1
|
March A, Hebner TS, Choe R, Benoit DSW. Leveraging the predictive power of a 3D in vitro vascularization screening assay for hydrogel-based tissue-engineered periosteum allograft healing. BIOMATERIALS ADVANCES 2025; 169:214187. [PMID: 39827700 PMCID: PMC11815559 DOI: 10.1016/j.bioadv.2025.214187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
A common strategy for promoting bone allograft healing is the design of tissue-engineered periosteum (TEP) to orchestrate host-tissue infiltration. However, evaluating requires costly and time-consuming in vivo studies. Therefore, in vitro assays are necessary to expedite TEP designs. Since angiogenesis is a critical process orchestrated by the periosteum, this study investigates in vitro 3D cell spheroid vascularization as a predictive tool for TEP-mediated in vivo healing. Spheroids of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) are encapsulated in enzymatically-degradable poly (ethylene glycol)-based hydrogels and sprout formation, network formation, and angiogenic growth factor secretion are quantified. Hydrogels are also evaluated as TEP-modified allografts for in vivo bone healing with graft vascularization, callus formation, and biomechanical strength quantified as healing metrics. Evaluation of hydrogels highlights the importance of degradation, with 24-fold greater day 1 sprouts observed in degradable hydrogels in vitro and 4-fold greater graft-localized vascular volume at 6-weeks in vivo compared to non-degradable hydrogels. Correlations between in vitro and in vivo studies elucidate linear relationships when comparing in vitro sprout formation and angiocrine production with 3- and 6-week in vivo graft vascularization, 3-week cartilage callus, and 6-week bone callus, with a Pearson's R2 value equal to 0.97 for the linear correlation between in vitro sprout formation and 6-week in vivo vascular volume. Non-linear relationships are found between in vitro measures and bone torque strength at week 6. These correlations suggest that the in vitro sprouting assay has predictive power for in vivo vascularization and bone allograft healing.
Collapse
Affiliation(s)
- Alyson March
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA
| | - Tayler S Hebner
- Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA; Davidson School of Chemical Engineering, Purdue University, 480 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Regine Choe
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, Center for Musculoskeletal Research, University of Rochester, 204 Robert B. Goergen Hall, Rochester, NY 14627, USA; Department of Bioengineering, Knight Campus for Accelerating Scientific Impact, University of Oregon, 6231 University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
2
|
Gonzalez‐Rubio J, Kubiza H, Xu Y, Koenigs‐Werner H, Schmitz MS, Schedel M, Apel C, Jockenhoevel S, Cornelissen CG, Thiebes AL. Pericytes Promote More Vascularization than Stromal Cells via an Interleukin-6-Dependent Mechanism in Microfluidic Chips. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408131. [PMID: 39887579 PMCID: PMC11984840 DOI: 10.1002/advs.202408131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Pericytes are a key player in vascularization, protecting endothelial cells from external harm and promoting the formation of new vessels when necessary. However, pericytic identity and its relationship with other cell types, such as mesenchymal stromal/stem cells, is highly debated. This study compares the role of pericytes and unselected stromal cells in vascularization using multichannel microfluidic chips. In both angiogenesis and vasculogenesis, pericytes promote more vessel formation than stromal cells. Pericytes can wrap around endothelial vessels acting as mural cells, while stromal cells remain separated. Whole-transcriptome sequencing confirms an upregulation of pro-vascularization genes in endothelial cell-pericyte co-cultures, while metabolism increases and inflammation decreases in stromal cell co-cultures. Treatment of stromal-endothelial cell co-cultures with either conditioned media or isolated extracellular vesicles from pericytes replicates the increase in vasculogenesis of the direct co-cultures. Cytokine quantification reveals that interleukin 6 (IL-6) is significantly increased in pericyte conditions. Blocking it with siltuximab results in a reduction of pericyte vasculogenic potential comparable to stromal cell levels, revealing that pericyte pro-vascularization is mediated by IL-6. This study provides new insights into the relationship between pericytes and endothelial cells and the elusive identity of mesenchymal stromal cells. These findings are relevant for both vascular biology and tissue engineering.
Collapse
Affiliation(s)
- Julian Gonzalez‐Rubio
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Hannah Kubiza
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Yong Xu
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Hiltrud Koenigs‐Werner
- Institute of PathologyElectron Microscopy FacilityRWTH Aachen University HospitalAachen, Pauwelsstrasse 3052074AachenGermany
| | - Mona Sophie Schmitz
- Department of Pulmonary MedicineUniversity Medicine Essen‐RuhrlandklinikTueschener Weg 4045239EssenGermany
- Department of Pulmonary MedicineUniversity Medicine EssenEssen, Hufelandstraße 5545147EssenGermany
| | - Michaela Schedel
- Department of Pulmonary MedicineUniversity Medicine Essen‐RuhrlandklinikTueschener Weg 4045239EssenGermany
- Department of Pulmonary MedicineUniversity Medicine EssenEssen, Hufelandstraße 5545147EssenGermany
| | - Christian Apel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Christian G. Cornelissen
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V)RWTH Aachen University HospitalPauwelsstrasse 3052074AachenGermany
| | - Anja Lena Thiebes
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| |
Collapse
|
3
|
Lin YY, Warren E, Macklin BL, Ramirez L, Gerecht S. Endothelial-pericyte interactions regulate angiogenesis via VEGFR2 signaling during retinal development and disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642174. [PMID: 40161680 PMCID: PMC11952325 DOI: 10.1101/2025.03.08.642174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Pericytes stabilize the microvasculature by enhancing endothelial barrier integrity, resulting in functional networks. During retinal development, pericyte recruitment is crucial for stabilizing nascent angiogenic vasculature. However, in adulthood, disrupted endothelial-pericyte interactions lead to vascular dropout and pathological angiogenesis in ocular microvascular diseases, and strategies to stabilize the retinal vasculature are lacking. We demonstrate that direct endothelial-pericyte contact downregulates pVEGFR2 in endothelial cells, which enhances pericyte migration and promotes endothelial cell barrier function. Intravitreal injection of a VEGFR2 inhibitor in mouse models of the developing retina and oxygen-induced retinopathy increased pericyte recruitment and aided vascular stability. The VEGFR2 inhibitor further rescued ischemic retinopathy by enhancing vascularization and tissue growth while reducing vascular permeability. Our findings offer a druggable target to support the growth of functional and mature microvasculature in ocular microvascular diseases and tissue regeneration overall.
Collapse
|
4
|
Zhou R, Brislinger D, Fuchs J, Lyons A, Langthaler S, Hauser CAE, Baumgartner C. Vascularised organoids: Recent advances and applications in cancer research. Clin Transl Med 2025; 15:e70258. [PMID: 40045486 PMCID: PMC11882480 DOI: 10.1002/ctm2.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Organoids are three-dimensional (3D) cellular models designed to replicate human tissues and organs while preserving their physiological complexity and functionality. Among these, vascularised organoids represent a groundbreaking advancement in 3D tissue engineering, incorporating vascular networks into engineered tissues to more accurately mimic the in vivo tumour microenvironment. These models offer significantly improved physiological relevance compared to conventional two-dimensional cultures or animal models, positioning them as invaluable tools in cancer research. Despite their potential, the rapid proliferation of techniques and materials for developing vascularised organoids presents challenges for researchers navigating this dynamic field. This systematic review provides a comprehensive examination of methodologies for fabricating vascularised organoids, with a focus on strategies that enhance vascularisation and support organoid growth. It critically evaluates the materials used, emphasising those that effectively mimic the extracellular matrix and facilitate vascular network formation. Key advancements in engineered organoids models are highlighted, emphasising their potential for studying interactions between vasculature and cancer cells, conducting drug screening, and understanding cytokine regulation. In summary, this review provides an in-depth overview of the current landscape of vascularised organoid fabrication and functionality, addressing challenges and opportunities within the field. A detailed understanding of the scope and future trajectories is essential for advancing organoid development and expanding their applications in both basic cancer research and clinical practice. KEY POINTS: Comparative analysis: Evaluation of organoids, animal models, and 2D models, highlighting their respective strengths and limitations in replicating physiological conditions and studying disease processes. Vascularisation techniques: Comparative evaluation of vascularised organoid fabrication methods, emphasising their efficiency, scalability and ability to replicate physiological vascular networks. Material selection: Thorough evaluation of materials for vascularised organoid culture system, focusing on those that effectively mimic the extracellular matrix and support vascular network formation. Applications: Overview of organoid applications in basic cancer research and clinical settings, with an emphasis on their potential in drug discovery, disease modelling and exploring complex biological processes.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Dagmar Brislinger
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Alicia Lyons
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Sonja Langthaler
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Charlotte A. E. Hauser
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| |
Collapse
|
5
|
Gomes KT, Prasad PR, Sandhu JS, Kumar A, Kumar NAN, Shridhar NB, Bisht B, Paul MK. Decellularization techniques: unveiling the blueprint for tracheal tissue engineering. Front Bioeng Biotechnol 2025; 13:1518905. [PMID: 40092377 PMCID: PMC11906413 DOI: 10.3389/fbioe.2025.1518905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Certain congenital or acquired diseases and defects such as tracheo-oesophageal fistula, tracheomalacia, tracheal stenosis, airway ischemia, infections, and tumours can cause damage to the trachea. Treatments available do not offer any permanent solutions. Moreover, long-segment defects in the trachea have no available surgical treatments. Tissue engineering has gained popularity in current regenerative medicine as a promising approach to bridge this gap. Among the various tissue engineering techniques, decellularization is a widely used approach that removes the cellular and nuclear contents from the tissue while preserving the native extracellular matrix components. The decellularized scaffolds exhibit significantly lower immunogenicity and retain the essential biomechanical and proangiogenic properties of native tissue, creating a foundation for trachea regeneration. The present review provides an overview of trachea decellularization advancements, exploring how recellularization approaches can be optimized by using various stem cells and tissue-specific cells to restore the scaffold's structure and function. We examine critical factors such as mechanical properties, revascularization, and immunogenicity involved in the transplantation of tissue-engineered grafts.
