1
|
Dravid AA, Singh A, García AJ. Biomaterial-Based Therapeutic Delivery of Immune Cells. Adv Healthc Mater 2025; 14:e2400586. [PMID: 38813869 PMCID: PMC11607182 DOI: 10.1002/adhm.202400586] [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] [Academic Contribution Register] [Received: 02/16/2024] [Revised: 05/15/2024] [Indexed: 05/31/2024]
Abstract
Immune cell therapy (ICT) is a transformative approach used to treat a wide range of diseases including type 1 diabetes, sickle cell disease, disorders of the hematopoietic system, and certain forms of cancers. Despite excellent clinical successes, the scope of adoptively transferred immune cells is limited because of toxicities like cytokine release syndrome and immune effector cell-associated neurotoxicity in patients. Furthermore, reports suggest that such treatment can impact major organ systems including cardiac, renal, pulmonary, and hepatic systems in the long term. Additionally, adoptively transferred immune cells cannot achieve significant penetration into solid tissues, thus limiting their therapeutic potential. Recent studies suggest that biomaterial-assisted delivery of immune cells can address these challenges by reducing toxicity, improving localization, and maintaining desired phenotypes to eventually regain tissue function. In this review, recent efforts in the field of biomaterial-based immune cell delivery for the treatment of diseases, their pros and cons, and where these approaches stand in terms of clinical treatment are highlighted.
Collapse
Affiliation(s)
- Ameya A. Dravid
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Ankur Singh
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA30332USA
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Andrés J. García
- Woodruff School of Mechanical EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Petit Institute for Bioengineering and BioscienceGeorgia Institute of TechnologyAtlantaGA30332USA
| |
Collapse
|
2
|
Yang H, Xu Y, Cheong S, Xie C, Zhu Y, Xu S, Lu F, He Y. Mobilization of subcutaneous fascia contributes to the vascularization and function of acellular adipose matrix via formation of vascular matrix complex. Mater Today Bio 2025; 30:101461. [PMID: 39866780 PMCID: PMC11764388 DOI: 10.1016/j.mtbio.2025.101461] [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] [Academic Contribution Register] [Received: 08/29/2024] [Revised: 12/11/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Regenerative biomaterials are commonly used for soft-tissue repair in both pre-clinical and clinical settings, but their effectiveness is often limited by poor regenerative outcomes and volume loss. Efficient vascularization is crucial for the long-term survival and function of these biomaterials in vivo. Despite numerous pro-vascularization strategies developed over the past decades, the fundamental mechanisms of vascularization in regenerative biomaterials remain largely unexplored. In this study, we employed matrix-tracing, vessel-tracing, cell-tracing, and matrix analysis techniques, etc. to investigate the vascularization process of acellular adipose matrix (AAM) implants in a murine model. Here, we show that the mobilization of subcutaneous fascia contributes to the vascularization in AAM implants. Tracing techniques revealed that the subcutaneous fascia migrates to encase the AAM implants, bringing along fascia-embedded blood vessels, thus forming a vascular matrix complex (VMC) on the implant surface. Restricting fascia mobilization or removing fascia tissue significantly reduced AAM vascularization and hindered the regenerative process, leading to implant collapse at a later stage. Notably, VMC exhibited a dynamic matrix remodeling process closely aligned with implant vascularization. Our findings highlight the crucial role of subcutaneous fascia mobility in facilitating the vascularization of AAM implants, offering a novel direction and target for guaranteeing long-term survival and function of regenerative biomaterials in vivo.
Collapse
Affiliation(s)
- Han Yang
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yidan Xu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Sousan Cheong
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Cuiying Xie
- Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yufan Zhu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Shujie Xu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Feng Lu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yunfan He
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| |
Collapse
|
3
|
Nguyen CT, Le VP, Le TH, Kim JS, Back SH, Koo KI. Sacrificing Alginate in Decellularized Extracellular Matrix Scaffolds for Implantable Artificial Livers. J Funct Biomater 2025; 16:35. [PMID: 39852591 PMCID: PMC11766338 DOI: 10.3390/jfb16010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
This research introduced a strategy to fabricate sub-millimeter-diameter artificial liver tissue by extruding a combination of a liver decellularized extracellular matrix (dECM), alginate, endothelial cells, and hepatocytes. Vascularization remains a critical challenge in liver tissue engineering, as replicating the liver's intricate vascular network is essential for sustaining cellular function and viability. Seven scaffold groups were evaluated, incorporating different cell compositions, scaffold materials, and structural configurations. The hepatocyte and endothelial cell scaffold treated with alginate lyase demonstrated the highest diffusion rate, along with enhanced albumin secretion (2.8 µg/mL) and urea synthesis (220 µg/mL) during the same period by day 10. A dense and interconnected endothelial cell network was observed as early as day 4 in the lyased coculture group. Furthermore, three-week implantation studies in rats showed a stable integration to the host with no adverse effects. This approach offers significant potential for advancing functional liver tissue replacements, combining accelerated diffusion, enhanced albumin secretion, improved urea synthesis, dense vascular network formation, and stable implantation outcomes.
Collapse
Affiliation(s)
- Chanh-Trung Nguyen
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Van Phu Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Thi Huong Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Jeong Sook Kim
- Department of Obstetrics and Gynecology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea;
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Kyo-in Koo
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| |
Collapse
|
4
|
Sueters J, van Heiningen R, de Vries R, Guler Z, Huirne J, Smit T. Advances in tissue engineering of peripheral nerve and tissue innervation - a systematic review. J Tissue Eng 2025; 16:20417314251316918. [PMID: 39911939 PMCID: PMC11795627 DOI: 10.1177/20417314251316918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/10/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Although various options are available to treat injured organs and peripheral nerves, none is without limitations. Auto- and allografts are the first choice of treatment, but tissue survival or functionality is not guaranteed due to often limited vascular and neural networks. In response, tissue-engineered solutions have been developed, yet clinical translations is rare. In this study, a systematic review was performed on tissue-engineered advancements for peripheral nerves and tissues, to aid future developments in bridging the gap toward the clinic by identifying high-potential solutions and unexplored areas. A systematic search was performed in PubMed, Embase, Web of Science, and Scopus until November 9, 2023. Search terms involved "tissue engineering," "guided," "tissue scaffold," and "tissue graft," together with "innervation" and "reinnervation." Original in vivo or in vitro studies meeting the inclusion criteria (tissue-engineered peripheral nerve/innervation of tissue) and no exclusion criteria (no full text available; written in foreign language; nonoriginal article; tissue-engineering of central nervous system; publication before 2012; insufficient study quality or reproducibility) were assessed. A total of 68 out of 3626 original studies were included. Data extraction was based on disease model, cell origin and host species, biomaterial nature and composition, and external stimuli of biological, chemical or physical origin. Although tissue engineering is still in its infancy, explored innervation strategies of today were highlighted with respect to biomaterials, cell types, and external stimuli. The findings emphasize that natural biomaterials, pre-seeding with autologous cell sources, and solutions for reproductive organs are beneficial for future research. Natural biomaterials possess important cues required for cell-material interaction and closely resemble native tissue in terms of biomechanical, geometrical and chemical composition. Autologous cells induce biomaterial functionalization. As these solutions pose no risk of immunorejection and have demonstrated good outcomes, they are most likely to fulfill the clinical demands.
Collapse
Affiliation(s)
- Jayson Sueters
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Rowan van Heiningen
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Angiogenesis Laboratory, Cancer Center Amsterdam, Department of Medical Oncology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Vrije Universiteit, Amsterdam, The Netherlands
| | - Zeliha Guler
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Obstetrics and Gynecology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| | - Judith Huirne
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
| | - Theo Smit
- Department of Gynaecology, Amsterdam UMC – location VUmc, Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Medical Biology, Amsterdam UMC – location AMC, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Nividha, Maiti A, Parihar K, Chakraborty R, Agarwala P, Sasmal DK, Radhakrishnan R, Bhatia D, Dey KK. Enzyme-Regulated Non-Thermal Fluctuations Enhance Ligand Diffusion and Receptor-Mediated Endocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627033. [PMID: 39713409 PMCID: PMC11661126 DOI: 10.1101/2024.12.05.627033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Indexed: 12/24/2024]
Abstract
Active enzymes during catalyzing chemical reactions, have been found to generate significant mechanical fluctuations, which can influence the dynamics of their surroundings. These phenomena open new avenues for controlling mass transport in complex and dynamically inhomogeneous environments through localized chemical reactions. To explore this potential, we studied the uptake of transferrin molecules in retinal pigment epithelium (RPE) cells via clathrin-mediated endocytosis. In the presence of enzyme catalysis in the extracellular matrix, we observed a significant enhancement in the transport of fluorophore-tagged transferrin inside the cells. Fluorescence correlation spectroscopy measurements showed substantial increase in transferrin diffusion in the presence of active fluctuations. This study sheds light on the possibility that enzyme-substrate reactions within the extracellular matrix may induce long-range mechanical influences, facilitating targeted material delivery within intracellular milieu more efficiently than passive diffusion. These insights are expected to contribute to the development of better therapeutic strategies by overcoming limitations imposed by slow molecular diffusion under complex environments.
Collapse
Affiliation(s)
- Nividha
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382055, India
| | - Arnab Maiti
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382055, India
| | - Kshitiz Parihar
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Rik Chakraborty
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382055, India
| | - Pratibha Agarwala
- Department of Chemistry, Indian Institute of Technology Jodhpur, Rajasthan 342037, India
| | - Dibyendu K. Sasmal
- Department of Chemistry, Indian Institute of Technology Jodhpur, Rajasthan 342037, India
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382055, India
| | - Krishna Kanti Dey
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar, Gujarat - 382055, India
| |
Collapse
|
6
|
Lee MC, Jodat YA, Endo Y, Rodríguez-delaRosa A, Zhang T, Karvar M, Tanoury ZA, Quint J, Kamperman T, Kiaee K, Ochoa SL, Shi K, Huang Y, Rosales MP, Lee H, Kim J, Ceron EL, Reyes IG, Panayi AC, Wang X, Kim KT, Moon JI, Park SG, Lee K, Calabrese MA, Lee J, Tamayol A, Lee L, Pourquié O, Kim WJ, Sinha I, Shin SR. Engineering large-scale hiPSC-derived vessel-integrated muscle-like lattices for enhanced volumetric muscle regeneration. Trends Biotechnol 2024; 42:1715-1744. [PMID: 39306493 PMCID: PMC11625013 DOI: 10.1016/j.tibtech.2024.08.001] [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] [Academic Contribution Register] [Received: 07/04/2024] [Revised: 07/31/2024] [Accepted: 08/02/2024] [Indexed: 12/08/2024]
Abstract
Engineering biomimetic tissue implants with human induced pluripotent stem cells (hiPSCs) holds promise for repairing volumetric tissue loss. However, these implants face challenges in regenerative capability, survival, and geometric scalability at large-scale injury sites. Here, we present scalable vessel-integrated muscle-like lattices (VMLs), containing dense and aligned hiPSC-derived myofibers alongside passively perfusable vessel-like microchannels inside an endomysium-like supporting matrix using an embedded multimaterial bioprinting technology. The contractile and millimeter-long myofibers are created in mechanically tailored and nanofibrous extracellular matrix-based hydrogels. Incorporating vessel-like lattice enhances myofiber maturation in vitro and guides host vessel invasion in vivo, improving implant integration. Consequently, we demonstrate successful de novo muscle formation and muscle function restoration through a combinatorial effect between improved graft-host integration and its increased release of paracrine factors within volumetric muscle loss injury models. The proposed modular bioprinting technology enables scaling up to centimeter-sized prevascularized hiPSC-derived muscle tissues with custom geometries for next-generation muscle regenerative therapies.
