. We systematically evaluate the biological, physicochemical, and wound healing properties of this scaffold. In vitro studies demonstrate that the hydrogel exhibits excellent biocompatibility and solid-like rheological properties, ensuring its successful printing. In vivo studies reveal that the composite hydrogel scaffolds effectively accelerate wound healing and achieve scar-free wound healing within three weeks. Histological and immunohistochemical (IHC) analyses show that the composite hydrogel scaffolds reduce the inflammatory response and inhibit excessive collagen accumulation. These combined effects underscore the potential of our approach in effectively inhibiting scar formation.
Collapse
Affiliation(s)
- Xusen Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Pengyu Wei
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Cewen Hu
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Huajing Zeng
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| | - Zengjie Fan
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing of Gansu Province, School of Stomatology, Lanzhou University, Lanzhou 730000, People's Republic of China.
| |
Collapse
|
13
|
Kosowska K, Korycka P, Jankowska-Snopkiewicz K, Gierałtowska J, Czajka M, Florys-Jankowska K, Dec M, Romanik-Chruścielewska A, Małecki M, Westphal K, Wszoła M, Klak M. Graphene Oxide (GO)-Based Bioink with Enhanced 3D Printability and Mechanical Properties for Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:760. [PMID: 38727354 PMCID: PMC11085087 DOI: 10.3390/nano14090760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
Currently, a major challenge in material engineering is to develop a cell-safe biomaterial with significant utility in processing technology such as 3D bioprinting. The main goal of this work was to optimize the composition of a new graphene oxide (GO)-based bioink containing additional extracellular matrix (ECM) with unique properties that may find application in 3D bioprinting of biomimetic scaffolds. The experimental work evaluated functional properties such as viscosity and complex modulus, printability, mechanical strength, elasticity, degradation and absorbability, as well as biological properties such as cytotoxicity and cell response after exposure to a biomaterial. The findings demonstrated that the inclusion of GO had no substantial impact on the rheological properties and printability, but it did enhance the mechanical properties. This enhancement is crucial for the advancement of 3D scaffolds that are resilient to deformation and promote their utilization in tissue engineering investigations. Furthermore, GO-based hydrogels exhibited much greater swelling, absorbability and degradation compared to non-GO-based bioink. Additionally, these biomaterials showed lower cytotoxicity. Due to its properties, it is recommended to use bioink containing GO for bioprinting functional tissue models with the vascular system, e.g., for testing drugs or hard tissue models.
Collapse
Affiliation(s)
- Katarzyna Kosowska
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Polbionica Sp. z o.o., 01-793 Warsaw, Poland
| | - Paulina Korycka
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
| | - Kamila Jankowska-Snopkiewicz
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
| | - Joanna Gierałtowska
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
| | - Milena Czajka
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Polbionica Sp. z o.o., 01-793 Warsaw, Poland
| | - Katarzyna Florys-Jankowska
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
| | - Magdalena Dec
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Polbionica Sp. z o.o., 01-793 Warsaw, Poland
| | - Agnieszka Romanik-Chruścielewska
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland
| | - Kinga Westphal
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, 6124 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Michał Wszoła
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Polbionica Sp. z o.o., 01-793 Warsaw, Poland
- Medispace Medical Centre, 01-044 Warsaw, Poland
| | - Marta Klak
- Foundation of Research and Science Development, 01-793 Warsaw, Poland; (P.K.); (K.J.-S.); (J.G.); (M.C.); (K.F.-J.); (M.D.); (A.R.-C.); (K.W.); (M.W.)
- Polbionica Sp. z o.o., 01-793 Warsaw, Poland
| |
Collapse
|
14
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
15
|
Smandri A, Al-Masawa ME, Hwei NM, Fauzi MB. ECM-derived biomaterials for regulating tissue multicellularity and maturation. iScience 2024; 27:109141. [PMID: 38405613 PMCID: PMC10884934 DOI: 10.1016/j.isci.2024.109141] [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] [Indexed: 02/27/2024] Open
Abstract
Recent breakthroughs in developing human-relevant organotypic models led to the building of highly resemblant tissue constructs that hold immense potential for transplantation, drug screening, and disease modeling. Despite the progress in fine-tuning stem cell multilineage differentiation in highly controlled spatiotemporal conditions and hosting microenvironments, 3D models still experience naive and incomplete morphogenesis. In particular, existing systems and induction protocols fail to maintain stem cell long-term potency, induce high tissue-level multicellularity, or drive the maturity of stem cell-derived 3D models to levels seen in their in vivo counterparts. In this review, we highlight the use of extracellular matrix (ECM)-derived biomaterials in providing stem cell niche-mimicking microenvironment capable of preserving stem cell long-term potency and inducing spatial and region-specific differentiation. We also examine the maturation of different 3D models, including organoids, encapsulated in ECM biomaterials and provide looking-forward perspectives on employing ECM biomaterials in building more innovative, transplantable, and functional organs.
Collapse
Affiliation(s)
- Ali Smandri
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Maimonah Eissa Al-Masawa
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Ng Min Hwei
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
16
|
Qi B, Ding Y, Zhang Y, Kou L, Zhao YZ, Yao Q. Biomaterial-assisted strategies to improve islet graft revascularization and transplant outcomes. Biomater Sci 2024; 12:821-836. [PMID: 38168805 DOI: 10.1039/d3bm01295f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Islet transplantation holds significant promise as a curative approach for type 1 diabetes (T1D). However, the transition of islet transplantation from the experimental phase to widespread clinical implementation has not occurred yet. One major hurdle in this field is the challenge of insufficient vascularization and subsequent early loss of transplanted islets, especially in non-intraportal transplantation sites. The establishment of a fully functional vascular system following transplantation is crucial for the survival and secretion function of islet grafts. This vascular network not only ensures the delivery of oxygen and nutrients, but also plays a critical role in insulin release and the timely removal of metabolic waste from the grafts. This review summarizes recent advances in effective strategies to improve graft revascularization and enhance islet survival. These advancements include the local release and regulation of angiogenic factors (e.g., vascular endothelial growth factor, VEGF), co-transplantation of vascular fragments, and pre-vascularization of the graft site. These innovative approaches pave the way for the development of effective islet transplantation therapies for individuals with T1D.
Collapse
Affiliation(s)
- Boyang Qi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Yang Ding
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Longfa Kou
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Qing Yao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| |
Collapse
|
17
|
Elomaa L, Almalla A, Keshi E, Hillebrandt KH, Sauer IM, Weinhart M. Rise of tissue- and species-specific 3D bioprinting based on decellularized extracellular matrix-derived bioinks and bioresins. BIOMATERIALS AND BIOSYSTEMS 2023; 12:100084. [PMID: 38035034 PMCID: PMC10685010 DOI: 10.1016/j.bbiosy.2023.100084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/26/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
Thanks to its natural complexity and functionality, decellularized extracellular matrix (dECM) serves as an excellent foundation for creating highly cell-compatible bioinks and bioresins. This enables the bioprinted cells to thrive in an environment that closely mimics their native ECM composition and offers customizable biomechanical properties. To formulate dECM bioinks and bioresins, one must first pulverize and/or solubilize the dECM into non-crosslinked fragments, which can then be chemically modified as needed. In bioprinting, the solubilized dECM-derived material is typically deposited and/or crosslinked in a layer-by-layer fashion to build 3D hydrogel structures. Since the introduction of the first liver-derived dECM-based bioinks, a wide variety of decellularized tissue have been employed in bioprinting, including kidney, heart, cartilage, and adipose tissue among others. This review aims to summarize the critical steps involved in tissue-derived dECM bioprinting, starting from the decellularization of the ECM to the standardized formulation of bioinks and bioresins, ultimately leading to the reproducible bioprinting of tissue constructs. Notably, this discussion also covers photocrosslinkable dECM bioresins, which are particularly attractive due to their ability to provide precise spatiotemporal control over the gelation in bioprinting. Both in extrusion printing and vat photopolymerization, there is a need for more standardized protocols to fully harness the unique properties of dECM-derived materials. In addition to mammalian tissues, the most recent bioprinting approaches involve the use of microbial extracellular polymeric substances in bioprinting of bacteria. This presents similar challenges as those encountered in mammalian cell printing and represents a fascinating frontier in bioprinting technology.
