1
|
Lee G, Kim YH, Kim D, Lee DH, Bhang SH, Lee K. PCL-fibrin-alginate hydrogel based cell co-culture system for improving angiogenesis and immune modulation in limb ischemia. Colloids Surf B Biointerfaces 2025; 250:114553. [PMID: 39921993 DOI: 10.1016/j.colsurfb.2025.114553] [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: 11/03/2024] [Revised: 12/10/2024] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
Stem cell therapy has demonstrated promise in regenerative medicine due to their ability to differentiate into various cell types and secrete growth factors. However, challenges such as poor survival rate of transplanted cells under ischemic and immune conditions limit its effectiveness. To address these issues, we developed a polycaprolactone (PCL)-fibrin-alginate matrix hydrogel, which combines adipose-derived stem cells and human umbilical vein endothelial cells with a PCL fiber, encapsulated within fibrin and alginate hydrogel to enhance cell survival, proliferation, and immune modulation. This structure offers protection to the encapsulated cells, supports angiogenesis, and modulates the immune response, significantly improving therapeutic outcomes in a mouse model of hindlimb ischemia. Our in vitro and in vivo results demonstrate the scaffold's ability to support cell viability, promote angiogenesis, and modulate inflammatory responses, indicating its potential as a promising platform for ischemic tissue repair and regenerative medicine. This innovative approach to cell-based therapy highlights the importance of scaffold design in enhancing the therapeutic efficacy of stem cell treatments for ischemic diseases.
Collapse
Affiliation(s)
- Gyubok Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dongwoo Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
2
|
Fowler M, Moreno Lozano A, Krause J, Bednarz P, Pandey S, Ghayour M, Zhang Q, Veiseh O. Guiding vascular infiltration through architected GelMA/PEGDA hydrogels: an in vivo study of channel diameter, length, and complexity. Biomater Sci 2025. [PMID: 40278545 DOI: 10.1039/d5bm00193e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Organ shortages for transplantation in the United States impact over 100 000 patients, with 17 dying daily due to the lack of available organs. This growing need is exacerbated by the limited functionality and disease risk of donated organs. Tissue-engineered organs present a promising alternative, requiring optimized scaffold architecture and cell integration. Vascular networks within organs are essential for supplying oxygen and nutrients to cells, with a critical distance between blood vessels and surrounding tissue to allow effective diffusion. Various microfabrication techniques, such as electrospinning, freeze-drying, and gas foaming, have been employed to develop engineered organs. However, these techniques often lack the complexity needed to support vascularization. 3D bioprinting, particularly digital light projection (DLP)-based stereolithography, offers a solution by enabling high-resolution control of both external and internal architectures. Gelatin methacrylate (GelMA) and polyethylene glycol diacrylate (PEGDA) hydrogels have shown potential for tissue integration in simple structures but require further optimization for vascularization in more complex constructs. This study utilizes DLP to 3D bioprint GelMA/PEGDA hydrogels, exploring various channel designs to enhance tissue infiltration and vascularization in rodent models, providing a potential platform for cell and tissue transplantation. We demonstrate that GelMA/PEGDA hydrogels are mechanically robust, biocompatible, and support in vivo vascular infiltration. Channel diameter significantly influenced vascularization, with 1 mm channels yielding the highest infiltration, while channel length had minimal impact. Among five tested architectures, one design (GEO3) promoted the greatest vascular ingrowth, establishing a tunable hydrogel platform for prevascularized tissue engineering applications.
Collapse
Affiliation(s)
- Martha Fowler
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | | | - Julian Krause
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | - Patrick Bednarz
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | - Shalini Pandey
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | - Mina Ghayour
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| | - Qixu Zhang
- Department of Plastic Surgery, MD Anderson, Houston, TX 77030, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX 77030, USA.
| |
Collapse
|
3
|
Guo Y, Wang Z, Zhang X, Li J, Gao S, Lv Y, Ouyang L. Microfiber-Templated Porogel Bioinks Enable Tubular Interfaces and Microvascularization Down to the Building Blocks for 3D Bioprinting. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501594. [PMID: 40099633 DOI: 10.1002/smll.202501594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/02/2025] [Indexed: 03/20/2025]
Abstract
Vascularization is key to the biofabrication of large-scale tissues. Despite the progress, there remain some outstanding challenges, such as limited vessel density, difficulty in fabricating microvasculatures, and inhomogeneity of post-seeding cells. Here, a new form of bioink called microfiber-templated porogel (µFTP) bioink is introduced to engineer vasculatures down to the filament building blocks of 3D bioprinted hydrogels. The cell-laden sacrificial microfibers (diameter ranges from 50-150 µm) are embedded in the bioink to template tubular voids and deliver endothelial cells for in-situ endothelialization. The inclusion of softening hydrogel microfibers retains the desirable rheological properties of the bioink for extrusion-based bioprinting and the microfibers are well inter-contacted in the extruded filament. Such bioinks can be printed into a well-defined 3D structure with tunable tubular porosities up to 55%. Compared to the conventional bulk bioink counterpart, the µFTP bioink supports the significant growth and spread of endothelial cells either embedded in the matrix or sacrificial fibers, free of the post-cell seeding procedure. Furthermore, the bioprinted scaffolds based on µFTP bioink are seen to significantly promote the in-growth of blood vessels and native tissues in vivo. The µFTP bioink approach enables the engineering of tubular bio-interfaces within the building blocks and contributes to the in-situ endothelialization of microvasculatures, providing a versatile tool for the construction of customized vascularized tissue models.
Collapse
Affiliation(s)
- Yuzhi Guo
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Ziyu Wang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Xuening Zhang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Jinghang Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Shan Gao
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Yang Lv
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Liliang Ouyang
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
- State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
4
|
Malkani S, Prado O, Stevens KR. Sacrificial Templating for Accelerating Clinical Translation of Engineered Organs. ACS Biomater Sci Eng 2025; 11:1-12. [PMID: 39701582 PMCID: PMC11733865 DOI: 10.1021/acsbiomaterials.4c01824] [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: 10/01/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Transplantable engineered organs could one day be used to treat patients suffering from end-stage organ failure. Yet, producing hierarchical vascular networks that sustain the viability and function of cells within human-scale organs remains a major challenge. Sacrificial templating has emerged as a promising biofabrication method that could overcome this challenge. Here, we explore and evaluate various strategies and materials that have been used for sacrificial templating. First, we emphasize fabrication approaches that use highly biocompatible sacrificial reagents and minimize the duration that cells spend in fabrication conditions without oxygen and nutrients. We then discuss strategies to create continuous, hierarchical vascular networks, both using biofabrication alone and using hybrid methods that integrate biologically driven vascular self-assembly into sacrificial templating workflows. Finally, we address the importance of structurally reinforcing engineered vessel walls to achieve stable blood flow in vivo, so that engineered organs remain perfused and functional long after implantation. Together, these sacrificial templating strategies have the potential to overcome many current limitations in biofabrication and accelerate clinical translation of transplantable, fully functional engineered organs to rescue patients from organ failure.
Collapse
Affiliation(s)
- Sherina Malkani
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Department
of Bioengineering, Rice University, Houston, Texas 77005, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Olivia Prado
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Kelly R. Stevens
- Department
of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Institute
for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington 98195, United States
- Department
of Laboratory Medicine and Pathology, University
of Washington, Seattle, Washington 98195, United States
- Brotman
Baty Institute for Precision Medicine, Seattle, Washington 98195, United States
| |
Collapse
|
5
|
Wen C, Lee K, Wang Y, Wang X, Wang Y. Bidirectional Enzyme Inhibition and Activation for In Situ Formation of Injectable Hydrogel Using a Bispecific Aptamer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26751-26759. [PMID: 39642164 DOI: 10.1021/acs.langmuir.4c03925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
In situ injectable hydrogels have been explored for biomedical applications, including regenerative medicine and drug delivery. However, controlling the kinetics of their gelation to facilitate easy injection remains a challenge. The purpose of this study was to demonstrate the potential of using bispecific aptamers and complementary sequences as a bidirectional modulation system for controlling enzyme-mediated hydrogel formation kinetics. The results show that a bispecific thrombin-binding aptamer effectively inhibits thrombin activity and significantly slowed fibrin hydrogel formation. Upon interaction with its complementary sequence, this inhibition could be reversed. As a result, the aptamer-bound thrombin was activated, leading to an acceleration of the fibrin formation kinetics. Thus, bispecific aptamers and complementary sequences can effectively function as dynamic control systems for enzyme-catalyzed in situ injectable hydrogel formation.
Collapse
Affiliation(s)
- Connie Wen
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Kyungsene Lee
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Yixun Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Xuelin Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| |
Collapse
|
6
|
Fan X, Su Z, Zhang W, Huang H, He C, Wu Z, Zhang P. An advanced chitosan based sponges dressing system with antioxidative, immunoregulation, angiogenesis and neurogenesis for promoting diabetic wound healing. Mater Today Bio 2024; 29:101361. [PMID: 39659839 PMCID: PMC11629240 DOI: 10.1016/j.mtbio.2024.101361] [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: 09/20/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/12/2024] Open
Abstract
Promoting wound nerve regeneration and synchronously initiating angiogenesis are critical factors in the healing process of diabetic wounds. However, existing research on diabetic wounds mainly focuses on angiogenesis, bacterial infection and reactive oxygen species, often failing to coordinate neurogenesis and angiogenesis. To coordinate the symbiosis of nerves and blood vessels in the diabetic wounds, we successfully designed a multifunctional chitosan (CS)-based sponges by regulating the structure of CS specifically for diabetic wound healing. This sponge, which facilitates effective exudate transfer and modulates the wound microenvironment, was constructed using hydroxybutyl CS grafted with thioctic acid (TA), named as HCT sponge. When applied in a humid environment, the hydrophobic side chains of the HCT sponge interact with self-assembled hydrophobic domains, forming gel-sponge composite. Experimental results showed that the adhesion strength of the HCT sponge to wet porcine skin was 70.3 kPa. Additionally, the sponge exhibited favorable degradability, cytocompatibility and antioxidant properties. As it is shown in the experiments in vitro, sponge can not only promote cell proliferation, migration, and blood vessel formation, but also promote M2 macrophage polarization. Moreover, the rat liver and femoral artery injury model validated that the HCT sponge can effectively treat heavy bleeding from wounds efficacy through quickly sealing wounds and the formation of multiple hemostatic dams. In vivo studies indicated that the HCT sponge significantly accelerated the diabetic wound healing process compared to the recombinant bovine basic fibroblast growth factor gel, achieving a better recovery from the HCT sponge after 15 days. Pathological results show that the designed novel sponge holds considerable promise for treating diabetic wound, allowing regenerative neurogenesis and angiogenesis at the wound site, which provides a significant potential for further improving clinical applications.