Collapse
Affiliation(s)
- Keisha T Gomes
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Palla Ranga Prasad
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Jagnoor Singh Sandhu
- Central Animal Research Facility, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, India
- Center for Animal Research, Ethics and Training (CARET), Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Ashwini Kumar
- Department of Forensic Medicine, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Naveena A N Kumar
- Department of Surgical Oncology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - N B Shridhar
- Department of Pharmacology and Toxicology, Obscure Disease Research Center, Veterinary College Campus, Shivamogga, Karnataka, India
| | - Bharti Bisht
- Department of Microbiology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States
| |
Collapse
|
6
|
Jun I, Choi H, Kim H, Chan Choi B, Chang HJ, Kim Y, Cho SW, Edwards JR, Hwang SW, Kim YC, Han HS, Jeon H. Exploring the potential of laser-textured metal alloys: Fine-tuning vascular cells responses through in vitro and ex vivo analysis. Bioact Mater 2025; 43:181-194. [PMID: 39386224 PMCID: PMC11462155 DOI: 10.1016/j.bioactmat.2024.09.019] [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: 03/14/2024] [Revised: 09/04/2024] [Accepted: 09/15/2024] [Indexed: 10/12/2024] Open
Abstract
Medical stents are vital for treating vascular complications and restoring blood flow in millions of patients. Despite its widespread effectiveness, restenosis, driven by the complex interplay of cellular responses, remains a concern. This study investigated the reactions of vascular cells to nano/microscale wrinkle (nano-W and micro-W) patterns created on laser-textured nitinol (NiTi) surfaces by adjusting laser processing parameters, such as spot overlap ratio and line overlap ratio. Evaluation of topographical effects on endothelial and smooth muscle cells (SMCs) revealed diverse morphologies, proliferation rates, and gene expressions. Notably, microscale wrinkle patterns exhibited reduced monocyte adhesion and inflammation-related gene expression, demonstrating their potential applications in mitigating vascular complications after stent insertion. Additionally, an ex vivo metatarsal assay was utilized to bridge the gap between in vitro and in vivo studies, demonstrating enhanced angiogenesis on laser-textured NiTi surfaces. Laser-textured NiTi exhibits a guided formation process, emphasizing their potential to promote swift endothelialization. These findings underscore the efficacy of laser texturing for tailored cellular interactions on metallic surfaces and offer valuable insights into optimizing biocompatibility and controlling cellular responses, which may pave the way for innovative advances in vascular care and contribute to the ongoing improvement of stent insertion.
Collapse
Affiliation(s)
- Indong Jun
- Environmental Safety Group, Korea Institute of Science & Technology Europe (KIST-EUROPE), Saarbrücken, 66123, Germany
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Haneul Choi
- Center for Hydrogen Energy Materials, Korea Institute of Science & Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyeok Kim
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Byoung Chan Choi
- Laser Surface Texturing Group, AYECLUS, Gyeonggi-do, 14255, Republic of Korea
| | - Hye Jung Chang
- Center for Hydrogen Energy Materials, Korea Institute of Science & Technology (KIST), Seoul, 02792, Republic of Korea
| | - Youngjun Kim
- Environmental Safety Group, Korea Institute of Science & Technology Europe (KIST-EUROPE), Saarbrücken, 66123, Germany
| | - Sung Woo Cho
- Division of Cardiology, Department of Internal Medicine, Inje University Ilsan Paik Hospital, College of Medicine, Inje University, Gyeonggi-do, 10380, Republic of Korea
| | - James R. Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, OX3 7LD, United Kingdom
| | - Suk-Won Hwang
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Integrative Energy Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yu-Chan Kim
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyung-Seop Han
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hojeong Jeon
- Biomaterials Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
7
|
Kong AM, Liu GS, Mitchell GM, Lim SY. Methods for Assessing Scaffold Vascularization with Human Endothelial Cells. Methods Mol Biol 2025; 2922:241-248. [PMID: 40208540 DOI: 10.1007/978-1-0716-4510-9_18] [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] [Indexed: 04/11/2025]
Abstract
The success of tissue engineering relies heavily on the efficient and rapid formation of blood vessels in the newly engineered tissues. To achieve this, it is crucial to use scaffolds that not only support the survival of vascular cells but also facilitate the development and maturation of a capillary network in vivo. Utilizing advanced biomaterials and scaffold designs is essential to ensure that the engineered tissues can integrate effectively with the host tissues and maintain their functionality over time. Here, we present a method for generating endothelial cells from induced pluripotent stem cells and pre-vascularizing scaffolds by seeding them with endothelial cells in a 3D porous structure. These pre-vascularized scaffolds can then be implanted in a rat subcutaneous model to create vascularized tissue constructs. This approach holds great promise for generating clinically viable constructs that can be used in regenerative medicine and drug development.
Collapse
Affiliation(s)
- Anne M Kong
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Guei-Sheung Liu
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia
- Center for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Geraldine M Mitchell
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Surgery and Medicine, University of Melbourne, East Melbourne, VIC, Australia
- Faculty of Health Sciences, Australian Catholic University, Melbourne, VIC, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, VIC, Australia.
- Department of Surgery and Medicine, University of Melbourne, East Melbourne, VIC, Australia.
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
- National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore.
| |
Collapse
|
8
|
Liang W, Zhou C, Liu X, Xie Q, Xia L, Liu L, Bao W, Lin H, Xiong X, Zhang H, Zheng Z, Zhao J. Current status of nano-embedded growth factors and stem cells delivery to bone for targeted repair and regeneration. J Orthop Translat 2025; 50:257-273. [PMID: 39902262 PMCID: PMC11788687 DOI: 10.1016/j.jot.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/01/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Bone-related diseases like osteoarthritis and osteoporosis impact millions globally, affecting quality of life. Osteoporosis considerably enhances the probability of bone fractures of the wrist, hip, and spine. Enhancement and acceleration of functional bone development can be achieved through the sustained delivery of growth factors (GFs) and cells in biomaterial carriers. The delivery of bioactive compounds in a targeted, spatiotemporal way that most closely resembles the natural defect repair process can be achieved by designing the carrier system with established release kinetics. Furthermore, the carrier can serve as a substrate that mimics the extracellular matrix, facilitating osteoprogenitor cell infiltration and growth for integrative tissue healing. In this report, we explore the significance of GFs within the realm of bone and cartilage tissue engineering, encompassing their encapsulation and delivery methodologies, the kinetics of release, and their amalgamation with biomaterials and stem cells (SCs) to facilitate the mending of bone fractures. Moreover, the significance of GFs in evaluating the microenvironment of bone tissue through reciprocal signaling with cells and biomaterial scaffolds is emphasized which will serve as the foundation for prospective advances in bone and cartilage tissue engineering as well as therapeutic equipment. Nanoparticles are being used in regenerative medicine to promote bone regeneration and repair by delivering osteoinductive growth factors like BMP-2, VEGF, TGF-β. These nanocarriers allow controlled release, minimizing adverse effects and ensuring growth factors are concentrated at the injury site. They are also mixed with mesenchymal stem cells (MSCs) to improve their engraftment, differentiation, and survival. This approach is a key step in developing multi-model systems that more efficiently facilitate bone regeneration. Researchers are exploring smart nanoparticles with immunomodulatory qualities to improve bonre regeneration and reduce inflammation in injury site. Despite promising preclinical results, challenges include cost management, regulatory approval, and long term safety. However, incorporating stem cell transport and growth factors in nanoparticles could revolutionize bone regeneration and offer more personalized therapies for complex bone disorders and accidents. The translational potential of this article Stem cell transport and growth factors encapsulated in nanoparticles are becoming revolutionary methods for bone regeneration and repair. By encouraging stem cells to develop into osteoblasts, osteoinductive GFs like BMP-2, VEGF, and TGF-β can be delivered under control due to nanomaterials like nanoparticles, nanofibers, and nanotubes. By ensuring sustained release, these nanocarriers lessen adverse effects and enhance therapeutic results. In order to prove their survival and development, MCSs, which are essential for bone regeneration, are mixed with nanoparticles, frequently using scaffolds that resemble the ECM of bone. Furthermore, by adjusting to the injured environment and lowering inflammation, immunomodulatory nanostructures and stimuli-responsive nanomaterials can further maximize. While there are still shotcomings to overcome, including managing expenses, negotiating regulatory processes, and guaranteeing long-term safety, this method promises to outperform traditional bone grafting by providing quicker, more individualized, and more efficient treatments. Nano-embedded growth factors and stem cell technologies have the potential to revolutionize orthopedic therapy and significantly enhance patient outcomes with further research.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Chao Zhou
- Department of Orthopedics, Zhoushan Guanghua Hospital, Zhoushan, 316000, China
| | - Xiankun Liu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Qiong Xie
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Linying Xia
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Lu Liu
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Wenwen Bao
- Medical Research Center, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hongming Lin
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Xiaochun Xiong
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Hao Zhang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Zeping Zheng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan, 316000, China
| |
Collapse
|
9
|
Li W, Li J, Pan C, Lee JS, Kim BS, Gao G. Light-based 3D bioprinting techniques for illuminating the advances of vascular tissue engineering. Mater Today Bio 2024; 29:101286. [PMID: 39435375 PMCID: PMC11492625 DOI: 10.1016/j.mtbio.2024.101286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/21/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Vascular tissue engineering faces significant challenges in creating in vitro vascular disease models, implantable vascular grafts, and vascularized tissue/organ constructs due to limitations in manufacturing precision, structural complexity, replicating the composited architecture, and mimicking the mechanical properties of natural vessels. Light-based 3D bioprinting, leveraging the unique advantages of light including high resolution, rapid curing, multi-material adaptability, and tunable photochemistry, offers transformative solutions to these obstacles. With the emergence of diverse light-based 3D bioprinting techniques and innovative strategies, the advances in vascular tissue engineering have been significantly accelerated. This review provides an overview of the human vascular system and its physiological functions, followed by an in-depth discussion of advancements in light-based 3D bioprinting, including light-dominated and light-assisted techniques. We explore the application of these technologies in vascular tissue engineering for creating in vitro vascular disease models recapitulating key pathological features, implantable blood vessel grafts, and tissue analogs with the integration of capillary-like vasculatures. Finally, we provide readers with insights into the future perspectives of light-based 3D bioprinting to revolutionize vascular tissue engineering.
Collapse
Affiliation(s)
- Wei Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology (BIT), Zhuhai 519088, China
| | - Chen Pan
- School of Mechanical Engineering, Beijing Institute of Technology, Beijing 100081, China
- School of Mechanical and Equipment Engineering, Hebei University of Engineering, Handan, 050024, China
| | - Jae-Seong Lee
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
- Department of Information Convergence Engineering, Pusan National University, Busan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
10
|
Kim B, Swain JWR, Fowler MJ, Yang CY, Vohidona D, Hartgerink JD, Veiseh O. Rapid method to screen biomaterial angiogenesis in vivo using fluorescence imaging in mice. Biomater Sci 2024; 12:5824-5833. [PMID: 39412699 DOI: 10.1039/d4bm00626g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2024]
Abstract
Effective vascularization is crucial for repairing and enhancing the longevity of engineered tissues and organs. As the field advances, there is a vital need for efficient and reliable methods for assessing vascularization in real-time. The integration and performance of constructed biomaterials in living organisms rely on angiogenesis and vascularization, making it essential to evaluate vascular development and networks within biomaterials. Current histology-based methods are limited and labor-intensive. On the other hand, fluorescence imaging offers promise for efficient, real-time evaluation of angiogenesis, reducing the time needed for screening many compounds and offering a high-throughput alternative to histology-based methods. Here, we investigated a novel, non-invasive method for quick and repeated analysis of the angiogenic and vascularization process in biomaterials via fluorescence IVIS imaging. Multi-domain peptides (MDPs), self-assembling peptide hydrogels that can possess pro-angiogenic properties depending on their primary sequence, were synthesized and utilized as angiogenic biomaterials and screened with a fluorescence IVIS probe to demonstrate real-time rapid angiogenesis in vivo. The fluorescence-based imaging showed the influence of the peptide chemistry, volume, and concentration on angiogenesis, with one particular MDP, SLanc, promoting robust angiogenesis after one week at 2 w/v%. Through this method, we were able to identify the optimal peptide for rapid and sustained angiogenesis. This approach enables real-time monitoring of angiogenic responses and vascularization processes in the same living subject. It promotes the development of new biomaterials that facilitate vascularization and validates an advanced in vivo screening technique for angiogenesis.