Collapse
Affiliation(s)
- Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Medicinal Materials Research Center, Korea Institute of Science and Technology, Seoul, 02792 Republic of Korea
| | - Yasamin A. Jodat
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yori Endo
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alejandra Rodríguez-delaRosa
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Ting Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Mehran Karvar
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ziad Al Tanoury
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Tom Kamperman
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kiavash Kiaee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Sofia Lara Ochoa
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Kun Shi
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Montserrat Pineda Rosales
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Hyeseon Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Jiseong Kim
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Eder Luna Ceron
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Isaac Garcia Reyes
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Adriana C. Panayi
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Xichi Wang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
| | - Ki-Tae Kim
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jae-I Moon
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seung Gwa Park
- Department of Molecular Genetics, School of Dentistry and Dental Research Institute, Dental Multi-omics Center, Seoul National University, Seoul, 08826, Republic of Korea
- Epigenetic Regulation of Aged Skeleto-Muscular System Laboratory, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kangju Lee
- Department of Healthcare and Medical Engineering, Chonnam National University, Yeosu 59626, South Korea
| | - Michelle A. Calabrese
- Chemical Engineering and Materials Science Department, University of Minnesota, Minneapolis, MN 55455, USA
| | - Junmin Lee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, Lincoln, NE, 68588, USA
| | - Luke Lee
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Olivier Pourquié
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA 02115
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Harvard Stem Cell Institute, Harvard University, Boston, MA 02138
| | - Woo-Jin Kim
- Correspondence: (I.S.), (W.J.K.), (S.R.S.), Twitter: Yasamin A. Jodat: @YasaminJodat
| | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA 02139, USA
- Lead contact
| |
Collapse
|
7
|
Werschler N, Quintard C, Nguyen S, Penninger J. Engineering next generation vascularized organoids. Atherosclerosis 2024; 398:118529. [PMID: 39304390 DOI: 10.1016/j.atherosclerosis.2024.118529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/20/2024] [Revised: 05/31/2024] [Accepted: 06/21/2024] [Indexed: 09/22/2024]
Abstract
Organoids are self-organizing 3D cell culture models that are valuable for studying the mechanisms underlying both development and disease in multiple species, particularly, in humans. These 3D engineered tissues can mimic the structure and function of human organs in vitro. Methods to generate organoids have substantially improved to better resemble, in various ways, their in vivo counterpart. One of the major limitations in current organoid models is the lack of a functional vascular compartment. Here we discuss methodological approaches to generating perfusable blood vessel networks in organoid systems. Inclusion of perfused vascular compartments markedly enhances the physiological relevance of organoid systems and is a critical step in the establishment of next generation, higher-complexity in vitro systems for use in developmental, clinical, and drug-development settings.
Collapse
Affiliation(s)
- Nicolas Werschler
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada.
| | - Clement Quintard
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada
| | - Stephanie Nguyen
- University of British Columbia, School of Biomedical Engineering, Vancouver, Canada
| | - Josef Penninger
- University of British Columbia, Life Sciences Institute, Vancouver, Canada; University of British Columbia, School of Biomedical Engineering, Vancouver, Canada; University of British Columbia, Medical Genetics, Vancouver, Canada; Helmholtz Centre for Infection Research, Germany; Eric Kandel Institute, Department of Laboratory Medicine, Medical University of Vienna, Austria; IMBA Institute of Molecular Biotechnology, Vienna, Austria
| |
Collapse
|
8
|
Yang T, Wang W, Xie L, Chen S, Ye X, Shen S, Chen H, Qi L, Cui Z, Xiong W, Guo Y, Chen J. Investigating retinal explant models cultured in static and perfused systems to test the performance of exosomes secreted from retinal organoids. J Neurosci Methods 2024; 408:110181. [PMID: 38823594 DOI: 10.1016/j.jneumeth.2024.110181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/15/2024] [Revised: 05/05/2024] [Accepted: 05/22/2024] [Indexed: 06/03/2024]
Abstract
BACKGROUND Ex vivo cultures of retinal explants are appropriate models for translational research. However, one of the difficult problems of retinal explants ex vivo culture is that their nutrient supply needs cannot be constantly met. NEW METHOD This study evaluated the effect of perfused culture on the survival of retinal explants, addressing the challenge of insufficient nutrition in static culture. Furthermore, exosomes secreted from retinal organoids (RO-Exos) were stained with PKH26 to track their uptake in retinal explants to mimic the efficacy of exosomal drugs in vivo. RESULTS We found that the retinal explants cultured with perfusion exhibited significantly higher viability, increased NeuN+ cells, and reduced apoptosis compared to the static culture group at Days Ex Vivo (DEV) 4, 7, and 14. The perfusion-cultured retinal explants exhibited reduced mRNA markers for gliosis and microglial activation, along with lower expression of GFAP and Iba1, as revealed by immunostaining. Additionally, RNA-sequencing analysis showed that perfusion culture mainly upregulated genes associated with visual perception and photoreceptor cell maintenance while downregulating the immune system process and immune response. RO-Exos promoted the uptake of PKH26-labelled exosomes and the growth of retinal explants in perfusion culture. COMPARISON WITH EXISTING METHODS Our perfusion culture system can provide a continuous supply of culture medium to achieve steady-state equilibrium in retinal explant culture. Compared to traditional static culture, it better preserves the vitality, provides better neuroprotection, and reduces glial activation. CONCLUSIONS This study provides a promising ex vivo model for further studies on degenerative retinal diseases and drug screening.
Collapse
Affiliation(s)
- Tingting Yang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China; Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Wenxuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Linyao Xie
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Sihui Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiuhong Ye
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Shuhao Shen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Hang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Ling Qi
- Central Laboratory, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Zekai Cui
- Aier Eye Institute, Changsha, Hunan, China
| | - Wei Xiong
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China
| | - Yonglong Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| | - Jiansu Chen
- Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China; Institute of Ophthalmology, Medical College, Jinan University, Guangzhou, China; Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, China; Aier Eye Institute, Changsha, Hunan, China.
| |
Collapse
|
9
|
Schot M, Becker M, Paggi CA, Gomes F, Koch T, Gensheimer T, Johnbosco C, Nogueira LP, van der Meer A, Carlson A, Haugen H, Leijten J. Photoannealing of Microtissues Creates High-Density Capillary Network Containing Living Matter in a Volumetric-Independent Manner. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308949. [PMID: 38095242 DOI: 10.1002/adma.202308949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 09/01/2023] [Revised: 11/21/2023] [Indexed: 07/13/2024]
Abstract
The vascular tree is crucial for the survival and function of large living tissues. Despite breakthroughs in 3D bioprinting to endow engineered tissues with large blood vessels, there is currently no approach to engineer high-density capillary networks into living tissues in a scalable manner. Here, photoannealing of living microtissue (PALM) is presented as a scalable strategy to engineer capillary-rich tissues. Specifically, in-air microfluidics is used to produce living microtissues composed of cell-laden microgels in ultrahigh throughput, which can be photoannealed into a monolithic living matter. Annealed microtissues inherently give rise to an open and interconnected pore network within the resulting living matter. Interestingly, utilizing soft microgels enables microgel deformation, which leads to the uniform formation of capillary-sized pores. Importantly, the ultrahigh throughput nature underlying the microtissue formation uniquely facilitates scalable production of living tissues of clinically relevant sizes (>1 cm3) with an integrated high-density capillary network. In short, PALM generates monolithic, microporous, modular tissues that meet the previously unsolved need for large engineered tissues containing high-density vascular networks, which is anticipated to advance the fields of engineered organs, regenerative medicine, and drug screening.
Collapse
Affiliation(s)
- Maik Schot
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| | - Malin Becker
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| | - Carlo Alberto Paggi
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| | - Francisca Gomes
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| | - Timo Koch
- Department of Mathematics, University of Oslo, Oslo, 0316, Norway
| | - Tarek Gensheimer
- Department of Applied Stem Cell Technology, TechMed Centre, University of Twente, Enschede, 7500AE, The Netherlands
| | - Castro Johnbosco
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| | | | - Andries van der Meer
- Department of Applied Stem Cell Technology, TechMed Centre, University of Twente, Enschede, 7500AE, The Netherlands
| | - Andreas Carlson
- Department of Mathematics, University of Oslo, Oslo, 0316, Norway
| | - Håvard Haugen
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, Oslo, 0316, Norway
| | - Jeroen Leijten
- Leijten lab, Department of Developmental BioEngineering, TechMed Centre, University of Twente, Enschede, 7522AE, The Netherlands
| |
Collapse
|
10
|
Bo T, Pascucci E, Capuani S, Campa-Carranza JN, Franco L, Farina M, Secco J, Becchi S, Cavazzana R, Joubert AL, Hernandez N, Chua CYX, Grattoni A. 3D bioprinted mesenchymal stem cell laden scaffold enhances subcutaneous vascularization for delivery of cell therapy. Biomed Microdevices 2024; 26:29. [PMID: 38888669 PMCID: PMC11189315 DOI: 10.1007/s10544-024-00713-2] [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] [Academic Contribution Register] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Subcutaneous delivery of cell therapy is an appealing minimally-invasive strategy for the treatment of various diseases. However, the subdermal site is poorly vascularized making it inadequate for supporting engraftment, viability, and function of exogenous cells. In this study, we developed a 3D bioprinted scaffold composed of alginate/gelatin (Alg/Gel) embedded with mesenchymal stem cells (MSCs) to enhance vascularization and tissue ingrowth in a subcutaneous microenvironment. We identified bio-ink crosslinking conditions that optimally recapitulated the mechanical properties of subcutaneous tissue. We achieved controlled degradation of the Alg/Gel scaffold synchronous with host tissue ingrowth and remodeling. Further, in a rat model, the Alg/Gel scaffold was superior to MSC-embedded Pluronic hydrogel in supporting tissue development and vascularization of a subcutaneous site. While the scaffold alone promoted vascular tissue formation, the inclusion of MSCs in the bio-ink further enhanced angiogenesis. Our findings highlight the use of simple cell-laden degradable bioprinted structures to generate a supportive microenvironment for cell delivery.