Collapse
Affiliation(s)
- Laura Elomaa
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Ahed Almalla
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
| | - Eriselda Keshi
- Experimental Surgery, Department of Surgery, CCM|CVK, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany
| | - Karl H. Hillebrandt
- Experimental Surgery, Department of Surgery, CCM|CVK, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Clinician Scientist Program, Charitéplatz 1, Berlin 10117, Germany
| | - Igor M. Sauer
- Experimental Surgery, Department of Surgery, CCM|CVK, Charité – Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany
- Cluster of Excellence Matters of Activity, Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany´s Excellence Strategy – EXC 2025, Germany
| | - Marie Weinhart
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, Berlin 14195, Germany
- Cluster of Excellence Matters of Activity, Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany´s Excellence Strategy – EXC 2025, Germany
- Institute of Physical Chemistry and Electrochemistry, Leibniz Universität Hannover, Callinstr. 3A, Hannover 30167, Germany
| |
Collapse
|
18
|
Abstract
Bioprinting, as a groundbreaking technology, enables the fabrication of biomimetic tissues and organs with highly complex structures, multiple cell types, mechanical heterogeneity, and diverse functional gradients. With the growing demand for organ transplantation and the limited number of organ donors, bioprinting holds great promise for addressing the organ shortage by manufacturing completely functional organs. While the bioprinting of complete organs remains a distant goal, there has been considerable progress in the development of bioprinted transplantable tissues and organs for regenerative medicine. This review article recapitulates the current achievements of organ 3D bioprinting, primarily encompassing five important organs in the human body (i.e., the heart, kidneys, liver, pancreas, and lungs). Challenges from cellular techniques, biomanufacturing technologies, and organ maturation techniques are also deliberated for the broad application of organ bioprinting. In addition, the integration of bioprinting with other cutting-edge technologies including machine learning, organoids, and microfluidics is envisioned, which strives to offer the reader the prospect of bioprinting in constructing functional organs.
Collapse
Affiliation(s)
- Yang Wu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Minghao Qin
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Xue Yang
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| |
Collapse
|
19
|
Pan C, Xu J, Gao Q, Li W, Sun T, Lu J, Shi Q, Han Y, Gao G, Li J. Sequentially suspended 3D bioprinting of multiple-layered vascular models with tunable geometries for in vitromodeling of arterial disorders initiation. Biofabrication 2023; 15:045017. [PMID: 37579751 DOI: 10.1088/1758-5090/aceffa] [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: 05/15/2023] [Accepted: 08/14/2023] [Indexed: 08/16/2023]
Abstract
As the main precursor of arterial disorders, endothelial dysfunction preferentially occurs in regions of arteries prone to generating turbulent flow, particularly in branched regions of vasculatures. Although various diseased models have been engineered to investigate arterial pathology, producing a multiple-layered vascular model with branched geometries that can recapitulate the critical physiological environments of human arteries, such as intercellular communications and local turbulent flows, remains challenging. This study develops a sequentially suspended three-dimensional bioprinting (SSB) strategy and a visible-light-curable decellularized extracellular matrix bioink (abbreviated as 'VCD bioink') to construct a biomimetic human arterial model with tunable geometries. The engineered multiple-layered arterial models with compartmentalized vascular cells can exhibit physiological functionality and pathological performance under defined physiological flows specified by computational fluid dynamics simulation. Using different configurations of the vascular models, we investigated the independent and synergetic effects of cellular crosstalk and abnormal hemodynamics on the initiation of endothelial dysfunction, a hallmark event of arterial disorder. The results suggest that the arterial model constructed using the SSB strategy and VCD bioinks has promise in establishing diagnostic/analytic platforms for understanding the pathophysiology of human arterial disorders and relevant abnormalities, such as atherosclerosis, aneurysms, and ischemic diseases.
Collapse
Affiliation(s)
- Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, People's Republic of China
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Jingwen Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, People's Republic of China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, People's Republic of China
| | - Qiqi Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Tao Sun
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, People's Republic of China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, People's Republic of China
| | - Jiping Lu
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Qing Shi
- School of Mechatronical Engineering, Beijing Institute of Technology, Beijing 100081, People's Republic of China
- Key Laboratory of Biomimetic Robots and Systems (Beijing Institute of Technology), Ministry of Education, Beijing 100081, People's Republic of China
| | - Yafeng Han
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, People's Republic of China
| |
Collapse
|
20
|
Wang X, Jin L, Liu W, Stingelin L, Zhang P, Tan Z. Construction of engineered 3D islet micro-tissue using porcine decellularized ECM for the treatment of diabetes. Biomater Sci 2023; 11:5517-5532. [PMID: 37387616 DOI: 10.1039/d3bm00346a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Islet transplantation improves diabetes patients' long-term blood glucose control, but its success and utility are limited by cadaver availability, quality, and considerable islet loss after transplantation due to ischemia and inadequate angiogenesis. This study used adipose, pancreatic, and liver tissue decellularized extracellular matrix (dECM) hydrogels in an effort to recapitulate the islet sites inside the pancreas in vitro, and successfully generated viable and functional heterocellular islet micro-tissues using islet cells, human umbilical vein endothelial cells, and adipose-derived mesenchymal stem cells. The three-dimensional (3D) islet micro-tissues maintained prolonged viability and normal secretory function, and showed high drug sensitivity in drug testing. Meanwhile, the 3D islet micro-tissues significantly enhanced survival and graft function in a mouse model of diabetes. These supportive 3D physiomimetic dECM hydrogels can be used not only for islet micro-tissue culture in vitro, but also have great promise for islet transplantation for the treatment of diabetes.
Collapse
Affiliation(s)
- Xiaocheng Wang
- Department of Infectious Diseases, Third Xiangya Hospital, Central South University, Changsha, 410008, China.
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Lijuan Jin
- College of Biology, Hunan University, Changsha, 410082, China.
- Shenzhen Institute, Hunan University, Shenzhen, 518000, China.
| | - Wenyu Liu
- College of Biology, Hunan University, Changsha, 410082, China.
- Shenzhen Institute, Hunan University, Shenzhen, 518000, China.
| | - Lukas Stingelin
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Pan Zhang
- Department of Infectious Diseases, Third Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, China.
- Shenzhen Institute, Hunan University, Shenzhen, 518000, China.
| |
Collapse
|
21
|
Glorieux L, Vandooren L, Derclaye S, Pyr Dit Ruys S, Oncina-Gil P, Salowka A, Herinckx G, Aajja E, Lemoine P, Spourquet C, Lefort H, Henriet P, Tyteca D, Spagnoli FM, Alsteens D, Vertommen D, Pierreux CE. In-Depth Analysis of the Pancreatic Extracellular Matrix during Development for Next-Generation Tissue Engineering. Int J Mol Sci 2023; 24:10268. [PMID: 37373416 DOI: 10.3390/ijms241210268] [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: 05/17/2023] [Revised: 06/04/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
The pancreas is a complex organ consisting of differentiated cells and extracellular matrix (ECM) organized adequately to enable its endocrine and exocrine functions. Although much is known about the intrinsic factors that control pancreas development, very few studies have focused on the microenvironment surrounding pancreatic cells. This environment is composed of various cells and ECM components, which play a critical role in maintaining tissue organization and homeostasis. In this study, we applied mass spectrometry to identify and quantify the ECM composition of the developing pancreas at the embryonic (E) day 14.5 and postnatal (P) day 1 stages. Our proteomic analysis identified 160 ECM proteins that displayed a dynamic expression profile with a shift in collagens and proteoglycans. Furthermore, we used atomic force microscopy to measure the biomechanical properties and found that the pancreatic ECM was soft (≤400 Pa) with no significant change during pancreas maturation. Lastly, we optimized a decellularization protocol for P1 pancreatic tissues, incorporating a preliminary crosslinking step, which effectively preserved the 3D organization of the ECM. The resulting ECM scaffold proved suitable for recellularization studies. Our findings provide insights into the composition and biomechanics of the pancreatic embryonic and perinatal ECM, offering a foundation for future studies investigating the dynamic interactions between the ECM and pancreatic cells.