Collapse
Affiliation(s)
- Boram Kim
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| | - Joseph W R Swain
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Martha J Fowler
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| | - Claire Y Yang
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | | | - Jeffrey D Hartgerink
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
- Department of Chemistry, Rice University, Houston, TX, 77005, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
11
|
Mikaeeli Kangarshahi B, Naghib SM, Rabiee N. 3D printing and computer-aided design techniques for drug delivery scaffolds in tissue engineering. Expert Opin Drug Deliv 2024; 21:1615-1636. [PMID: 39323396 DOI: 10.1080/17425247.2024.2409913] [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: 01/20/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The challenge in tissue engineering lies in replicating the intricate structure of the native extracellular matrix. Recent advancements in AM, notably 3D printing, offer unprecedented capabilities to tailor scaffolds precisely, controlling properties like structure and bioactivity. CAD tools complement this by facilitating design using patient-specific data. AREA’S COVERED This review introduces additive manufacturing (AM) and computer-aided design (CAD) as pivotal tools in advancing tissue engineering, particularly cartilage regeneration. This article explores various materials utilized in AM, focusing on polymers and hydrogels for their advantageous properties in tissue engineering applications. Integrating bioactive molecules, including growth factors, into scaffolds to promote tissue regeneration is discussed alongside strategies involving different cell sources, such as stem cells, to enhance tissue development within scaffold matrices. EXPERT OPINION Applications of AM and CAD in addressing specific challenges like osteochondral defects and osteoarthritis in cartilage tissue engineering are highlighted. This review consolidates current research findings, offering expert insights into the evolving landscape of AM and CAD technologies in advancing tissue engineering, particularly in cartilage regeneration.
Collapse
Affiliation(s)
- Babak Mikaeeli Kangarshahi
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Seyed Morteza Naghib
- Nanotechnology Department, School of Advanced Technologies, Iran University of Science and Technology, Tehran, Iran
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai, India
| |
Collapse
|
12
|
Snyder CA, Dwyer KD, Coulombe KLK. Advancing Human iPSC-Derived Cardiomyocyte Hypoxia Resistance for Cardiac Regenerative Therapies through a Systematic Assessment of In Vitro Conditioning. Int J Mol Sci 2024; 25:9627. [PMID: 39273573 PMCID: PMC11395605 DOI: 10.3390/ijms25179627] [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: 08/07/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Acute myocardial infarction (MI) is a sudden, severe cardiac ischemic event that results in the death of up to one billion cardiomyocytes (CMs) and subsequent decrease in cardiac function. Engineered cardiac tissues (ECTs) are a promising approach to deliver the necessary mass of CMs to remuscularize the heart. However, the hypoxic environment of the heart post-MI presents a critical challenge for CM engraftment. Here, we present a high-throughput, systematic study targeting several physiological features of human induced pluripotent stem cell-derived CMs (hiPSC-CMs), including metabolism, Wnt signaling, substrate, heat shock, apoptosis, and mitochondrial stabilization, to assess their efficacy in promoting ischemia resistance in hiPSC-CMs. The results of 2D experiments identify hypoxia preconditioning (HPC) and metabolic conditioning as having a significant influence on hiPSC-CM function in normoxia and hypoxia. Within 3D engineered cardiac tissues (ECTs), metabolic conditioning with maturation media (MM), featuring high fatty acid and calcium concentration, results in a 1.5-fold increase in active stress generation as compared to RPMI/B27 control ECTs in normoxic conditions. Yet, this functional improvement is lost after hypoxia treatment. Interestingly, HPC can partially rescue the function of MM-treated ECTs after hypoxia. Our systematic and iterative approach provides a strong foundation for assessing and leveraging in vitro culture conditions to enhance the hypoxia resistance, and thus the successful clinical translation, of hiPSC-CMs in cardiac regenerative therapies.
Collapse
Affiliation(s)
- Caroline A Snyder
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kiera D Dwyer
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Kareen L K Coulombe
- Institute for Biology, Engineering and Medicine, School of Engineering, Brown University, Providence, RI 02912, USA
| |
Collapse
|
13
|
Kim J, Ro J, Cho YK. Vascularized platforms for investigating cell communication via extracellular vesicles. BIOMICROFLUIDICS 2024; 18:051504. [PMID: 39323481 PMCID: PMC11421861 DOI: 10.1063/5.0220840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
The vascular network plays an essential role in the maintenance of all organs in the body via the regulated delivery of oxygen and nutrients, as well as tissue communication via the transfer of various biological signaling molecules. It also serves as a route for drug administration and affects pharmacokinetics. Due to this importance, engineers have sought to create physiologically relevant and reproducible vascular systems in tissue, considering cell-cell and extracellular matrix interaction with structural and physical conditions in the microenvironment. Extracellular vesicles (EVs) have recently emerged as important carriers for transferring proteins and genetic material between cells and organs, as well as for drug delivery. Vascularized platforms can be an ideal system for studying interactions between blood vessels and EVs, which are crucial for understanding EV-mediated substance transfer in various biological situations. This review summarizes recent advances in vascularized platforms, standard and microfluidic-based techniques for EV isolation and characterization, and studies of EVs in vascularized platforms. It provides insights into EV-related (patho)physiological regulations and facilitates the development of EV-based therapeutics.
Collapse
|
14
|
Zhang S, Yu M, Li M, He M, Xie L, Huo F, Tian W. Notch Signaling Hydrogels Enable Rapid Vascularization and Promote Dental Pulp Tissue Regeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310285. [PMID: 39013081 PMCID: PMC11425206 DOI: 10.1002/advs.202310285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/07/2024] [Indexed: 07/18/2024]
Abstract
Successful dental pulp regeneration is closely associated with rapid revascularization and angiogenesis, processes driven by the Jagged1(JAG1)/Notch signaling pathway. However, soluble Notch ligands have proven ineffective in activating this pathway. To overcome this limitation, a Notch signaling hydrogel is developed by indirectly immobilizing JAG1, aimed at precisely directing the regeneration of vascularized pulp tissue. This hydrogel displays favorable mechanical properties and biocompatibility. Cultivating dental pulp stem cells (DPSCs) and endothelial cells (ECs) on this hydrogel significantly upregulate Notch target genes and key proangiogenic markers expression. Three-dimensional (3D) culture assays demonstrate Notch signaling hydrogels improve effectiveness by facilitating encapsulated cell differentiation, enhancing their paracrine functions, and promoting capillary lumen formation. Furthermore, it effectively communicates with the Wnt signaling pathway, creating an odontoinductive microenvironment for pulp-dentin complex formation. In vivo studies show that short-term transplantation of the Notch signaling hydrogel accelerates angiogenesis, stabilizes capillary-like structures, and improves cell survival. Long-term transplantation further confirms its capability to promote the formation of pulp-like tissues rich in blood vessels and peripheral nerve-like structures. In conclusion, this study introduces a feasible and effective hydrogel tailored to specifically regulate the JAG1/Notch signaling pathway, showing potential in advancing regenerative strategies for dental pulp tissue.
Collapse
Affiliation(s)
- Siyuan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Mei Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Maojiao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Min He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Li Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Fangjun Huo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Oral Regenerative Medicine, Engineering Research Center of Oral Translational Medicine Ministry of Education, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
15
|
Nain A, Joshi A, Debnath S, Choudhury S, Thomas J, Satija J, Huang CC, Chatterjee K. A 4D printed nanoengineered super bioactive hydrogel scaffold with programmable deformation for potential bifurcated vascular channel construction. J Mater Chem B 2024; 12:7604-7617. [PMID: 38984474 DOI: 10.1039/d4tb00498a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Four-dimensional (4D) printing of hydrogels enabled the fabrication of complex scaffold geometries out of static parts. Although current 4D fabrication strategies are promising for creating vascular parts such as tubes, developing branched networks or tubular junctions is still challenging. Here, for the first time, a 4D printing approach is employed to fabricate T-shaped perfusable bifurcation using an extrusion-based multi-material 3D printing process. An alginate/methylcellulose-based dual-component hydrogel system (with defined swelling behavior) is nanoengineered with carbonized alginate (∼100 nm) to introduce anti-oxidative, anti-inflammatory, and anti-thrombotic properties and shape-shifting properties. A computational model to predict shape deformations in the printed hydrogels with defined infill angles was designed and further validated experimentally. Shape deformations of the 3D-printed flat sheets were achieved by ionic cross-linking. An undisrupted perfusion of a dye solution through a T-junction with minimal leakage mimicking blood flow through vessels is also demonstrated. Moreover, human umbilical vein endothelial and fibroblast cells seeded with printed constructs show intact morphology and excellent cell viability. Overall, the developed strategy paves the way for manufacturing self-actuated vascular bifurcations with remarkable anti-thrombotic properties to potentially treat coronary artery diseases.
Collapse
Affiliation(s)
- Amit Nain
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
- Department of Applied Mechanics & Biomedical Engineering, Indian Institute of Technology-Madras, 600036, Tamil Nadu, India
| | - Akshat Joshi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90024, USA
| | - Souvik Debnath
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
| | - Saswat Choudhury
- Department of Bioengineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| | - Jobin Thomas
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Jitendra Satija
- Centre for Nanobiotechnology, Vellore Institute of Technology, Vellore, Tamil Nadu 632014, India
| | - Chih-Ching Huang
- Department of Bioscience and Biotechnology and Centre of Excellence for the Oceans, National Taiwan Ocean University, Keelung, 202301, Taiwan
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Kaushik Chatterjee
- Department of Material Engineering, Indian Institute of Science, Bangalore, Karnataka 560012, India.
- Department of Bioengineering, Indian Institute of Science, Bangalore, Karnataka 560012, India
| |
Collapse
|
16
|
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
|
17
|
Al-Badri G, Phillips JB, Shipley RJ, Ovenden NC. Formation of vascular-like structures using a chemotaxis-driven multiphase model. Math Biosci 2024; 372:109183. [PMID: 38554855 DOI: 10.1016/j.mbs.2024.109183] [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: 11/28/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
We propose a continuum model for pattern formation, based on the multiphase model framework, to explore in vitro cell patterning within an extracellular matrix (ECM). We demonstrate that, within this framework, chemotaxis-driven cell migration can lead to the formation of cell clusters and vascular-like structures in 1D and 2D respectively. The influence on pattern formation of additional mechanisms commonly included in multiphase tissue models, including cell-matrix traction, contact inhibition, and cell-cell aggregation, are also investigated. Using sensitivity analysis, the relative impact of each model parameter on the simulation outcomes is assessed to identify the key parameters involved. Chemoattractant-matrix binding is further included, motivated by previous experimental studies, and found to reduce the spatial scale of patterning to within a biologically plausible range for capillary structures. Key findings from the in-depth parameter analysis of the 1D models, both with and without chemoattractant-matrix binding, are demonstrated to translate well to the 2D model, obtaining vascular-like cell patterning for multiple parameter regimes. Overall, we demonstrate a biologically-motivated multiphase model capable of generating long-term pattern formation on a biologically plausible spatial scale both in 1D and 2D, with applications for modelling in vitro vascular network formation.