Collapse
Affiliation(s)
- Xianmou Fan
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, 524000, China
| | - Zhihong Su
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Wanjun Zhang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Haili Huang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Chengzhang He
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Zeyong Wu
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
| | - Peihua Zhang
- Department of Plastic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524000, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, 524000, China
| |
Collapse
|
7
|
Liu S, Chen Y, Wang Z, Liu M, Zhao Y, Tan Y, Qu Z, Du L, Wu C. The cutting-edge progress in bioprinting for biomedicine: principles, applications, and future perspectives. MedComm (Beijing) 2024; 5:e753. [PMID: 39314888 PMCID: PMC11417428 DOI: 10.1002/mco2.753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Bioprinting is a highly promising application area of additive manufacturing technology that has been widely used in various fields, including tissue engineering, drug screening, organ regeneration, and biosensing. Its primary goal is to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices through software-layered discrete and numerical control molding. Despite its immense potential, bioprinting technology still faces several challenges. It requires concerted efforts from researchers, engineers, regulatory bodies, and industry stakeholders are principal to overcome these challenges and unlock the full potential of bioprinting. This review systematically discusses bioprinting principles, applications, and future perspectives while also providing a topical overview of research progress in bioprinting over the past two decades. The most recent advancements in bioprinting are comprehensively reviewed here. First, printing techniques and methods are summarized along with advancements related to bioinks and supporting structures. Second, interesting and representative cases regarding the applications of bioprinting in tissue engineering, drug screening, organ regeneration, and biosensing are introduced in detail. Finally, the remaining challenges and suggestions for future directions of bioprinting technology are proposed and discussed. Bioprinting is one of the most promising application areas of additive manufacturing technology that has been widely used in various fields. It aims to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices. This review systematically discusses bioprinting principles, applications, and future perspectives, which provides a topical description of the research progress of bioprinting.
Collapse
Affiliation(s)
- Shuge Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yating Chen
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhiyao Wang
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Minggao Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yundi Zhao
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yushuo Tan
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhan Qu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Liping Du
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Chunsheng Wu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| |
Collapse
|
8
|
Fantini DA, Yang G, Khanna A, Subramanian D, Phillippi JA, Huang NF. Overcoming big bottlenecks in vascular regeneration. Commun Biol 2024; 7:876. [PMID: 39020071 PMCID: PMC11255241 DOI: 10.1038/s42003-024-06567-x] [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: 12/04/2023] [Accepted: 07/05/2024] [Indexed: 07/19/2024] Open
Abstract
Bioengineering and regenerative medicine strategies are promising for the treatment of vascular diseases. However, current limitations inhibit the ability of these approaches to be translated to clinical practice. Here we summarize some of the big bottlenecks that inhibit vascular regeneration in the disease applications of aortic aneurysms, stroke, and peripheral artery disease. We also describe the bottlenecks preventing three-dimensional bioprinting of vascular networks for tissue engineering applications. Finally, we describe emerging technologies and opportunities to overcome these challenges to advance vascular regeneration.
Collapse
Affiliation(s)
- Dalia A Fantini
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Guang Yang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Epicrispr Biotechnologies, Inc, South San Francisco, CA, USA
| | | | - Divya Subramanian
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Julie A Phillippi
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Ngan F Huang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| |
Collapse
|
9
|
Baek S, Ha HS, Park JS, Cho MJ, Kim HS, Yu SE, Chung S, Kim C, Kim J, Lee JY, Lee Y, Kim H, Nam Y, Cho S, Lee K, Yoon JK, Choi JS, Han DH, Sung HJ. Chip collection of hepatocellular carcinoma based on O 2 heterogeneity from patient tissue. Nat Commun 2024; 15:5117. [PMID: 38879551 PMCID: PMC11180182 DOI: 10.1038/s41467-024-49386-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 06/04/2024] [Indexed: 06/19/2024] Open
Abstract
Hepatocellular carcinoma frequently recurs after surgery, necessitating personalized clinical approaches based on tumor avatar models. However, location-dependent oxygen concentrations resulting from the dual hepatic vascular supply drive the inherent heterogeneity of the tumor microenvironment, which presents challenges in developing an avatar model. In this study, tissue samples from 12 patients with hepatocellular carcinoma are cultured directly on a chip and separated based on preference of oxygen concentration. Establishing a dual gradient system with drug perfusion perpendicular to the oxygen gradient enables the simultaneous separation of cells and evaluation of drug responsiveness. The results are further cross-validated by implanting the chips into mice at various oxygen levels using a patient-derived xenograft model. Hepatocellular carcinoma cells exposed to hypoxia exhibit invasive and recurrent characteristics that mirror clinical outcomes. This chip provides valuable insights into treatment prognosis by identifying the dominant hepatocellular carcinoma type in each patient, potentially guiding personalized therapeutic interventions.
Collapse
Affiliation(s)
- Sewoom Baek
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun-Su Ha
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong Su Park
- Department of Severance Biomedical Science Institute, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Min Jeong Cho
- Department of Clinical Pharmacology & Therapeutics, Catholic University of Korea, Seoul St. Mary's Hospital, 222, BanpoDaero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seung Eun Yu
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seyong Chung
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Chansik Kim
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jueun Kim
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Ji Youn Lee
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yerin Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyunjae Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yujin Nam
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sungwoo Cho
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyubae Lee
- Department of Biomedical Materials, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, Republic of Korea
| | - Ja Kyung Yoon
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Center for Clinical Imaging Data Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jin Sub Choi
- Department of Surgery, Division of Hepato-biliary and Pancreatic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dai Hoon Han
- Department of Surgery, Division of Hepato-biliary and Pancreatic Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| | - Hak-Joon Sung
- Department of Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
10
|
Lammers A, Hsu HH, Sundaram S, Gagnon KA, Kim S, Lee JH, Tung YC, Eyckmans J, Chen CS. Rapid Tissue Perfusion Using Sacrificial Percolation of Anisotropic Networks. MATTER 2024; 7:2184-2204. [PMID: 39221109 PMCID: PMC11360881 DOI: 10.1016/j.matt.2024.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Tissue engineering has long sought to rapidly generate perfusable vascularized tissues with vessel sizes spanning those seen in humans. Current techniques such as biological 3D printing (top-down) and cellular self-assembly (bottom-up) are resource intensive and have not overcome the inherent tradeoff between vessel resolution and assembly time, limiting their utility and scalability for engineering tissues. We present a flexible and scalable technique termed SPAN - Sacrificial Percolation of Anisotropic Networks, where a network of perfusable channels is created throughout a tissue in minutes, irrespective of its size. Conduits with length scales spanning arterioles to capillaries are generated using pipettable alginate fibers that interconnect above a percolation density threshold and are then degraded within constructs of arbitrary size and shape. SPAN is readily used within common tissue engineering processes, can be used to generate endothelial cell-lined vasculature in a multi-cell type construct, and paves the way for rapid assembly of perfusable tissues.
Collapse
Affiliation(s)
- Alex Lammers
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Heng-Hua Hsu
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Subramanian Sundaram
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Keith A. Gagnon
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sudong Kim
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Joshua H. Lee
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Yi-Chung Tung
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Jeroen Eyckmans
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Christopher S. Chen
- The Biological Design Center and Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
- Lead contact
| |
Collapse
|
11
|
Pelin G, Sonmez M, Pelin CE. The Use of Additive Manufacturing Techniques in the Development of Polymeric Molds: A Review. Polymers (Basel) 2024; 16:1055. [PMID: 38674976 PMCID: PMC11054453 DOI: 10.3390/polym16081055] [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: 03/09/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
The continuous growth of additive manufacturing in worldwide industrial and research fields is driven by its main feature which allows the customization of items according to the customers' requirements and limitations. There is an expanding competitiveness in the product development sector as well as applicative research that serves special-use domains. Besides the direct use of additive manufacturing in the production of final products, 3D printing is a viable solution that can help manufacturers and researchers produce their support tooling devices (such as molds and dies) more efficiently, in terms of design complexity and flexibility, timeframe, costs, and material consumption reduction as well as functionality and quality enhancements. The compatibility of the features of 3D printing of molds with the requirements of low-volume production and individual-use customized items development makes this class of techniques extremely attractive to a multitude of areas. This review paper presents a synthesis of the use of 3D-printed polymeric molds in the main applications where molds exhibit a major role, from industrially oriented ones (injection, casting, thermoforming, vacuum forming, composite fabrication) to research or single-use oriented ones (tissue engineering, biomedicine, soft lithography), with an emphasis on the benefits of using 3D-printed polymeric molds, compared to traditional tooling.
Collapse
Affiliation(s)
- George Pelin
- INCAS—National Institute for Aerospace Research “Elie Carafoli”, Bd. Iuliu Maniu 220, 061126 Bucharest, Romania;
| | - Maria Sonmez
- INCDTP-ICPI—National Research and Development Institute for Textile and Leather—Division Leather and Footwear Research Institute, Ion Minulescu St. 93, 031215 Bucharest, Romania;
| | - Cristina-Elisabeta Pelin
- INCAS—National Institute for Aerospace Research “Elie Carafoli”, Bd. Iuliu Maniu 220, 061126 Bucharest, Romania;
| |
Collapse
|
12
|
Nishiguchi A, Ito S, Nagasaka K, Komatsu H, Uto K, Taguchi T. Injectable microcapillary network hydrogels engineered by liquid-liquid phase separation for stem cell transplantation. Biomaterials 2024; 305:122451. [PMID: 38169189 DOI: 10.1016/j.biomaterials.2023.122451] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/17/2023] [Accepted: 12/22/2023] [Indexed: 01/05/2024]
Abstract
Injectable hydrogels are promising carriers for cell delivery in regenerative medicine. However, injectable hydrogels composed of crosslinked polymer networks are often non-microporous and prevent biological communication with host tissues through signals, nutrients, oxygen, and cells, thereby limiting graft survival and tissue integration. Here we report injectable hydrogels with liquid-liquid phase separation-induced microcapillary networks (μCN) as stem cell-delivering scaffolds. The molecular modification of gelatin with hydrogen bonding moieties induced liquid-liquid phase separation when mixed with unmodified gelatin to form μCN structures in the hydrogels. Through spatiotemporally controlled covalent crosslinking and dissolution processes, porous μCN structures were formed in the hydrogels, which can enhance mass transport and cellular activity. The encapsulation of cells with injectable μCN hydrogels improved cellular spreading, migration, and proliferation. Transplantation of mesenchymal stem cells with injectable μCN hydrogels enhanced graft survival and recovered hindlimb ischemia by enhancing material-tissue communication with biological signals and cells through μCN. This facile approach may serve as an advanced scaffold for improving stem cell transplantation therapies in regenerative medicine.
