1
|
Trevizani M, Leal LL, da Silva Barros RJ, de Paoli F, Nogueira BV, Costa FF, de Aguiar JAK, da Costa Maranduba CM. Effects of decellularization on glycosaminoglycans and collagen macromolecules in bovine bone extracellular matrix. Int J Biol Macromol 2025; 307:141007. [PMID: 39971037 DOI: 10.1016/j.ijbiomac.2025.141007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/27/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
Bovine bones were decellularized to obtain extracellular matrices with potential use for Tissue Bioengineering. The objective of the present study was to develop a decellularization protocol for bovine trabecular bone while maintaining the integrity of the extracellular matrix (ECM). The protocol proved to be effective in significantly reducing the Amount of genetic material and cellular content, and it was considered innovative, being filed as a patent. The scaffold obtained showed a reduction in the content of glycosaminoglycans (GAGs) and collagen. Even with the loss of these ECM components, the material obtained can be considered an alternative for use in Tissue Engineering and Regenerative Medicine.
Collapse
Affiliation(s)
- Marizia Trevizani
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Laís Lopardi Leal
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Rodolpho José da Silva Barros
- Carlos Alberto Redins Cellular Ultrastructure Laboratory (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Flávia de Paoli
- Department of Morphology, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Breno Valentim Nogueira
- Carlos Alberto Redins Cellular Ultrastructure Laboratory (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espírito Santo, Espírito Santo, Brazil
| | - Fabiano Freire Costa
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Minas Gerais, Brazil
| | - Jair Adriano Kopke de Aguiar
- Department of Biochemistry, Glycoconjugate Analysis Laboratory, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil.
| | - Carlos Magno da Costa Maranduba
- Department of Biology, Laboratory of Human Genetics and Cell Therapy, Institute of Biological Sciences, Federal University of Juiz de Fora, Minas Gerais, Brazil.
| |
Collapse
|
2
|
Wang Z, Liang W, Ao R, An Y. Adipose Decellularized Matrix: A Promising Skeletal Muscle Tissue Engineering Material for Volume Muscle Loss. Biomater Res 2025; 29:0174. [PMID: 40248249 PMCID: PMC12003953 DOI: 10.34133/bmr.0174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/06/2025] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Volume muscle loss is a severe injury often caused by trauma, fracture, tumor resection, or degenerative disease, leading to long-term dysfunction or disability. The current gold-standard treatment is autologous muscle tissue transplantation, with limitations due to donor site restrictions, complications, and low regeneration efficiency. Tissue engineering shows potential to overcome these challenges and achieve optimal muscle regeneration, vascularization, nerve repair, and immunomodulation. In the field of muscle tissue engineering, skeletal muscle decellularized matrices are regarded as an ideal material due to their similarity to the defect site environment, yet they suffer from difficulties in preparation, severe fibrosis, and inconsistent experimental findings. Adipose decellularized matrices (AdECMs) have demonstrated consistent efficacy in promoting muscle regeneration, and their ease of preparation and abundant availability make them even more attractive. The full potential of AdECMs for muscle regeneration remains to be explored. The aim of this review is to summarize the relevant studies on using AdECMs to promote muscle regeneration, to summarize the preparation methods of various applied forms, and to analyze their advantages and shortcomings, as well as to further explore their mechanisms and to propose possible improvements, so as to provide new ideas for the clinical solution of the problem of volume muscle loss.
Collapse
Affiliation(s)
| | - Wei Liang
- Address correspondence to: (W.L.); (Y.A.)