Collapse
Affiliation(s)
- Laura Glorieux
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Laura Vandooren
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Sylvie Derclaye
- Nanobiophysics Lab, Louvain Institute of Biomolecular Science and Technology, UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | | | - Paloma Oncina-Gil
- Nanobiophysics Lab, Louvain Institute of Biomolecular Science and Technology, UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Anna Salowka
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - Gaëtan Herinckx
- de Duve Institute and MASSPROT Platform, UCLouvain, 1200 Brussels, Belgium
| | - Elias Aajja
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Pascale Lemoine
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | | | - Hélène Lefort
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Patrick Henriet
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Donatienne Tyteca
- Cell Biology Unit, de Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | - Francesca M Spagnoli
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Great Maze Pond, London SE1 9RT, UK
| | - David Alsteens
- Nanobiophysics Lab, Louvain Institute of Biomolecular Science and Technology, UCLouvain, 1348 Louvain-la-Neuve, Belgium
| | - Didier Vertommen
- de Duve Institute and MASSPROT Platform, UCLouvain, 1200 Brussels, Belgium
| | | |
Collapse
|
22
|
Zhe M, Wu X, Yu P, Xu J, Liu M, Yang G, Xiang Z, Xing F, Ritz U. Recent Advances in Decellularized Extracellular Matrix-Based Bioinks for 3D Bioprinting in Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2023; 16:3197. [PMID: 37110034 PMCID: PMC10143913 DOI: 10.3390/ma16083197] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/30/2023] [Accepted: 04/15/2023] [Indexed: 06/19/2023]
Abstract
In recent years, three-dimensional (3D) bioprinting has been widely utilized as a novel manufacturing technique by more and more researchers to construct various tissue substitutes with complex architectures and geometries. Different biomaterials, including natural and synthetic materials, have been manufactured into bioinks for tissue regeneration using 3D bioprinting. Among the natural biomaterials derived from various natural tissues or organs, the decellularized extracellular matrix (dECM) has a complex internal structure and a variety of bioactive factors that provide mechanistic, biophysical, and biochemical signals for tissue regeneration and remodeling. In recent years, more and more researchers have been developing the dECM as a novel bioink for the construction of tissue substitutes. Compared with other bioinks, the various ECM components in dECM-based bioink can regulate cellular functions, modulate the tissue regeneration process, and adjust tissue remodeling. Therefore, we conducted this review to discuss the current status of and perspectives on dECM-based bioinks for bioprinting in tissue engineering. In addition, the various bioprinting techniques and decellularization methods were also discussed in this study.
Collapse
Affiliation(s)
- Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyu Wu
- West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Jiawei Xu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming Liu
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Guang Yang
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhou Xiang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fei Xing
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
23
|
Lam EHY, Yu F, Zhu S, Wang Z. 3D Bioprinting for Next-Generation Personalized Medicine. Int J Mol Sci 2023; 24:ijms24076357. [PMID: 37047328 PMCID: PMC10094501 DOI: 10.3390/ijms24076357] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023] Open
Abstract
In the past decade, immense progress has been made in advancing personalized medicine to effectively address patient-specific disease complexities in order to develop individualized treatment strategies. In particular, the emergence of 3D bioprinting for in vitro models of tissue and organ engineering presents novel opportunities to improve personalized medicine. However, the existing bioprinted constructs are not yet able to fulfill the ultimate goal: an anatomically realistic organ with mature biological functions. Current bioprinting approaches have technical challenges in terms of precise cell deposition, effective differentiation, proper vascularization, and innervation. This review introduces the principles and realizations of bioprinting with a strong focus on the predominant techniques, including extrusion printing and digital light processing (DLP). We further discussed the applications of bioprinted constructs, including the engraftment of stem cells as personalized implants for regenerative medicine and in vitro high-throughput drug development models for drug discovery. While no one-size-fits-all approach to bioprinting has emerged, the rapid progress and promising results of preliminary studies have demonstrated that bioprinting could serve as an empowering technology to resolve critical challenges in personalized medicine.
Collapse
Affiliation(s)
- Ethan Hau Yin Lam
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Nutritional Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Fengqing Yu
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Computer Science, University of Toronto, Toronto, ON M5S 1A4, Canada
| | - Sabrina Zhu
- Faculty of Arts and Science, University of Toronto, Toronto, ON M5S 3G3, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Zongjie Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3E1, Canada
- McCormick School of Engineering, Northwestern University, Chicago, IL 60611, USA
- Correspondence: or
| |
Collapse
|
24
|
Samadi A, Moammeri A, Pourmadadi M, Abbasi P, Hosseinpour Z, Farokh A, Shamsabadipour A, Heydari M, Mohammadi MR. Cell Encapsulation and 3D Bioprinting for Therapeutic Cell Transplantation. ACS Biomater Sci Eng 2023; 9:1862-1890. [PMID: 36877212 DOI: 10.1021/acsbiomaterials.2c01183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
The promise of cell therapy has been augmented by introducing biomaterials, where intricate scaffold shapes are fabricated to accommodate the cells within. In this review, we first discuss cell encapsulation and the promising potential of biomaterials to overcome challenges associated with cell therapy, particularly cellular function and longevity. More specifically, cell therapies in the context of autoimmune disorders, neurodegenerative diseases, and cancer are reviewed from the perspectives of preclinical findings as well as available clinical data. Next, techniques to fabricate cell-biomaterials constructs, focusing on emerging 3D bioprinting technologies, will be reviewed. 3D bioprinting is an advancing field that enables fabricating complex, interconnected, and consistent cell-based constructs capable of scaling up highly reproducible cell-biomaterials platforms with high precision. It is expected that 3D bioprinting devices will expand and become more precise, scalable, and appropriate for clinical manufacturing. Rather than one printer fits all, seeing more application-specific printer types, such as a bioprinter for bone tissue fabrication, which would be different from a bioprinter for skin tissue fabrication, is anticipated in the future.
Collapse
Affiliation(s)
- Amirmasoud Samadi
- Department of Chemical and Biomolecular Engineering, 6000 Interdisciplinary Science & Engineering Building (ISEB), Irvine, California 92617, United States
| | - Ali Moammeri
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Mehrab Pourmadadi
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Parisa Abbasi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Avenue, Tehran 1458889694, Iran
| | - Zeinab Hosseinpour
- Biotechnology Research Laboratory, Faculty of Chemical Engineering, Babol Noshirvani University of Technology, Babol 4714871167, Mazandaran Province, Iran
| | - Arian Farokh
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Amin Shamsabadipour
- School of Chemical Engineering, College of Engineering, University of Tehran, Enghelab Square, 16 Azar Street, Tehran 1417935840, Iran
| | - Maryam Heydari
- Department of Cell and Molecular Biology, Faculty of Biological Science, University of Kharazmi, Tehran 199389373, Iran
| | - M Rezaa Mohammadi
- Dale E. and Sarah Ann Fowler School of Engineering, Chapman University, Orange, California 92866, United States
| |
Collapse
|
25
|
de Carvasal KP, Vergoten G, Vasseur JJ, Smietana M, Morvan F. Supramolecular Recognition of Phosphodiester-Based Donor and Acceptor Oligomers Forming Gels in Water. Biomacromolecules 2023; 24:756-765. [PMID: 36724436 DOI: 10.1021/acs.biomac.2c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Inspired by automated DNA synthesis, electron-rich dialkoxynaphthalene (DAN) donor and electron-deficient naphthalene-tetracarboxylic diimide (NDI) acceptor phosphodiester-linked homohexamers were synthesized by the phosphoramidite method. Two types of hexamers were prepared, one with only one phosphodiester between the aromatics (i.e., DAN or NDI) and a second with two phosphodiesters around a propanediol between the aromatics, leading to the latter more flexible and more hydrophilic hexamers. The folding properties of these homohexamers alone or mixed together, in water only, were studied by UV-visible absorption spectroscopy and atomic force microscopy (AFM). AFM imaging revealed that a 1:1 mixture of hexaDAN and hexaNDI formed fibers by charge transfer donor-acceptor recognition leading to a hydrogel after drying. The organization of the resulting structures is strongly dependent on the nature of the complementary partner, leading to the formation of mono- or multilayer hydrogel networks with different compactness.