Collapse
Affiliation(s)
- Tommaso Bo
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
| | - Elia Pascucci
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
- Department of Applied Science and Technology, Politecnico Di Torino, Turin, Italy
| | - Simone Capuani
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
| | - Jocelyn Nikita Campa-Carranza
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey, NL, Mexico
| | - Letizia Franco
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
- Department of Applied Science and Technology, Politecnico Di Torino, Turin, Italy
| | - Marco Farina
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
| | - Jacopo Secco
- Department of Electronics and Telecommunications, Politecnico Di Torino, Turin, Italy
| | - Sara Becchi
- Department of Electronics and Telecommunications, Politecnico Di Torino, Turin, Italy
| | - Rosanna Cavazzana
- Department of Electronics and Telecommunications, Politecnico Di Torino, Turin, Italy
| | - Ashley L Joubert
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
| | - Nathanael Hernandez
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
| | - Corrine Ying Xuan Chua
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX77030, , R8-111, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
11
|
Butch E, Prideaux M, Holland M, Phan JT, Trent C, Soon V, Hutchins G, Smith L. The 'bIUreactor': An Open-Source 3D Tissue Research Platform. Ann Biomed Eng 2024; 52:1678-1692. [PMID: 38532173 PMCID: PMC11082015 DOI: 10.1007/s10439-024-03481-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/21/2023] [Accepted: 02/16/2024] [Indexed: 03/28/2024]
Abstract
We developed the open-source bIUreactor research platform for studying 3D structured tissues. The versatile and modular platform allows a researcher to generate 3D tissues, culture them with oxygenated perfusion, and provide cyclic loading, all in their own lab (in laboratorium) for an all in cost of $8,000 including 3D printer, printing resin, and electronics. We achieved this by applying a design philosophy that leverages 3D printing, open-source software and hardware, and practical techniques to produce the following: 1. perfusible 3D tissues, 2. a bioreactor chamber for tissue culture, 3. a module for applying cyclic compression, 4. a peristaltic pump for providing oxygenated perfusion to 3D tissues, 5. motor control units, and 6. open-source code for running the control units. By making it widely available for researchers to investigate 3D tissue models and easy for them to use, we intend for the bIUreactor to democratize 3D tissue research, therefore increasing the pace and scale of biomedical research discoveries using 3D tissue models.
Collapse
Affiliation(s)
- Elizabeth Butch
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mark Holland
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Justin-Thuy Phan
- Smith BioFab Lab, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cole Trent
- Smith BioFab Lab, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Victor Soon
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gary Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lester Smith
- Smith BioFab Lab, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Xu Z, Wang B, Huang R, Guo M, Han D, Yin L, Zhang X, Huang Y, Li X. Efforts to promote osteogenesis-angiogenesis coupling for bone tissue engineering. Biomater Sci 2024; 12:2801-2830. [PMID: 38683241 DOI: 10.1039/d3bm02017g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/01/2024]
Abstract
Repair of bone defects exceeding a critical size has been always a big challenge in clinical practice. Tissue engineering has exhibited great potential to effectively repair the defects with less adverse effect than traditional bone grafts, during which how to induce vascularized bone formation has been recognized as a critical issue. Therefore, recently many studies have been launched to attempt to promote osteogenesis-angiogenesis coupling. This review summarized comprehensively and explored in depth current efforts to ameliorate the coupling of osteogenesis and angiogenesis from four aspects, namely the optimization of scaffold components, modification of scaffold structures, loading strategies for bioactive substances, and employment tricks for appropriate cells. Especially, the advantages and the possible reasons for every strategy, as well as the challenges, were elaborated. Furthermore, some promising research directions were proposed based on an in-depth analysis of the current research. This paper will hopefully spark new ideas and approaches for more efficiently boosting new vascularized bone formations.
Collapse
Affiliation(s)
- Zhiwei Xu
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Bingbing Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Ruoyu Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Mengyao Guo
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| | - Di Han
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Lan Yin
- Key Laboratory of Advanced Materials of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Xiaoyun Zhang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Yong Huang
- College of Lab Medicine, Hebei North University, Zhangjiakou 075000, China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, 37 Xueyuan Rd, Haidian District, Beijing, 100083, China.
| |
Collapse
|
13
|
Bupphathong S, Lim J, Fang HW, Tao HY, Yeh CE, Ku TA, Huang W, Kuo TY, Lin CH. Enhanced Vascular-like Network Formation of Encapsulated HUVECs and ADSCs Coculture in Growth Factors Conjugated GelMA Hydrogels. ACS Biomater Sci Eng 2024; 10:3306-3315. [PMID: 38634810 PMCID: PMC11094682 DOI: 10.1021/acsbiomaterials.4c00465] [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] [Academic Contribution Register] [Received: 03/09/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
Tissue engineering primarily aimed to alleviate the insufficiency of organ donations worldwide. Nonetheless, the survival of the engineered tissue is often compromised due to the complexity of the natural organ architectures, especially the vascular system inside the organ, which allows food-waste transfer. Thus, vascularization within the engineered tissue is of paramount importance. A critical aspect of this endeavor is the ability to replicate the intricacies of the extracellular matrix and promote the formation of functional vascular networks within engineered constructs. In this study, human adipose-derived stem cells (hADSCs) and human umbilical vein endothelial cells (HUVECs) were cocultured in different types of gelatin methacrylate (GelMA). In brief, pro-angiogenic signaling growth factors (GFs), vascular endothelial growth factor (VEGF165) and basic fibroblast growth factor (bFGF), were conjugated onto GelMA via an EDC/NHS coupling reaction. The GelMA hydrogels conjugated with VEGF165 (GelMA@VEGF165) and bFGF (GelMA@bFGF) showed marginal changes in the chemical and physical characteristics of the GelMA hydrogels. Moreover, the conjugation of these growth factors demonstrated improved cell viability and cell proliferation within the hydrogel construct. Additionally, vascular-like network formation was observed predominantly on GelMA@GrowthFactor (GelMA@GF) hydrogels, particularly on GelMA@bFGF. This study suggests that growth factor-conjugated GelMA hydrogels would be a promising biomaterial for 3D vascular tissue engineering.
Collapse
Affiliation(s)
- Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
- High-Value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Hsu-Wei Fang
- High-Value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Hsuan-Ya Tao
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Tian-An Ku
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Ting-Yu Kuo
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
14
|
Maiti A, Koyano Y, Kitahata H, Dey KK. Activity-induced diffusion recovery in crowded colloidal suspensions. Phys Rev E 2024; 109:054607. [PMID: 38907422 DOI: 10.1103/physreve.109.054607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/07/2023] [Accepted: 03/12/2024] [Indexed: 06/24/2024]
Abstract
We show that the forces generated by active enzyme molecules are strong enough to influence the dynamics of their surroundings under artificial crowded environments. We measured the behavior of polymer microparticles in a quasi-two-dimensional system under aqueous environment, at various area fraction values of particles. In the presence of enzymatic activity, not only was the diffusion of the suspended particles enhanced at shorter time-scales, but the system also showed a transition from subdiffusive to diffusive dynamics at longer time-scale limits. Similar observations were also recorded with enzyme-functionalized microparticles. Brownian dynamics simulations have been performed to support the experimental observations.
Collapse
Affiliation(s)
- Arnab Maiti
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat 382055, India
| | - Yuki Koyano
- Graduate School of Human Development and Environment, Kobe University, 3-11 Tsurukabuto, Nada-ku, Kobe, Hyogo 657-0011, Japan
| | - Hiroyuki Kitahata
- Department of Physics, Graduate School of Science, Chiba University, Yayoi-cho 1-33, Inage-ku, Chiba 263-8522, Japan
| | - Krishna Kanti Dey
- Laboratory of Soft and Living Materials, Department of Physics, Indian Institute of Technology Gandhinagar, Gandhinagar, Gujarat 382055, India
| |
Collapse
|
15
|
Bargavi P, Balakumar S, Raghunandhakumar S. Multi-functional bandage - bioactive glass/metal oxides/alginate composites based regenerative membrane facilitating re-epithelialization in diabetic wounds with sustained drug delivery and anti-bactericidal efficacy. Int J Biol Macromol 2024; 262:130054. [PMID: 38342258 DOI: 10.1016/j.ijbiomac.2024.130054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/17/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
Chronic wounds, especially diabetic, foot and pressure ulcers are a major health problem affecting >10 % of the world's populace. Calcium phosphate materials, particularly, bioactive glasses (BG), used as a potential material for hard and soft tissue repair. This study combines nanostructured 45S5 BG with titania (TiO2) and alumina (Al2O3) into a composite via simple sol-gel method. Prepared composites with alginate (Alg) formed a bioactive nanocomposite hydrogel membrane via freezing method. X-ray diffraction revealed formation of two phases such as Na1.8Ca1.1Si6O14 and β-Na2Ca4(PO4)2SiO4 in the silica network. Fourier transformed InfraRed spectroscopy confirmed the network formation and cross-linking between composite and alginate. <2 % hemolysis, optimal in vitro degradation and porosity was systematically evaluated up to 7 days, resulting in increasing membrane bioactivity. Significant cytocompatibility, cell migration and proliferation and a 3-4-fold increase in Collagen (Col) and Vascular Endothelial Growth Factor (VEGF) expression were obtained. Sustained delivery of 80 % Dox in 24 h and effective growth reduction of S. aureus and destruction of biofilm development against E. coli and S. aureus within 24 h. Anatomical fin regeneration, rapid re-epithelialization and wound closure were achieved within 14 days in both zebrafish and in streptozotocin (STZ) induced rat in vivo animal models with optimal blood glucose levels. Hence, the fabricated bioactive membrane can act as effective wound dressing material, for diabetic chronic infectious wounds.
Collapse
Affiliation(s)
- P Bargavi
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India; Department of Oral Pathology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, India
| | - S Balakumar
- National Centre for Nanoscience and Nanotechnology, University of Madras, Guindy Campus, Chennai 600025, Tamil Nadu, India.
| | - S Raghunandhakumar
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, India
| |
Collapse
|
16
|
He ZJ, Chu C, Dickson R, Okuda K, Cai LH. A gel-coated air-liquid-interface culture system with tunable substrate stiffness matching healthy and diseased lung tissues. Am J Physiol Lung Cell Mol Physiol 2024; 326:L292-L302. [PMID: 38252871 PMCID: PMC11280679 DOI: 10.1152/ajplung.00153.2023] [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] [Academic Contribution Register] [Received: 05/15/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Since its invention in the late 1980s, the air-liquid-interface (ALI) culture system has been the standard in vitro model for studying human airway biology and pulmonary diseases. However, in a conventional ALI system, cells are cultured on a porous plastic membrane that is much stiffer than human airway tissues. Here, we develop a gel-ALI culture system by simply coating the plastic membrane with a thin layer of hydrogel with tunable stiffness matching that of healthy and fibrotic airway tissues. We determine the optimum gel thickness that does not impair the transport of nutrients and biomolecules essential to cell growth. We show that the gel-ALI system allows human bronchial epithelial cells (HBECs) to proliferate and differentiate into pseudostratified epithelium. Furthermore, we discover that HBECs migrate significantly faster on hydrogel substrates with stiffness matching that of fibrotic lung tissues, highlighting the importance of mechanical cues in human airway remodeling. The developed gel-ALI system provides a facile approach to studying the effects of mechanical cues in human airway biology and in modeling pulmonary diseases.NEW & NOTEWORTHY In a conventional ALI system, cells are cultured on a plastic membrane that is much stiffer than human airway tissues. We develop a gel-ALI system by coating the plastic membrane with a thin layer of hydrogel with tunable stiffness matching that of healthy and fibrotic airway tissues. We discover that human bronchial epithelial cells migrate significantly faster on hydrogel substrates with pathological stiffness, highlighting the importance of mechanical cues in human airway remodeling.