| | - Rigele Ao
- Department of Plastic Surgery,
Peking University Third Hospital, Beijing 100191, China
| | - Yang An
- Department of Plastic Surgery,
Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
3
|
Kang S, Shi X, Chen Y, Zhang L, Liu Q, Lin Z, Lu H, Pan H. Injectable decellularized Wharton's jelly hydrogel containing CD56 + umbilical cord mesenchymal stem cell-derived exosomes for meniscus tear healing and cartilage protection. Mater Today Bio 2024; 29:101258. [PMID: 39347017 PMCID: PMC11437876 DOI: 10.1016/j.mtbio.2024.101258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Traditional meniscectomy or suture for meniscal tear usually leads to failed self-healing, cartilage degeneration and worse osteoarthritis. The strategies that facilitate the healing process of torn meniscus and safeguard knee cartilage against degeneration will be promising for clinical therapy. The CD56+ umbilical cord mesenchymal stem cells (UCSCs) (CD56+UCSCs) were sorted from Wharton's jelly using flow cytometer. Then, the modified decellularized Wharton's Jelly hydrogel (DWJH) was combined with isolated CD56+Exos from CD56+UCSCs to fabricate DWJH/CD56+Exos. The in vitro studies were performed to characterize the DWJ (decellularized Wharton's Jelly). The injectability and rheological properties were assessed by shear rate and frequency sweep analysis. The biocompatibility and chondrogenic differentiation inducibility of DWJH/CD56+Exos were performed on human bone marrow mesenchymal stem cells (hBMSCs) and RAW 264.7 cells. The release dynamics was evaluated in vitro and in vivo experiments. As for the in vivo experiments, the operated rats that subjected to a 2 mm full-thickness longitudinal tear in right medial anterior meniscus were injected a single dose of DWJH/CD56+Exos. At 4 and 8 weeks postoperatively, torn meniscus healing and articular cartilage degeneration were evaluated by hematoxylin and eosin (H&E), safranin O/fast green (SO&FG), and Sirius red staining. In in vitro experiments, the injectable DWJH/CD56+Exos demonstrated excellent biocompatibility, exosome releasing efficiency, injectable property and chondrogenic inducibility. The results of in vivo experiments revealed that DWJH/CD56+Exos degraded over time, promoted meniscal chondrogenesis, organized meniscal extracellular matrix remodeling, safeguard articular cartilage and inhibited secondary cartilage degeneration, which accelerated further facilitated torn meniscus healing. The novel injectable DWJH/CD56+Exos promoted meniscal tear healing by promoting meniscal chondrogenesis, safeguarding articular cartilage, and inhibiting secondary cartilage degeneration.
Collapse
Affiliation(s)
- Simiao Kang
- Department of Sports Medicine and Joint Arthroplasty, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Xin Shi
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Chen
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Zhang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Quanbo Liu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyang Lin
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, China
- Hunan Engineering Research Center of Sports and Health, Changsha, China
- Mobile Health Ministry of Education, China Mobile Joint Laboratory, Changsha, China
- Xiangya Hospital International Chinese Musculoskeletal Research Society Sports Medicine Research Centre, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Haile Pan
- Department of Sports Medicine and Joint Arthroplasty, Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| |
Collapse
|
4
|
Jones S, VandenHeuvel S, Luengo Martinez A, Birur R, Burgeson E, Gilbert I, Baker A, Wolf M, Raghavan SA, Rogers S, Cosgriff-Hernandez E. Suspension electrospinning of decellularized extracellular matrix: A new method to preserve bioactivity. Bioact Mater 2024; 41:640-656. [PMID: 39280898 PMCID: PMC11401211 DOI: 10.1016/j.bioactmat.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/18/2024] Open
Abstract
Decellularized extracellular matrices (dECM) have strong regenerative potential as tissue engineering scaffolds; however, current clinical options for dECM scaffolds are limited to freeze-drying its native form into sheets. Electrospinning is a versatile scaffold fabrication technique that allows control of macro- and microarchitecture. It remains challenging to electrospin dECM, which has led researchers to either blend it with synthetic materials or use enzymatic digestion to fully solubilize the dECM. Both strategies reduce the innate bioactivity of dECM and limit its regenerative potential. Herein, we developed a new suspension electrospinning method to fabricate a pure dECM fibrous mesh that retains its innate bioactivity. Systematic investigation of suspension parameters was used to identify critical rheological properties required to instill "spinnability," including homogenization, concentration, and particle size. Homogenization enhanced particle interaction to impart the requisite elastic behavior to withstand electrostatic drawing without breaking. A direct correlation between concentration and viscosity was observed that altered fiber morphology; whereas, particle size had minimal impact on suspension properties and fiber morphology. The versatility of this new method was demonstrated by electrospinning dECM with three common decellularization techniques (Abraham, Badylak, Luo) and tissue sources (intestinal submucosa, heart, skin). Bioactivity retention after electrospinning was confirmed using cell proliferation, angiogenesis, and macrophage polarization assays. Collectively, these findings provide a framework for researchers to electrospin dECM for diverse tissue engineering applications.