Collapse
Affiliation(s)
- Kévan Pérez de Carvasal
- Université de Montpellier, CNRS, ENSCM, Institut des Biomolécules Max Mousseron, Montpellier 34293, France
| | - Gérard Vergoten
- Université de Lille, Inserm, INFINITE - U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, Lille 59006, France
| | - Jean-Jacques Vasseur
- Université de Montpellier, CNRS, ENSCM, Institut des Biomolécules Max Mousseron, Montpellier 34293, France
| | - Michael Smietana
- Université de Montpellier, CNRS, ENSCM, Institut des Biomolécules Max Mousseron, Montpellier 34293, France
| | - François Morvan
- Université de Montpellier, CNRS, ENSCM, Institut des Biomolécules Max Mousseron, Montpellier 34293, France
| |
Collapse
|
26
|
Kort-Mascort J, Flores-Torres S, Peza-Chavez O, Jang JH, Pardo LA, Tran SD, Kinsella J. Decellularized ECM hydrogels: prior use considerations, applications, and opportunities in tissue engineering and biofabrication. Biomater Sci 2023; 11:400-431. [PMID: 36484344 DOI: 10.1039/d2bm01273a] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Tissue development, wound healing, pathogenesis, regeneration, and homeostasis rely upon coordinated and dynamic spatial and temporal remodeling of extracellular matrix (ECM) molecules. ECM reorganization and normal physiological tissue function, require the establishment and maintenance of biological, chemical, and mechanical feedback mechanisms directed by cell-matrix interactions. To replicate the physical and biological environment provided by the ECM in vivo, methods have been developed to decellularize and solubilize tissues which yield organ and tissue-specific bioactive hydrogels. While these biomaterials retain several important traits of the native ECM, the decellularizing process, and subsequent sterilization, and solubilization result in fragmented, cleaved, or partially denatured macromolecules. The final product has decreased viscosity, moduli, and yield strength, when compared to the source tissue, limiting the compatibility of isolated decellularized ECM (dECM) hydrogels with fabrication methods such as extrusion bioprinting. This review describes the physical and bioactive characteristics of dECM hydrogels and their role as biomaterials for biofabrication. In this work, critical variables when selecting the appropriate tissue source and extraction methods are identified. Common manual and automated fabrication techniques compatible with dECM hydrogels are described and compared. Fabrication and post-manufacturing challenges presented by the dECM hydrogels decreased mechanical and structural stability are discussed as well as circumvention strategies. We further highlight and provide examples of the use of dECM hydrogels in tissue engineering and their role in fabricating complex in vitro 3D microenvironments.
Collapse
Affiliation(s)
| | | | - Omar Peza-Chavez
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | - Joyce H Jang
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| | | | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Joseph Kinsella
- Department of Bioengineering, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
27
|
Wang Z, Xiang L, Lin F, Tang Y, Cui W. 3D bioprinting of emulating homeostasis regulation for regenerative medicine applications. J Control Release 2023; 353:147-165. [PMID: 36423869 DOI: 10.1016/j.jconrel.2022.11.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/25/2022]
Abstract
Homeostasis is the most fundamental mechanism of physiological processes, occurring simultaneously as the production and outcomes of pathological procedures. Accompanied by manufacture and maturation of intricate and highly hierarchical architecture obtained from 3D bioprinting (three-dimension bioprinting), homeostasis has substantially determined the quality of printed tissues and organs. Instead of only shape imitation that has been the remarkable advances, fabrication for functionality to make artificial tissues and organs that act as real ones in vivo has been accepted as the optimized strategy in 3D bioprinting for the next several years. Herein, this review aims to provide not only an overview of 3D bioprinting, but also the main strategies used for homeostasis bioprinting. This paper briefly introduces the principles of 3D bioprinting system applied in homeostasis regulations firstly, and then summarizes the specific strategies and potential trend of homeostasis regulations using multiple types of stimuli-response biomaterials to maintain auto regulation, specifically displaying a brilliant prospect in hormone regulation of homeostasis with the most recently outbreak of vasculature fabrication. Finally, we discuss challenges and future prospects of homeostasis fabrication based on 3D bioprinting in regenerative medicine, hoping to further inspire the development of functional fabrication in 3D bioprinting.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Lei Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Feng Lin
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai 200025, PR China.
| |
Collapse
|
28
|
Kim H, Jang JH, Han W, Hwang HJ, Jang J, Kim JY, Cho DW. Extracellular matrix-based sticky sealants for scar-free corneal tissue reconstruction. Biomaterials 2023; 292:121941. [PMID: 36495802 DOI: 10.1016/j.biomaterials.2022.121941] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/05/2022]
Abstract
Regenerative medicine requires both tissue restoration and ease of compliance for clinical application. Considering this, sticky tissue sealants have been shown to have great potentials over surgical suturing and wound treatment. However, tissue sealants currently used pose challenges such as uncontrollable adhesion formation, mechanical mismatch, and lack of tissue restoration. A new sticky sealant based on gelatinized cornea-derived extracellular matrix (GelCodE) with a visible light-activating system is firstly being introduced in this study. De novo tissue regeneration relies on the matrisome in charge of tissue-organization and development within GelCodE while visible light-based photopolymerization with ruthenium/sodium persulfate rapidly induces covalent bonds with the adjacent tissues. The ease of not only in vivo application, biocompatibility, and biointegration, but also exceptional de novo tissue formation is demonstrated in this study. Interestingly, newly regenerated tissues were shown to have normal tissue-like matrices with little scar formation. Hence, this work presents a promising strategy to meet clinical demands for scar-free tissue recovery with superior ease of clinical application.
Collapse
Affiliation(s)
- Hyeonji Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, 37673, Pohang, Kyungbuk, Republic of Korea
| | - Je-Hwan Jang
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, 05029, Seoul, Republic of Korea
| | - Wonil Han
- Division of Integrative Bioscience and Biotechnology, POSTECH, 77 Cheongam-ro, Nam-gu, 37673, Pohang, Kyungbuk, Republic of Korea
| | - Hyun-Jeong Hwang
- Department of Clinical Pathology, College of Veterinary Medicine, Konkuk University, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, 37673, Pohang, Kyungbuk, Republic of Korea; Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, 03722, Seoul, Republic of Korea; Department of Convergence IT Engineering, 77 Cheongam-ro, Nam-gu, POSTECH, 37673, Pohang, Kyungbuk, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, 77 Cheongam-ro, Nam-gu, POSTECH, 37673, Pohang, Kyungbuk, Republic of Korea.
| | - Joon Young Kim
- Department of Veterinary Ophthalmology, College of Veterinary Medicine, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, 05029, Seoul, Republic of Korea; KU Center for Animal Blood Medical Science, Konkuk University, 120, Neungdong-ro, Gwangjin-gu, 05029, Seoul, Republic of Korea.
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, 37673, Pohang, Kyungbuk, Republic of Korea; Institute of Convergence Science, Yonsei University, 50 Yonsei-ro, 03722, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Challenges with Cell-based Therapies for Type 1 Diabetes Mellitus. Stem Cell Rev Rep 2022; 19:601-624. [PMID: 36434300 DOI: 10.1007/s12015-022-10482-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2022] [Indexed: 11/27/2022]
Abstract
Type 1 diabetes (T1D) is a chronic, lifelong metabolic disease. It is characterised by the autoimmune-mediated loss of insulin-producing pancreatic β cells in the islets of Langerhans (β-islets), resulting in disrupted glucose homeostasis. Administration of exogenous insulin is the most common management method for T1D, but this requires lifelong reliance on insulin injections and invasive blood glucose monitoring. Replacement therapies with beta cells are being developed as an advanced curative treatment for T1D. Unfortunately, this approach is limited by the lack of donated pancreatic tissue, the difficulties in beta cell isolation and viability maintenance, the longevity of the transplanted cells in vivo, and consequently high costs. Emerging approaches to address these limitations are under intensive investigations, including the production of insulin-producing beta cells from various stem cells, and the development of bioengineered devices including nanotechnologies for improving islet transplantation efficacy without the need for recipients taking toxic anti-rejection drugs. These emerging approaches present promising prospects, while the challenges with the new techniques need to be tackled for ultimately clinical treatment of T1D. This review discussed the benefits and limitations of the cell-based therapies for beta cell replacement as potential curative treatment for T1D, and the applications of bioengineered devices including nanotechnology to overcome the challenges associated with beta cell transplantation.