Collapse
Affiliation(s)
- Akihiro Nishiguchi
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan.
| | - Shima Ito
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Kazuhiro Nagasaka
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Hiyori Komatsu
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan
| | - Koichiro Uto
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan
| | - Tetsushi Taguchi
- Biomaterials Field, Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki, 305-0044, Japan; Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
13
|
Jang HJ, Yoon JK. The Role of Vasculature and Angiogenic Strategies in Bone Regeneration. Biomimetics (Basel) 2024; 9:75. [PMID: 38392121 PMCID: PMC10887147 DOI: 10.3390/biomimetics9020075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/24/2024] Open
Abstract
Bone regeneration is a complex process that involves various growth factors, cell types, and extracellular matrix components. A crucial aspect of this process is the formation of a vascular network, which provides essential nutrients and oxygen and promotes osteogenesis by interacting with bone tissue. This review provides a comprehensive discussion of the critical role of vasculature in bone regeneration and the applications of angiogenic strategies, from conventional to cutting-edge methodologies. Recent research has shifted towards innovative bone tissue engineering strategies that integrate vascularized bone complexes, recognizing the significant role of vasculature in bone regeneration. The article begins by examining the role of angiogenesis in bone regeneration. It then introduces various in vitro and in vivo applications that have achieved accelerated bone regeneration through angiogenesis to highlight recent advances in bone tissue engineering. This review also identifies remaining challenges and outlines future directions for research in vascularized bone regeneration.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
| |
Collapse
|
14
|
Mohanto S, Narayana S, Merai KP, Kumar JA, Bhunia A, Hani U, Al Fatease A, Gowda BHJ, Nag S, Ahmed MG, Paul K, Vora LK. Advancements in gelatin-based hydrogel systems for biomedical applications: A state-of-the-art review. Int J Biol Macromol 2023; 253:127143. [PMID: 37793512 DOI: 10.1016/j.ijbiomac.2023.127143] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/06/2023]
Abstract
A gelatin-based hydrogel system is a stimulus-responsive, biocompatible, and biodegradable polymeric system with solid-like rheology that entangles moisture in its porous network that gradually protrudes to assemble a hierarchical crosslinked arrangement. The hydrolysis of collagen directs gelatin construction, which retains arginyl glycyl aspartic acid and matrix metalloproteinase-sensitive degeneration sites, further confining access to chemicals entangled within the gel (e.g., cell encapsulation), modulating the release of encapsulated payloads and providing mechanical signals to the adjoining cells. The utilization of various types of functional tunable biopolymers as scaffold materials in hydrogels has become highly attractive due to their higher porosity and mechanical ability; thus, higher loading of proteins, peptides, therapeutic molecules, etc., can be further modulated. Furthermore, a stimulus-mediated gelatin-based hydrogel with an impaired concentration of gellan demonstrated great shear thinning and self-recovering characteristics in biomedical and tissue engineering applications. Therefore, this contemporary review presents a concise version of the gelatin-based hydrogel as a conceivable biomaterial for various biomedical applications. In addition, the article has recapped the multiple sources of gelatin and their structural characteristics concerning stimulating hydrogel development and delivery approaches of therapeutic molecules (e.g., proteins, peptides, genes, drugs, etc.), existing challenges, and overcoming designs, particularly from drug delivery perspectives.
Collapse
Affiliation(s)
- Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India.
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Khushboo Paresh Merai
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Jahanvee Ashok Kumar
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Adrija Bhunia
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Umme Hani
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - B H Jaswanth Gowda
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India; School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK.
| | - Sagnik Nag
- Department of Bio-Sciences, School of Biosciences & Technology, Vellore Institute of Technology (VIT), Tiruvalam Rd, 632014, Tamil Nadu, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, Karnataka, India
| | - Karthika Paul
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru 570015, Karnataka, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK
| |
Collapse
|
15
|
Curry CW, Sturgeon SM, O'Grady BJ, Yates A, Kjar A, Paige H, Mowery LS, Katdare KA, Patel R, Mlouk K, Stiefbold MR, Vafaie-Partin S, Kawabata A, McKee R, Moore-Lotridge S, Hawkes A, Kusunose J, Gibson-Corley KN, Schmeckpeper J, Schoenecker JG, Caskey CF, Lippmann ES. Growth factor free, peptide-functionalized gelatin hydrogel promotes arteriogenesis and attenuates tissue damage in a murine model of critical limb ischemia. Biomaterials 2023; 303:122397. [PMID: 37979513 PMCID: PMC10843678 DOI: 10.1016/j.biomaterials.2023.122397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/20/2023]
Abstract
Critical limb ischemia (CLI) occurs when blood flow is restricted through the arteries, resulting in ulcers, necrosis, and chronic wounds in the downstream extremities. The development of collateral arterioles (i.e. arteriogenesis), either by remodeling of pre-existing vascular networks or de novo growth of new vessels, can prevent or reverse ischemic damage, but it remains challenging to stimulate collateral arteriole development in a therapeutic context. Here, we show that a gelatin-based hydrogel, devoid of growth factors or encapsulated cells, promotes arteriogenesis and attenuates tissue damage in a murine CLI model. The gelatin hydrogel is functionalized with a peptide derived from the extracellular epitope of Type 1 cadherins. Mechanistically, these "GelCad" hydrogels promote arteriogenesis by recruiting smooth muscle cells to vessel structures in both ex vivo and in vivo assays. In a murine femoral artery ligation model of CLI, delivery of in situ crosslinking GelCad hydrogels was sufficient to restore limb perfusion and maintain tissue health for 14 days, whereas mice treated with gelatin hydrogels had extensive necrosis and autoamputated within 7 days. A small cohort of mice receiving the GelCad hydrogels were aged out to 5 months and exhibited no decline in tissue quality, indicating durability of the collateral arteriole networks. Overall, given the simplicity and off-the-shelf format of the GelCad hydrogel platform, we suggest it could have utility for CLI treatment and potentially other indications that would benefit from arteriole development.
Collapse
Affiliation(s)
- Corinne W Curry
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sarah M Sturgeon
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Brian J O'Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alexis Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hayden Paige
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lucas S Mowery
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Riya Patel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kate Mlouk
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Madison R Stiefbold
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sidney Vafaie-Partin
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Atsuyuki Kawabata
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel McKee
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Adrienne Hawkes
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine N Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Schmeckpeper
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Charles F Caskey
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
16
|
Liu J, Tang C, Huang J, Gu J, Yin J, Xu G, Yan S. Nanofiber Composite Microchannel-Containing Injectable Hydrogels for Cartilage Tissue Regeneration. Adv Healthc Mater 2023; 12:e2302293. [PMID: 37689993 DOI: 10.1002/adhm.202302293] [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: 07/19/2023] [Revised: 09/05/2023] [Indexed: 09/11/2023]
Abstract
Articular cartilage tissue is incapable of self-repair and therapies for cartilage defects are still lacking. Injectable hydrogels have drawn much attention in the field of cartilage regeneration. Herein, the novel design of nanofiber composite microchannel-containing hydrogels inspired by the tunnel-piled structure of subway tunnels is proposed. Based on the aldehydized polyethylene glycol/carboxymethyl chitosan (APA/CMCS) hydrogels, thermosensitive gelatin microrods (GMs) are used as a pore-forming agent, and coaxial electrospinning polylactic acid/gelatin fibers (PGFs) loaded with kartogenin (KGN) are used as a reinforcing agent and a drug delivery system to construct the nanofiber composite microchannel-containing injectable hydrogels (APA/CMCS/KGN@PGF/GM hydrogels). The in situ formation, micromorphology and porosity, swelling and degradation, mechanical properties, self-healing behavior, as well as drug release of the nanofiber composite microchannel-containing hydrogels are investigated. The hydrogel exhibits good self-healing ability, and the introduction of PGF nanofibers can significantly improve the mechanical properties. The drug delivery system can realize sustained release of KGN to match the process of cartilage repair. The microchannel structure effectively promotes bone marrow mesenchymal stem cell (BMSC) proliferation and ingrowth within the hydrogels. In vitro and animal experiments indicate that the APA/CMCS/KGN@PGF/GM hydrogels can enhance the chondrogenesis of BMSCs and promote neocartilage formation in the rabbit cartilage defect model.
Collapse
Affiliation(s)
- Jia Liu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Chen Tang
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Huang
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Jinhong Gu
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Jingbo Yin
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Guohua Xu
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, 200003, P. R. China
| | - Shifeng Yan
- Department of Polymer Materials, School of Materials Science and Engineering, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
17
|
Yu Y, Wang S, Chen X, Gao Z, Dai K, Wang J, Liu C. Sulfated oligosaccharide activates endothelial Notch for inducing macrophage-associated arteriogenesis to treat ischemic diseases. Proc Natl Acad Sci U S A 2023; 120:e2307480120. [PMID: 37943835 PMCID: PMC10655224 DOI: 10.1073/pnas.2307480120] [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: 05/04/2023] [Accepted: 10/01/2023] [Indexed: 11/12/2023] Open
Abstract
Ischemic diseases lead to considerable morbidity and mortality, yet conventional clinical treatment strategies for therapeutic angiogenesis fall short of being impactful. Despite the potential of biomaterials to deliver pro-angiogenic molecules at the infarct site to induce angiogenesis, their efficacy has been impeded by aberrant vascular activation and off-target circulation. Here, we present a semisynthetic low-molecular sulfated chitosan oligosaccharide (SCOS) that efficiently induces therapeutic arteriogenesis with a spontaneous generation of collateral circulation and blood reperfusion in rodent models of hind limb ischemia and myocardial infarction. SCOS elicits anti-inflammatory macrophages' (Mφs') differentiation into perivascular Mφs, which in turn directs artery formation via a cell-to-cell communication rather than secretory factor regulation. SCOS-mediated arteriogenesis requires a canonical Notch signaling pathway in Mφs via the glycosylation of protein O-glucosyltransferases 2, which results in promoting arterial differentiation and tissue repair in ischemia. Thus, this highly bioactive oligosaccharide can be harnessed to direct efficiently therapeutic arteriogenesis and perfusion for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Shuang Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Xinye Chen
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Zehua Gao
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Kai Dai
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Changsheng Liu
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| |
Collapse
|
18
|
Hao M, Wang D, Duan M, Kan S, Li S, Wu H, Xiang J, Liu W. Functional drug-delivery hydrogels for oral and maxillofacial wound healing. Front Bioeng Biotechnol 2023; 11:1241660. [PMID: 37600316 PMCID: PMC10434880 DOI: 10.3389/fbioe.2023.1241660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 07/21/2023] [Indexed: 08/22/2023] Open
Abstract
The repair process for oral and maxillofacial injuries involves hemostasis, inflammation, proliferation, and remodeling. Injury repair involves a variety of cells, including platelets, immune cells, fibroblasts, and various cytokines. Rapid and adequate healing of oral and maxillofacial trauma is a major concern to patients. Functional drug-delivery hydrogels play an active role in promoting wound healing and have shown unique advantages in wound dressings. Functional hydrogels promote wound healing through their adhesive, anti-inflammatory, antioxidant, antibacterial, hemostatic, angiogenic, and re-epithelialization-promoting properties, effectively sealing wounds and reducing inflammation. In addition, functional hydrogels can respond to changes in temperature, light, magnetic fields, pH, and reactive oxygen species to release drugs, enabling precise treatment. Furthermore, hydrogels can deliver various cargos that promote healing, including nucleic acids, cytokines, small-molecule drugs, stem cells, exosomes, and nanomaterials. Therefore, functional drug-delivery hydrogels have a positive impact on the healing of oral and maxillofacial injuries. This review describes the oral mucosal structure and healing process and summarizes the currently available responsive hydrogels used to promote wound healing.