Collapse
Affiliation(s)
- Georgina Al-Badri
- Department of Mathematics, University College London, London, UK; Centre for Nerve Engineering, University College London, London, UK.
| | - James B Phillips
- Centre for Nerve Engineering, University College London, London, UK; Department of Pharmacology, University College London, London, UK
| | - Rebecca J Shipley
- Centre for Nerve Engineering, University College London, London, UK; Department of Mechanical Engineering, University College London, London, UK
| | - Nicholas C Ovenden
- Department of Mathematics, University College London, London, UK; Centre for Nerve Engineering, University College London, London, UK
| |
Collapse
|
18
|
Zhang Y, Liu J, de Souza Araujo I, Bahammam L, Munn L, Huang G. Neovascularization by DPSC-ECs in a Tube Model for Pulp Regeneration Study. J Dent Res 2024; 103:652-661. [PMID: 38716736 PMCID: PMC11122093 DOI: 10.1177/00220345241236392] [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] [Indexed: 05/24/2024] Open
Abstract
The process of neovascularization during cell-based pulp regeneration is difficult to study. Here we developed a tube model that simulates root canal space and allows direct visualization of the vascularization process in vitro. Endothelial-like cells (ECs) derived from guiding human dental pulp stem cells (DPSCs) into expressing endothelial cell markers CD144, vWF, VEGFR1, and VEGFR2 were used. Human microvascular endothelial cells (hMVECs) were used as a positive control. DPSC-ECs formed tubules on Matrigel similar to hMVECs. Cells were mixed in fibrinogen/thrombin or mouse blood and seeded into wells of 96-well plates or injected into a tapered plastic tube (14 mm in length and 1 or 2 mm diameter of the apex opening) with the larger end sealed with MTA to simulate root canal space. Cells/gels in wells or tubes were incubated for various times in vitro and observed under the microscope for morphological changes. Samples were then fixed and processed for histological analysis to determine vessel formation. Vessel-like networks were observed in culture from 1 to 3 d after cell seeding. Cells/gels in 96-well plates were maintained up to 25 d. Histologically, both hMVECs and DPSC-ECs in 96-well plates or tubes showed intracellular vacuole formation. Some cells showed merged large vacuoles indicating the lumenization. Tubular structures were also observed resembling blood vessels. Cells appeared healthy throughout the tube except some samples (1 mm apical diameter) in the coronal third. Histological analysis also showed pulp-like soft tissue throughout the tube samples with vascular-like structures. hMVECs formed larger vascular lumen size than DPSC-ECs while the latter tended to have more lumen and tubular structure counts. We conclude that DPSC-ECs can form vascular structures and sustained in the 3-dimensional fibrin gel system in vitro. The tube model appears to be a proper and simple system simulating the root canal space for vascular formation and pulp regeneration studies.
Collapse
Affiliation(s)
- Y. Zhang
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - J. Liu
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - I.J. de Souza Araujo
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - L. Bahammam
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Faculty of Dentistry, King Abdulaziz University, Jeddah, Makkah, Kingdom of Saudi Arabia*
| | - L.L. Munn
- Radiation Oncology, Massachusetts General Hospital Research Institute, Harvard Medical School, Charlestown, MA, USA
| | - G.T.J. Huang
- Departments of Bioscience Research and Endodontics, College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
19
|
Davoodi P, Rezaei N, Hassan M, Hay DC, Vosough M. Bioengineering vascularized liver tissue for biomedical research and application. Scand J Gastroenterol 2024; 59:623-629. [PMID: 38319110 DOI: 10.1080/00365521.2024.2310172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/18/2024] [Accepted: 01/20/2024] [Indexed: 02/07/2024]
Abstract
The liver performs a wide range of biological functions that are essential to body homeostasis. Damage to liver tissue can result in reduced organ function, and if chronic in nature can lead to organ scarring and progressive disease. Currently, donor liver transplantation is the only longterm treatment for end-stage liver disease. However, orthotopic organ transplantation suffers from several drawbacks that include organ scarcity and lifelong immunosuppression. Therefore, new therapeutic strategies are required. One promising strategy is the engineering of implantable and vascularized liver tissue. This resource could also be used to build the next generation of liver tissue models to better understand human health, disease and aging in vitro. This article reviews recent progress in the field of liver tissue bioengineering, including microfluidic-based systems, bio-printed vascularized tissue, liver spheroids and organoid models, and the induction of angiogenesis in vivo.
Collapse
Affiliation(s)
- Parsa Davoodi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Niloofar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David C Hay
- Centre for Regenerative Medicine, Institute for Repair and Regeneration, University of Edinburgh, Edinburgh, UK
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
Żuchowska A, Baranowska P, Flont M, Brzózka Z, Jastrzębska E. Review: 3D cell models for organ-on-a-chip applications. Anal Chim Acta 2024; 1301:342413. [PMID: 38553129 DOI: 10.1016/j.aca.2024.342413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 04/02/2024]
Abstract
Two-dimensional (2D) cultures do not fully reflect the human organs' physiology and the real effectiveness of the used therapy. Therefore, three-dimensional (3D) models are increasingly used in bioanalytical science. Organ-on-a-chip systems are used to obtain cellular in vitro models, better reflecting the human body's in vivo characteristics and allowing us to obtain more reliable results than standard preclinical models. Such 3D models can be used to understand the behavior of tissues/organs in response to selected biophysical and biochemical factors, pathological conditions (the mechanisms of their formation), drug screening, or inter-organ interactions. This review characterizes 3D models obtained in microfluidic systems. These include spheroids/aggregates, hydrogel cultures, multilayers, organoids, or cultures on biomaterials. Next, the methods of formation of different 3D cultures in Organ-on-a-chip systems are presented, and examples of such Organ-on-a-chip systems are discussed. Finally, current applications of 3D cell-on-a-chip systems and future perspectives are covered.
Collapse
Affiliation(s)
- Agnieszka Żuchowska
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Patrycja Baranowska
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822, Warsaw, Poland
| | - Magdalena Flont
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822, Warsaw, Poland
| | - Zbigniew Brzózka
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland.
| |
Collapse
|
21
|
Klak M, Rachalewski M, Filip A, Dobrzański T, Berman A, Wszoła M. Bioprinting of Perfusable, Biocompatible Vessel-like Channels with dECM-Based Bioinks and Living Cells. Bioengineering (Basel) 2024; 11:439. [PMID: 38790306 PMCID: PMC11117567 DOI: 10.3390/bioengineering11050439] [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: 03/28/2024] [Revised: 04/14/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
There is a growing interest in the production of bioinks that on the one hand, are biocompatible and, on the other hand, have mechanical properties that allow for the production of stable constructs that can survive for a long time after transplantation. While the selection of the right material is crucial for bioprinting, there is another equally important issue that is currently being extensively researched-the incorporation of the vascular system into the fabricated scaffolds. Therefore, in the following manuscript, we present the results of research on bioink with unique physico-chemical and biological properties. In this article, two methods of seeding cells were tested using bioink B and seeding after bioprinting the whole model. After 2, 5, 8, or 24 h of incubation, the flow medium was used in the tested systems. At the end of the experimental trial, for each time variant, the canals were stored in formaldehyde, and immunohistochemical staining was performed to examine the presence of cells on the canal walls and roof. Cells adhered to both ways of fiber arrangement; however, a parallel bioprint with the 5 h incubation and the intermediate plating of cells resulted in better adhesion efficiency. For this test variant, the percentage of cells that adhered was at least 20% higher than in the other analyzed variants. In addition, it was for this variant that the lowest percentage of viable cells was found that were washed out of the tested model. Importantly, hematoxylin and eosin staining showed that after 8 days of culture, the cells were evenly distributed throughout the canal roof. Our study clearly shows that neovascularization-promoting cells effectively adhere to ECM-based pancreatic bioink. Summarizing the presented results, it was demonstrated that the proposed bioink compositions can be used for bioprinting bionic organs with a vascular system formed by endothelial cells and fibroblasts.
Collapse
Affiliation(s)
- Marta Klak
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
- Polbionica sp. z o.o., 01-242 Warsaw, Poland
| | - Michał Rachalewski
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
| | - Anna Filip
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
| | | | | | - Michał Wszoła
- Foundation of Research and Science Development, 01-242 Warsaw, Poland or (M.W.)
- Polbionica sp. z o.o., 01-242 Warsaw, Poland
| |
Collapse
|
22
|
Jackson CE, Green NH, English WR, Claeyssens F. The use of microphysiological systems to model metastatic cancer. Biofabrication 2024; 16:032002. [PMID: 38579739 DOI: 10.1088/1758-5090/ad3b70] [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: 11/13/2023] [Accepted: 04/05/2024] [Indexed: 04/07/2024]
Abstract
Cancer is one of the leading causes of death in the 21st century, with metastasis of cancer attributing to 90% of cancer-related deaths. Therefore, to improve patient outcomes there is a need for better preclinical models to increase the success of translating oncological therapies into the clinic. Current traditional staticin vitromodels lack a perfusable network which is critical to overcome the diffusional mass transfer limit to provide a mechanism for the exchange of essential nutrients and waste removal, and increase their physiological relevance. Furthermore, these models typically lack cellular heterogeneity and key components of the immune system and tumour microenvironment. This review explores rapidly developing strategies utilising perfusable microphysiological systems (MPS) for investigating cancer cell metastasis. In this review we initially outline the mechanisms of cancer metastasis, highlighting key steps and identifying the current gaps in our understanding of the metastatic cascade, exploring MPS focused on investigating the individual steps of the metastatic cascade before detailing the latest MPS which can investigate multiple components of the cascade. This review then focuses on the factors which can affect the performance of an MPS designed for cancer applications with a final discussion summarising the challenges and future directions for the use of MPS for cancer models.