Collapse
Affiliation(s)
- Zhi-Jian He
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Catherine Chu
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Riley Dickson
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States
| | - Kenichi Okuda
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina, Chapel Hill, North Carolina, United States
| | - Li-Heng Cai
- Soft Biomatter Laboratory, Department of Materials Science and Engineering, University of Virginia, Charlottesville, Virginia, United States
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, United States
| |
Collapse
|
17
|
Orlowska MK, Krycer JR, Reid JD, Mills RJ, Doran MR, Hudson JE. A miniaturized culture platform for control of the metabolic environment. BIOMICROFLUIDICS 2024; 18:024101. [PMID: 38434908 PMCID: PMC10908563 DOI: 10.1063/5.0169143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 09/18/2023] [Accepted: 02/05/2024] [Indexed: 03/05/2024]
Abstract
The heart is a metabolic "omnivore" and adjusts its energy source depending on the circulating metabolites. Human cardiac organoids, a three-dimensional in vitro model of the heart wall, are a useful tool to study cardiac physiology and pathology. However, cardiac tissue naturally experiences shear stress and nutrient fluctuations via blood flow in vivo, whilst in vitro models are conventionally cultivated in a static medium. This necessitates the regular refreshing of culture media, which creates acute cellular disturbances and large metabolic fluxes. To culture human cardiac organoids in a more physiological manner, we have developed a perfused bioreactor for cultures in a 96-well plate format. The designed bioreactor is easy to fabricate using a common culture plate and a 3D printer. Its open system allows for the use of traditional molecular biology techniques, prevents flow blockage issues, and provides easy access for sampling and cell assays. We hypothesized that a perfused culture would create more stable environment improving cardiac function and maturation. We found that lactate is rapidly produced by human cardiac organoids, resulting in large fluctuations in this metabolite under static culture. Despite this, neither medium perfusion in bioreactor culture nor lactate supplementation improved cardiac function or maturation. In fact, RNA sequencing revealed little change across the transcriptome. This demonstrates that cardiac organoids are robust in response to fluctuating environmental conditions under normal physiological conditions. Together, we provide a framework for establishing an easily accessible perfusion system that can be adapted to a range of miniaturized cell culture systems.
Collapse
|
18
|
Gapinske L, Clark L, Caro-Rivera LM, Bashir R. Cryopreservation Alters Tissue Structure and Improves Differentiation of Engineered Skeletal Muscle. Tissue Eng Part A 2023; 29:557-568. [PMID: 37463097 DOI: 10.1089/ten.tea.2023.0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 07/20/2023] Open
Abstract
Tissue-engineered skeletal muscle can play an important role in regenerative medicine, disease modeling, drug testing, as well as the actuation of biohybrid machines. As the applications of engineered muscle tissues expand, there exists a growing need to cryopreserve and store these tissues without impairing function. In a previous study, we developed a cryopreservation protocol in which engineered skeletal muscle tissues are frozen before myogenic differentiation. In that study, we found that this cryopreservation process led to a three-fold increase in the force generation of the differentiated muscle. Here, we perform further testing to determine the mechanisms by which cryopreservation enhances engineered skeletal muscle function. We found that cryopreservation alters the microstructure of the tissue by increasing pore size and decreasing elastic modulus of the extracellular matrix (ECM), which leads to increased expression of genes related to cell migration, cell-matrix adhesion, ECM secretion, and protease activity. Specifically, cryopreservation leads to the upregulation of many ECM proteins, including laminin, fibronectin, and several types of collagens, as well as integrins and matrix metalloproteinases. These changes to ECM structure and composition were associated with enhanced myogenic differentiation, as evidenced by the upregulation of late-stage myogenic markers and increased force generation. These results highlight the need to understand the effects of cryopreservation on the ECM of other tissues as we strive to advance tissue and organ cryopreservation protocols for regenerative medicine.
Collapse
Affiliation(s)
- Lauren Gapinske
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Lindsay Clark
- HPCBio, Roy J. Carver Biotechnology Center, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Lourdes Marinna Caro-Rivera
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Rashid Bashir
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
19
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
20
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
21
|
Cheng KC, Theato P, Hsu SH. 3D-bioprintable endothelial cell-laden sacrificial ink for fabrication of microvessel networks. Biofabrication 2023; 15:045026. [PMID: 37722376 DOI: 10.1088/1758-5090/acfac1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 09/20/2023]
Abstract
Although various research efforts have been made to produce a vascular-like network structure as scaffolds for tissue engineering, there are still several limitations. Meanwhile, no articles have been published on the direct embedding of cells within a glucose sensitive sacrificial hydrogel followed by three-dimensional (3D) bioprinting to fabricate vascular structures. In this study, the hydrogel composed of reversibly crosslinked poly(ethylene glycol) diacrylate and dithiothreitol with borax and branched polyethylenimine was used as the sacrificial hydrogel to fabricate vascular-like network structure. The component proportion ratio of the sacrificial hydrogel was optimized to achieve proper self-healing, injectable, glucose-sensitive, and 3D printing properties through the balance of boronate ester bond, hydrogen bond, and steric hinderance effect. The endothelial cells (ECs) can be directly embedded into sacrificial hydrogel and then bioprinted through a 110μm nozzle into the neural stem cell (NSC)-laden non-sacrificial hydrogel, forming the customized EC-laden vascularized microchannel (one-step). The EC-laden sacrificial hydrogel was dissolved immediately in the medium while cells kept growing. The ECs proliferated well within the vascularized microchannel structure and were able to migrate to the non-sacrificial hydrogel in one day. ECs and NSCs interacted around the vascularized microchannel to form capillary-like structure and vascular-like structure expressing CD31 in 14 d. The sacrificial hydrogel conveniently prepared from commercially available chemicals through simple mixing can be used in 3D bioprinting to create customized and complex but easily removable vascularized structure for tissue engineering applications.
Collapse
Affiliation(s)
- Kun-Chih Cheng
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Patrick Theato
- Institute for Chemical Technology and Polymer Chemistry, Karlsruhe Institute of Technology (KIT), Engesser Str. 18, D-76131 Karlsruhe, Germany
- Institute for Biological Interfaces III (IBG3), Hermann-von-Helmholtz-Platz 1, D-76344 Eggenstein-Leopoldshafen, Germany
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, Taipei, Taiwan, R.O.C
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan, R.O.C
| |
Collapse
|
22
|
Di Gravina GM, Loi G, Auricchio F, Conti M. Computer-aided engineering and additive manufacturing for bioreactors in tissue engineering: State of the art and perspectives. BIOPHYSICS REVIEWS 2023; 4:031303. [PMID: 38510707 PMCID: PMC10903388 DOI: 10.1063/5.0156704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/02/2023] [Accepted: 07/21/2023] [Indexed: 03/22/2024]
Abstract
Two main challenges are currently present in the healthcare world, i.e., the limitations given by transplantation and the need to have available 3D in vitro models. In this context, bioreactors are devices that have been introduced in tissue engineering as a support for facing the mentioned challenges by mimicking the cellular native microenvironment through the application of physical stimuli. Bioreactors can be divided into two groups based on their final application: macro- and micro-bioreactors, which address the first and second challenge, respectively. The bioreactor design is a crucial step as it determines the way in which physical stimuli are provided to cells. It strongly depends on the manufacturing techniques chosen for the realization. In particular, in bioreactor prototyping, additive manufacturing techniques are widely used nowadays as they allow the fabrication of customized shapes, guaranteeing more degrees of freedom. To support the bioreactor design, a powerful tool is represented by computational simulations that allow to avoid useless approaches of trial-and-error. In the present review, we aim to discuss the general workflow that must be carried out to develop an optimal macro- and micro-bioreactor. Accordingly, we organize the discussion by addressing the following topics: general and stimulus-specific (i.e., perfusion, mechanical, and electrical) requirements that must be considered during the design phase based on the tissue target; computational models as support in designing bioreactors based on the provided stimulus; manufacturing techniques, with a special focus on additive manufacturing techniques; and finally, current applications and new trends in which bioreactors are involved.
Collapse
Affiliation(s)
| | - Giada Loi
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
23
|
Yamada S, Ockermann PN, Schwarz T, Mustafa K, Hansmann J. Translation of biophysical environment in bone into dynamic cell culture under flow for bone tissue engineering. Comput Struct Biotechnol J 2023; 21:4395-4407. [PMID: 37711188 PMCID: PMC10498129 DOI: 10.1016/j.csbj.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Bone is a dynamic environment where osteocytes, osteoblasts, and mesenchymal stem/progenitor cells perceive mechanical cues and regulate bone metabolism accordingly. In particular, interstitial fluid flow in bone and bone marrow serves as a primary biophysical stimulus, which regulates the growth and fate of the cellular components of bone. The processes of mechano-sensory and -transduction towards bone formation have been well studied mainly in vivo as well as in two-dimensional (2D) dynamic cell culture platforms, which elucidated mechanically induced osteogenesis starting with anabolic responses, such as production of nitrogen oxide and prostaglandins followed by the activation of canonical Wnt signaling, upon mechanosensation. The knowledge has been now translated into regenerative medicine, particularly into the field of bone tissue engineering, where multipotent stem cells are combined with three-dimensional (3D) scaffolding biomaterials to produce transplantable constructs for bone regeneration. In the presence of 3D scaffolds, the importance of suitable dynamic cell culture platforms increases further not only to improve mass transfer inside the scaffolds but to provide appropriate biophysical cues to guide cell fate. In principle, the concept of dynamic cell culture platforms is rooted to bone mechanobiology. Therefore, this review primarily focuses on biophysical environment in bone and its translation into dynamic cell culture platforms commonly used for 2D and 3D cell expansion, including their advancement, challenges, and future perspectives. Additionally, it provides the literature review of recent empirical studies using 2D and 3D flow-based dynamic cell culture systems for bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Philipp Niklas Ockermann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Kamal Mustafa
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
- Department of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Germany
| |
Collapse
|
24
|
Abstract
Regeneration of large bone defects is a significant clinical challenge with variable success, but tissue engineering strategies are promising for rapid and effective bone regeneration. Maintaining an adequate oxygen level within implanted scaffolds is a major obstacle in bone tissue engineering. We developed a new oxygen-generating scaffold by electrospinning polycaprolactone with calcium peroxide (CaO2) nanocuboids (CPNCs) and characterized the physical, chemical, and biological properties of the resulting composites. Our scaffolds are highly porous and composed of submicron fibers that include CPNC as confirmed with XRD and FTIR analyses. Scaffolds containing CPNC provided controlled oxygen release for 14-days and supported cell proliferation while protecting preosteoblasts from hypoxia-induced cell death. Oxygen-generating scaffolds also facilitated bone mimetic defect contraction in vitro. The results suggest that our approach can be used to develop tissue-engineered products which target bone defects.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
| | - Gulden Camci-Unal
- Department of Chemical Engineering, University of Massachusetts, Lowell, Massachusetts 01854, USA.