Collapse
Affiliation(s)
- Sarah Jones
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Sabrina VandenHeuvel
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Andres Luengo Martinez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Ruchi Birur
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Eric Burgeson
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | - Isabelle Gilbert
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Aaron Baker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Matthew Wolf
- Cancer Biomaterials Engineering Section, Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, 21702, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Simon Rogers
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Champaign, IL, 61820, USA
| | | |
Collapse
|
5
|
Ostadi Y, Khanali J, Tehrani FA, Yazdanpanah G, Bahrami S, Niazi F, Niknejad H. Decellularized Extracellular Matrix Scaffolds for Soft Tissue Augmentation: From Host-Scaffold Interactions to Bottlenecks in Clinical Translation. Biomater Res 2024; 28:0071. [PMID: 39247652 PMCID: PMC11378302 DOI: 10.34133/bmr.0071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/29/2024] [Indexed: 09/10/2024] Open
Abstract
Along with a paradigm shift in looking at soft tissue fillers from space-filling to bioactive materials, decellularized extracellular matrix (DEM) fillers have gained more attention considering their superior bioactivity. However, the complex mechanisms that govern the interaction between host tissues and DEMs have been partially understood. This review first covers the mechanisms that determine immunogenicity, angiogenesis and vasculogenesis, and recellularization and remodeling after DEM implantation into host tissue, with a particular focus on related findings from filler materials. Accordingly, the review delves into the dual role of macrophages and their M1/M2 polarization paradigm to form both constructive and destructive immune responses to DEM implants. Moreover, the contribution of macrophages in angiogenesis has been elucidated, which includes but is not limited to the secretion of angiogenic growth factors and extracellular matrix (ECM) remodeling. The findings challenge the traditional view of immune cells as solely destructive entities in biomaterials and indicate their multifaceted roles in tissue regeneration. Furthermore, the review discusses how the compositional factors of DEMs, such as the presence of growth factors and matrikines, can influence angiogenesis, cell fate, and differentiation during the recellularization process. It is also shown that the biomechanical properties of DEMs, including tissue stiffness, modulate cell responses through mechanotransduction pathways, and the structural properties of DEMs, such as scaffold porosity, impact cell-cell and cell-ECM interactions. Finally, we pointed out the current clinical applications, the bottlenecks in the clinical translation of DEM biomaterials into soft tissue fillers, as well as the naïve research areas of the field.
Collapse
Affiliation(s)
- Yasamin Ostadi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Khanali
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh A Tehrani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Yazdanpanah
- Department of Ophthalmology and Visual Sciences, Illinois Eye and Ear Infirmary, University of Illinois at Chicago, Chicago, IL, USA
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Feizollah Niazi
- Department of Plastic and Reconstructive Surgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Omidian H, Wilson RL, Dey Chowdhury S. Injectable Biomimetic Gels for Biomedical Applications. Biomimetics (Basel) 2024; 9:418. [PMID: 39056859 PMCID: PMC11274625 DOI: 10.3390/biomimetics9070418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Biomimetic gels are synthetic materials designed to mimic the properties and functions of natural biological systems, such as tissues and cellular environments. This manuscript explores the advancements and future directions of injectable biomimetic gels in biomedical applications and highlights the significant potential of hydrogels in wound healing, tissue regeneration, and controlled drug delivery due to their enhanced biocompatibility, multifunctionality, and mechanical properties. Despite these advancements, challenges such as mechanical resilience, controlled degradation rates, and scalable manufacturing remain. This manuscript discusses ongoing research to optimize these properties, develop cost-effective production techniques, and integrate emerging technologies like 3D bioprinting and nanotechnology. Addressing these challenges through collaborative efforts is essential for unlocking the full potential of injectable biomimetic gels in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (R.L.W.); (S.D.C.)
| | | | | |
Collapse
|
7
|
Mancini A, Gentile MT, Pentimalli F, Cortellino S, Grieco M, Giordano A. Multiple aspects of matrix stiffness in cancer progression. Front Oncol 2024; 14:1406644. [PMID: 39015505 PMCID: PMC11249764 DOI: 10.3389/fonc.2024.1406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
The biophysical and biomechanical properties of the extracellular matrix (ECM) are crucial in the processes of cell differentiation and proliferation. However, it is unclear to what extent tumor cells are influenced by biomechanical and biophysical changes of the surrounding microenvironment and how this response varies between different tumor forms, and over the course of tumor progression. The entire ensemble of genes encoding the ECM associated proteins is called matrisome. In cancer, the ECM evolves to become highly dysregulated, rigid, and fibrotic, serving both pro-tumorigenic and anti-tumorigenic roles. Tumor desmoplasia is characterized by a dramatic increase of α-smooth muscle actin expressing fibroblast and the deposition of hard ECM containing collagen, fibronectin, proteoglycans, and hyaluronic acid and is common in many solid tumors. In this review, we described the role of inflammation and inflammatory cytokines, in desmoplastic matrix remodeling, tumor state transition driven by microenvironment forces and the signaling pathways in mechanotransduction as potential targeted therapies, focusing on the impact of qualitative and quantitative variations of the ECM on the regulation of tumor development, hypothesizing the presence of matrisome drivers, acting alongside the cell-intrinsic oncogenic drivers, in some stages of neoplastic progression and in some tumor contexts, such as pancreatic carcinoma, breast cancer, lung cancer and mesothelioma.