Collapse
Affiliation(s)
- Caitlin E Jackson
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - Nicola H Green
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| | - William R English
- Norwich Medical School, University of East Anglia, Norwich NR3 7TJ, United Kingdom
| | - Frederik Claeyssens
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, United Kingdom
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, United Kingdom
| |
Collapse
|
23
|
Mamidi N, Ijadi F, Norahan MH. Leveraging the Recent Advancements in GelMA Scaffolds for Bone Tissue Engineering: An Assessment of Challenges and Opportunities. Biomacromolecules 2024; 25:2075-2113. [PMID: 37406611 DOI: 10.1021/acs.biomac.3c00279] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
The field of bone tissue engineering has seen significant advancements in recent years. Each year, over two million bone transplants are performed globally, and conventional treatments, such as bone grafts and metallic implants, have their limitations. Tissue engineering offers a new level of treatment, allowing for the creation of living tissue within a biomaterial framework. Recent advances in biomaterials have provided innovative approaches to rebuilding bone tissue function after damage. Among them, gelatin methacryloyl (GelMA) hydrogel is emerging as a promising biomaterial for supporting cell proliferation and tissue regeneration, and GelMA has exhibited exceptional physicochemical and biological properties, making it a viable option for clinical translation. Various methods and classes of additives have been used in the application of GelMA for bone regeneration, with the incorporation of nanofillers or other polymers enhancing its resilience and functional performance. Despite promising results, the fabrication of complex structures that mimic the bone architecture and the provision of balanced physical properties for both cell and vasculature growth and proper stiffness for load bearing remain as challenges. In terms of utilizing osteogenic additives, the priority should be on versatile components that promote angiogenesis and osteogenesis while reinforcing the structure for bone tissue engineering applications. This review focuses on recent efforts and advantages of GelMA-based composite biomaterials for bone tissue engineering, covering the literature from the last five years.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Fatemeh Ijadi
- Department of Chemistry and Nanotechnology, School of Engineering and Science, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| | - Mohammad Hadi Norahan
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Nuevo León 64849, México
| |
Collapse
|
24
|
Sun T, Xiang Y, Turner F, Bao X. Integrated Experimental and Mathematical Exploration of Modular Tissue Cultures for Developmental Engineering. Int J Mol Sci 2024; 25:2987. [PMID: 38474234 DOI: 10.3390/ijms25052987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/06/2024] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developmental engineering (DE) involves culturing various cells on modular scaffolds (MSs), yielding modular tissues (MTs) assembled into three-dimensional (3D) tissues, mimicking developmental biology. This study employs an integrated approach, merging experimental and mathematical methods to investigate the biological processes in MT cultivation and assembly. Human dermal fibroblasts (HDFs) were cultured on tissue culture plastics, poly(lactic acid) (PLA) discs with regular open structures, or spherical poly(methyl methacrylate) (PMMA) MSs, respectively. Notably, HDFs exhibited flattened spindle shapes when adhered to solid surfaces, and complex 3D structures when migrating into the structured voids of PLA discs or interstitial spaces between aggregated PMMA MSs, showcasing coordinated colonization of porous scaffolds. Empirical investigations led to power law models simulating density-dependent cell growth on solid surfaces or voids. Concurrently, a modified diffusion model was applied to simulate oxygen diffusion within tissues cultured on solid surfaces or porous structures. These mathematical models were subsequently combined to explore the influences of initial cell seeding density, culture duration, and oxygen diffusion on MT cultivation and assembly. The findings underscored the intricate interplay of factors influencing MT design for tissue assembly. The integrated approach provides insights into mechanistic aspects, informing bioprocess design for manufacturing MTs and 3D tissues in DE.
Collapse
Affiliation(s)
- Tao Sun
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Yu Xiang
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Freya Turner
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| |
Collapse
|
25
|
Li H, Shang Y, Zeng J, Matsusaki M. Technology for the formation of engineered microvascular network models and their biomedical applications. NANO CONVERGENCE 2024; 11:10. [PMID: 38430377 PMCID: PMC10908775 DOI: 10.1186/s40580-024-00416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024]
Abstract
Tissue engineering and regenerative medicine have made great progress in recent decades, as the fields of bioengineering, materials science, and stem cell biology have converged, allowing tissue engineers to replicate the structure and function of various levels of the vascular tree. Nonetheless, the lack of a fully functional vascular system to efficiently supply oxygen and nutrients has hindered the clinical application of bioengineered tissues for transplantation. To investigate vascular biology, drug transport, disease progression, and vascularization of engineered tissues for regenerative medicine, we have analyzed different approaches for designing microvascular networks to create models. This review discusses recent advances in the field of microvascular tissue engineering, explores potential future challenges, and offers methodological recommendations.
Collapse
Affiliation(s)
- He Li
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yucheng Shang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
26
|
Conci C, Sironi L, Jacchetti E, Panzeri D, Inverso D, Martínez Vázquez R, Osellame R, Collini M, Cerullo G, Chirico G, Raimondi MT. In vivo label-free tissue histology through a microstructured imaging window. APL Bioeng 2024; 8:016102. [PMID: 38222895 PMCID: PMC10787586 DOI: 10.1063/5.0165411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 11/06/2023] [Indexed: 01/16/2024] Open
Abstract
Tissue histopathology, based on hematoxylin and eosin (H&E) staining of thin tissue slices, is the gold standard for the evaluation of the immune reaction to the implant of a biomaterial. It is based on lengthy and costly procedures that do not allow longitudinal studies. The use of non-linear excitation microscopy in vivo, largely label-free, has the potential to overcome these limitations. With this purpose, we develop and validate an implantable microstructured device for the non-linear excitation microscopy assessment of the immune reaction to an implanted biomaterial label-free. The microstructured device, shaped as a matrix of regular 3D lattices, is obtained by two-photon laser polymerization. It is subsequently implanted in the chorioallantoic membrane (CAM) of embryonated chicken eggs for 7 days to act as an intrinsic 3D reference frame for cell counting and identification. The histological analysis based on H&E images of the tissue sections sampled around the implanted microstructures is compared to non-linear excitation and confocal images to build a cell atlas that correlates the histological observations to the label-free images. In this way, we can quantify the number of cells recruited in the tissue reconstituted in the microstructures and identify granulocytes on label-free images within and outside the microstructures. Collagen and microvessels are also identified by means of second-harmonic generation and autofluorescence imaging. The analysis indicates that the tissue reaction to implanted microstructures is like the one typical of CAM healing after injury, without a massive foreign body reaction. This opens the path to the use of similar microstructures coupled to a biomaterial, to image in vivo the regenerating interface between a tissue and a biomaterial with label-free non-linear excitation microscopy. This promises to be a transformative approach, alternative to conventional histopathology, for the bioengineering and the validation of biomaterials in in vivo longitudinal studies.
Collapse
Affiliation(s)
- Claudio Conci
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Laura Sironi
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Emanuela Jacchetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Davide Panzeri
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Donato Inverso
- Division of Immunology, Transplantation and Infectious Diseases IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Maddalena Collini
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Giulio Cerullo
- Institute for Photonics and Nanotechnologies (IFN), CNR and Department of Physics, Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| | - Giuseppe Chirico
- Department of Physics, Università di Milano-Bicocca, Piazza della Scienza 3, 20126 Milan, Italy
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta,” Politecnico di Milano, Piazza L. da Vinci 32, 20133 Milan, Italy
| |
Collapse
|
27
|
Utagawa Y, Ino K, Takinoue M, Shiku H. Fabrication and Cell Culture Applications of Core-Shell Hydrogel Fibers Composed of Chitosan/DNA Interfacial Polyelectrolyte Complexation and Calcium Alginate: Straight and Beaded Core Variations. Adv Healthc Mater 2023; 12:e2302011. [PMID: 37478383 PMCID: PMC11468996 DOI: 10.1002/adhm.202302011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Indexed: 07/23/2023]
Abstract
Core-shell hydrogel fibers are widely used in cell culture applications. A simple and rapid method is presented for fabricating core-shell hydrogel fibers, consisting of straight or beaded core fibers, for cell culture applications. The core fibers are prepared using interfacial polyelectrolyte complexation (IPC) with chitosan and DNA. Briefly, two droplets of chitosan and DNA are brought in contact to form an IPC film, which is dragged to prepare an IPC fiber. The incubation time and DNA concentration are adjusted to prepare straight and beaded IPC fibers. The fibers with Ca2+ are immersed in an alginate solution to form calcium alginate shell hydrogels around the core IPC fibers. To the best of the knowledge, this is the first report of core-shell hydrogel fibers with IPC fiber cores. To demonstrate cell culture, straight hydrogel fibers are applied to fabricate hepatic models consisting of HepG2 and 3T3 fibroblasts, and vascular models consisting of human umbilical vein endothelial cells and 3T3 fibroblasts. To evaluate the effect of co-culture, albumin secretion, and angiogenesis are evaluated. Beaded hydrogel fibers are used to fabricate many size-controlled spheroids for fiber and cloning applications. This method can be widely applied in tissue engineering and cell analysis.
Collapse
Affiliation(s)
| | - Kosuke Ino
- Graduate School of EngineeringTohoku UniversitySendai980–8579Japan
| | - Masahiro Takinoue
- Department of Computer ScienceTokyo Institute of TechnologyYokohama226–8502Japan
| | - Hitoshi Shiku
- Graduate School of EngineeringTohoku UniversitySendai980–8579Japan
- Graduate School of Environmental StudiesTohoku UniversitySendai980–8579Japan
| |
Collapse
|
28
|
Duong VT, Lin CC. Digital Light Processing 3D Bioprinting of Gelatin-Norbornene Hydrogel for Enhanced Vascularization. Macromol Biosci 2023; 23:e2300213. [PMID: 37536347 PMCID: PMC10837335 DOI: 10.1002/mabi.202300213] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Indexed: 08/05/2023]
Abstract
Digital light processing (DLP) bioprinting can be used to fabricate volumetric scaffolds with intricate internal structures, such as perfusable vascular channels. The successful implementation of DLP bioprinting in tissue fabrication requires using suitable photo-reactive bioinks. Norbornene-based bioinks have emerged as an attractive alternative to (meth)acrylated macromers in 3D bioprinting owing to their mild and rapid reaction kinetics, high cytocompatibility for in situ cell encapsulation, and adaptability for post-printing modification or conjugation of bioactive motifs. In this contribution, the development of gelatin-norbornene (GelNB) is reported as a photo-cross-linkable bioink for DLP 3D bioprinting. Low concentrations of GelNB (2-5 wt.%) and poly(ethylene glycol)-tetra-thiol (PEG4SH) are DLP-printed with a wide range of stiffness (G' ≈120 to 4000 Pa) and with perfusable channels. DLP-printed GelNB hydrogels are highly cytocompatible, as demonstrated by the high viability of the encapsulated human umbilical vein endothelial cells (HUVECs). The encapsulated HUVECs formed an interconnected microvascular network with lumen structures. Notably, the GelNB bioink permitted both in situ tethering and secondary conjugation of QK peptide, a vascular endothelial growth factor (VEGF)-mimetic peptide. Incorporation of QK peptide significantly improved endothelialization and vasculogenesis of the DLP-printed GelNB hydrogels, reinforcing the applicability of this bioink system in diverse biofabrication applications.
Collapse
Affiliation(s)
- Van Thuy Duong
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Chien-Chi Lin
- Department of Biomedical Engineering, Purdue School of Engineering & Technology, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
29
|
Downer M, Berry CE, Parker JB, Kameni L, Griffin M. Current Biomaterials for Wound Healing. Bioengineering (Basel) 2023; 10:1378. [PMID: 38135969 PMCID: PMC10741152 DOI: 10.3390/bioengineering10121378] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/20/2023] [Accepted: 11/23/2023] [Indexed: 12/24/2023] Open
Abstract
Wound healing is the body's process of injury recovery. Skin healing is divided into four distinct overlapping phases: hemostasis, inflammation, proliferation, and remodeling. Cell-to-cell interactions mediated by both cytokines and chemokines are imperative for the transition between these phases. Patients can face difficulties in the healing process due to the wound being too large, decreased vascularization, infection, or additional burdens of a systemic illness. The field of tissue engineering has been investigating biomaterials as an alternative for skin regeneration. Biomaterials used for wound healing may be natural, synthetic, or a combination of both. Once a specific biomaterial is selected, it acts as a scaffold for skin regeneration. When the scaffold is applied to a wound, it allows for the upregulation of distinct molecular signaling pathways important for skin repair. Although tissue engineering has made great progress, more research is needed in order to support the use of biomaterials for wound healing for clinical translation.
Collapse
Affiliation(s)
- Mauricio Downer
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.D.); (C.E.B.); (J.B.P.); (L.K.)
| | - Charlotte E. Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.D.); (C.E.B.); (J.B.P.); (L.K.)
| | - Jennifer B. Parker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.D.); (C.E.B.); (J.B.P.); (L.K.)