Collapse
Affiliation(s)
- Ming Hao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Dongxu Wang
- Laboratory Animal Center, College of Animal Science, Jilin University, Changchun, China
| | - Mengna Duan
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shaoning Kan
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Shuangji Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Han Wu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Jingcheng Xiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Weiwei Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
19
|
Curry CW, Sturgeon SM, O’Grady BJ, Yates AK, Kjar A, Paige HA, Mowery LS, Katdare KA, Patel RV, Mlouk K, Stiefbold MR, Vafaie-Partin S, Kawabata A, McKee RM, Moore-Lotridge S, Hawkes A, Kusunose J, Gibson-Corley KN, Schmeckpeper J, Schoenecker JG, Caskey CF, Lippmann ES. Growth factor-free, peptide-functionalized gelatin hydrogel promotes arteriogenesis and attenuates tissue damage in a murine model of critical limb ischemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.542150. [PMID: 37292898 PMCID: PMC10245920 DOI: 10.1101/2023.05.24.542150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Critical limb ischemia (CLI) occurs when blood flow is restricted through the arteries, resulting in ulcers, necrosis, and chronic wounds in the downstream extremities. The development of collateral arterioles (i.e. arteriogenesis), either by remodeling of pre-existing vascular networks or de novo growth of new vessels, can prevent or reverse ischemic damage, but it remains challenging to stimulate collateral arteriole development in a therapeutic context. Here, we show that a gelatin-based hydrogel, devoid of growth factors or encapsulated cells, promotes arteriogenesis and attenuates tissue damage in a murine CLI model. The gelatin hydrogel is functionalized with a peptide derived from the extracellular epitope of Type 1 cadherins. Mechanistically, these "GelCad" hydrogels promote arteriogenesis by recruiting smooth muscle cells to vessel structures in both ex vivo and in vivo assays. In a murine femoral artery ligation model of CLI, delivery of in situ crosslinking GelCad hydrogels was sufficient to restore limb perfusion and maintain tissue health for 14 days, whereas mice treated with gelatin hydrogels had extensive necrosis and autoamputated within 7 days. A small cohort of mice receiving the GelCad hydrogels were aged out to 5 months and exhibited no decline in tissue quality, indicating durability of the collateral arteriole networks. Overall, given the simplicity and off-the-shelf format of the GelCad hydrogel platform, we suggest it could have utility for CLI treatment and potentially other indications that would benefit from arteriole development.
Collapse
Affiliation(s)
- Corinne W. Curry
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sarah M. Sturgeon
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Brian J. O’Grady
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Alexis K. Yates
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
| | - Andrew Kjar
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Hayden A. Paige
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Lucas S. Mowery
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Ketaki A. Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Riya V. Patel
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kate Mlouk
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Madison R. Stiefbold
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Sidney Vafaie-Partin
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Atsuyuki Kawabata
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel M. McKee
- Department of Orthopedics, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Adrienne Hawkes
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology and Immunology, Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Schmeckpeper
- Department of Cardiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Charles F. Caskey
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ethan S. Lippmann
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Interdisciplinary Materials Science Program, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
20
|
Jung SH, Jang BH, Kwon S, Park SJ, Park TE, Kang JH. Nematic Fibrin Fibers Enabling Vascularized Thrombus Implants Facilitate Scarless Cutaneous Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2211149. [PMID: 37052392 DOI: 10.1002/adma.202211149] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/15/2023] [Indexed: 06/19/2023]
Abstract
Autologous implantable scaffolds that induce vasculogenesis have shown great potential in tissue regeneration; however, previous attempts mainly relied on cell-laden hydrogel patches using fat tissues or platelet-rich plasma, which are insufficient for generating a uniform vasculature in a scalable manner. Here, implantable vascularized engineered thrombi (IVETs) are presented using autologous whole blood, which potentiate effective skin wound healing by constructing robust microcapillary vessel networks at the wound site. Microfluidic shear stresses enable the alignment of bundled fibrin fibers along the direction of the blood flow streamlines and the activation of platelets, both of which offer moderate stiffness of the microenvironment optimal for facilitating endothelial cell maturation and vascularization. Rodent dorsal skin wounds patched with IVET present superior wound closure rates (96.08 ± 1.58%), epidermis thickness, collagen deposition, hair follicle numbers, and neutrophil infiltration, which are permitted by enhanced microvascular circulation. Moreover, IVET treatment accelerates wound healing by recruiting M2 phenotype macrophages.
Collapse
Affiliation(s)
- Su Hyun Jung
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| | - Bong Hwan Jang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| | - Seyong Kwon
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| | - Sung Jin Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| | - Joo H Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), UNIST gil 50, Ulsan, 44919, Republic of Korea
| |
Collapse
|
21
|
Yang X, Ma Y, Wang X, Yuan S, Huo F, Yi G, Zhang J, Yang B, Tian W. A 3D-Bioprinted Functional Module Based on Decellularized Extracellular Matrix Bioink for Periodontal Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205041. [PMID: 36516309 PMCID: PMC9929114 DOI: 10.1002/advs.202205041] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/15/2022] [Indexed: 05/14/2023]
Abstract
Poor fiber orientation and mismatched bone-ligament interface fusion have plagued the regeneration of periodontal defects by cell-based scaffolds. A 3D bioprinted biomimetic periodontal module is designed with high architectural integrity using a methacrylate gelatin/decellularized extracellular matrix (GelMA/dECM) cell-laden bioink. The module presents favorable mechanical properties and orientation guidance by high-precision topographical cues and provides a biochemical environment conducive to regulating encapsulated cell behavior. The dECM features robust immunomodulatory activity, reducing the release of proinflammatory factors by M1 macrophages and decreasing local inflammation in Sprague Dawley rats. In a clinically relevant critical-size periodontal defect model, the bioprinted module significantly enhances the regeneration of hybrid periodontal tissues in beagles, especially the anchoring structures of the bone-ligament interface, well-aligned periodontal fibers, and highly mineralized alveolar bone. This demonstrates the effectiveness and feasibility of 3D bioprinting combined with a dental follicle-specific dECM bioink for periodontium regeneration, providing new avenues for future clinical practice.
Collapse
Affiliation(s)
- Xueting Yang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Yue Ma
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Xiuting Wang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Shengmeng Yuan
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Fangjun Huo
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Genzheng Yi
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Jingyi Zhang
- Chengdu Shiliankangjian Biotechnology Co., Ltd.Chengdu610041P. R. China
| | - Bo Yang
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| | - Weidong Tian
- State Key Laboratory of Oral DiseasesNational Engineering Laboratory for Oral Regenerative MedicineEngineering Research Center of Oral Translational MedicineMinistry of EducationDepartment of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengdu610041P. R. China
| |
Collapse
|
22
|
Wang L, Zhao W, Zhao Y, Li W, Wang G, Zhang Q. Enzymatically-mineralized double-network hydrogels with ultrahigh mechanical strength, toughness, and stiffness. Theranostics 2023; 13:673-684. [PMID: 36632214 PMCID: PMC9830447 DOI: 10.7150/thno.77417] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 12/07/2022] [Indexed: 01/04/2023] Open
Abstract
Background: Synthetic hydrogels are commonly mechanically weak which limits the scope of their applications. Methods: In this study, we synthesized an organic-inorganic hybrid hydrogel with ultrahigh strength, stiffness, and toughness via enzyme-induced mineralization of calcium phosphate in a double network of bacterial cellulose nanofibers and alginate-Ca2+. Results: Cellulose nanofibers formed the first rigid network via hydrogen binding and templated the deposition of calcium phosphate, while alginate-Ca2+ formed the second energy-dissipating network via ionic interaction. The two networks created a brick-mortar-like structure, in which the "tortuous fracture path" mechanism by breaking the interlaced calcium phosphate-coated bacterial cellulose nanofibers and the hysteresis by unzipping the ionic alginate-Ca2+ network made a great contribution to the mechanical properties of the hydrogels. Conclusion: The optimized hydrogel exhibited ultrahigh fracture stress of 48 MPa, Young's modulus of 1329 MPa, and fracture energy of 3013 J/m2, which are barely possessed by the reported synthetic hydrogels. Finally, the hydrogel represented potential use in subchondral bone defect repair in an ex vivo model.