Collapse
Affiliation(s)
- Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- BioUp Sagl, Lugano, Switzerland
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University “Giuseppe De Gennaro,” Casamassima, Bari, Italy
| | - Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, NA, Italy
- Sbarro Health Research Organization (S.H.R.O.) Italia Foundation ETS, Candiolo, TO, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
8
|
Zhu T, Alves SM, Adamo A, Wen X, Corn KC, Shostak A, Johnson S, Shaub ND, Martello SE, Hacker BC, D'Amore A, Bardhan R, Rafat M. Mammary tissue-derived extracellular matrix hydrogels reveal the role of irradiation in driving a pro-tumor and immunosuppressive microenvironment. Biomaterials 2024; 308:122531. [PMID: 38531198 PMCID: PMC11065579 DOI: 10.1016/j.biomaterials.2024.122531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 03/03/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Radiation therapy (RT) is essential for triple negative breast cancer (TNBC) treatment. However, patients with TNBC continue to experience recurrence after RT. The role of the extracellular matrix (ECM) of irradiated breast tissue in tumor recurrence is still unknown. In this study, we evaluated the structure, molecular composition, and mechanical properties of irradiated murine mammary fat pads (MFPs) and developed ECM hydrogels from decellularized tissues (dECM) to assess the effects of RT-induced ECM changes on breast cancer cell behavior. Irradiated MFPs were characterized by increased ECM deposition and fiber density compared to unirradiated controls, which may provide a platform for cell invasion and proliferation. ECM component changes in collagens I, IV, and VI, and fibronectin were observed following irradiation in both MFPs and dECM hydrogels. Encapsulated TNBC cell proliferation and invasive capacity was enhanced in irradiated dECM hydrogels. In addition, TNBC cells co-cultured with macrophages in irradiated dECM hydrogels induced M2 macrophage polarization and exhibited further increases in proliferation. Our study establishes that the ECM in radiation-damaged sites promotes TNBC invasion and proliferation as well as an immunosuppressive microenvironment. This work represents an important step toward elucidating how changes in the ECM after RT contribute to breast cancer recurrence.
Collapse
Affiliation(s)
- Tian Zhu
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Steven M Alves
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Arianna Adamo
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaona Wen
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin C Corn
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Anastasia Shostak
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Nicholas D Shaub
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Shannon E Martello
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Benjamin C Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Antonio D'Amore
- Ri.MED Foundation, Palermo, Italy; McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; Nanovaccine Institute, Iowa State University, Ames, IA, USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
9
|
Liang W, Han M, Li G, Dang W, Wu H, Meng X, Zhen Y, Lin W, Ao R, Hu X, An Y. Perfusable adipose decellularized extracellular matrix biological scaffold co-recellularized with adipose-derived stem cells and L6 promotes functional skeletal muscle regeneration following volumetric muscle loss. Biomaterials 2024; 307:122529. [PMID: 38489911 DOI: 10.1016/j.biomaterials.2024.122529] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/02/2024] [Accepted: 03/08/2024] [Indexed: 03/17/2024]
Abstract
Muscle tissue engineering is a promising therapeutic strategy for volumetric muscle loss (VML). Among them, decellularized extracellular matrix (dECM) biological scaffolds have shown certain effects in restoring muscle function. However, researchers have inconsistent or even contradictory results on whether dECM biological scaffolds can efficiently regenerate muscle fibers and restore muscle function. This suggests that therapeutic strategies based on dECM biological scaffolds need to be further optimized and developed. In this study, we used a recellularization method of perfusing adipose-derived stem cells (ASCs) and L6 into adipose dECM (adECM) through vascular pedicles. On one hand, this strategy ensures sufficient quantity and uniform distribution of seeded cells inside scaffold. On the other hand, auxiliary L6 cells addresses the issue of low myogenic differentiation efficiency of ASCs. Subsequently, the treatment of VML animal experiments showed that the combined recellularization strategy can improve muscle regeneration and angiogenesis than the single ASCs recellularization strategy, and the TA of former had greater muscle contraction strength. Further single-nucleus RNA sequencing (snRNA-seq) analysis found that L6 cells induced ASCs transform into a new subpopulation of cells highly expressing Mki67, CD34 and CDK1 genes, which had stronger ability of oriented myogenic differentiation. This study demonstrates that co-seeding ASCs and L6 cells through vascular pedicles is a promising recellularization strategy for adECM biological scaffolds, and the engineered muscle tissue constructed based on this has significant therapeutic effects on VML. Overall, this study provides a new paradigm for optimizing and developing dECM-based therapeutic strategies.