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lionel Kameni
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.D.); (C.E.B.); (J.B.P.); (L.K.)
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.D.); (C.E.B.); (J.B.P.); (L.K.)
- Hagey Laboratory for Pediatric Regenerative Medicine, 257 Campus Drive, MC 5148, Stanford, CA 94305, USA
| |
Collapse
|
30
|
Gupta S, Sharma A, Petrovski G, Verma RS. Vascular reconstruction of the decellularized biomatrix for whole-organ engineering-a critical perspective and future strategies. Front Bioeng Biotechnol 2023; 11:1221159. [PMID: 38026872 PMCID: PMC10680456 DOI: 10.3389/fbioe.2023.1221159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Whole-organ re-engineering is the most challenging goal yet to be achieved in tissue engineering and regenerative medicine. One essential factor in any transplantable and functional tissue engineering is fabricating a perfusable vascular network with macro- and micro-sized blood vessels. Whole-organ development has become more practical with the use of the decellularized organ biomatrix (DOB) as it provides a native biochemical and structural framework for a particular organ. However, reconstructing vasculature and re-endothelialization in the DOB is a highly challenging task and has not been achieved for constructing a clinically transplantable vascularized organ with an efficient perfusable capability. Here, we critically and articulately emphasized factors that have been studied for the vascular reconstruction in the DOB. Furthermore, we highlighted the factors used for vasculature development studies in general and their application in whole-organ vascular reconstruction. We also analyzed in detail the strategies explored so far for vascular reconstruction and angiogenesis in the DOB for functional and perfusable vasculature development. Finally, we discussed some of the crucial factors that have been largely ignored in the vascular reconstruction of the DOB and the future directions that should be addressed systematically.
Collapse
Affiliation(s)
- Santosh Gupta
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Akriti Sharma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| | - Goran Petrovski
- Center for Eye Research and Innovative Diagnostics, Department of Ophthalmology, Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, University of Split School of Medicine and University Hospital Centre, Split, Croatia
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology, Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences. Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
31
|
Bülow A, Schäfer B, Beier JP. Three-Dimensional Bioprinting in Soft Tissue Engineering for Plastic and Reconstructive Surgery. Bioengineering (Basel) 2023; 10:1232. [PMID: 37892962 PMCID: PMC10604458 DOI: 10.3390/bioengineering10101232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Skeletal muscle tissue engineering (TE) and adipose tissue engineering have undergone significant progress in recent years. This review focuses on the key findings in these areas, particularly highlighting the integration of 3D bioprinting techniques to overcome challenges and enhance tissue regeneration. In skeletal muscle TE, 3D bioprinting enables the precise replication of muscle architecture. This addresses the need for the parallel alignment of cells and proper innervation. Satellite cells (SCs) and mesenchymal stem cells (MSCs) have been utilized, along with co-cultivation strategies for vascularization and innervation. Therefore, various printing methods and materials, including decellularized extracellular matrix (dECM), have been explored. Similarly, in adipose tissue engineering, 3D bioprinting has been employed to overcome the challenge of vascularization; addressing this challenge is vital for graft survival. Decellularized adipose tissue and biomimetic scaffolds have been used as biological inks, along with adipose-derived stem cells (ADSCs), to enhance graft survival. The integration of dECM and alginate bioinks has demonstrated improved adipocyte maturation and differentiation. These findings highlight the potential of 3D bioprinting techniques in skeletal muscle and adipose tissue engineering. By integrating specific cell types, biomaterials, and printing methods, significant progress has been made in tissue regeneration. However, challenges such as fabricating larger constructs, translating findings to human models, and obtaining regulatory approvals for cellular therapies remain to be addressed. Nonetheless, these advancements underscore the transformative impact of 3D bioprinting in tissue engineering research and its potential for future clinical applications.
Collapse
Affiliation(s)
- Astrid Bülow
- Department of Plastic Surgery, Hand Surgery, Burn Center, University Hospital RWTH Aachen, 52074 Aachen, Germany; (B.S.); (J.P.B.)
| | | | | |
Collapse
|
32
|
Covani U, Giammarinaro E, Di Pietro N, Boncompagni S, Rastelli G, Romasco T, Velasco-Ortega E, Jimenez-Guerra A, Iezzi G, Piattelli A, Marconcini S. Electron Microscopy (EM) Analysis of Collagen Fibers in the Peri-Implant Soft Tissues around Two Different Abutments. J Funct Biomater 2023; 14:445. [PMID: 37754859 PMCID: PMC10532031 DOI: 10.3390/jfb14090445] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
The design of the implant prosthesis-abutment complex appears crucial for shaping healthy and stable peri-implant soft tissues. The aim of the present animal study was to compare two implants with different healing abutment geometries: a concave design (TEST) and a straight one (CTRL). Transmission electron microscopy (TEM) was used to quantify the three-dimensional topography and morphological properties of collagen at nanoscale resolution. 2 swine were included in the experiment and 6 implants per animal were randomly placed in the left or right hemimandible in either the physiologically mature bone present between the lower canine and first premolar or in the mandibular premolar area, within tooth extraction sites. Each CTRL implant was positioned across from its respective TEST implant on the other side of the jaw. After 12 weeks of healing, 8 specimens (4 CTRL and 4 TEST) were retrieved and prepared for histological and TEM analysis. The results showed a significantly higher percentage of area covered by collagen bundles and average bundle size in TEST implants, as well as a significant decrease in the number of longitudinally oriented bundles with respect to CTRL implants, which is potentially due to the larger size of TEST bundles. These data suggest that a concave transmucosal abutment design serves as a scaffold, favoring the deposition and growth of a well-organized peri-implant collagen structure over the implant platform in the early healing phase, also promoting the convergence of collagen fibers toward the abutment collar.
Collapse
Affiliation(s)
- Ugo Covani
- Department of Stomatology, Tuscan Dental Institute, 55041 Lido di Camaiore, Italy; (U.C.); (E.G.); (S.M.)
| | - Enrica Giammarinaro
- Department of Stomatology, Tuscan Dental Institute, 55041 Lido di Camaiore, Italy; (U.C.); (E.G.); (S.M.)
| | - Natalia Di Pietro
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (T.R.); (G.I.)
- Center for Advanced Studies and Technology—CAST, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.B.); (G.R.)
| | - Simona Boncompagni
- Center for Advanced Studies and Technology—CAST, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.B.); (G.R.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgia Rastelli
- Center for Advanced Studies and Technology—CAST, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.B.); (G.R.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Tea Romasco
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (T.R.); (G.I.)
- Center for Advanced Studies and Technology—CAST, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (S.B.); (G.R.)
| | - Eugenio Velasco-Ortega
- Department of Stomatology, Faculty of Dentistry, University of Seville, 41013 Seville, Spain; (E.V.-O.); (A.J.-G.)
| | - Alvaro Jimenez-Guerra
- Department of Stomatology, Faculty of Dentistry, University of Seville, 41013 Seville, Spain; (E.V.-O.); (A.J.-G.)
| | - Giovanna Iezzi
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (T.R.); (G.I.)
| | - Adriano Piattelli
- School of Dentistry, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy;
- Facultad de Medicina, UCAM Universidad Católica San Antonio de Murcia, 30107 Murcia, Spain
| | - Simone Marconcini
- Department of Stomatology, Tuscan Dental Institute, 55041 Lido di Camaiore, Italy; (U.C.); (E.G.); (S.M.)
| |
Collapse
|
33
|
Hartmann J, Mayor R. Self-organized collective cell behaviors as design principles for synthetic developmental biology. Semin Cell Dev Biol 2023; 141:63-73. [PMID: 35450765 DOI: 10.1016/j.semcdb.2022.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Over the past two decades, molecular cell biology has graduated from a mostly analytic science to one with substantial synthetic capability. This success is built on a deep understanding of the structure and function of biomolecules and molecular mechanisms. For synthetic biology to achieve similar success at the scale of tissues and organs, an equally deep understanding of the principles of development is required. Here, we review some of the central concepts and recent progress in tissue patterning, morphogenesis and collective cell migration and discuss their value for synthetic developmental biology, emphasizing in particular the power of (guided) self-organization and the role of theoretical advances in making developmental insights applicable in synthesis.
Collapse
Affiliation(s)
- Jonas Hartmann
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
34
|
Park D, Lee SJ, Choi DK, Park JW. Therapeutic Agent-Loaded Fibrous Scaffolds for Biomedical Applications. Pharmaceutics 2023; 15:pharmaceutics15051522. [PMID: 37242764 DOI: 10.3390/pharmaceutics15051522] [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: 04/05/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Tissue engineering is a sophisticated field that involves the integration of various disciplines, such as clinical medicine, material science, and life science, to repair or regenerate damaged tissues and organs. To achieve the successful regeneration of damaged or diseased tissues, it is necessary to fabricate biomimetic scaffolds that provide structural support to the surrounding cells and tissues. Fibrous scaffolds loaded with therapeutic agents have shown considerable potential in tissue engineering. In this comprehensive review, we examine various methods for fabricating bioactive molecule-loaded fibrous scaffolds, including preparation methods for fibrous scaffolds and drug-loading techniques. Additionally, we delved into the recent biomedical applications of these scaffolds, such as tissue regeneration, inhibition of tumor recurrence, and immunomodulation. The aim of this review is to discuss the latest research trends in fibrous scaffold manufacturing methods, materials, drug-loading methods with parameter information, and therapeutic applications with the goal of contributing to the development of new technologies or improvements to existing ones.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Dong Kyu Choi
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
35
|
Hu Y, Becker ML, Willits RK. Quantification of cell migration: metrics selection to model application. Front Cell Dev Biol 2023; 11:1155882. [PMID: 37255596 PMCID: PMC10225508 DOI: 10.3389/fcell.2023.1155882] [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: 02/01/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Cell migration plays an essential role in physiological and pathological states, such as immune response, tissue generation and tumor development. This phenomenon can occur spontaneously or it can be triggered by an external stimuli, including biochemical, mechanical, or electrical cues that induce or direct cells to migrate. The migratory response to these cues is foundational to several fields including neuroscience, cancer and regenerative medicine. Various platforms are available to qualitatively and quantitatively measure cell migration, making the measurements of cell motility straight-forward. Migratory behavior must be analyzed by multiple metrics and then models to connect the measurements to physiological meaning. This review will focus on describing and quantifying cell movement for individual cell migration.