Collapse
Affiliation(s)
- Li Wang
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P.R. China
| | - Wei Zhao
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P. R. China
| | - Yining Zhao
- Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P.R. China
| | - Wei Li
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P. R. China.,✉ Corresponding authors: Q. Z. (E-mail: ); G. W. (E-mail: ); W. L. (E-mail: li_wei_sh@hotmail. com)
| | - Guodong Wang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P. R. China.,✉ Corresponding authors: Q. Z. (E-mail: ); G. W. (E-mail: ); W. L. (E-mail: li_wei_sh@hotmail. com)
| | - Qiang Zhang
- Department of Stomatology, Changzheng Hospital, Naval Medical University, Shanghai, 200003, P. R. China.,Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai, 200241, P.R. China.,✉ Corresponding authors: Q. Z. (E-mail: ); G. W. (E-mail: ); W. L. (E-mail: li_wei_sh@hotmail. com)
| |
Collapse
|
23
|
Lee S, Lee J, Baek J, Park CH, Kim CS. Design of Volumetric Nanolayers via Rapid Proteolysis of Silk Fibroin for Tissue Engineering. Biomacromolecules 2022; 23:4995-5006. [PMID: 36367817 DOI: 10.1021/acs.biomac.2c00802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Various methods have been studied to make a regenerated silk fibroin solution. However, most of them take too much time and effort to liquefy. Here, we report that a regenerated silk fibroin solution could be prepared within seconds through acid proteolysis for the first time. The solubilized fibroin could be applied to advanced tissue engineering. Our method shortened the production time to one day (more than 10 times) compared to the general fibroin solution preparation method. It was confirmed that the initial protein affinity nearly doubled from 0.028 to 0.076 μg·mm-2 in FF(ac) compared to FF(aq). A fibroin nanofiber layer having a volumetric hierarchical structure was prepared by electrospinning an acid-proteolyzed fibroin solution, followed by gas foaming. In vitro results of cell adhesion and proliferation capacity of the gas-foamed scaffold were not significantly different compared to the two-dimensional (2D) fibroin nanofiber membrane, overcoming the limitations of volumetric nanofiber scaffolds. We are confident that our research will greatly contribute to the development of regenerative engineering using other proteins.
Collapse
Affiliation(s)
- Sunny Lee
- Department of Bionanosystem Engineering, Jeonbuk National University, Jeonju561-756, Jeonbuk, Republic of Korea
| | - Joshua Lee
- Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju561-756, Republic of Korea
| | - Jiwon Baek
- Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju561-756, Republic of Korea
| | - Chan Hee Park
- Department of Bionanosystem Engineering, Jeonbuk National University, Jeonju561-756, Jeonbuk, Republic of Korea.,Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju561-756, Republic of Korea.,Division of Mechanical Design Engineering, Jeonbuk National University, Jeonju561-756, Jeonbuk, Republic of Korea
| | - Cheol Sang Kim
- Department of Bionanosystem Engineering, Jeonbuk National University, Jeonju561-756, Jeonbuk, Republic of Korea.,Department of Bionanotechnology and Bioconvergence Engineering, Graduate School, Jeonbuk National University, Jeonju561-756, Republic of Korea.,Division of Mechanical Design Engineering, Jeonbuk National University, Jeonju561-756, Jeonbuk, Republic of Korea
| |
Collapse
|
24
|
Yang Z, Shi C, Cheng D, Wang Y, Xing Y, Du F, Wu F, Jin Y, Dong Y, Li M. Biomimetic nanomaterial-facilitated oxygen generation strategies for enhancing tumour treatment outcomes. Front Bioeng Biotechnol 2022; 10:1007960. [PMID: 36277398 PMCID: PMC9581162 DOI: 10.3389/fbioe.2022.1007960] [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: 07/31/2022] [Accepted: 09/20/2022] [Indexed: 12/05/2022] Open
Abstract
Hypoxia, as a typical hallmark of the tumour microenvironment (TME), has been verified to exist in most malignancies and greatly hinders the outcome of tumour treatments, including chemotherapy, photodynamic therapy, radiotherapy, and immunotherapy. Various approaches to alleviate tumour hypoxia have been reported. Among them, biomimetic nanomaterial-facilitated tumour oxygenation strategies, based on the engagement of human endogenous proteins, red blood cells, the cell membrane, and catalase, are the most impressive due to their excellent tumour active-targeting ability and superior tumour-selective capability, which, however, have not yet been systematically reviewed. Herein, we are ready to describe the current progress in biomimetic nanomaterial-facilitated tumour oxygenation strategies and corresponding improvements in tumour treatment outputs. In this review, the underlying mechanism behind the superior effect of these biomimetic nanomaterials, compared with other materials, on alleviating the hypoxic TME is highlighted. Additionally, the ongoing problems and potential solutions are also discussed.
Collapse
|
25
|
Pan Y, Luo Y, Hong J, He H, Dai L, Zhu H, Wu J. Advances for the treatment of lower extremity arterial disease associated with diabetes mellitus. Front Mol Biosci 2022; 9:929718. [PMID: 36060247 PMCID: PMC9429832 DOI: 10.3389/fmolb.2022.929718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Lower extremity arterial disease (LEAD) is a major vascular complication of diabetes. Vascular endothelial cells dysfunction can exacerbate local ischemia, leading to a significant increase in amputation, disability, and even mortality in patients with diabetes combined with LEAD. Therefore, it is of great clinical importance to explore proper and effective treatments. Conventional treatments of diabetic LEAD include lifestyle management, medication, open surgery, endovascular treatment, and amputation. As interdisciplinary research emerges, regenerative medicine strategies have provided new insights to treat chronic limb threatening ischemia (CLTI). Therapeutic angiogenesis strategies, such as delivering growth factors, stem cells, drugs to ischemic tissues, have also been proposed to treat LEAD by fundamentally stimulating multidimensional vascular regeneration. Recent years have seen the rapid growth of tissue engineering technology; tissue-engineered biomaterials have been used to study the treatment of LEAD, such as encapsulation of growth factors and drugs in hydrogel to facilitate the restoration of blood perfusion in ischemic tissues of animals. The primary purpose of this review is to introduce treatments and novel biomaterials development in LEAD. Firstly, the pathogenesis of LEAD is briefly described. Secondly, conventional therapies and therapeutic angiogenesis strategies of LEAD are discussed. Finally, recent research advances and future perspectives on biomaterials in LEAD are proposed.
Collapse
Affiliation(s)
- Yang Pan
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuting Luo
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jing Hong
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huacheng He
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, China
- *Correspondence: Huacheng He, ; Hong Zhu,
| | - Lu Dai
- The Fourth Outpatient Department, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Zhu
- Department of Endocrinology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Huacheng He, ; Hong Zhu,
| | - Jiang Wu
- Key Laboratory of Biotechnology and Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
26
|
Baek S, Yu SE, Deng Y, Lee Y, Lee DG, Kim S, Yoon S, Kim H, Park J, Lee CH, Lee JB, Kong HJ, Kang S, Shin YM, Sung H. Quenching Epigenetic Drug Resistance Using Antihypoxic Microparticles in Glioblastoma Patient-Derived Chips. Adv Healthc Mater 2022; 11:e2102226. [PMID: 34963195 PMCID: PMC11468717 DOI: 10.1002/adhm.202102226] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 12/22/2021] [Indexed: 11/06/2022]
Abstract
Glioblastoma (GBM) is one of the most intractable tumor types due to the progressive drug resistance upon tumor mass expansion. Incremental hypoxia inside the growing tumor mass drives epigenetic drug resistance by activating nongenetic repair of antiapoptotic DNA, which could be impaired by drug treatment. Hence, rescuing intertumor hypoxia by oxygen-generating microparticles may promote susceptibility to antitumor drugs. Moreover, a tumor-on-a-chip model enables user-specified alternation of clinic-derived samples. This study utilizes patient-derived glioblastoma tissue to generate cell spheroids with size variations in a 3D microchannel network chip (GBM chip). As the spheroid size increases, epigenetic drug resistance is promoted with inward hypoxia severance, as supported by the spheroid size-proportional expression of hypoxia-inducible factor-1a in the chip. Loading antihypoxia microparticles onto the spheroid surface significantly reduces drug resistance by silencing the expression of critical epigenetic factor, resulting in significantly decreased cell invasiveness. The results are confirmed in vitro using cell line and patient samples in the chip as well as chip implantation into a hypoxic hindlimb ischemia model in mice, which is an unprecedented approach in the field.
Collapse
Affiliation(s)
- Sewoom Baek
- Department of Brain Korea 21 FOUR Project for Medical ScienceMedical Device Engineering and ManagementDepartment of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Seung Eun Yu
- Department of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Yu‐Heng Deng
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Yong‐Jae Lee
- Department of Obstetrics and GynecologyYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Dong Gue Lee
- Department of NeurosurgeryYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Surim Kim
- Department of Bio‐convergenceYonsei University Underwood International College50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Seonjin Yoon
- Department of NeurosurgeryYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Hye‐Seon Kim
- Department of Brain Korea 21 FOUR Project for Medical ScienceMedical Device Engineering and ManagementDepartment of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Jeongeun Park
- Department of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Chan Hee Lee
- Department of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Jung Bok Lee
- Department of Biological ScienceSookmyung Women's University25, Cheongpa‐ro 47ga‐gil, Yongsan‐guSeoul04314Republic of Korea
| | - Hyun Joon Kong
- Department of Chemical and Biomolecular EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIL61801USA
| | - Seok‐Gu Kang
- Department of NeurosurgeryYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Young Min Shin
- Department of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| | - Hak‐Joon Sung
- Department of Medical EngineeringYonsei University College of Medicine50‐1 Yonsei‐ro, Seodaemun‐guSeoul03722Republic of Korea
| |
Collapse
|
27
|
Li R, Liu K, Huang X, Li D, Ding J, Liu B, Chen X. Bioactive Materials Promote Wound Healing through Modulation of Cell Behaviors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105152. [PMID: 35138042 PMCID: PMC8981489 DOI: 10.1002/advs.202105152] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/24/2021] [Indexed: 05/13/2023]
Abstract
Skin wound repair is a multistage process involving multiple cellular and molecular interactions, which modulate the cell behaviors and dynamic remodeling of extracellular matrices to maximize regeneration and repair. Consequently, abnormalities in cell functions or pathways inevitably give rise to side effects, such as dysregulated inflammation, hyperplasia of nonmigratory epithelial cells, and lack of response to growth factors, which impedes angiogenesis and fibrosis. These issues may cause delayed wound healing or even non-healing states. Current clinical therapeutic approaches are predominantly dedicated to preventing infections and alleviating topical symptoms rather than addressing the modulation of wound microenvironments to achieve targeted outcomes. Bioactive materials, relying on their chemical, physical, and biological properties or as carriers of bioactive substances, can affect wound microenvironments and promote wound healing at the molecular level. By addressing the mechanisms of wound healing from the perspective of cell behaviors, this review discusses how bioactive materials modulate the microenvironments and cell behaviors within the wounds during the stages of hemostasis, anti-inflammation, tissue regeneration and deposition, and matrix remodeling. A deeper understanding of cell behaviors during wound healing is bound to promote the development of more targeted and efficient bioactive materials for clinical applications.