- Department of Surgery, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA
| |
Collapse
|
25
|
Fadl A, Leask A. Hiding in Plain Sight: Human Gingival Fibroblasts as an Essential, Yet Overlooked, Tool in Regenerative Medicine. Cells 2023; 12:2021. [PMID: 37626831 PMCID: PMC10453328 DOI: 10.3390/cells12162021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/01/2023] [Revised: 08/01/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Adult human gingival fibroblasts (HGFs), the most abundant cells in the oral cavity, are essential for maintaining oral homeostasis. Compared with other tissues, adult oral mucosal wounds heal regeneratively, without scarring. Relative to fibroblasts from other locations, HGFs are relatively refractory to myofibroblast differentiation, immunomodulatory, highly regenerative, readily obtained via minimally invasive procedures, easily and rapidly expanded in vitro, and highly responsive to growth factors and cytokines. Consequently, HGFs might be a superior, yet perhaps underappreciated, source of adult mesenchymal progenitor cells to use in tissue engineering and regeneration applications, including the treatment of fibrotic auto-immune connective tissue diseases such as scleroderma. Herein, we highlight in vitro and translational studies that have investigated the regenerative and differentiation potential of HGFs, with the objective of outlining current limitations and inspiring future research that could facilitate translating the regenerative potential of HGFs into the clinic.
Collapse
Affiliation(s)
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, 105 Wiggins Road, Saskatoon, SK S7N 5A2, Canada;
| |
Collapse
|
26
|
ten Dam MJ, Frederix GW, ten Ham RM, van der Laan LJ, Schneeberger K. Toward Transplantation of Liver Organoids: From Biology and Ethics to Cost-effective Therapy. Transplantation 2023; 107:1706-1717. [PMID: 36757819 PMCID: PMC10358442 DOI: 10.1097/tp.0000000000004520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/16/2022] [Revised: 11/25/2022] [Accepted: 12/15/2022] [Indexed: 02/10/2023]
Abstract
Liver disease is a common cause of morbidity and mortality, and many patients would benefit from liver transplantation. However, because of a shortage of suitable donor livers, even of those patients who are placed on the donor liver waiting list, many do not survive the waiting time for transplantation. Therefore, alternative treatments for end-stage liver disease need to be explored. Recent advances in organoid technology might serve as a solution to overcome the donor liver shortage in the future. In this overview, we highlight the potential of organoid technology for cell therapy and tissue engineering approaches. Both organoid-based approaches could be used as treatment for end-stage liver disease patients. Additionally, organoid-based cell therapy can also be used to repair liver grafts ex vivo to increase the supply of transplantable liver tissue. The potential of both approaches to become clinically available is carefully assessed, including their clinical, ethical, and economic implications. We provide insight into what aspects should be considered further to allow alternatives to donor liver transplantation to be successfully clinically implemented.
Collapse
Affiliation(s)
- Marjolein J.M. ten Dam
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Geert W.J. Frederix
- Department of Public Health, Healthcare Innovation and Evaluation and Medical Humanities, Julius Center, Utrecht University, Utrecht, The Netherlands
| | - Renske M.T. ten Ham
- Department of Public Health, Healthcare Innovation and Evaluation and Medical Humanities, Julius Center, Utrecht University, Utrecht, The Netherlands
| | - Luc J.W. van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | - Kerstin Schneeberger
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
27
|
Licata JP, Schwab KH, Har-El YE, Gerstenhaber JA, Lelkes PI. Bioreactor Technologies for Enhanced Organoid Culture. Int J Mol Sci 2023; 24:11427. [PMID: 37511186 PMCID: PMC10380004 DOI: 10.3390/ijms241411427] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
An organoid is a 3D organization of cells that can recapitulate some of the structure and function of native tissue. Recent work has seen organoids gain prominence as a valuable model for studying tissue development, drug discovery, and potential clinical applications. The requirements for the successful culture of organoids in vitro differ significantly from those of traditional monolayer cell cultures. The generation and maturation of high-fidelity organoids entails developing and optimizing environmental conditions to provide the optimal cues for growth and 3D maturation, such as oxygenation, mechanical and fluidic activation, nutrition gradients, etc. To this end, we discuss the four main categories of bioreactors used for organoid culture: stirred bioreactors (SBR), microfluidic bioreactors (MFB), rotating wall vessels (RWV), and electrically stimulating (ES) bioreactors. We aim to lay out the state-of-the-art of both commercial and in-house developed bioreactor systems, their benefits to the culture of organoids derived from various cells and tissues, and the limitations of bioreactor technology, including sterilization, accessibility, and suitability and ease of use for long-term culture. Finally, we discuss future directions for improvements to existing bioreactor technology and how they may be used to enhance organoid culture for specific applications.
Collapse
Affiliation(s)
- Joseph P Licata
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Kyle H Schwab
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yah-El Har-El
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Jonathan A Gerstenhaber
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
28
|
Yamada S, Yassin MA, Torelli F, Hansmann J, Green JBA, Schwarz T, Mustafa K. Unique osteogenic profile of bone marrow stem cells stimulated in perfusion bioreactor is Rho-ROCK-mediated contractility dependent. Bioeng Transl Med 2023; 8:e10509. [PMID: 37206242 PMCID: PMC10189446 DOI: 10.1002/btm2.10509] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
The fate determination of bone marrow mesenchymal stem/stromal cells (BMSC) is tightly regulated by mechanical cues, including fluid shear stress. Knowledge of mechanobiology in 2D culture has allowed researchers in bone tissue engineering to develop 3D dynamic culture systems with the potential for clinical translation in which the fate and growth of BMSC are mechanically controlled. However, due to the complexity of 3D dynamic cell culture compared to the 2D counterpart, the mechanisms of cell regulation in the dynamic environment remain relatively undescribed. In the present study, we analyzed the cytoskeletal modulation and osteogenic profiles of BMSC under fluid stimuli in a 3D culture condition using a perfusion bioreactor. BMSC subjected to fluid shear stress (mean 1.56 mPa) showed increased actomyosin contractility, accompanied by the upregulation of mechanoreceptors, focal adhesions, and Rho GTPase-mediated signaling molecules. Osteogenic gene expression profiling revealed that fluid shear stress promoted the expression of osteogenic markers differently from chemically induced osteogenesis. Osteogenic marker mRNA expression, type 1 collagen formation, ALP activity, and mineralization were promoted in the dynamic condition, even in the absence of chemical supplementation. The inhibition of cell contractility under flow by Rhosin chloride, Y27632, MLCK inhibitor peptide-18, or Blebbistatin revealed that actomyosin contractility was required for maintaining the proliferative status and mechanically induced osteogenic differentiation in the dynamic culture. The study highlights the cytoskeletal response and unique osteogenic profile of BMSC in this type of dynamic cell culture, stepping toward the clinical translation of mechanically stimulated BMCS for bone regeneration.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Mohammed A. Yassin
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Francesco Torelli
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Jan Hansmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital WürzburgWürzburgGermany
- Department of Electrical EngineeringUniversity of Applied Sciences Würzburg‐SchweinfurtSchweinfurtGermany
| | - Jeremy B. A. Green
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonUK
| | - Thomas Schwarz
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
| | - Kamal Mustafa
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| |
Collapse
|
29
|
Nguyen HT, Peirsman A, Tirpakova Z, Mandal K, Vanlauwe F, Maity S, Kawakita S, Khorsandi D, Herculano R, Umemura C, Yilgor C, Bell R, Hanson A, Li S, Nanda HS, Zhu Y, Najafabadi AH, Jucaud V, Barros N, Dokmeci MR, Khademhosseini A. Engineered Vasculature for Cancer Research and Regenerative Medicine. MICROMACHINES 2023; 14:978. [PMID: 37241602 PMCID: PMC10221678 DOI: 10.3390/mi14050978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/19/2023] [Revised: 04/10/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023]
Abstract
Engineered human tissues created by three-dimensional cell culture of human cells in a hydrogel are becoming emerging model systems for cancer drug discovery and regenerative medicine. Complex functional engineered tissues can also assist in the regeneration, repair, or replacement of human tissues. However, one of the main hurdles for tissue engineering, three-dimensional cell culture, and regenerative medicine is the capability of delivering nutrients and oxygen to cells through the vasculatures. Several studies have investigated different strategies to create a functional vascular system in engineered tissues and organ-on-a-chips. Engineered vasculatures have been used for the studies of angiogenesis, vasculogenesis, as well as drug and cell transports across the endothelium. Moreover, vascular engineering allows the creation of large functional vascular conduits for regenerative medicine purposes. However, there are still many challenges in the creation of vascularized tissue constructs and their biological applications. This review will summarize the latest efforts to create vasculatures and vascularized tissues for cancer research and regenerative medicine.
Collapse
Affiliation(s)
- Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Zuzana Tirpakova
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Florian Vanlauwe
- Plastic, Reconstructive and Aesthetic Surgery, Ghent University Hospital, 9000 Ghent, Belgium
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Rondinelli Herculano
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Bioengineering & Biomaterials Group, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800-903, SP, Brazil
| | - Christian Umemura
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Remy Bell
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Adrian Hanson
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Shaopei Li
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Himansu Sekhar Nanda
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
- Biomedical Engineering and Technology Laboratory, PDPM—Indian Institute of Information Technology Design Manufacturing, Jabalpur 482005, Madhya Pradesh, India
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | - Natan Barros
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90064, USA
| |
Collapse
|
30
|
Alkildani S, Ren Y, Liu L, Rimashevskiy D, Schnettler R, Radenković M, Najman S, Stojanović S, Jung O, Barbeck M. Analyses of the Cellular Interactions between the Ossification of Collagen-Based Barrier Membranes and the Underlying Bone Defects. Int J Mol Sci 2023; 24:ijms24076833. [PMID: 37047808 PMCID: PMC10095555 DOI: 10.3390/ijms24076833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/04/2023] [Revised: 03/27/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
Barrier membranes are an essential tool in guided bone Regeneration (GBR), which have been widely presumed to have a bioactive effect that is beyond their occluding and space maintenance functionalities. A standardized calvaria implantation model was applied for 2, 8, and 16 weeks on Wistar rats to test the interactions between the barrier membrane and the underlying bone defects which were filled with bovine bone substitute materials (BSM). In an effort to understand the barrier membrane’s bioactivity, deeper histochemical analyses, as well as the immunohistochemical detection of macrophage subtypes (M1/M2) and vascular endothelial cells, were conducted and combined with histomorphometric and statistical approaches. The native collagen-based membrane was found to have ossified due to its potentially osteoconductive and osteogenic properties, forming a “bony shield” overlying the bone defects. Histomorphometrical evaluation revealed the resorption of the membranes and their substitution with bone matrix. The numbers of both M1- and M2-macrophages were significantly higher within the membrane compartments compared to the underlying bone defects. Thereby, M2-macrophages significantly dominated the tissue reaction within the membrane compartments. Statistically, a correlation between M2-macropahges and bone regeneration was only found at 2 weeks post implantationem, while the pro-inflammatory limb of the immune response correlated with the two processes at 8 weeks. Altogether, this study elaborates on the increasingly described correlations between barrier membranes and the underlying bone regeneration, which sheds a light on the understanding of the immunomodulatory features of biomaterials.