Collapse
|
30
|
Xu Y, Song D, Wang X. 3D Bioprinting for Pancreas Engineering/Manufacturing. Polymers (Basel) 2022; 14:polym14235143. [PMID: 36501537 PMCID: PMC9741443 DOI: 10.3390/polym14235143] [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: 10/11/2022] [Revised: 10/29/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetes is the most common chronic disease in the world, and it brings a heavy burden to people's health. Against this background, diabetic research, including islet functionalization has become a hot topic in medical institutions all over the world. Especially with the rapid development of microencapsulation and three-dimensional (3D) bioprinting technologies, organ engineering and manufacturing have become the main trends for disease modeling and drug screening. Especially the advanced 3D models of pancreatic islets have shown better physiological functions than monolayer cultures, suggesting their potential in elucidating the behaviors of cells under different growth environments. This review mainly summarizes the latest progress of islet capsules and 3D printed pancreatic organs and introduces the activities of islet cells in the constructs with different encapsulation technologies and polymeric materials, as well as the vascularization and blood glucose control capabilities of these constructs after implantation. The challenges and perspectives of the pancreatic organ engineering/manufacturing technologies have also been demonstrated.
Collapse
|
31
|
McInnes AD, Moser MAJ, Chen X. Preparation and Use of Decellularized Extracellular Matrix for Tissue Engineering. J Funct Biomater 2022; 13:jfb13040240. [PMID: 36412881 PMCID: PMC9680265 DOI: 10.3390/jfb13040240] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/22/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022] Open
Abstract
The multidisciplinary fields of tissue engineering and regenerative medicine have the potential to revolutionize the practise of medicine through the abilities to repair, regenerate, or replace tissues and organs with functional engineered constructs. To this end, tissue engineering combines scaffolding materials with cells and biologically active molecules into constructs with the appropriate structures and properties for tissue/organ regeneration, where scaffolding materials and biomolecules are the keys to mimic the native extracellular matrix (ECM). For this, one emerging way is to decellularize the native ECM into the materials suitable for, directly or in combination with other materials, creating functional constructs. Over the past decade, decellularized ECM (or dECM) has greatly facilitated the advance of tissue engineering and regenerative medicine, while being challenged in many ways. This article reviews the recent development of dECM for tissue engineering and regenerative medicine, with a focus on the preparation of dECM along with its influence on cell culture, the modification of dECM for use as a scaffolding material, and the novel techniques and emerging trends in processing dECM into functional constructs. We highlight the success of dECM and constructs in the in vitro, in vivo, and clinical applications and further identify the key issues and challenges involved, along with a discussion of future research directions.
Collapse
Affiliation(s)
- Adam D. McInnes
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Correspondence: ; Tel.: +1-306-966-5435
| | - Michael A. J. Moser
- Department of Surgery, Health Sciences Building, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| |
Collapse
|
32
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
33
|
Brown M, Li J, Moraes C, Tabrizian M, Li-Jessen NY. Decellularized extracellular matrix: New promising and challenging biomaterials for regenerative medicine. Biomaterials 2022; 289:121786. [DOI: 10.1016/j.biomaterials.2022.121786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022]
|
34
|
Pignatelli C, Campo F, Neroni A, Piemonti L, Citro A. Bioengineering the Vascularized Endocrine Pancreas: A Fine-Tuned Interplay Between Vascularization, Extracellular-Matrix-Based Scaffold Architecture, and Insulin-Producing Cells. Transpl Int 2022; 35:10555. [PMID: 36090775 PMCID: PMC9452644 DOI: 10.3389/ti.2022.10555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Intrahepatic islet transplantation is a promising β-cell replacement strategy for the treatment of type 1 diabetes. Instant blood-mediated inflammatory reactions, acute inflammatory storm, and graft revascularization delay limit islet engraftment in the peri-transplant phase, hampering the success rate of the procedure. Growing evidence has demonstrated that islet engraftment efficiency may take advantage of several bioengineering approaches aimed to recreate both vascular and endocrine compartments either ex vivo or in vivo. To this end, endocrine pancreas bioengineering is an emerging field in β-cell replacement, which might provide endocrine cells with all the building blocks (vascularization, ECM composition, or micro/macro-architecture) useful for their successful engraftment and function in vivo. Studies on reshaping either the endocrine cellular composition or the islet microenvironment have been largely performed, focusing on a single building block element, without, however, grasping that their synergistic effect is indispensable for correct endocrine function. Herein, the review focuses on the minimum building blocks that an ideal vascularized endocrine scaffold should have to resemble the endocrine niche architecture, composition, and function to foster functional connections between the vascular and endocrine compartments. Additionally, this review highlights the possibility of designing bioengineered scaffolds integrating alternative endocrine sources to overcome donor organ shortages and the possibility of combining novel immune-preserving strategies for long-term graft function.
Collapse
Affiliation(s)
- Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
35
|
Wang D, Guo Y, Zhu J, Liu F, Xue Y, Huang Y, Zhu B, Wu D, Pan H, Gong T, Lu Y, Yang Y, Wang Z. Hyaluronic acid methacrylate/pancreatic extracellular matrix as a potential 3D printing bioink for constructing islet organoids. Acta Biomater 2022:S1742-7061(22)00375-0. [PMID: 35803504 DOI: 10.1016/j.actbio.2022.06.036] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/01/2022]
Abstract
Islet transplantation has poor long-term efficacy because of the lack of extracellular matrix support and neovascularization; this limits its wide application in diabetes research. In this study, we develop a 3D-printed islet organoid by combining a pancreatic extracellular matrix (pECM) and hyaluronic acid methacrylate (HAMA) as specific bioinks. The HAMA/pECM hydrogel was validated in vitro to maintain islet cell adhesion and morphology through the Rac1/ROCK/MLCK signaling pathway, which helps improve islet function and activity. Further, in vivo experiments confirmed that the 3D-printed islet-encapsulated HAMA/pECM hydrogel increases insulin levels in diabetic mice, maintains blood glucose levels within a normal range for 90 days, and rapidly secretes insulin in response to blood glucose stimulation. In addition, the HAMA/pECM hydrogel can facilitate the attachment and growth of new blood vessels and increase the density of new vessels. Meanwhile, the designed 3D-printed structure was conducive to the formation of vascular networks and it promoted the construction of 3D-printed islet organoids. In conclusion, our experiments optimized the HAMA/pECM bioink composition and 3D-printed structure of islet organoids with promising therapeutic effects compared with the HAMA hydrogel group that can be potentially used in clinical applications to improve the effectiveness and safety of islet transplantation in vivo. STATEMENT OF SIGNIFICANCE: The extraction process of pancreatic islets can easily cause damage to the extracellular matrix and vascular system, resulting in poor islet transplantation efficiency. We developed a new tissue-specific bioink by combining pancreatic extracellular matrix (pECM) and hyaluronic acid methacrylate (HAMA). The islet organoids constructed by 3D printing can mimic the microenvironment of the pancreas and maintain islet cell adhesion and morphology through the Rac1/ROCK/MLCK signaling pathway, thereby improving islet function and activity. In addition, the 3D-printed structures we designed are favorable for the formation of new blood vessel networks, bringing hope for the long-term efficacy of islet transplantation.
Collapse
Affiliation(s)
- Dongzhi Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Jiacheng Zhu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China
| | - Fang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China
| | - Yan Xue
- Department of Internal Medicine, Nantong Health College of Jiangsu Province, Nantong, 226010, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Biwen Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Di Wu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Haopeng Pan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China
| | - Tiancheng Gong
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226006, China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China.