Collapse
Affiliation(s)
- Ruotao Li
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Kai Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Xu Huang
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| | - Bin Liu
- Department of Hand and Foot SurgeryThe First Hospital of Jilin University1 Xinmin StreetChangchun130065P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin StreetChangchun130022P. R. China
| |
Collapse
|
28
|
Xing Z, Zhao C, Wu S, Yang D, Zhang C, Wei X, Wei X, Su H, Liu H, Fan Y. Hydrogel Loaded with VEGF/TFEB-Engineered Extracellular Vesicles for Rescuing Critical Limb Ischemia by a Dual-Pathway Activation Strategy. Adv Healthc Mater 2022; 11:e2100334. [PMID: 34297471 DOI: 10.1002/adhm.202100334] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/03/2021] [Indexed: 02/05/2023]
Abstract
Critical limb ischemia (CLI) is the most severe clinical manifestation of peripheral arterial disease, which causes many amputations and deaths. Conventional treatment strategies for CLI (e.g., stent implantation and vascular surgery) bring surgical risk, which are not suitable for each patient. Extracellular vesicles (EVs) can be a potential solution for CLI. Herein, vascular endothelial growth factor (VEGF; i.e., a crucial molecule related to angiogenesis) and transcription factor EB (TFEB; i.e., a pivotal regulator of autophagy) are chosen as the target gene to improve the bioactivity of EVs derived from endothelial cells. The VEGF/TFEB-engineered EVs (Engineered-EVs) are fabricated by genetically engineering the parent cells, and their versatile functions are confirmed using three cell models (human umbilical vein endothelial cells, myoblast, and monocytes). Injectable thermal-responsive hydrogel are then combined with Engineered-EVs to combat CLI. These results reveal that the hydrogel can enhance the stability of Engineered-EVs in vivo and release EVs at different temperatures. Moreover, the results of animal studies indicate that Engineered-EV/Hydrogel can significantly improve neovascularization, attenuate muscle injury, and recover limb function after CLI. Finally, mechanistic studies shed light on the therapeutic effect of Engineered-EV/Hydrogel due to the activated VEGF/VEGFR pathway and autophagy-lysosomal pathway.
Collapse
Affiliation(s)
- Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| | - Chen Zhao
- School of Pharmaceutical Sciences Tsinghua University Beijing 100084 P. R. China
| | - Siwen Wu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy West China Hospital Sichuan University Chengdu 610041 P. R. China
| | - Depeng Yang
- School of Life Sciences and Technology Harbin Institute of Technology Harbin Heilongjiang 150001 P. R. China
| | - Chunchen Zhang
- Key Laboratory of Biomedical Engineering of Ministry of Education Zhejiang University Hangzhou 310027 China
| | - Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| | - Xinran Wei
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| | - Haoran Su
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education Beijing Advanced Innovation Centre for Biomedical Engineering School of Biological Science and Medical Engineering Beihang University Beijing 100191 P. R. China
| |
Collapse
|
29
|
Costa ALR, Willerth SM, de la Torre LG, Han SW. Trends in hydrogel-based encapsulation technologies for advanced cell therapies applied to limb ischemia. Mater Today Bio 2022; 13:100221. [PMID: 35243296 PMCID: PMC8866736 DOI: 10.1016/j.mtbio.2022.100221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/28/2022] [Accepted: 02/12/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Ana Letícia Rodrigues Costa
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, V8W 2Y2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, V8W 2Y2, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Lucimara Gaziola de la Torre
- Department of Materials and Bioprocesses Engineering, School of Chemical Engineering, University of Campinas, Campinas, SP, Brazil
| | - Sang Won Han
- Department of Biophysics, Escola Paulista de Medicina, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
- Corresponding author.
| |
Collapse
|
30
|
Soliman BG, Major GS, Atienza-Roca P, Murphy CA, Longoni A, Alcala-Orozco CR, Rnjak-Kovacina J, Gawlitta D, Woodfield TBF, Lim KS. Development and Characterization of Gelatin-Norbornene Bioink to Understand the Interplay between Physical Architecture and Micro-Capillary Formation in Biofabricated Vascularized Constructs. Adv Healthc Mater 2022; 11:e2101873. [PMID: 34710291 DOI: 10.1002/adhm.202101873] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/21/2021] [Indexed: 12/12/2022]
Abstract
The principle challenge for engineering viable, cell-laden hydrogel constructs of clinically-relevant size, is rapid vascularization, in order to moderate the finite capacity of passive nutrient diffusion. A multiscale vascular approach, with large open channels and bulk microcapillaries may be an admissible approach to accelerate this process, promoting overall pre-vascularization for long-term viability of constructs. However, the limited availability of bioinks that possess suitable characteristics that support both fabrication of complex architectures and formation of microcapillaries, remains a barrier to advancement in this space. In this study, gelatin-norbornene (Gel-NOR) is investigated as a vascular bioink with tailorable physico-mechanical properties, which promoted the self-assembly of human stromal and endothelial cells into microcapillaries, as well as being compatible with extrusion and lithography-based biofabrication modalities. Gel-NOR constructs containing self-assembled microcapillaries are successfully biofabricated with varying physical architecture (fiber diameter, spacing, and orientation). Both channel sizes and cell types affect the overall structural changes of the printed constructs, where cross-signaling between both human stromal and endothelial cells may be responsible for the reduction in open channel lumen observed over time. Overall, this work highlights an exciting three-way interplay between bioink formulation, construct design, and cell-mediated response that can be exploited towards engineering vascular tissues.
Collapse
Affiliation(s)
- Bram G Soliman
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Gretel S Major
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Pau Atienza-Roca
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Caroline A Murphy
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Alessia Longoni
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Cesar R Alcala-Orozco
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, 2006, Australia
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery and Special Dental Care, University Medical Center Utrecht, Utrecht, GA, 3508, The Netherlands
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Light Activated Biomaterials (LAB) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, Christchurch, 8011, New Zealand
| |
Collapse
|
31
|
Yoon H, Chung YS, Lee YJ, Yu SE, Baek S, Kim H, Kim SW, Lee J, Kim S, Sung H. Cancer Patient Tissueoid with Self-Homing Nano-Targeting of Metabolic Inhibitor. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102640. [PMID: 34664430 PMCID: PMC8596099 DOI: 10.1002/advs.202102640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/02/2021] [Indexed: 05/11/2023]
Abstract
The current paradigm of cancer medicine focuses on patient- and/or cancer-specific treatments, which has led to continuous progress in the development of patient representatives (e.g., organoids) and cancer-targeting carriers for drug screening. As breakthrough concepts, i) living cancer tissues convey intact profiles of patient-specific microenvironmental signatures. ii) The growth mechanisms of cancer mass with intense cell-cell interactions can be harnessed to develop self-homing nano-targeting by using cancer cell-derived nanovesicles (CaNVs). Hence, a tissueoid model of ovarian cancer (OC) is developed by culturing OC patient tissues in a 3D gel chip, whose microchannel networks enable perfusion to maintain tissue viability. A novel model of systemic cancer responses is approached by xenografting OC tissueoids into ischaemic hindlimbs in nude mice. CaNVs are produced to carry general chemotherapeutics or new drugs under pre/clinical studies that target the BRCA mutation or energy metabolism, thereby increasing the test scope. This pioneer study cross-validates drug responses from the OC clinic, tissueoid, and animal model by demonstrating the alignment of results in drug type-specific efficiency, BRCA mutation-dependent drug efficiency, and metabolism inhibition-based anti-cancer effects. Hence, this study provides a directional foundation to accelerate the discovery of patient-specific drugs with CaNV application towards future precision medicine.
Collapse
Affiliation(s)
- Hyo‐Jin Yoon
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Young Shin Chung
- Department of Obstetrics and GynecologyInstitution of Women's Life Medical ScienceSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Yong Jae Lee
- Department of Obstetrics and GynecologyInstitution of Women's Life Medical ScienceSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Seung Eun Yu
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Sewoom Baek
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hye‐Seon Kim
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| | - Sang Wun Kim
- Department of Obstetrics and GynecologyInstitution of Women's Life Medical ScienceSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Jung‐Yun Lee
- Department of Obstetrics and GynecologyInstitution of Women's Life Medical ScienceSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Sunghoon Kim
- Department of Obstetrics and GynecologyInstitution of Women's Life Medical ScienceSeverance HospitalYonsei University College of MedicineSeoul03722Republic of Korea
| | - Hak‐Joon Sung
- Department of Medical EngineeringYonsei University College of MedicineSeoul03722Republic of Korea
| |
Collapse
|
32
|
Cho S, Jang J. Recent Trends in Biofabrication Technologies for Studying Skeletal Muscle Tissue-Related Diseases. Front Bioeng Biotechnol 2021; 9:782333. [PMID: 34778240 PMCID: PMC8578921 DOI: 10.3389/fbioe.2021.782333] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 01/15/2023] Open
Abstract
In native skeletal muscle, densely packed myofibers exist in close contact with surrounding motor neurons and blood vessels, which are embedded in the fibrous connective tissue. In comparison to conventional two-dimensional (2D) cultures, the three-dimensional (3D) engineered skeletal muscle models allow structural and mechanical resemblance with native skeletal muscle tissue by providing geometric confinement and physiological matrix stiffness to the cells. In addition, various external stimuli applied to these models enhance muscle maturation along with cell-cell and cell-extracellular matrix interaction. Therefore, 3D in vitro muscle models can adequately recapitulate the pathophysiologic events occurring in tissue-tissue interfaces inside the native skeletal muscle such as neuromuscular junction. Moreover, 3D muscle models can induce pathological phenotype of human muscle dystrophies such as Duchenne muscular dystrophy by incorporating patient-derived induced pluripotent stem cells and human primary cells. In this review, we discuss the current biofabrication technologies for modeling various skeletal muscle tissue-related diseases (i.e., muscle diseases) including muscular dystrophies and inflammatory muscle diseases. In particular, these approaches would enable the discovery of novel phenotypic markers and the mechanism study of human muscle diseases with genetic mutations.
Collapse
Affiliation(s)
- Seungyeun Cho
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, South Korea
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, South Korea
| |
Collapse
|
33
|
Mavris SM, Hansen LM. Optimization of Oxygen Delivery Within Hydrogels. J Biomech Eng 2021; 143:101004. [PMID: 33973004 PMCID: PMC8299803 DOI: 10.1115/1.4051119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/05/2021] [Indexed: 12/19/2022]
Abstract
The field of tissue engineering has been continuously evolving since its inception over three decades ago with numerous new advancements in biomaterials and cell sources and widening applications to most tissues in the body. Despite the substantial promise and great opportunities for the advancement of current medical therapies and procedures, the field has yet to capture wide clinical translation due to some remaining challenges, including oxygen availability within constructs, both in vitro and in vivo. While this insufficiency of nutrients, specifically oxygen, is a limitation within the current frameworks of this field, the literature shows promise in new technological advances to efficiently provide adequate delivery of nutrients to cells. This review attempts to capture the most recent advances in the field of oxygen transport in hydrogel-based tissue engineering, including a comparison of current research as it pertains to the modeling, sensing, and optimization of oxygen within hydrogel constructs as well as new technological innovations to overcome traditional diffusion-based limitations. The application of these findings can further the advancement and development of better hydrogel-based tissue engineered constructs for future clinical translation and adoption.