Collapse
Affiliation(s)
- Yang Hu
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States
| | - Matthew L. Becker
- Departments of Chemistry, Mechanical Engineering and Materials Science, Biomedical Engineering and Orthopedic Surgery, Duke University, Durham, NC, United States
| | - Rebecca Kuntz Willits
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, United States
| |
Collapse
|
36
|
Karanfil AS, Louis F, Matsusaki M. Biofabrication of vascularized adipose tissues and their biomedical applications. MATERIALS HORIZONS 2023; 10:1539-1558. [PMID: 36789675 DOI: 10.1039/d2mh01391f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in adipose tissue engineering and cell biology have led to the development of innovative therapeutic strategies in regenerative medicine for adipose tissue reconstruction. To date, the many in vitro and in vivo models developed for vascularized adipose tissue engineering cover a wide range of research areas, including studies with cells of various origins and types, polymeric scaffolds of natural and synthetic derivation, models presented using decellularized tissues, and scaffold-free approaches. In this review, studies on adipose tissue types with different functions, characteristics and body locations have been summarized with 3D in vitro fabrication approaches. The reason for the particular focus on vascularized adipose tissue models is that current liposuction and fat transplantation methods are unsuitable for adipose tissue reconstruction as the lack of blood vessels results in inadequate nutrient and oxygen delivery, leading to necrosis in situ. In the first part of this paper, current studies and applications of white and brown adipose tissues are presented according to the polymeric materials used, focusing on the studies which could show vasculature in vitro and after in vivo implantation, and then the research on adipose tissue fabrication and applications are explained.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
37
|
Cell–scaffold interactions in tissue engineering for oral and craniofacial reconstruction. Bioact Mater 2023; 23:16-44. [DOI: 10.1016/j.bioactmat.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
|
38
|
Ren B, Jiang Z, Murfee WL, Katz AJ, Siemann D, Huang Y. Realizations of vascularized tissues: From in vitro platforms to in vivo grafts. BIOPHYSICS REVIEWS 2023; 4:011308. [PMID: 36938117 PMCID: PMC10015415 DOI: 10.1063/5.0131972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023]
Abstract
Vascularization is essential for realizing thick and functional tissue constructs that can be utilized for in vitro study platforms and in vivo grafts. The vasculature enables the transport of nutrients, oxygen, and wastes and is also indispensable to organ functional units such as the nephron filtration unit, the blood-air barrier, and the blood-brain barrier. This review aims to discuss the latest progress of organ-like vascularized constructs with specific functionalities and realizations even though they are not yet ready to be used as organ substitutes. First, the human vascular system is briefly introduced and related design considerations for engineering vascularized tissues are discussed. Second, up-to-date creation technologies for vascularized tissues are summarized and classified into the engineering and cellular self-assembly approaches. Third, recent applications ranging from in vitro tissue models, including generic vessel models, tumor models, and different human organ models such as heart, kidneys, liver, lungs, and brain, to prevascularized in vivo grafts for implantation and anastomosis are discussed in detail. The specific design considerations for the aforementioned applications are summarized and future perspectives regarding future clinical applications and commercialization are provided.
Collapse
Affiliation(s)
- Bing Ren
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Zhihua Jiang
- Department of Surgery, University of Florida, Gainesville, Florida 32610, USA
| | - Walter Lee Murfee
- Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, USA
| | - Adam J. Katz
- Department of Plastic and Reconstructive Surgery, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Dietmar Siemann
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Yong Huang
- Author to whom correspondence should be addressed:
| |
Collapse
|
39
|
Dixit K, Bora H, Lakshmi Parimi J, Mukherjee G, Dhara S. Biomaterial mediated immunomodulation: An interplay of material environment interaction for ameliorating wound regeneration. J Biomater Appl 2023; 37:1509-1528. [PMID: 37069479 DOI: 10.1177/08853282231156484] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Chronic wounds are the outcome of an imbalanced inflammatory response caused by sustenance of immune microenvironment. In this context, tissue engineered graft played great role in healing wounds but faced difficulty in scar remodelling, immune rejection and poor vascularization. All the limitations faced are somewhere linked with the immune cells involved in healing. In this consideration, immunomodulatory biomaterials bridge a large gap with the delivery of modulating factors for triggering key inflammatory cells responsible towards interplay in the wound micro-environment. Inherent physico-chemical properties of biomaterials substantially determine the nature of cell-materials interaction thereby facilitating differential cytokine gradient involved in activation or suppression of inflammatory signalling pathways, and followed by surface marker expression. This review aims to systematically describe the interplay of immune cells involved in different phases in the wound microenvironment and biomaterials. Additionally, it also focuses on modulating innate immune cell responses in the context of triggering the halted phase of the wound healing, i.e., inflammatory phase. The various strategies are highlighted for modulation of wound microenvironment towards wound regeneration including stem cells, cytokines, growth factors, vitamins, and anti-inflammatory agents to induce interactive ability of biomaterials with immune cells. The last section focuses on prospective approaches and current potential strategies for wound regeneration. This includes the development of different models to bridge the gap between mouse models and human patients. Emerging new tools to study inflammatory response owing to biomaterials and novel strategies for modulation of monocyte and macrophage behaviour in the wound environment are also discussed.
Collapse
Affiliation(s)
- Krishna Dixit
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
- Immunology and Inflammation Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Hema Bora
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Jhansi Lakshmi Parimi
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Gayatri Mukherjee
- Immunology and Inflammation Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Santanu Dhara
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| |
Collapse
|
40
|
Quezada A, Ward C, Bader ER, Zolotavin P, Altun E, Hong S, Killian NJ, Xie C, Batista-Brito R, Hébert JM. An In Vivo Platform for Rebuilding Functional Neocortical Tissue. Bioengineering (Basel) 2023; 10:263. [PMID: 36829757 PMCID: PMC9952056 DOI: 10.3390/bioengineering10020263] [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: 12/29/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Recent progress in cortical stem cell transplantation has demonstrated its potential to repair the brain. However, current transplant models have yet to demonstrate that the circuitry of transplant-derived neurons can encode useful function to the host. This is likely due to missing cell types within the grafts, abnormal proportions of cell types, abnormal cytoarchitecture, and inefficient vascularization. Here, we devised a transplant platform for testing neocortical tissue prototypes. Dissociated mouse embryonic telencephalic cells in a liquid scaffold were transplanted into aspiration-lesioned adult mouse cortices. The donor neuronal precursors differentiated into upper and deep layer neurons that exhibited synaptic puncta, projected outside of the graft to appropriate brain areas, became electrophysiologically active within one month post-transplant, and responded to visual stimuli. Interneurons and oligodendrocytes were present at normal densities in grafts. Grafts became fully vascularized by one week post-transplant and vessels in grafts were perfused with blood. With this paradigm, we could also organize cells into layers. Overall, we have provided proof of a concept for an in vivo platform that can be used for developing and testing neocortical-like tissue prototypes.
Collapse
Affiliation(s)
- Alexandra Quezada
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Claire Ward
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Edward R. Bader
- Department of Neurological Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pavlo Zolotavin
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Esra Altun
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Sarah Hong
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nathaniel J. Killian
- Department of Neurological Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chong Xie
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Renata Batista-Brito
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jean M. Hébert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Stem Cell Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
41
|
Shrestha B, Tang L, Hood RL. Nanotechnology for Personalized Medicine. Nanomedicine (Lond) 2023. [DOI: 10.1007/978-981-16-8984-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
42
|
Chinnadurai R. Advanced Technologies for Potency Assay Measurement. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1420:81-95. [PMID: 37258785 DOI: 10.1007/978-3-031-30040-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Crucial for their application, cell products need to be well-characterized in the cell manufacturing facilities and conform to regulatory approval criteria before infusion into the patients. Mesenchymal Stromal Cells (MSCs) are the leading cell therapy candidate in clinical trials worldwide. Early phase clinical trials have demonstrated that MSCs display an excellent safety profile and are well tolerated. However, MSCs have also exhibited contradictory efficacy in later-phase clinical trials with reasons for this discrepancy including poorly understood mechanism of MSC therapeutic action. With likelihood that a number of attributes are involved in MSC derived clinical benefit, an assay that measures a single quality of may not adequately reflect potency, thus a combination of bioassays and analytical methods, collectively called "assay matrix" are favoured for defining the potency of MSC more adequately. This chapter highlights advanced technologies and targets that can achieve quantitative measurement for a range of MSC attributes, including immunological, genomic, secretome, phosphorylation, morphological, biomaterial, angiogenic and metabolic assays.
Collapse
Affiliation(s)
- Raghavan Chinnadurai
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA.
| |
Collapse
|
43
|
Li M, Gao L, Zhao L, Zou T, Xu H. Toward the next generation of vascularized human neural organoids. Med Res Rev 2023; 43:31-54. [PMID: 35993813 DOI: 10.1002/med.21922] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/22/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Thanks to progress in the development of three-dimensional (3D) culture technologies, human central nervous system (CNS) development and diseases have been gradually deciphered by using organoids derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). Selforganized neural organoids (NOs) have been used to mimic morphogenesis and functions of specific organs in vitro. Many NOs have been reproduced in vitro, such as those mimicking the human brain, retina, and spinal cord. However, NOs fail to capitulate to the maturation and complexity of in vivo neural tissues. The persistent issues with current NO cultivation protocols are inadequate oxygen supply and nutrient diffusion due to the absence of vascular networks. In vivo, the developing CNS is interpenetrated by vasculature that not only supplies oxygen and nutrients but also provides a structural template for neuronal growth. To address these deficiencies, recent studies have begun to couple NO culture with bioengineering techniques and methodologies, including genetic engineering, coculture, multidifferentiation, microfluidics and 3D bioprinting, and transplantation, which might promote NO maturation and create more functional NOs. These cutting-edge methods could generate an ever more reliable NO model in vitro for deciphering the codes of human CNS development, disease progression, and translational application. In this review, we will summarize recent technological advances in culture strategies to generate vascularized NOs (vNOs), with a special focus on cerebral- and retinal-organoid models.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing, China
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
44
|
Bolívar-Monsalve EJ, Ceballos-González CF, Chávez-Madero C, de la Cruz-Rivas BG, Velásquez Marín S, Mora-Godínez S, Reyes-Cortés LM, Khademhosseini A, Weiss PS, Samandari M, Tamayol A, Alvarez MM, Trujillo-de Santiago G. One-Step Bioprinting of Multi-Channel Hydrogel Filaments Using Chaotic Advection: Fabrication of Pre-Vascularized Muscle-Like Tissues. Adv Healthc Mater 2022; 11:e2200448. [PMID: 35930168 DOI: 10.1002/adhm.202200448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/07/2022] [Indexed: 01/28/2023]
Abstract
The biofabrication of living constructs containing hollow channels is critical for manufacturing thick tissues. However, current technologies are limited in their effectiveness in the fabrication of channels with diameters smaller than hundreds of micrometers. It is demonstrated that the co-extrusion of cell-laden hydrogels and sacrificial materials through printheads containing Kenics static mixing elements enables the continuous and one-step fabrication of thin hydrogel filaments (1 mm in diameter) containing dozens of hollow microchannels with widths as small as a single cell. Pre-vascularized skeletal muscle-like filaments are bioprinted by loading murine myoblasts (C2C12 cells) in gelatin methacryloyl - alginate hydrogels and using hydroxyethyl cellulose as a sacrificial material. Higher viability and metabolic activity are observed in filaments with hollow multi-channels than in solid constructs. The presence of hollow channels promotes the expression of Ki67 (a proliferation biomarker), mitigates the expression of hypoxia-inducible factor 1-alpha , and markedly enhances cell alignment (i.e., 82% of muscle myofibrils aligned (in ±10°) to the main direction of the microchannels after seven days of culture). The emergence of sarcomeric α-actin is verified through immunofluorescence and gene expression. Overall, this work presents an effective and practical tool for the fabrication of pre-vascularized engineered tissues.