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China.
| |
Collapse
|
36
|
Urbanczyk M, Zbinden A, Schenke-Layland K. Organ-specific endothelial cell heterogenicity and its impact on regenerative medicine and biomedical engineering applications. Adv Drug Deliv Rev 2022; 186:114323. [PMID: 35568103 DOI: 10.1016/j.addr.2022.114323] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 04/23/2022] [Accepted: 05/05/2022] [Indexed: 02/08/2023]
Abstract
Endothelial cells (ECs) are a key cellular component of the vascular system as they form the inner lining of the blood vessels. Recent findings highlight that ECs express extensive phenotypic heterogenicity when following the vascular tree from the major vasculature down to the organ capillaries. However, in vitro models, used for drug development and testing, or to study the role of ECs in health and disease, rarely acknowledge this EC heterogenicity. In this review, we highlight the main differences between different EC types, briefly summarize their different characteristics and focus on the use of ECs in in vitro models. We introduce different approaches on how ECs can be utilized in co-culture test systems in the field of brain, pancreas, and liver research to study the role of the endothelium in health and disease. Finally, we discuss potential improvements to current state-of-the-art in vitro models and future directions.
Collapse
|
37
|
Viability and Functionality of Neonatal Porcine Islet-like Cell Clusters Bioprinted in Alginate-Based Bioinks. Biomedicines 2022; 10:biomedicines10061420. [PMID: 35740440 PMCID: PMC9220255 DOI: 10.3390/biomedicines10061420] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
The transplantation of pancreatic islets can prevent severe long-term complications in diabetes mellitus type 1 patients. With respect to a shortage of donor organs, the transplantation of xenogeneic islets is highly attractive. To avoid rejection, islets can be encapsulated in immuno-protective hydrogel-macrocapsules, whereby 3D bioprinted structures with macropores allow for a high surface-to-volume ratio and reduced diffusion distances. In the present study, we applied 3D bioprinting to encapsulate the potentially clinically applicable neonatal porcine islet-like cell clusters (NICC) in alginate-methylcellulose. The material was additionally supplemented with bovine serum albumin or the human blood plasma derivatives platelet lysate and fresh frozen plasma. NICC were analysed for viability, proliferation, the presence of hormones, and the release of insulin in reaction to glucose stimulation. Bioprinted NICC are homogeneously distributed, remain morphologically intact, and show a comparable viability and proliferation to control NICC. The number of insulin-positive cells is comparable between the groups and over time. The amount of insulin release increases over time and is released in response to glucose stimulation over 4 weeks. In summary, we show the successful bioprinting of NICC and could demonstrate functionality over the long-term in vitro. Supplementation resulted in a trend for higher viability, but no additional benefit on functionality was observed.
Collapse
|
38
|
Chen S, Luo J, Shen L, Liu X, Wang W, Xu J, Ren Y, Ye Y, Shi G, Cheng F, Cheng L, Su X, Dai L, Gou M, Deng H. 3D Printing Mini-Capsule Device for Islet Delivery to Treat Type 1 Diabetes. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23139-23151. [PMID: 35544723 DOI: 10.1021/acsami.2c02487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Transplantation of encapsulated islets has been shown to hold a promising potential treatment for type 1 diabetes (T1D). However, there are several obstacles to overcome, such as immune rejection by the host of the grafts, sustainability of islet function, and retrievability or replacement of the encapsulated system, hinder their clinical applications. In this study, mini-capsule devices containing islets were fabricated by using digital light processing (DLP) 3D printing. To ensure a high survival rate and low immunogenicity of the fabricated islets, 20s was selected as the most suitable printing condition. Meanwhile, the mini-capsule devices with a groove structure were fabricated to prevent islet cells leakage. Subcutaneous transplantations of encapsulated islets in immunocompetent C57BL/6 mice indicated significant improvement in the symptoms of streptozotocin-induced hyperglycemia without any immunosuppression treatment for at least 15 weeks. In vivo intraperitoneal glucose tolerance tests (IPGTT) performed at different time points demonstrated therapeutically relevant glycemic ameliorate of the device. The implants retrieved after 15 weeks still contained viable and adequate numbers of islet cells. The results of this study indicate that the proposed mini-capsule device can deliver sufficient islet cell mass, prevent islet cells leakage, and maintain long-term cell survival while allowing easy retrieval. Furthermore, the proposed encapsulated islets may help with T1D cellular treatment by overcoming the obstacles of islet transplantation.
Collapse
Affiliation(s)
- Shuang Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lanlin Shen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xuan Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wenshuang Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jia Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yushuang Ren
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yixin Ye
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Maling Gou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
39
|
Fatimi A, Okoro OV, Podstawczyk D, Siminska-Stanny J, Shavandi A. Natural Hydrogel-Based Bio-Inks for 3D Bioprinting in Tissue Engineering: A Review. Gels 2022; 8:179. [PMID: 35323292 PMCID: PMC8948717 DOI: 10.3390/gels8030179] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Three-dimensional (3D) printing is well acknowledged to constitute an important technology in tissue engineering, largely due to the increasing global demand for organ replacement and tissue regeneration. In 3D bioprinting, which is a step ahead of 3D biomaterial printing, the ink employed is impregnated with cells, without compromising ink printability. This allows for immediate scaffold cellularization and generation of complex structures. The use of cell-laden inks or bio-inks provides the opportunity for enhanced cell differentiation for organ fabrication and regeneration. Recognizing the importance of such bio-inks, the current study comprehensively explores the state of the art of the utilization of bio-inks based on natural polymers (biopolymers), such as cellulose, agarose, alginate, decellularized matrix, in 3D bioprinting. Discussions regarding progress in bioprinting, techniques and approaches employed in the bioprinting of natural polymers, and limitations and prospects concerning future trends in human-scale tissue and organ fabrication are also presented.
Collapse
Affiliation(s)
- Ahmed Fatimi
- Department of Chemistry, Polydisciplinary Faculty, Sultan Moulay Slimane University, P.O. Box 592 Mghila, Beni-Mellal 23000, Morocco
- ERSIC, Polydisciplinary Faculty, Sultan Moulay Slimane University, P.O. Box 592 Mghila, Beni-Mellal 23000, Morocco
| | - Oseweuba Valentine Okoro
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium; (O.V.O.); (J.S.-S.)
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland;
| | - Julia Siminska-Stanny
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium; (O.V.O.); (J.S.-S.)
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373 Wroclaw, Poland;
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), Avenue F.D. Roosevelt, 50-CP 165/61, 1050 Brussels, Belgium; (O.V.O.); (J.S.-S.)
| |
Collapse
|
40
|
Chae S, Kim J, Yi HG, Cho DW. 3D Bioprinting of an In Vitro Model of a Biomimetic Urinary Bladder with a Contract-Release System. MICROMACHINES 2022; 13:277. [PMID: 35208401 PMCID: PMC8877589 DOI: 10.3390/mi13020277] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 01/01/2023]
Abstract
The development of curative therapy for bladder dysfunction is usually hampered owing to the lack of reliable ex vivo human models that can mimic the complexity of the human bladder. To overcome this issue, 3D in vitro model systems offering unique opportunities to engineer realistic human tissues/organs have been developed. However, existing in vitro models still cannot entirely reflect the key structural and physiological characteristics of the native human bladder. In this study, we propose an in vitro model of the urinary bladder that can create 3D biomimetic tissue structures and dynamic microenvironments to replicate the smooth muscle functions of an actual human urinary bladder. In other words, the proposed biomimetic model system, developed using a 3D bioprinting approach, can recreate the physiological motion of the urinary bladder by incorporating decellularized extracellular matrix from the bladder tissue and introducing cyclic mechanical stimuli. The results showed that the developed bladder tissue models exhibited high cell viability and proliferation rate and promoted myogenic differentiation potential given dynamic mechanical cues. We envision the developed in vitro bladder mimicry model can serve as a research platform for fundamental studies on human disease modeling and pharmaceutical testing.
Collapse
Affiliation(s)
- Suhun Chae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.C.); (J.K.)
| | - Jaewook Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.C.); (J.K.)
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea; (S.C.); (J.K.)