Collapse
Affiliation(s)
- Wei Liang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Meng Han
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Guan Li
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Wanwen Dang
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Huiting Wu
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoyu Meng
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Yonghuan Zhen
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Weibo Lin
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Rigele Ao
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Beijing, 100191, China.
| | - Yang An
- Department of Plastic Surgery, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
10
|
Teixeira Polez R, Huynh N, Pridgeon CS, Valle-Delgado JJ, Harjumäki R, Österberg M. Insights into spheroids formation in cellulose nanofibrils and Matrigel hydrogels using AFM-based techniques. Mater Today Bio 2024; 26:101065. [PMID: 38706731 PMCID: PMC11066555 DOI: 10.1016/j.mtbio.2024.101065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/30/2024] [Accepted: 04/15/2024] [Indexed: 05/07/2024] Open
Abstract
The recent FDA decision to eliminate animal testing requirements emphasises the role of cell models, such as spheroids, as regulatory test alternatives for investigations of cellular behaviour, drug responses, and disease modelling. The influence of environment on spheroid formation are incompletely understood, leading to uncertainty in matrix selection for scaffold-based 3D culture. This study uses atomic force microscopy-based techniques to quantify cell adhesion to Matrigel and cellulose nanofibrils (CNF), and cell-cell adhesion forces, and their role in spheroid formation of hepatocellular carcinoma (HepG2) and induced pluripotent stem cells (iPS(IMR90)-4). Results showed different cell behaviour in CNF and Matrigel cultures. Both cell lines formed compact spheroids in CNF but loose cell aggregates in Matrigel. Interestingly, the type of cell adhesion protein, and not the bond strength, appeared to be a key factor in the formation of compact spheroids. The gene expression of E- and N-cadherins, proteins on cell membrane responsible for cell-cell interactions, was increased in CNF culture, leading to formation of compact spheroids while Matrigel culture induced integrin-laminin binding and downregulated E-cadherin expression, resulting in looser cell aggregates. These findings enhance our understanding of cell-biomaterial interactions in 3D cultures and offer insights for improved 3D cell models, culture biomaterials, and applications in drug research.
Collapse
Affiliation(s)
- Roberta Teixeira Polez
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Ngoc Huynh
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Chris S. Pridgeon
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Juan José Valle-Delgado
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| | - Riina Harjumäki
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00790, Helsinki, Finland
| | - Monika Österberg
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, FI-00076, Aalto, Finland
| |
Collapse
|
11
|
Cicuéndez M, García-Lizarribar A, Casarrubios L, Feito MJ, Fernández-San-Argimiro FJ, García-Urkia N, Murua O, Madarieta I, Olalde B, Diez-Orejas R, Portolés MT. Functionality of macrophages encapsulated in porcine decellularized adipose matrix hydrogels and interaction with Candida albicans. BIOMATERIALS ADVANCES 2024; 159:213794. [PMID: 38367317 DOI: 10.1016/j.bioadv.2024.213794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/19/2024]
Abstract
Extracellular matrix hydrogels are considered one of the most suitable biomaterials for tissue regeneration due to their similarity with the extracellular microenvironment of the native tissue. Their properties are dependent on their composition, material concentration, fiber density and the fabrication approaches, among other factors. The encapsulation of immune cells in this kind of hydrogels, both in absence or presence of a pathogen, represents a promising strategy for the development of platforms that mimic healthy and infected tissues, respectively. In this work, we have encapsulated macrophages in 3D hydrogels of porcine decellularized adipose matrices (pDAMs) without and with the Candida albicans fungus, as 3D experimental models to study the macrophage immunocompetence in a closer situation to the physiological conditions and to mimic an infection scenario. Our results indicate that encapsulated macrophages preserve their functionality within these pDAM hydrogels and phagocytose live pathogens. In addition, their behavior is influenced by the hydrogel pore size, inversely related to the hydrogel concentration. Thus, larger pore size promotes the polarization of macrophages towards M2 phenotype along the time and enhances their phagocytosis capability. It is important to point out that encapsulated macrophages in absence of pathogen showed an M2 phenotype, but macrophages coencapsulated with C. albicans can switch towards an M1 inflammatory phenotype to resolve the infection, depending on the fungus quantity. The present study reveals that pDAM hydrogels preserve the macrophage plasticity, demonstrating their relevance as new models for macrophage-pathogen interaction studies that mimic an infection scenario with application in regenerative medicine research.