Collapse
Affiliation(s)
- Sophia M. Mavris
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332
| | - Laura M. Hansen
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA 30322
| |
Collapse
|
34
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
35
|
Chang CW, Yeh YC. Poly(glycerol sebacate)-co-poly(ethylene glycol)/Gelatin Hybrid Hydrogels as Biocompatible Biomaterials for Cell Proliferation and Spreading. Macromol Biosci 2021; 21:e2100248. [PMID: 34514730 DOI: 10.1002/mabi.202100248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/06/2021] [Indexed: 01/05/2023]
Abstract
Synthetic polymers have been widely employed to prepare hydrogels for biomedical applications, such as cell culture, drug delivery, and tissue engineering. However, the activity of cells cultured in the synthetic polymer-based hydrogels faces the challenges of limited cell proliferation and spreading compared to cells cultured in natural polymer-based hydrogels. To address this concern, a hybrid hydrogel strategy is demonstrated by incorporating thiolated gelatin (GS) into the norbornene-functionalized poly (glycerol sebacate)-co-polyethylene glycol (Nor_PGS-co-PEG, NPP) network to prepare highly biocompatible NPP/GS_UV hydrogels after the thiol-ene photo-crosslinking reaction. The GS introduces several desirable features (i.e., enhanced water content, enlarged pore size, increased mechanical property, and more cell adhesion sites) to the NPP/GS_UV hydrogels, facilitating the cell proliferation and spreading inside the network. Thus, the highly biocompatible NPP/GS_UV hydrogels are promising materials for cell encapsulation and tissue engineering applications. Taken together, the hybrid hydrogel strategy is demonstrated as a powerful approach to fabricate hydrogels with a highly friendly environment for cell culture, expanding the biomedical applications of hydrogels.
Collapse
Affiliation(s)
- Chun-Wei Chang
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan
| | - Yi-Cheun Yeh
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, 10617, Taiwan
| |
Collapse
|
36
|
Blake C, Massey O, Boyd-Moss M, Firipis K, Rifai A, Franks S, Quigley A, Kapsa R, Nisbet DR, Williams RJ. Replace and repair: Biomimetic bioprinting for effective muscle engineering. APL Bioeng 2021; 5:031502. [PMID: 34258499 PMCID: PMC8270648 DOI: 10.1063/5.0040764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
The debilitating effects of muscle damage, either through ischemic injury or volumetric muscle loss (VML), can have significant impacts on patients, and yet there are few effective treatments. This challenge arises when function is degraded due to significant amounts of skeletal muscle loss, beyond the regenerative ability of endogenous repair mechanisms. Currently available surgical interventions for VML are quite invasive and cannot typically restore function adequately. In response to this, many new bioengineering studies implicate 3D bioprinting as a viable option. Bioprinting for VML repair includes three distinct phases: printing and seeding, growth and maturation, and implantation and application. Although this 3D bioprinting technology has existed for several decades, the advent of more advanced and novel printing techniques has brought us closer to clinical applications. Recent studies have overcome previous limitations in diffusion distance with novel microchannel construct architectures and improved myotubule alignment with highly biomimetic nanostructures. These structures may also enhance angiogenic and nervous ingrowth post-implantation, though further research to improve these parameters has been limited. Inclusion of neural cells has also shown to improve myoblast maturation and development of neuromuscular junctions, bringing us one step closer to functional, implantable skeletal muscle constructs. Given the current state of skeletal muscle 3D bioprinting, the most pressing future avenues of research include furthering our understanding of the physical and biochemical mechanisms of myotube development and expanding our control over macroscopic and microscopic construct structures. Further to this, current investigation needs to be expanded from immunocompromised rodent and murine myoblast models to more clinically applicable human cell lines as we move closer to viable therapeutic implementation.
Collapse
Affiliation(s)
- Cooper Blake
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Oliver Massey
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | | | | | | | - Stephanie Franks
- Laboratory of Advanced Biomaterials, The Australian National University, Canberra, ACT 2601, Australia
| | | | | | | | | |
Collapse
|
37
|
Zhang B, Su Y, Zhou J, Zheng Y, Zhu D. Toward a Better Regeneration through Implant-Mediated Immunomodulation: Harnessing the Immune Responses. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100446. [PMID: 34117732 PMCID: PMC8373114 DOI: 10.1002/advs.202100446] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/08/2021] [Indexed: 05/06/2023]
Abstract
Tissue repair/regeneration, after implantation or injury, involves comprehensive physiological processes wherein immune responses play a crucial role to enable tissue restoration, amidst the immune cells early-stage response to tissue damages. These cells break down extracellular matrix, clear debris, and secret cytokines to orchestrate regeneration. However, the immune response can also lead to abnormal tissue healing or scar formation if not well directed. This review first introduces the general immune response post injury, with focus on the major immune cells including neutrophils, macrophages, and T cells. Next, a variety of implant-mediated immunomodulation strategies to regulate immune response through physical, chemical, and biological cues are discussed. At last, various scaffold-facilitated regenerations of different tissue types, such as, bone, cartilage, blood vessel, and nerve system, by harnessing the immunomodulation are presented. Therefore, the most recent data in biomaterials and immunomodulation is presented here in a bid to shape expert perspectives, inspire researchers to go in new directions, and drive development of future strategies focusing on targeted, sequential, and dynamic immunomodulation elicited by implants.
Collapse
Affiliation(s)
- Ben Zhang
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yingchao Su
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Juncen Zhou
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| | - Yufeng Zheng
- Department of Materials Science and EngineeringCollege of EngineeringPeking UniversityBeijing100871China
| | - Donghui Zhu
- Department of Biomedical EngineeringStony Brook UniversityStony BrookNew York11794USA
| |
Collapse
|
38
|
Abstract
The blood-brain barrier (BBB) is one of the most selective endothelial barriers. An understanding of its cellular, morphological, and biological properties in health and disease is necessary to develop therapeutics that can be transported from blood to brain. In vivo models have provided some insight into these features and transport mechanisms adopted at the brain, yet they have failed as a robust platform for the translation of results into clinical outcomes. In this article, we provide a general overview of major BBB features and describe various models that have been designed to replicate this barrier and neurological pathologies linked with the BBB. We propose several key parameters and design characteristics that can be employed to engineer physiologically relevant models of the blood-brain interface and highlight the need for a consensus in the measurement of fundamental properties of this barrier.
Collapse
Affiliation(s)
- Cynthia Hajal
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Baptiste Le Roi
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ben M Maoz
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
39
|
Effects of Macro-/Micro-Channels on Vascularization and Immune Response of Tissue Engineering Scaffolds. Cells 2021; 10:cells10061514. [PMID: 34208449 PMCID: PMC8235743 DOI: 10.3390/cells10061514] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 01/07/2023] Open
Abstract
Although the use of porous scaffolds in tissue engineering has been relatively successful, there are still many limitations that need to be addressed, such as low vascularization, low oxygen and nutrient levels, and immune-induced inflammation. As a result, the current porous scaffolds are insufficient when treating large defects. This paper analyzed scientific research pertaining to the effects of macro-/micro-channels on the cell recruitment, vascularization, and immune response of tissue engineering scaffolds. Most of the studies contained either cell culturing experimentation or experimentation on small animals such as rats and mice. The sacrificial template method, template casting method, and 3D printing method were the most common methods in the fabrication of channeled scaffolds. Some studies combine the sacrificial and 3D printing methods to design and create their scaffold with channels. The overall results from these studies showed that the incorporation of channels within scaffolds greatly increased vascularization, reduced immune response, and was much more beneficial for cell and growth factor recruitment compared with control groups that contained no channels. More research on the effect of micro-/macro-channels on vascularization or immune response in animal models is necessary in the future in order to achieve clinical translation.
Collapse
|
40
|
Xing Z, Zhao C, Wu S, Zhang C, Liu H, Fan Y. Hydrogel-based therapeutic angiogenesis: An alternative treatment strategy for critical limb ischemia. Biomaterials 2021; 274:120872. [PMID: 33991951 DOI: 10.1016/j.biomaterials.2021.120872] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 04/24/2021] [Accepted: 05/02/2021] [Indexed: 02/08/2023]
Abstract
Critical limb ischemia (CLI) is the most severe clinical manifestation of peripheral arterial disease (PAD), resulting in the total or partial loss of limb function. Although the conventional treatment strategy of CLI (e.g., medical treatment and surgery) can improve blood perfusion and restore limb function, many patients are unsuitable for these strategies and they still face the threats of amputation or death. Therapeutic angiogenesis, as a potential solution for these problems, attempts to manipulate blood vessel growth in vivo for augment perfusion without the help of extra pharmaceutics and surgery. With the rise of interdisciplinary research, regenerative medicine strategies provide new possibilities for treating many clinical diseases. Hydrogel, as an excellent biocompatibility material, is an ideal candidate for delivering bioactive molecules and cells for therapeutic angiogenesis. Besides, hydrogel could precisely deliver, control release, and keep the bioactivity of cargos, making hydrogel-based therapeutic angiogenesis a new strategy for CLI therapy. In this review, we comprehensively discuss the approaches of hydrogel-based strategy for CLI treatment as well as their challenges, and future directions.
Collapse
Affiliation(s)
- Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, PR China
| | - Chen Zhao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, PR China
| | - Siwen Wu
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Chunchen Zhang
- Key Laboratory for Biomedical Engineering of Education Ministry of China, Zhejiang University, Hangzhou, 310027, PR China; Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, 310027, PR China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, PR China.