Collapse
Affiliation(s)
| | - Yanru Ren
- BerlinAnalytix GmbH, 12109 Berlin, Germany
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Luo Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100013, China
| | - Denis Rimashevskiy
- Department of Traumatology and Orthopedics, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - Reinhard Schnettler
- University Medical Centre, Justus Liebig University of Giessen, 35390 Giessen, Germany
| | - Milena Radenković
- Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Stevo Najman
- Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Sanja Stojanović
- Department for Cell and Tissue Engineering, Scientific Research Center for Biomedicine, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
- Department of Biology and Human Genetics, Faculty of Medicine, University of Niš, 18000 Niš, Serbia
| | - Ole Jung
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany
| | - Mike Barbeck
- BerlinAnalytix GmbH, 12109 Berlin, Germany
- Clinic and Policlinic for Dermatology and Venereology, University Medical Center Rostock, 18057 Rostock, Germany
| |
Collapse
|
31
|
A 3D multi-cellular tissue model of the human omentum to study the formation of ovarian cancer metastasis. Biomaterials 2023; 294:121996. [PMID: 36689832 DOI: 10.1016/j.biomaterials.2023.121996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2022] [Revised: 11/10/2022] [Accepted: 01/05/2023] [Indexed: 01/15/2023]
Abstract
Reliable and predictive experimental models are urgently needed to study metastatic mechanisms of ovarian cancer cells in the omentum. Although models for ovarian cancer cell adhesion and invasion were previously investigated, the lack of certain omental cell types, which influence the metastatic behavior of cancer cells, limits the application of these tissue models. Here, we describe a 3D multi-cellular human omentum tissue model, which considers the spatial arrangement of five omental cell types. Reproducible tissue models were fabricated combining permeable cell culture inserts and bioprinting technology to mimic metastatic processes of immortalized and patient-derived ovarian cancer cells. The implementation of an endothelial barrier further allowed studying the interaction between cancer and endothelial cells during hematogenous dissemination and the impact of chemotherapeutic drugs. This proof-of-concept study may serve as a platform for patient-specific investigations in personalized oncology in the future.
Collapse
|
32
|
Annunziata C, Fattahpour H, Fong D, Hadjiargyrou M, Sanaei P. Effects of Elasticity on Cell Proliferation in a Tissue-Engineering Scaffold Pore. Bull Math Biol 2023; 85:25. [PMID: 36826607 DOI: 10.1007/s11538-023-01134-7] [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] [Academic Contribution Register] [Received: 08/16/2022] [Accepted: 02/07/2023] [Indexed: 02/25/2023]
Abstract
Scaffolds engineered for in vitro tissue engineering consist of multiple pores where cells can migrate along with nutrient-rich culture medium. The presence of the nutrient medium throughout the scaffold pores promotes cell proliferation, and this process depends on several factors such as scaffold geometry, nutrient medium flow rate, shear stress, cell-scaffold focal adhesions and elastic properties of the scaffold material. While numerous studies have addressed the first four factors, the mathematical approach described herein focuses on cell proliferation rate in elastic scaffolds, under constant flux of nutrients. As cells proliferate, the scaffold pores radius shrinks and thus, in order to sustain the nutrient flux, the inlet applied pressure on the upstream side of the scaffold pore must be increased. This results in expansion of the elastic scaffold pore, which in turn further increases the rate of cell proliferation. Considering the elasticity of the scaffold, the pore deformation allows further cellular growth beyond that of inelastic conditions. In this paper, our objectives are as follows: (i) Develop a mathematical model for describing fluid dynamics, scaffold elasticity and cell proliferation for scaffolds consist of identical nearly cylindrical pores; (ii) Solve the models and then simulate cellular proliferation within an elastic pore. The simulation can emulate real life tissue growth in a scaffold and offer a solution which reduces the numerical burdens. Lastly, our results demonstrated are in qualitative agreement with experimental observations reported in the literature.
Collapse
Affiliation(s)
- Carlyn Annunziata
- Department of Biomedical Engineering, New York Institute of Technology, Old Westbury, NY, 11568, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Haniyeh Fattahpour
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, 30303, USA
| | - Daniel Fong
- Department of Mathematics and Science, U.S. Merchant Marine Academy, Kings Point, NY, 11024, USA
| | - Michael Hadjiargyrou
- Department of Biological and Chemical Sciences, New York Institute of Technology, Old Westbury, NY, 11568, USA
| | - Pejman Sanaei
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
33
|
Human in vitro spermatogenesis as a regenerative therapy - where do we stand? Nat Rev Urol 2023:10.1038/s41585-023-00723-4. [PMID: 36750655 DOI: 10.1038/s41585-023-00723-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 01/04/2023] [Indexed: 02/09/2023]
Abstract
Spermatogenesis involves precise temporal and spatial gene expression and cell signalling to reach a coordinated balance between self-renewal and differentiation of spermatogonial stem cells through various germ cell states including mitosis, and meiosis I and II, which result in the generation of haploid cells with a unique genetic identity. Subsequently, these round spermatids undergo a series of morphological changes to shed excess cytoplast, develop a midpiece and tail, and undergo DNA repackaging to eventually form millions of spermatozoa. The goal of recreating this process in vitro has been pursued since the 1920s as a tool to treat male factor infertility in patients with azoospermia. Continued advances in reproductive bioengineering led to successful generation of mature, functional sperm in mice and, in the past 3 years, in humans. Multiple approaches to study human in vitro spermatogenesis have been proposed, but technical and ethical obstacles have limited the ability to complete spermiogenesis, and further work is needed to establish a robust culture system for clinical application.
Collapse
|
34
|
de Silva L, Bernal PN, Rosenberg A, Malda J, Levato R, Gawlitta D. Biofabricating the vascular tree in engineered bone tissue. Acta Biomater 2023; 156:250-268. [PMID: 36041651 DOI: 10.1016/j.actbio.2022.08.051] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/28/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 01/18/2023]
Abstract
The development of tissue engineering strategies for treatment of large bone defects has become increasingly relevant, given the growing demand for bone substitutes. Native bone is composed of a dense vascular network necessary for the regulation of bone development, regeneration and homeostasis. A major obstacle in fabricating living, clinically relevant-sized bone mimics (1-10 cm3) is the limited supply of nutrients, including oxygen to the core of the construct. Therefore, strategies to support vascularization are pivotal for the development of tissue engineered bone constructs. Creating a functional bone construct integrated with a vascular network, capable of delivering the necessary nutrients for optimal tissue development is imperative for translation into the clinics. The vascular system is composed of a complex network that runs throughout the body in a tree-like hierarchical branching fashion. A significant challenge for tissue engineering approaches lies in mimicking the intricate, multi-scale structures consisting of larger vessels (macro-vessels) which interconnect with multiple sprouting vessels (microvessels) in a closed network. The advent of biofabrication has enabled complex, out of plane channels to be generated and has laid the groundwork for the creation of multi-scale vasculature in recent years. This review highlights the key state-of-the-art achievements for the development of vascular networks of varying scales in the field of biofabrication with a particular focus for its application in developing a functional tissue engineered bone construct. STATEMENT OF SIGNIFICANCE: There is a growing need for bone substitutes to overcome the limited supply of patient-derived bone. Bone tissue engineering aims to overcome this by combining stem cells with scaffolds to restore missing bone. The current bottleneck in upscaling is the lack of an integrated vascular network, required for the delivery of nutrients to cells. 3D bioprinting techniques has enabled the creation of complex hollow structures of varying dimensions that resemble native blood vessels. The convergence of multiple materials, cell types and fabrication approaches, opens the possibility of developing clinically-relevant sized vascularized bone constructs. This review provides an up-to-date insight of the technologies currently available for the generation of complex vascular networks, with a focus on their application in bone tissue engineering.
Collapse
Affiliation(s)
- Leanne de Silva
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands.
| | - Paulina N Bernal
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Ajw Rosenberg
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands
| | - Jos Malda
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands; Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, the Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, 3508 GA, the Netherlands; Regenerative Medicine Center Utrecht, Utrecht, 3584 CT, the Netherlands
| |
Collapse
|
35
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
36
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
37
|
Development of Tumor-Vasculature Interaction on Chip Mimicking Vessel Co-Option of Glioblastoma. BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00090-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/24/2022]
|
38
|
Dorison A, Forbes TA, Little MH. What can we learn from kidney organoids? Kidney Int 2022; 102:1013-1029. [PMID: 35970244 DOI: 10.1016/j.kint.2022.06.032] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 12/14/2022]
Abstract
The ability to generate 3-dimensional models of the developing human kidney via the directed differentiation of pluripotent stem cells-termed kidney organoids-has been hailed as a major advance in experimental nephrology. Although these provide an opportunity to interrogate human development, model-specific kidney diseases facilitate drug screening and even deliver bioengineered tissue; most of these prophetic end points remain to be realized. Indeed, at present we are still finding out what we can learn and what we cannot learn from this approach. In this review, we will summarize the approaches available to generate models of the human kidney from stem cells, the existing successful applications of kidney organoids, their limitations, and remaining challenges.
Collapse
Affiliation(s)
- Aude Dorison
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Thomas A Forbes
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Department of Nephrology, Royal Children's Hospital, Parkville, Melbourne, Australia
| | - Melissa H Little
- Murdoch Children's Research Institute, Parkville, Melbourne, Australia; Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Melbourne, Australia; Novo Nordisk Foundation Centre for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| |
Collapse
|
39
|
Freitas-Ribeiro S, Diogo GS, Oliveira C, Martins A, Silva TH, Jarnalo M, Horta R, Reis RL, Pirraco RP. Growth Factor-Free Vascularization of Marine-Origin Collagen Sponges Using Cryopreserved Stromal Vascular Fractions from Human Adipose Tissue. Mar Drugs 2022; 20:md20100623. [PMID: 36286447 PMCID: PMC9604698 DOI: 10.3390/md20100623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/31/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
The successful integration of transplanted three-dimensional tissue engineering (TE) constructs depends greatly on their rapid vascularization. Therefore, it is essential to address this vascularization issue in the initial design of constructs for perfused tissues. Two of the most important variables in this regard are scaffold composition and cell sourcing. Collagens with marine origins overcome some issues associated with mammal-derived collagen while maintaining their advantages in terms of biocompatibility. Concurrently, the freshly isolated stromal vascular fraction (SVF) of adipose tissue has been proposed as an advantageous cell fraction for vascularization purposes due to its highly angiogenic properties, allowing extrinsic angiogenic growth factor-free vascularization strategies for TE applications. In this study, we aimed at understanding whether marine collagen 3D matrices could support cryopreserved human SVF in maintaining intrinsic angiogenic properties observed for fresh SVF. For this, cryopreserved human SVF was seeded on blue shark collagen sponges and cultured up to 7 days in a basal medium. The secretome profile of several angiogenesis-related factors was studied throughout culture times and correlated with the expression pattern of CD31 and CD146, which showed the formation of a prevascular network. Upon in ovo implantation, increased vessel recruitment was observed in prevascularized sponges when compared with sponges without SVF cells. Immunohistochemistry for CD31 demonstrated the improved integration of prevascularized sponges within chick chorioalantoic membrane (CAM) tissues, while in situ hybridization showed human cells lining blood vessels. These results demonstrate the potential of using cryopreserved SVF combined with marine collagen as a streamlined approach to improve the vascularization of TE constructs.