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
41
|
Papoz A, Clément F, Laporte C, Tubbs E, Gidrol X, Pitaval A. [Generating pancreatic islets organoids: Langerhanoids]. Med Sci (Paris) 2022; 38:52-58. [PMID: 35060887 DOI: 10.1051/medsci/2021244] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The extension of islet transplantation to a wider number of Type 1 diabetic patients is compromised by the scarcity of donors, the reduced ex vivo survival of pancreatic islets and the use of immunosuppressive treatments. Islets of Langerhans isolated from brain-dead donors are currently the only cell source for transplantation. Thus, it is crucial to find an alternative and an abundant source of functional insulin secreting cells not only for clinical use but also for the development of research dedicated to the screening of drugs and to the development of new therapeutic targets. Several groups around the world, including ours, develop 3D culture models as Langerhanoids that closely mimick human pancreatic islets physiology. In this review, we describe recent advances to mimic the pancreatic niche (extracellular matrix, vascularization, microfluidics) allowing better functionality of Langerhanoids.
Collapse
Affiliation(s)
- Anastasia Papoz
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Flora Clément
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Camille Laporte
- Univ. Grenoble Alpes, CEA, Leti, Division for biology and healthcare technologies, Microfluidic systems and bioengineering Lab, F-38000, Grenoble, France
| | - Emily Tubbs
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France - Univ. Grenoble Alpes, LBFA et BEeSy, Inserm U1055, F-38000, Grenoble, France
| | - Xavier Gidrol
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| | - Amandine Pitaval
- Univ. Grenoble Alpes, CEA, Inserm, IRIG, Biomics, F-38000, Grenoble, France
| |
Collapse
|
42
|
Mansoor S, Kondiah PPD, Choonara YE. Advanced Hydrogels for the Controlled Delivery of Insulin. Pharmaceutics 2021; 13:2113. [PMID: 34959394 PMCID: PMC8703368 DOI: 10.3390/pharmaceutics13122113] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/02/2023] Open
Abstract
Insulin is a peptide hormone that is key to regulating physiological glucose levels. Its molecular size and susceptibility to conformational change under physiological pH make it challenging to orally administer insulin in diabetes. The most effective route for insulin delivery remains daily injection. Unfortunately, this results in poor patient compliance and increasing the risk of micro- and macro-vascular complications and thus rising morbidity and mortality rates in diabetics. The use of 3D hydrogels has been used with much interest for various biomedical applications. Hydrogels can mimic the extracellular matrix (ECM) and retain large quantities of water with tunable properties, which renders them suitable for administering a wide range of sensitive therapeutics. Several studies have demonstrated the fixation of insulin within the structural mesh of hydrogels as a bio-scaffold for the controlled delivery of insulin. This review provides a concise incursion into recent developments for the safe and effective controlled delivery of insulin using advanced hydrogel platforms with a special focus on sustained release injectable formulations. Various hydrogel platforms in terms of their methods of synthesis, properties, and unique features such as stimuli responsiveness for the treatment of Type 1 diabetes mellitus are critically appraised. Key criteria for classifying hydrogels are also outlined together with future trends in the field.
Collapse
Affiliation(s)
| | | | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa; (S.M.); (P.P.D.K.)
| |
Collapse
|
43
|
Kim M, Jang J. Construction of 3D hierarchical tissue platforms for modeling diabetes. APL Bioeng 2021; 5:041506. [PMID: 34703970 PMCID: PMC8530538 DOI: 10.1063/5.0055128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/27/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) is one of the most serious systemic diseases worldwide, and the majority of DM patients face severe complications. However, many of underlying disease mechanisms related to these complications are difficult to understand with the use of currently available animal models. With the urgent need to fundamentally understand DM pathology, a variety of 3D biomimetic platforms have been generated by the convergence of biofabrication and tissue engineering strategies for the potent drug screening platform of pre-clinical research. Here, we suggest key requirements for the fabrication of physiomimetic tissue models in terms of recapitulating the cellular organization, creating native 3D microenvironmental niches for targeted tissue using biomaterials, and applying biofabrication technologies to implement tissue-specific geometries. We also provide an overview of various in vitro DM models, from a cellular level to complex living systems, which have been developed using various bioengineering approaches. Moreover, we aim to discuss the roadblocks facing in vitro tissue models and end with an outlook for future DM research.
Collapse
Affiliation(s)
- Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, POSTECH, 77 Cheongam-ro, Namgu, Pohang, Kyungbuk, 37673, Republic of Korea
| | - Jinah Jang
- Author to whom correspondence should be addressed:
| |
Collapse
|
44
|
Akhavan S, Tutunchi S, Malmir A, Ajorlou P, Jalili A, Panahi G. Molecular study of the proliferation process of beta cells derived from pluripotent stem cells. Mol Biol Rep 2021; 49:1429-1436. [PMID: 34734370 DOI: 10.1007/s11033-021-06892-y] [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: 07/11/2021] [Accepted: 10/28/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Diabetes mellitus (DM) is a chronic metabolic disorder, increasing in the number of patients and poses a severe threat to human health. Significant advances have been made in DM treatment; the most important of which is differentiation and proliferation of beta cells from IPSCs. METHODS Data were collected from PUBMED at various time points up to the academic year of 2020. The related keywords are listed as follows: "Induced pluripotent stem cell", "Proliferation", "Growth factor", "Small molecule", "cardiotoxicity" and "Scaffold." RESULT The use of growth factors along with small molecules can be a good strategy for beta-cell proliferation. Also, proliferation of beta cells on nanofibers scaffolds can create a similar in vivo environment, that leads to increased function of beta-cell. Some transcription factors that cause beta cells proliferation play an important role in inflammation; so, it is essential to monitor them to prevent inflammation. CONCLUSION Finally, the simultaneous use of growth factors, micronutrients and scaffolds can be an excellent strategy to increase the proliferation and function of beta cells derived from IPSCs.
Collapse
Affiliation(s)
- Saeedeh Akhavan
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University (IAU), Tehran, Iran
| | - Sara Tutunchi
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ali Malmir
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Parisa Ajorlou
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACER, Tehran, Iran
| | - Ghodratollah Panahi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
In Vitro Disease Models of the Endocrine Pancreas. Biomedicines 2021; 9:biomedicines9101415. [PMID: 34680532 PMCID: PMC8533367 DOI: 10.3390/biomedicines9101415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022] Open
Abstract
The ethical constraints and shortcomings of animal models, combined with the demand to study disease pathogenesis under controlled conditions, are giving rise to a new field at the interface of tissue engineering and pathophysiology, which focuses on the development of in vitro models of disease. In vitro models are defined as synthetic experimental systems that contain living human cells and mimic tissue- and organ-level physiology in vitro by taking advantage of recent advances in tissue engineering and microfabrication. This review provides an overview of in vitro models and focuses specifically on in vitro disease models of the endocrine pancreas and diabetes. First, we briefly review the anatomy, physiology, and pathophysiology of the human pancreas, with an emphasis on islets of Langerhans and beta cell dysfunction. We then discuss different types of in vitro models and fundamental elements that should be considered when developing an in vitro disease model. Finally, we review the current state and breakthroughs in the field of pancreatic in vitro models and conclude with some challenges that need to be addressed in the future development of in vitro models.
Collapse
|
46
|
Hwang DG, Jo Y, Kim M, Yong U, Cho S, Choi YM, Kim J, Jang J. A 3D bioprinted hybrid encapsulation system for delivery of human pluripotent stem cell-derived pancreatic islet-like aggregates. Biofabrication 2021; 14. [PMID: 34479233 DOI: 10.1088/1758-5090/ac23ac] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/03/2021] [Indexed: 11/12/2022]
Abstract
Islet transplantation is a promising treatment for type 1 diabetes. However, treatment failure can result from loss of functional cells associated with cell dispersion, low viability, and severe immune response. To overcome these limitations, various islet encapsulation approaches have been introduced. Among them, macroencapsulation offers the advantages of delivering and retrieving a large volume of islets in one system. In this study, we developed a hybrid encapsulation system composed of a macroporous polymer capsule with stagger-type membrane and assemblable structure, and a nanoporous decellularized extracellular matrix (dECM) hydrogel containing pancreatic islet-like aggregates using 3D bioprinting technique. The outer part (macroporous polymer capsule) was designed to have an interconnected porous architecture, which allows insulin-producingβ-cells encapsulated in the hybrid encapsulation system to maintain their cellular behaviors, including viability, cell proliferation, and insulin-producing function. The inner part (nanoporous dECM hydrogel), composed of the 3D biofabricated pancreatic islet-like aggregates, was simultaneously placed into the macroporous polymer capsule in one step. The developed hybrid encapsulation system exhibited biocompatibilityin vitroandin vivoin terms of M1 macrophage polarization. Furthermore, by controlling the printing parameters, we generated islet-like aggregates, improving cell viability and functionality. Moreover, the 3D bioprinted pancreatic islet-like aggregates exhibited structural maturation and functional enhancement associated with intercellular interaction occurring at theβ-cell edges. In addition, we also investigated the therapeutic potential of a hybrid encapsulation system by integrating human pluripotent stem cell-derived insulin-producing cells, which are promising to overcome the donor shortage problem. In summary, these results demonstrated that the 3D bioprinting approach facilitates the fabrication of a hybrid islet encapsulation system with multiple materials and potentially improves the clinical outcomes by driving structural maturation and functional improvement of cells.