Collapse
Affiliation(s)
- Mónica Cicuéndez
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Laura Casarrubios
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María José Feito
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | | | - Nerea García-Urkia
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Olatz Murua
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Iratxe Madarieta
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Beatriz Olalde
- TECNALIA, Basque Research and Technology Alliance (BRTA), E20009 Donostia-San Sebastian, Spain
| | - Rosalía Diez-Orejas
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain.
| | - María Teresa Portolés
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, 28040 Madrid, Spain.
| |
Collapse
|
12
|
Gadre M, Kasturi M, Agarwal P, Vasanthan KS. Decellularization and Their Significance for Tissue Regeneration in the Era of 3D Bioprinting. ACS OMEGA 2024; 9:7375-7392. [PMID: 38405516 PMCID: PMC10883024 DOI: 10.1021/acsomega.3c08930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/19/2023] [Accepted: 01/10/2024] [Indexed: 02/27/2024]
Abstract
Three-dimensional bioprinting is an emerging technology that has high potential application in tissue engineering and regenerative medicine. Increasing advancement and improvement in the decellularization process have led to an increase in the demand for using a decellularized extracellular matrix (dECM) to fabricate tissue engineered products. Decellularization is the process of retaining the extracellular matrix (ECM) while the cellular components are completely removed to harvest the ECM for the regeneration of various tissues and across different sources. Post decellularization of tissues and organs, they act as natural biomaterials to provide the biochemical and structural support to establish cell communication. Selection of an effective method for decellularization is crucial, and various factors like tissue density, geometric organization, and ECM composition affect the regenerative potential which has an impact on the end product. The dECM is a versatile material which is added as an important ingredient to formulate the bioink component for constructing tissue and organs for various significant studies. Bioink consisting of dECM from various sources is used to generate tissue-specific bioink that is unique and to mimic different biometric microenvironments. At present, there are many different techniques applied for decellularization, and the process is not standardized and regulated due to broad application. This review aims to provide an overview of different decellularization procedures, and we also emphasize the different dECM-derived bioinks present in the current global market and the major clinical outcomes. We have also highlighted an overview of benefits and limitations of different decellularization methods and various characteristic validations of decellularization and dECM-derived bioinks.
Collapse
Affiliation(s)
- Mrunmayi Gadre
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Meghana Kasturi
- Department
of Mechanical Engineering, University of
Michigan, Dearborn, Michigan 48128, United States
| | - Prachi Agarwal
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Kirthanashri S. Vasanthan
- Manipal
Centre for Biotherapeutics Research, Manipal
Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
13
|
Qi J, Li Z, Li S, Fu S, Luan J. Effectiveness of a New Enzyme-Free Method for the Preparation of a Decellularized Adipose-Derived Matrix. Aesthet Surg J 2024; 44:NP184-NP192. [PMID: 37715728 DOI: 10.1093/asj/sjad307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/02/2023] [Accepted: 09/14/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Decellularized adipose-derived matrix (DAM) represents a new alternative to tissue fillers. The function of DAM is closely associated with the decellularization technique used for its preparation. However, most techniques are time-consuming and expensive, and this might reduce the popularity of DAM. OBJECTIVES The study aimed to investigate an enzyme-free adipose decellularization method and generate a DAM capable of adipose tissue regeneration. METHODS DAMs prepared by the enzyme-free and Flynn's methods were compared and co-cultured with human adipose-derived stem cells (hADSCs) to investigate cytocompatibility. Adipose tissue formation was evaluated by injecting the DAMs into the backs of nude mice over 4 weeks. Samples were harvested for gross and perilipin immunohistochemistry analysis at 1 and 4 weeks. RESULTS The enzyme-free method is effective for adipose decellularization because it removes adipocytes and preserves the microstructure. In vitro, the DAM made by the enzyme-free method could support the attachment, growth, proliferation, and differentiation of hADSCs, and promote the enhanced secretion of vascular endothelial growth factor by hADSCs; this DAM also induced the formation and maturity of adipocytes in vivo. CONCLUSIONS This study describes a highly effective enzyme-free method for adipose tissue decellularization that also promotes adipocyte formation and adipose tissue volume stability in vitro and in vivo, resulting in a new alternative tissue filler.