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, PR China.
| |
Collapse
|
41
|
Yoon HJ, Lee YJ, Baek S, Chung YS, Kim DH, Lee JH, Shin YC, Shin YM, Ryu C, Kim HS, Ahn SH, Kim H, Won YB, Lee I, Jeon MJ, Cho SH, Lee BS, Sung HJ, Choi YS. Hormone autocrination by vascularized hydrogel delivery of ovary spheroids to rescue ovarian dysfunctions. SCIENCE ADVANCES 2021; 7:7/18/eabe8873. [PMID: 33910892 PMCID: PMC8081364 DOI: 10.1126/sciadv.abe8873] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/18/2021] [Indexed: 05/25/2023]
Abstract
The regeneration potential of implantable organ model hydrogels is applied to treat a loss of ovarian endocrine function in women experiencing menopause and/or cancer therapy. A rat ovariectomy model is used to harvest autologous ovary cells while subsequently producing a layer-by-layer form of follicle spheroids. Implantation of a microchannel network hydrogel with cell spheroids [vascularized hydrogel with ovarian spheroids (VHOS)] into an ischemic hindlimb of ovariectomized rats significantly aids the recovery of endocrine function with hormone release, leading to full endometrium regeneration. The VHOS implantation effectively suppresses the side effects observed with synthetic hormone treatment (i.e., tissue overgrowth, hyperplasia, cancer progression, deep vein thrombosis) to the normal levels, while effectively preventing the representative aftereffects of menopause (i.e., gaining fatty weight, inducing osteoporosis). These results highlight the unprecedented therapeutic potential of an implantable VHOS against menopause and suggest that it may be used as an alternative approach to standard hormone therapy.
Collapse
Affiliation(s)
- Hyo-Jin Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yong Jae Lee
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sewoom Baek
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young Shin Chung
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dae-Hyun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jae Hoon Lee
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Yong Cheol Shin
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chungsoon Ryu
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - So Hyun Ahn
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heeyon Kim
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young Bin Won
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Inha Lee
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Myung Jae Jeon
- Department of Obstetrics and Gynecology, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Si Hyun Cho
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea
| | - Byung Seok Lee
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Young Sik Choi
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
- Department of Obstetrics and Gynecology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
42
|
Barrett-Catton E, Ross ML, Asuri P. Multifunctional Hydrogel Nanocomposites for Biomedical Applications. Polymers (Basel) 2021; 13:856. [PMID: 33799539 PMCID: PMC8001467 DOI: 10.3390/polym13060856] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/05/2021] [Accepted: 03/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hydrogels are used for various biomedical applications due to their biocompatibility, capacity to mimic the extracellular matrix, and ability to encapsulate and deliver cells and therapeutics. However, traditional hydrogels have a few shortcomings, especially regarding their physical properties, thereby limiting their broad applicability. Recently, researchers have investigated the incorporation of nanoparticles (NPs) into hydrogels to improve and add to the physical and biochemical properties of hydrogels. This brief review focuses on papers that describe the use of nanoparticles to improve more than one property of hydrogels. Such multifunctional hydrogel nanocomposites have enhanced potential for various applications including tissue engineering, drug delivery, wound healing, bioprinting, and biowearable devices.
Collapse
Affiliation(s)
| | | | - Prashanth Asuri
- Department of Bioengineering, Santa Clara University, Santa Clara, CA 95053, USA; (E.B.-C.); (M.L.R.)
| |
Collapse
|
43
|
Li C, Kitzerow O, Nie F, Dai J, Liu X, Carlson MA, Casale GP, Pipinos II, Li X. Bioengineering strategies for the treatment of peripheral arterial disease. Bioact Mater 2021; 6:684-696. [PMID: 33005831 PMCID: PMC7511653 DOI: 10.1016/j.bioactmat.2020.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/12/2020] [Accepted: 09/12/2020] [Indexed: 12/21/2022] Open
Abstract
Peripheral arterial disease (PAD) is a progressive atherosclerotic disorder characterized by narrowing and occlusion of arteries supplying the lower extremities. Approximately 200 million people worldwide are affected by PAD. The current standard of operative care is open or endovascular revascularization in which blood flow restoration is the goal. However, many patients are not appropriate candidates for these treatments and are subject to continuous ischemia of their lower limbs. Current research in the therapy of PAD involves developing modalities that induce angiogenesis, but the results of simple cell transplantation or growth factor delivery have been found to be relatively poor mainly due to difficulties in stem cell retention and survival and rapid diffusion and enzymolysis of growth factors following injection of these agents in the affected tissues. Biomaterials, including hydrogels, have the capability to protect stem cells during injection and to support cell survival. Hydrogels can also provide a sustained release of growth factors at the injection site. This review will focus on biomaterial systems currently being investigated as carriers for cell and growth factor delivery, and will also discuss biomaterials as a potential stand-alone method for the treatment of PAD. Finally, the challenges of development and use of biomaterials systems for PAD treatment will be reviewed.
Collapse
Affiliation(s)
- Cui Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Oliver Kitzerow
- Department of Genetics Cell Biology and Anatomy, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Fujiao Nie
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Jingxuan Dai
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaoyan Liu
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Mark A. Carlson
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, 68198, United States
- Omaha VA Medical Center, Omaha, NE, 68105, United States
| | - George P. Casale
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Iraklis I. Pipinos
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| | - Xiaowei Li
- Mary & Dick Holland Regenerative Medicine Program and Department of Neurological Sciences, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, United States
| |
Collapse
|
44
|
Li C, Kuss M, Kong Y, Nie F, Liu X, Liu B, Dunaevsky A, Fayad P, Duan B, Li X. 3D Printed Hydrogels with Aligned Microchannels to Guide Neural Stem Cell Migration. ACS Biomater Sci Eng 2021; 7:690-700. [PMID: 33507749 DOI: 10.1021/acsbiomaterials.0c01619] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Following traumatic or ischemic brain injury, rapid cell death and extracellular matrix degradation lead to the formation of a cavity at the brain lesion site, which is responsible for prolonged neurological deficits and permanent disability. Transplantation of neural stem/progenitor cells (NSCs) represents a promising strategy for reconstructing the lesion cavity and promoting tissue regeneration. In particular, the promotion of neuronal migration, organization, and integration of transplanted NSCs is critical to the success of stem cell-based therapy. This is particularly important for the cerebral cortex, the most common area involved in brain injuries, because the highly organized structure of the cerebral cortex is essential to its function. Biomaterials-based strategies show some promise for conditioning the lesion site microenvironment to support transplanted stem cells, but the progress in demonstrating organized cell engraftment and integration into the brain is very limited. An effective approach to sufficiently address these challenges has not yet been developed. Here, we have implemented a digital light-processing-based 3D printer and printed hydrogel scaffolds with a designed shape, uniaxially aligned microchannels, and tunable mechanical properties. We demonstrated the capacity to achieve high shape precision to the lesion site with brain tissue-matching mechanical properties. We also established spatial control of bioactive molecule distribution within 3D printed hydrogel scaffolds. These printed hydrogel scaffolds have shown high neuro-compatibility with aligned neuronal outgrowth along with the microchannels. This study will provide a biomaterial-based approach that can serve as a protective and guidance vehicle for transplanted NSC organization and integration for brain tissue regeneration after injuries.
Collapse
Affiliation(s)
- Cui Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.,Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Fujiao Nie
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Xiaoyan Liu
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bo Liu
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Anna Dunaevsky
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Pierre Fayad
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Xiaowei Li
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
45
|
Zhang C, Yan Q, Li J, Zhu Y, Zhang Y. Nanoenabled Tumor Oxygenation Strategies for Overcoming Hypoxia-Associated Immunosuppression. ACS APPLIED BIO MATERIALS 2021; 4:277-294. [PMID: 35014284 DOI: 10.1021/acsabm.0c01328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy, which initiates or strengthens innate immune responses to attack cancer cells, has shown great promise in cancer treatment. However, low immune response impacted by immunosuppressive tumor microenvironment (TME) remains a key challenge, which has been found related to tumor hypoxia. Recently, nanomaterial systems are proving to be excellent platforms for tumor oxygenation, which can reverse hypoxia-associated immunosuppression, strengthen the systemic antitumor immune responses, and thus afford a striking abscopal effect to clear metastatic cancer cells. In this review, we would like to survey recent progress in utilizing nanomaterials for tumor oxygenation through approaches such as in situ O2 generation, O2 delivery, tumor vasculature normalization, and mitochondrial-respiration inhibition. Their effects on tumor hypoxia-associated immunosuppression are highlighted. We also discuss the ongoing challenges and how to further improve the clinical prospect of cancer immunotherapy.
Collapse
Affiliation(s)
- Chao Zhang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acids Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qinglong Yan
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Jiang Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Ying Zhu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.,Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| | - Yu Zhang
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, The Interdisciplinary Research Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.,Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
| |
Collapse
|
46
|
Ichanti H, Sladic S, Kalies S, Haverich A, Andrée B, Hilfiker A. Characterization of Tissue Engineered Endothelial Cell Networks in Composite Collagen-Agarose Hydrogels. Gels 2020; 6:gels6030027. [PMID: 32899293 PMCID: PMC7559300 DOI: 10.3390/gels6030027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Scaffolds constitute an important element in vascularized tissues and are therefore investigated for providing the desired mechanical stability and enabling vasculogenesis and angiogenesis. In this study, supplementation of hydrogels containing either MatrigelTM and rat tail collagen I (MatrigelTM/rCOL) or human collagen (hCOL) with SeaPlaqueTM agarose were analyzed with regard to construct thickness and formation and characteristics of endothelial cell (EC) networks compared to constructs without agarose. Additionally, the effect of increased rCOL content in MatrigelTM/rCOL constructs was studied. An increase of rCOL content from 1 mg/mL to 3 mg/mL resulted in an increase of construct thickness by approximately 160%. The high rCOL content, however, impaired the formation of an EC network. The supplementation of MatrigelTM/rCOL with agarose increased the thickness of the hydrogel construct by approximately 100% while supporting the formation of a stable EC network. The use of hCOL/agarose composite hydrogels led to a slight increase in the thickness of the 3D hydrogel construct and supported the formation of a multi-layered EC network compared to control constructs. Our findings suggest that agarose/collagen-based composite hydrogels are promising candidates for tissue engineering of vascularized constructs as cell viability is maintained and the formation of a stable and multi-layered EC network is supported.
Collapse
Affiliation(s)
- Houda Ichanti
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Sanja Sladic
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Stefan Kalies
- Institute of Quantum Optics, Leibniz University Hannover, 30167 Hannover, Germany;
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development, 30625 Hannover, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
| | - Birgit Andrée
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
- Correspondence: (B.A.); (A.H.); Tel.: +49-511-532-8913 (B.A.); +49-511-532-8998 (A.H.)
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, 30625 Hannover, Germany; (H.I.); (S.S.); (A.H.)
- Correspondence: (B.A.); (A.H.); Tel.: +49-511-532-8913 (B.A.); +49-511-532-8998 (A.H.)
| |
Collapse
|