Collapse
Affiliation(s)
| | | | - Carolina Chávez-Madero
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Brenda Guadalupe de la Cruz-Rivas
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Silvana Velásquez Marín
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Shirley Mora-Godínez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA, 90024, USA
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, Department of Bioengineering, Department of Materials Science and Engineering, California NanoSystems Institute (CNSI), University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Mario Moisés Alvarez
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| | - Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnologico de Monterrey, Monterrey, NL, 64849, México.,Departamento de Ingeniería Mecatrónica y Eléctrica, Escuela de Ingeniería y Ciencias, Tecnologico de Monterrey, Monterrey, NL, 64849, México
| |
Collapse
|
45
|
Recent trends in bioartificial muscle engineering and their applications in cultured meat, biorobotic systems and biohybrid implants. Commun Biol 2022; 5:737. [PMID: 35869250 PMCID: PMC9307618 DOI: 10.1038/s42003-022-03593-5] [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: 07/20/2021] [Accepted: 06/16/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractRecent advances in tissue engineering and biofabrication technology have yielded a plethora of biological tissues. Among these, engineering of bioartificial muscle stands out for its exceptional versatility and its wide range of applications. From the food industry to the technology sector and medicine, the development of this tissue has the potential to affect many different industries at once. However, to date, the biofabrication of cultured meat, biorobotic systems, and bioartificial muscle implants are still considered in isolation by individual peer groups. To establish common ground and share advances, this review outlines application-specific requirements for muscle tissue generation and provides a comprehensive overview of commonly used biofabrication strategies and current application trends. By solving the individual challenges and merging various expertise, synergetic leaps of innovation that inspire each other can be expected in all three industries in the future.
Collapse
|
46
|
Guerrero J, Ghayor C, Bhattacharya I, Weber FE. Osteoconductivity of bone substitutes with filament-based microarchitectures: Influence of directionality, filament dimension, and distance. Int J Bioprint 2022; 9:626. [PMID: 36844242 PMCID: PMC9947381 DOI: 10.18063/ijb.v9i1.626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
63Additive manufacturing can be applied to produce personalized bone substitutes. At present, the major three-dimensional (3D) printing methodology relies on filament extrusion. In bioprinting, the extruded filament consists mainly of hydrogels, in which growth factors and cells are embedded. In this study, we used a lithography-based 3D printing methodology to mimic filament-based microarchitectures by varying the filament dimension and the distance between the filaments. In the first set of scaffolds, all filaments were aligned toward bone ingrowth direction. In a second set of scaffolds, which were derived from the identical microarchitecture but tilted by 90°, only 50% of the filaments were in line with the bone ingrowth direction. Testing of all tricalcium phosphate-based constructs for osteoconduction and bone regeneration was performed in a rabbit calvarial defect model. The results revealed that if all filaments are in line with the direction of bone ingrowth, filament size and distance (0.40-1.25 mm) had no significant influence on defect bridging. However, with 50% of filaments aligned, osteoconductivity declined significantly with an increase in filament dimension and distance. Therefore, for filament-based 3D- or bio-printed bone substitutes, the distance between the filaments should be 0.40 to 0.50 mm irrespective of the direction of bone ingrowth or up to 0.83 mm if perfectly aligned to it.
Collapse
Affiliation(s)
- Julien Guerrero
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zurich, 8032 Zurich, Switzerland
| | - Chafik Ghayor
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zurich, 8032 Zurich, Switzerland
| | - Indranil Bhattacharya
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zurich, 8032 Zurich, Switzerland
| | - Franz E. Weber
- Center of Dental Medicine, Oral Biotechnology & Bioengineering, University of Zurich, 8032 Zurich, Switzerland,CABMM, Center for Applied Biotechnology and Molecular Medicine, University of Zurich, 8057 Zurich, Switzerland,Corresponding author: Franz E. Weber ()
| |
Collapse
|
47
|
Advances in Biomaterials for Promoting Vascularization. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00217-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
48
|
Asrar H, Tucker AS. Endothelial cells during craniofacial development: Populating and patterning the head. Front Bioeng Biotechnol 2022; 10:962040. [PMID: 36105604 PMCID: PMC9465086 DOI: 10.3389/fbioe.2022.962040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/28/2022] [Indexed: 11/26/2022] Open
Abstract
Major organs and tissues require close association with the vasculature during development and for later function. Blood vessels are essential for efficient gas exchange and for providing metabolic sustenance to individual cells, with endothelial cells forming the basic unit of this complex vascular framework. Recent research has revealed novel roles for endothelial cells in mediating tissue morphogenesis and differentiation during development, providing an instructive role to shape the tissues as they form. This highlights the importance of providing a vasculature when constructing tissues and organs for tissue engineering. Studies in various organ systems have identified important signalling pathways crucial for regulating the cross talk between endothelial cells and their environment. This review will focus on the origin and migration of craniofacial endothelial cells and how these cells influence the development of craniofacial tissues. For this we will look at research on the interaction with the cranial neural crest, and individual organs such as the salivary glands, teeth, and jaw. Additionally, we will investigate the methods used to understand and manipulate endothelial networks during the development of craniofacial tissues, highlighting recent advances in this area.
Collapse
|
49
|
Tan XH, Liu L, Mitryashkin A, Wang Y, Goh JCH. Silk Fibroin as a Bioink - A Thematic Review of Functionalization Strategies for Bioprinting Applications. ACS Biomater Sci Eng 2022; 8:3242-3270. [PMID: 35786841 DOI: 10.1021/acsbiomaterials.2c00313] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bioprinting is an emerging tissue engineering technique that has attracted the attention of researchers around the world, for its ability to create tissue constructs that recapitulate physiological function. While the technique has been receiving hype, there are still limitations to the use of bioprinting in practical applications, much of which is due to inappropriate bioink design that is unable to recapitulate complex tissue architecture. Silk fibroin (SF) is an exciting and promising bioink candidate that has been increasingly popular in bioprinting applications because of its processability, biodegradability, and biocompatibility properties. However, due to its lack of optimum gelation properties, functionalization strategies need to be employed so that SF can be effectively used in bioprinting applications. These functionalization strategies are processing methods which allow SF to be compatible with specific bioprinting techniques. Previous literature reviews of SF as a bioink mainly focus on discussing different methods to functionalize SF as a bioink, while a comprehensive review on categorizing SF functional methods according to their potential applications is missing. This paper seeks to discuss and compartmentalize the different strategies used to functionalize SF for bioprinting and categorize the strategies for each bioprinting method (namely, inkjet, extrusion, and light-based bioprinting). By compartmentalizing the various strategies for each printing method, the paper illustrates how each strategy is better suited for a target tissue application. The paper will also discuss applications of SF bioinks in regenerating various tissue types and the challenges and future trends that SF can take in its role as a bioink material.
Collapse
Affiliation(s)
- Xuan Hao Tan
- Department of Biomedical Engineering, College of Engineering and Design, National University of Singapore, 15 Kent Ridge Crescent, E7-06-03, Singapore 119276, Singapore.,Integrative Sciences and Engineering Programme, National University of Singapore, University Hall, Tan Chin Tuan Wing, #05-03, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
| | - Ling Liu
- Department of Biomedical Engineering, College of Engineering and Design, National University of Singapore, 15 Kent Ridge Crescent, E7-06-03, Singapore 119276, Singapore
| | - Alexander Mitryashkin
- Department of Biomedical Engineering, College of Engineering and Design, National University of Singapore, 15 Kent Ridge Crescent, E7-06-03, Singapore 119276, Singapore
| | - Yunyun Wang
- Department of Biomedical Engineering, College of Engineering and Design, National University of Singapore, 15 Kent Ridge Crescent, E7-06-03, Singapore 119276, Singapore
| | - James Cho Hong Goh
- Department of Biomedical Engineering, College of Engineering and Design, National University of Singapore, 15 Kent Ridge Crescent, E7-06-03, Singapore 119276, Singapore.,Integrative Sciences and Engineering Programme, National University of Singapore, University Hall, Tan Chin Tuan Wing, #05-03, 21 Lower Kent Ridge Road, Singapore 119077, Singapore.,Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, NUHS Tower Block, Level 11, 1E Kent Ridge Road, Singapore 119288, Singapore
| |
Collapse
|
50
|
Ryma M, Genç H, Nadernezhad A, Paulus I, Schneidereit D, Friedrich O, Andelovic K, Lyer S, Alexiou C, Cicha I, Groll J. A Print-and-Fuse Strategy for Sacrificial Filaments Enables Biomimetically Structured Perfusable Microvascular Networks with Functional Endothelium Inside 3D Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2200653. [PMID: 35595711 DOI: 10.1002/adma.202200653] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/29/2022] [Indexed: 06/15/2023]
Abstract
A facile and flexible approach for the integration of biomimetically branched microvasculature within bulk hydrogels is presented. For this, sacrificial scaffolds of thermoresponsive poly(2-cyclopropyl-2-oxazoline) (PcycloPrOx) are created using melt electrowriting (MEW) in an optimized and predictable way and subsequently placed into a customized bioreactor system, which is then filled with a hydrogel precursor solution. The aqueous environment above the lower critical solution temperature (LCST) of PcycloPrOx at 25 °C swells the polymer without dissolving it, resulting in fusion of filaments that are deposited onto each other (print-and-fuse approach). Accordingly, an adequate printing pathway design results in generating physiological-like branchings and channel volumes that approximate Murray's law in the geometrical ratio between parent and daughter vessels. After gel formation, a temperature decrease below the LCST produces interconnected microchannels with distinct inlet and outlet regions. Initial placement of the sacrificial scaffolds in the bioreactors in a pre-defined manner directly yields perfusable structures via leakage-free fluid connections in a reproducible one-step procedure. Using this approach, rapid formation of a tight and biologically functional endothelial layer, as assessed not only through fluorescent dye diffusion, but also by tumor necrosis factor alpha (TNF-α) stimulation, is obtained within three days.
Collapse
Affiliation(s)
- Matthias Ryma
- Chair for Functional Materials for Medicine and Dentistry at the Institute for Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Hatice Genç
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glueckstr. 10a, 91054, Erlangen, Germany
| | - Ali Nadernezhad
- Chair for Functional Materials for Medicine and Dentistry at the Institute for Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Ilona Paulus
- Chair for Functional Materials for Medicine and Dentistry at the Institute for Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| | - Dominik Schneidereit
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulrich-Schalk-Str. 3, 91056, Erlangen, Germany
| | - Oliver Friedrich
- Institute of Medical Biotechnology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Ulrich-Schalk-Str. 3, 91056, Erlangen, Germany
| | - Kristina Andelovic
- (Chair of) Experimental Biomedicine II, University Hospital Würzburg, Josef-Schneider-Straße 2, 97080, Würzburg, Germany
| | - Stefan Lyer
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glueckstr. 10a, 91054, Erlangen, Germany
| | - Christoph Alexiou
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glueckstr. 10a, 91054, Erlangen, Germany
| | - Iwona Cicha
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-endowed Professorship for Nanomedicine, ENT Department, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Glueckstr. 10a, 91054, Erlangen, Germany
| | - Jürgen Groll
- Chair for Functional Materials for Medicine and Dentistry at the Institute for Functional Materials and Biofabrication (IFB) and Bavarian Polymer Institute (BPI), University of Würzburg, Pleicherwall 2, 97070, Würzburg, Germany
| |
Collapse
|