Collapse
Affiliation(s)
- Sara Freitas-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Gabriela S. Diogo
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Catarina Oliveira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Albino Martins
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Tiago H. Silva
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, 4200-319 Porto, Portugal
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
| | - Rogério P. Pirraco
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Barco, 4805-017 Guimarães, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, Braga, 4710-057 Guimarães, Portugal
- Correspondence:
| |
Collapse
|
40
|
Zhang W, Rau S, Kotzagiorgis K, Rothweiler R, Nahles S, Gottwald E, Rolauffs B, Steinberg T, Nelson K, Altmann B. A matter of origin - identification of SEMA3A, BGLAP, SPP1 and PHEX as distinctive molecular features between bone site-specific human osteoblasts on transcription level. Front Bioeng Biotechnol 2022; 10:918866. [PMID: 36246375 PMCID: PMC9554416 DOI: 10.3389/fbioe.2022.918866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
In oral and maxillofacial bone reconstruction, autografts from the iliac crest represent the gold standard due to their superior clinical performance, compared to autografts derived from other extraoral regions. Thus, the aim of our study was to identify putative differences between osteoblasts derived from alveolar (hOB-A) and iliac crest (hOB-IC) bone of the same donor (nine donors) by means of their molecular properties in 2D and 3D culture. We thereby focused on the gene expression of biomarkers involved in osteogenic differentiation, matrix formation and osteoclast modulation. Furthermore, we examined the transcriptional response to Vit.D3 in hOB-A and hOB-IC. Our results revealed different modulation modes of the biomarker expression in osteoblasts, namely cell origin/bone entity-dependent, and culture configuration- and/or time-dependent modulations. SEMA3A, SPP1, BGLAP and PHEX demonstrated the strongest dependence on cell origin. With respect to Vit.D3-effects, BGLAP, SPP1 and ALPL displayed the highest Vit.D3-responsiveness. In this context we demonstrated that the transcriptional Vit.D3-response concerning SPP1 and ALPL in human osteoblasts depended on the cell origin. The results indicate a higher bone remodeling activity of iliac crest than alveolar osteoblasts and support the growing evidence that a high osteoclast activity at the host-/donor bone interface may support graft integration.
Collapse
Affiliation(s)
- Weiping Zhang
- G.E.R.N Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Oral- and Craniomaxillofacial Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sibylle Rau
- Department of Oral and Craniomaxillofacial Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Konstantinos Kotzagiorgis
- G.E.R.N Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Prosthetic Dentistry, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - René Rothweiler
- Department of Oral and Craniomaxillofacial Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susanne Nahles
- Department of Oral and Maxillofacial Surgery, Berlin Institute of Health, Corporate Member of Freie Universität Berlin, Charité - Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Eric Gottwald
- Group 3D Cell Culture Systems, Institute of Functional Interfaces, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - Bernd Rolauffs
- G.E.R.N Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thorsten Steinberg
- Department of Oral Biotechnology, Center for Dental Medicine, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katja Nelson
- Department of Oral and Craniomaxillofacial Surgery, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Brigitte Altmann
- G.E.R.N Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Prosthetic Dentistry, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- *Correspondence: Brigitte Altmann,
| |
Collapse
|
41
|
Wang Z, Tian X, Wang C, Qi X, Gracia‐Sancho J, Dong L. Transforming one organ into another to overcome challenges in tissue engineering. PORTAL HYPERTENSION & CIRRHOSIS 2022; 1:116-124. [DOI: 10.1002/poh2.26] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 04/11/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2025]
Abstract
AbstractTissue engineering (TE) is promising for the regeneration of failed organs. However, immune rejection, shortage of seed cells, and unintegrated blood vessels restrict the development and clinical application of TE. The last factor is the most challenging and intractable. Harnessing the mature blood vessel network in existing dispensable organs could be a powerful approach to effectively overcome the obstacles. After being remodeled to harbor an immunosuppressive and proregenerative niche, these potential target organs can be transformed into other organs with specific physiological functions, compensating the latter's failed native functions. Organ transformation, such as a hepatized spleen, represents an effective and encouraging TE strategy. In this review, we discuss the current development and obstacles of TE and its feasibility and superiority in organ transformation.
Collapse
Affiliation(s)
- Zhenzhen Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Xuejiao Tian
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences University of Macau Taipa Macau SAR China
| | - Xiaolong Qi
- CHESS Center, Institute of Portal Hypertension The First Hospital of Lanzhou University Lanzhou Gansu China
| | - Jordi Gracia‐Sancho
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital University of Bern Bern Switzerland
- Liver Vascular Biology Research Group IDIBAPS Research Institute, CIBEREHD Barcelona Spain
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences Nanjing University Nanjing Jiangsu China
- Chemistry and Biomedicine Innovative Center Nanjing University Nanjing Jiangsu China
| |
Collapse
|
42
|
Scalable macroporous hydrogels enhance stem cell treatment of volumetric muscle loss. Biomaterials 2022; 290:121818. [PMID: 36209578 DOI: 10.1016/j.biomaterials.2022.121818] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2022] [Revised: 08/15/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022]
Abstract
Volumetric muscle loss (VML), characterized by an irreversible loss of skeletal muscle due to trauma or surgery, is accompanied by severe functional impairment and long-term disability. Tissue engineering strategies combining stem cells and biomaterials hold great promise for skeletal muscle regeneration. However, scaffolds, including decellularized extracellular matrix (dECM), hydrogels, and electrospun fibers, used for VML applications generally lack macroporosity. As a result, the scaffolds used typically delay host cell infiltration, transplanted cell proliferation, and new tissue formation. To overcome these limitations, we engineered a macroporous dECM-methacrylate (dECM-MA) hydrogel, which we will refer to as a dECM-MA sponge, and investigated its therapeutic potential in vivo. Our results demonstrate that dECM-MA sponges promoted early cellularization, endothelialization, and establishment of a pro-regenerative immune microenvironment in a mouse VML model. In addition, dECM-MA sponges enhanced the proliferation of transplanted primary muscle stem cells, muscle tissue regeneration, and functional recovery four weeks after implantation. Finally, we investigated the scale-up potential of our scaffolds using a rat VML model and found that dECM-MA sponges significantly improved transplanted cell proliferation and muscle regeneration compared to conventional dECM scaffolds. Together, these results validate macroporous hydrogels as novel scaffolds for VML treatment and skeletal muscle regeneration.
Collapse
|
43
|
Hoyle H, Stenger C, Przyborski S. Design considerations of benchtop fluid flow bioreactors for bio-engineered tissue equivalents in vitro. BIOMATERIALS AND BIOSYSTEMS 2022; 8:100063. [PMID: 36824373 PMCID: PMC9934498 DOI: 10.1016/j.bbiosy.2022.100063] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2022] [Revised: 07/08/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022] Open
Abstract
One of the major aims of bio-engineering tissue equivalents in vitro is to create physiologically relevant culture conditions to accurately recreate the cellular microenvironment. This often includes incorporation of factors such as the extracellular matrix, co-culture of multiple cell types and three-dimensional culture techniques. These advanced techniques can recapitulate some of the properties of tissue in vivo, however fluid flow is a key aspect that is often absent. Fluid flow can be introduced into cell and tissue culture using bioreactors, which are becoming increasingly common as we seek to produce increasingly accurate tissue models. Bespoke technology is continuously being developed to tailor systems for specific applications and to allow compatibility with a range of culture techniques. For effective perfusion of a tissue culture many parameters can be controlled, ranging from impacts of the fluid flow such as increased shear stress and mass transport, to potentially unwanted side effects such as temperature fluctuations. A thorough understanding of these properties and their implications on the culture model can aid with a more accurate interpretation of results. Improved and more complete characterisation of bioreactor properties will also lead to greater accuracy when reporting culture conditions in protocols, aiding experimental reproducibility, and allowing more precise comparison of results between different systems. In this review we provide an analysis of the different factors involved in the development of benchtop flow bioreactors and their potential biological impacts across a range of applications.
Collapse
Key Words
- 3D, three-dimensional
- ABS, acrylonitrile butadiene styrene
- ALI, air-liquid interface
- Bioreactors
- CFD, computational fluid dynamics
- Cell culture
- ECM, extracellular matrix
- FDM, fused deposition modelling
- Fluid flow
- PC, polycarbonate
- PET, polyethylene terephthalate
- PLA, polylactic acid
- PTFE, polytetrafluoroethylene
- SLA, stereolithography
- Tissue engineering
- UL, unstirred layer
- UV, ultraviolet light
Collapse
Affiliation(s)
- H.W. Hoyle
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - C.M.L. Stenger
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK
| | - S.A. Przyborski
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK,NETPark Incubator, Reprocell Europe Ltd., Thomas Wright Way, Sedgefield TS21 3FD, UK,Corresponding author at: Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK.
| |
Collapse
|
44
|
Sachdeva P, M AR, Shukla R, Sahani A. A review on artificial pancreas and regenerative medicine used in the management of Type 1 diabetes mellitus. J Med Eng Technol 2022; 46:693-702. [PMID: 35801984 DOI: 10.1080/03091902.2022.2095049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023]
Abstract
Diabetes mellitus is one of the fastest-growing lifestyle disorders in the world. While numerous regimes have been developed to manage diabetes, there continue to be high numbers of diabetes-related deaths worldwide. The review gives a brief introduction to the pathology and aetiology of the disorder, different solutions developed over time with their advantages and disadvantages, and highlights the technological components and challenges of the latest technologies: artificial pancreas and regenerative medicine. The study is restricted to a set of high-quality publications from the last decade.
Collapse
Affiliation(s)
- Pallavi Sachdeva
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Ashrit R M
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Rahul Shukla
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Ashish Sahani
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| |
Collapse
|
45
|
Seiler C, Luepke M, Bach JP, Seifert H. Preparation of artificial vascularised tissue and the indirect determination of its void volume using μCT. VET MED-CZECH 2022; 67:387-394. [PMID: 39161852 PMCID: PMC11333037 DOI: 10.17221/100/2020-vetmed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/01/2020] [Accepted: 02/24/2022] [Indexed: 08/21/2024] Open
Abstract
The non-invasive determination of the vasculature volume would be very useful in many fields of medicine such as oncology and implantation. The purpose of this research was, therefore, to develop a methodology to investigate vascularisation in phantoms using microcomputed tomography (μCT) without having to visualise the single vessels. Epoxy resin and cotton candy were used to form the phantoms with microchannels. The size of the channels was measured via microscopy and the proportion of the void volume (PVV) was calculated. The phantoms were placed in contrast agent solutions of different concentrations and scanned in μCT. The mean CT numbers of the phantoms were calculated with the Amira software and displayed as a function of the determined PVV and the contrast agent concentration (CAC). The fabricated microchannels had the size of biological capillaries (diameter: 5 μm to 15 μm) and the phantoms showed a microchannel density of 5 to15 microchannels per mm². With an increasing CAC, the CT numbers increased significantly. Additionally, the phantoms with a higher PVV also had a higher CT number. The CT numbers and the PVV correlated moderately together, but significantly. The slope of the regression line increased with an increasing CAC.