Collapse
Affiliation(s)
- Dong Gyu Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Seungyeon Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Yoo-Mi Choi
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jaewook Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
47
|
Kupikowska-Stobba B, Grzeczkowicz M, Lewińska D. A one-step in vitro continuous flow assessment of protein release from core-shell polymer microcapsules designed for therapeutic protein delivery. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Kant RJ, Bare CF, Coulombe KL. Tissues with Patterned Vessels or Protein Release Induce Vascular Chemotaxis in an In Vitro Platform. Tissue Eng Part A 2021; 27:1290-1304. [PMID: 33472529 PMCID: PMC8610033 DOI: 10.1089/ten.tea.2020.0269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Engineered tissues designed for translational applications in regenerative medicine require vascular networks to deliver oxygen and nutrients rapidly to the implanted cells. A limiting factor of in vivo translation is the rapid and successful inosculation, or connection, of host and implanted vascular networks and subsequent perfusion of the implant. An approach gaining favor in vascular tissue engineering is to provide instructive cues from the engineered tissue to enhance host vascular penetration and connection with the implant. Here, we use a novel in vitro platform based on the aortic ring assay to evaluate the impact of patterned, endothelialized vessels or growth factor release from engineered constructs on preinosculative vascular cell outgrowth from surrogate host tissue in a controlled, defined environment, and introduce robust tools for evaluating vascular morphogenesis and chemotaxis. We demonstrate the creation of engineered vessels at the arteriole scale, which develop basement membrane, exhibit tight junctions, and actively sprout into the surrounding bulk hydrogel. Vessel-containing constructs are co-cultured adjacent to rodent aortic rings, and the resulting heterocellular outgrowth is quantified. Cells originating from the aortic ring migrate preferentially toward constructs containing engineered vessels with 1.5-fold faster outgrowth kinetics, 2.5-fold increased cellular density, and 1.6-fold greater network formation versus control (no endothelial cells and growth factor-reduced culture medium). Growth factor release from constructs with nonendothelialized channels and in reduced factor medium equivalently stimulates sustained vascular outgrowth distance, cellular density, and network formation, akin to engineered vessels in endothelial growth medium 2 (EGM-2) medium. In conclusion, we show that three-dimensional endothelialized patterned vessels or growth factor release stimulate a robust, host-derived vascular cell chemotactic response at early time points critical for instructive angiogenic cues. Further, we developed robust, unbiased tools to quantify metrics of vascular morphogenesis and preinosculative heterocellular outgrowth from rat aortic rings and demonstrated the utility of our complex, controlled environment, heterocellular in vitro platform. Impact statement Using a novel in vitro platform, we show that engineered constructs with patterned vessels or angiogenic growth factor release, two methods of instructing host revascularization responses, equivalently improve early host-derived vascular outgrowth. Our platform leverages the aortic ring assay in a tissue engineering context to study preinosculative vascular cell chemotaxis from surrogate host vascular cells in response to paracrine cues from co-cultured engineered tissues using robust, open-source quantification tools. Our accessible and flexible platform enables translationally focused studies in revascularization using implantable therapeutics containing prepatterned vessels with greater environmental control than in vivo studies to advance vascular tissue engineering.
Collapse
Affiliation(s)
- Rajeev J. Kant
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Colette F. Bare
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Kareen L.K. Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
49
|
Zhao F, Cheng J, Zhang J, Yu H, Dai W, Yan W, Sun M, Ding G, Li Q, Meng Q, Liu Q, Duan X, Hu X, Ao Y. Comparison of three different acidic solutions in tendon decellularized extracellular matrix bio-ink fabrication for 3D cell printing. Acta Biomater 2021; 131:262-275. [PMID: 34157451 DOI: 10.1016/j.actbio.2021.06.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 06/05/2021] [Accepted: 06/15/2021] [Indexed: 12/13/2022]
Abstract
Decellularized extracellular matrix (dECM) hydrogels are being increasingly investigated for use in bio-inks for three-dimensional cell printing given their good cytocompatibility and biomimetic properties. The osmotic pressure and stiffness of bio-ink are important factors affecting the biological functions of printed cells. However, little attention has been given to the osmotic pressure and stiffness of the dECM bio-inks. Here, we compared three types of commonly used acidic solutions in the bio-fabrication of a tendon derived dECM bio-ink for 3D cell printing (0.5 M acetic acid, 0.1 M hydrochloric acid and 0.02 M hydrochloric acid). We found that low pH value of 0.1 M hydrochloric acid could accelerate the digestion process for dECM powders. This could lead to a much softer dECM hydrogel with storage modulus less than 100 Pa. This soft dECM hydrogel facilitated the spreading and proliferation of stem cells encapsulated within it. It also showed better tendon-inducing ability compared with two others much stiffer dECM hydrogels. However, this over-digested dECM hydrogel was more unstable as it could shrink with the culture time going on. For 0.5 M acetic acid made dECM bio-ink, the hyperosmotic state of the bio-ink led to much lower cellular viability rates. Postprocess (Dilution or dialysis) to tailor the osmotic pressure of hydrogels could be a necessary step before mixed with cells. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation. And a balance should be made between the digestion period, strength of acidic solution, as well as the size and concentration of the dECM powders. STATEMENT OF SIGNIFICANCE: The dECM bio-ink has been widely used in 3D cell printing for tissue engineering and organ modelling. In this study, we found that different types of acid have different digestion and dissolution status for the dECM materials. A much softer tendon derived dECM hydrogel with lower stiffness could facilitate the cellular spreading, proliferation and tendon differentiation. We also demonstrated that the osmotic pressure should be taken care of in the preparation of dECM bio-ink with 0.5 M acetic acid. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation.
Collapse
|
50
|
Gao G, Ahn M, Cho WW, Kim BS, Cho DW. 3D Printing of Pharmaceutical Application: Drug Screening and Drug Delivery. Pharmaceutics 2021; 13:1373. [PMID: 34575448 PMCID: PMC8465948 DOI: 10.3390/pharmaceutics13091373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022] Open
Abstract
Advances in three-dimensional (3D) printing techniques and the development of tailored biomaterials have facilitated the precise fabrication of biological components and complex 3D geometrics over the past few decades. Moreover, the notable growth of 3D printing has facilitated pharmaceutical applications, enabling the development of customized drug screening and drug delivery systems for individual patients, breaking away from conventional approaches that primarily rely on transgenic animal experiments and mass production. This review provides an extensive overview of 3D printing research applied to drug screening and drug delivery systems that represent pharmaceutical applications. We classify several elements required by each application for advanced pharmaceutical techniques and briefly describe state-of-the-art 3D printing technology consisting of cells, bioinks, and printing strategies that satisfy requirements. Furthermore, we discuss the limitations of traditional approaches by providing concrete examples of drug screening (organoid, organ-on-a-chip, and tissue/organ equivalent) and drug delivery systems (oral/vaginal/rectal and transdermal/surgical drug delivery), followed by the introduction of recent pharmaceutical investigations using 3D printing-based strategies to overcome these challenges.
Collapse
Affiliation(s)
- Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Minjun Ahn
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Won-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Kyungbuk, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| |
Collapse
|