Collapse
Affiliation(s)
- Christian Seiler
- Institute of General Radiology and Medical Physics, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Matthias Luepke
- Institute of General Radiology and Medical Physics, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Jan-Peter Bach
- Small Animal Clinic, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Hermann Seifert
- Institute of General Radiology and Medical Physics, University of Veterinary Medicine Foundation, Hannover, Germany
| |
Collapse
|
46
|
Smirani R, Rémy M, Devillard R, Naveau A. Use of Human Gingival Fibroblasts for Pre-Vascularization Strategies in Oral Tissue Engineering. Tissue Eng Regen Med 2022; 19:525-535. [PMID: 35048331 PMCID: PMC9130389 DOI: 10.1007/s13770-021-00415-3] [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] [Academic Contribution Register] [Received: 10/10/2021] [Revised: 11/06/2021] [Accepted: 11/22/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cocultures of human gingival fibrobasts (hGF) and endothelial cells could enhance regeneration and repair models as well as improve vascularization limitations in tissue engineering. The aim of this study was to assess if hGF could support formation of stable vessel-like networks. METHODS Explant primary hGF were isolated from gum surgical wastes collected from healthy patients with no history of periodontitis. Human umbilical vein endothelial cells (HUVEC) were two-dimensional (2D) and three-dimensional (3D) cocultured in vitro with hGF at a cell ratio of 1:1 and medium of 1:1 of their respective media during at least 31 days. Vessel quantification of HUVEC networks was performed. In order to investigate the pericyte-like properties of hGF, the expression of perivascular markers α-SMA, NG2, CD146 and PDGFR-β was studied using immunocytochemistry and flow cytometry on 2D cultures. RESULTS hGF were able to support a long-lasting HUVEC network at least 31 days, even in the absence of a bioreactor with flow. As observed, HUVEC started to communicate with each other from day 7, constructing a network. Their interconnection increased significantly between day 2 and day 21 and lasted beyond the 31 days of observation. Moreover, we tried to explain the stability of the networks obtained and showed that a small population of hGF in close vicinity of HUVEC networks expressed perivascular markers. CONCLUSION These findings highlight a new interesting property concerning hGF, accentuating their relevance in tissue engineering and periodontal regeneration. These promising results need to be confirmed using more 3D applications and in vivo testing.
Collapse
Affiliation(s)
- Rawen Smirani
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, Univ. Bordeaux, 33 076, Bordeaux, France.
| | - Murielle Rémy
- Univ. Bordeaux, CNRS, Chimie et Biologie des Membranes et des Nanoobjets (CBMN), U5248, Univ. Bordeaux, 33600, Pessac, France
| | - Raphaël Devillard
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, Univ. Bordeaux, 33 076, Bordeaux, France
| | - Adrien Naveau
- Univ. Bordeaux, INSERM, Laboratoire Bioingénierie Tissulaire (BioTis), U1026, CHU Bordeaux, Univ. Bordeaux, 33 076, Bordeaux, France
| |
Collapse
|
47
|
Yu Y, Xie R, He Y, Zhao F, Zhang Q, Wang W, Zhang Y, Hu J, Luo D, Peng W. Dual-core coaxial bioprinting of double-channel constructs with a potential for perfusion and interaction of cells. Biofabrication 2022; 14. [PMID: 35616388 DOI: 10.1088/1758-5090/ac6e88] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/13/2021] [Accepted: 05/11/2022] [Indexed: 11/11/2022]
Abstract
Coaxial bioprinting of hydrogel tubes has tremendous potential in the fabrication of highly complex large-scale vascularized structures, however, constructs with bioinks of simultaneous weak printability and perfusable networks have not been reported. Here, we report a coaxial printing method in which double-channel filaments are three-dimensional (3D) extrusion-bioprinted using a customized dual-core coaxial nozzle. The filament in one channel can perform core/shell role and the other channel can play a role in perfusion. These parallel channels within filaments are separated by an interval wall of alginate, whose thickness (∼50μm) is beneficial to supplement nutrients via perfusion. Different cell-laden hydrogels of weak mechanics were used to test the adaptability and perfusability of our method, and the results showed that dynamic perfusion maintained higher viability and functions than static culture. By combining with a bioprinter, 8-layer perfusable double-channel constructs were fabricated, and the cell viabilities gradually decreased with the reduction in nutrients and oxygen in the downstream medium. Furthermore, the double-channel filaments were tested as a platform to mimic dynamic functions between cells through sequential perfusion by using Mouse insulinoma 6 (Min6) and Hepatocellular carcinoma (HepG2) as the model cells. These results demonstrated the insulin secreted by Min6 upstream simulated and increased the uptake of glucose by the downstream HepG2 cells. In conclusion, our study provided evidence for the probability of all-in-one fabrication of 3D double-channel perfusable constructs with high simplicity, expansibility, and versability. Our strategy has significant potential for building large-scale tissue constructs for applications in tissue engineering, possibly even in drug screening and regenerative medicine.
Collapse
Affiliation(s)
- Yanrong Yu
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, School of Pharmacy, Nanchang University, Nanchang, People's Republic of China.,Jiangxi Provincal Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, People's Republic of China
| | - Renjian Xie
- Jiangxi Provincal Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, People's Republic of China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People's Republic of China.,School of Medical Information Engineering, Gannan Medical University, Ganzhou, People's Republic of China
| | - Yueteng He
- Jiangxi Provincial Key Laboratory of Basic Pharmacology, School of Pharmacy, Nanchang University, Nanchang, People's Republic of China.,Jiangxi Provincal Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, People's Republic of China
| | - Furong Zhao
- Department of Pharmacy, Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Quan Zhang
- Jiangxi Academy of Medical Science, Nanchang University, Nanchang, People's Republic of China
| | - Wei Wang
- Jiangxi Academy of Medical Science, Nanchang University, Nanchang, People's Republic of China
| | - Yong Zhang
- Jiangxi Academy of Medical Science, Nanchang University, Nanchang, People's Republic of China
| | - Jiawei Hu
- Jiangxi Provincal Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, People's Republic of China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People's Republic of China
| | - Dan Luo
- Department of Physiology, School of Basic Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Weijie Peng
- Jiangxi Provincal Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering, Gannan Medical University, Ganzhou, People's Republic of China.,Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, People's Republic of China.,Jiangxi Academy of Medical Science, Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
48
|
Sayer S, Zandrini T, Markovic M, Van Hoorick J, Van Vlierberghe S, Baudis S, Holnthoner W, Ovsianikov A. Guiding cell migration in 3D with high-resolution photografting. Sci Rep 2022; 12:8626. [PMID: 35606455 PMCID: PMC9126875 DOI: 10.1038/s41598-022-11612-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2021] [Accepted: 03/24/2022] [Indexed: 11/09/2022] Open
Abstract
Multi-photon lithography (MPL) has proven to be a suitable tool to precisely control the microenvironment of cells in terms of the biochemical and biophysical properties of the hydrogel matrix. In this work, we present a novel method, based on multi-photon photografting of 4,4′-diazido-2,2′-stilbenedisulfonic acid (DSSA), and its capabilities to induce cell alignment, directional cell migration and endothelial sprouting in a gelatin-based hydrogel matrix. DSSA-photografting allows for the fabrication of complex patterns at a high-resolution and is a biocompatible, universally applicable and straightforward process that is comparably fast. We have demonstrated the preferential orientation of human adipose-derived stem cells (hASCs) in response to a photografted pattern. Co-culture spheroids of hASCs and human umbilical vein endothelial cells (HUVECs) have been utilized to study the directional migration of hASCs into the modified regions. Subsequently, we have highlighted the dependence of endothelial sprouting on the presence of hASCs and demonstrated the potential of photografting to control the direction of the sprouts. MPL-induced DSSA-photografting has been established as a promising method to selectively alter the microenvironment of cells.
Collapse
Affiliation(s)
- Simon Sayer
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Tommaso Zandrini
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Marica Markovic
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria.,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria
| | - Jasper Van Hoorick
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Polymer Chemistry and Technology Group, Institute of Applied Synthetic Chemistry, TU Wien, Vienna, Austria
| | - Wolfgang Holnthoner
- Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.,Ludwig-Boltzmann-Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Aleksandr Ovsianikov
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien, Vienna, Austria. .,Austrian Cluster for Tissue Regeneration (https://www.tissue-regeneration.at), Vienna, Austria.
| |
Collapse
|
49
|
Parimala Chelvi Ratnamani M, Zhang X, Wang H. A Comprehensive Assessment on the Pivotal Role of Hydrogels in Scaffold-Based Bioprinting. Gels 2022; 8:gels8040239. [PMID: 35448140 PMCID: PMC9028353 DOI: 10.3390/gels8040239] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/24/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/05/2023] Open
Abstract
The past a few decades have seen exponential growth in the field of regenerative medicine. What began as extirpative (complete tissue or organ removal), with little regard to the effects of tissue loss and/or disfigurement, has evolved towards fabricating engineered tissues using personalized living cells (e.g., stem cells), and customizing a matrix or structural organization to support and guide tissue development. Biofabrication, largely accomplished through three-dimensional (3D) printing technology, provides precise, controlled, and layered assemblies of cells and biomaterials, emulating the heterogenous microenvironment of the in vivo tissue architecture. This review provides a concise framework for the bio-manufacturing process and addresses the contributions of hydrogels to biological modeling. The versatility of hydrogels in bioprinting is detailed along with an extensive elaboration of their physical, mechanical, and biological properties, as well as their assets and limitations in bioprinting. The scope of various hydrogels in tissue formation has been discussed through the case studies of biofabricated 3D constructs in order to provide the readers with a glimpse into the barrier-breaking accomplishments of biomedical sciences. In the end, the restraints of bioprinting itself are discussed, accompanied with the identification of available engineering strategies to overcome them.
Collapse
Affiliation(s)
| | - Xinping Zhang
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA;
| | - Hongjun Wang
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA;
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
- Correspondence:
| |
Collapse
|
50
|
Ze Y, Li Y, Huang L, Shi Y, Li P, Gong P, Lin J, Yao Y. Biodegradable Inks in Indirect Three-Dimensional Bioprinting for Tissue Vascularization. Front Bioeng Biotechnol 2022; 10:856398. [PMID: 35402417 PMCID: PMC8990266 DOI: 10.3389/fbioe.2022.856398] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/17/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Mature vasculature is important for the survival of bioengineered tissue constructs, both in vivo and in vitro; however, the fabrication of fully vascularized tissue constructs remains a great challenge in tissue engineering. Indirect three-dimensional (3D) bioprinting refers to a 3D printing technique that can rapidly fabricate scaffolds with controllable internal pores, cavities, and channels through the use of sacrificial molds. It has attracted much attention in recent years owing to its ability to create complex vascular network-like channels through thick tissue constructs while maintaining endothelial cell activity. Biodegradable materials play a crucial role in tissue engineering. Scaffolds made of biodegradable materials act as temporary templates, interact with cells, integrate with native tissues, and affect the results of tissue remodeling. Biodegradable ink selection, especially the choice of scaffold and sacrificial materials in indirect 3D bioprinting, has been the focus of several recent studies. The major objective of this review is to summarize the basic characteristics of biodegradable materials commonly used in indirect 3D bioprinting for vascularization, and to address recent advances in applying this technique to the vascularization of different tissues. Furthermore, the review describes how indirect 3D bioprinting creates blood vessels and vascularized tissue constructs by introducing the methodology and biodegradable ink selection. With the continuous improvement of biodegradable materials in the future, indirect 3D bioprinting will make further contributions to the development of this field.
Collapse
Affiliation(s)
- Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|