Collapse
|
14
|
Yang J, Tang J, Dang J, Rong X, Wang K, Zhang Z, Hou M, Yu Z, Yi C. Bioactive decellularized adipose matrix prepared using a rapid, nonchemical/enzymatic method for adipogenesis. Biotechnol Bioeng 2024; 121:157-175. [PMID: 37691171 DOI: 10.1002/bit.28547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/12/2023]
Abstract
Recent developments in the field of regenerative surgeries and medical applications have led to a renewed interest in adipose tissue-enriched mesenchymal stem cell scaffolds. Various advantages declared for the decellularized adipose matrix (DAM) have caused its extensive use in the transfer of stem cells or growth factors for soft tissue regeneration induction. Meanwhile, the long-term application of detergents toward DAM regeneration has been assumed as a risky obstacle in this era. Herein, a rapid, mechanical protocol was developed to prepare DAM (M-DAM) without chemicals/enzymes and was comprehensively compared with the ordinary DAM (traditional chemical method). Accordingly, this method could effectively hinder oils and cells, sustain the structural and biological elements, and contain a superior level of collagen content. In addition, more protein numbers, as well as higher basement membrane elements, glycoproteins, and extracellular matrix-related proteins were detected in the regenerated M-DAM. Also, superior adipogenesis and angiogenesis proteins were distinguished. The noncytotoxicity of the M-DAM was also approved, and a natural ecological niche was observed for the proliferation and differentiation of stem cells, confirming its great potential for vascularization and adipogenesis in vivo. The suggested technique could effectively prepare the modified DAM in variant constructions of tablets, powders, emulsions, hydrogels, and different three-dimensional-printed structures. Hence, this rapid, mechanical process can produce bioactive DAM, which has the potential to be widely used in various research fields of regenerative medicine.
Collapse
Affiliation(s)
- Jizhong Yang
- Department of Plastic Surgery, The Second Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Jiezhang Tang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Juanli Dang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiangke Rong
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Kai Wang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Zhaoxiang Zhang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mengmeng Hou
- Department of Plastic Surgery, The Second Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, China
| | - Zhou Yu
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chenggang Yi
- Department of Plastic Surgery, The Second Affiliated Hospital, Medical School, Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Gujjar S, Tyagi A, Sainger S, Bharti P, Nain V, Sood P, Jayabal P, Sharma JC, Sharma P, Rajput S, Pandey AK, Pandey AK, Abnave P, Mathapati S. Biocompatible Human Placental Extracellular Matrix Derived Hydrogels. Adv Biol (Weinh) 2024; 8:e2300349. [PMID: 37786307 DOI: 10.1002/adbi.202300349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/29/2023] [Indexed: 10/04/2023]
Abstract
Solubilizing extracellular matrix (ECM) materials and transforming them into hydrogels has expanded their potential applications both in vitro and in vivo. In this study, hydrogels are prepared by decellularization of human placental tissue using detergent and enzymes and by the subsequent creation of a homogenized acellular placental tissue powder (P-ECM). A perfusion-based decellularization approach is employed using detergent and enzymes. The P-ECM with and without gamma irradiation is then utilized to prepare P-ECM hydrogels. Physical and biological evaluations are conducted to assess the suitability of the P-ECM hydrogels for biocompatibility. The decellularized tissue has significantly reduced cellular content and retains the major ECM proteins. Increasing the concentration of P-ECM leads to improved mechanical properties of the P-ECM hydrogels. The biocompatibility of the P-ECM hydrogel is demonstrated through cell proliferation and viability assays. Notably, gamma-sterilized P-ECM does not support the formation of a stable hydrogel. Nonetheless, the use of HCl during the digestion process effectively decreases spore growth and bacterial bioburden. The study demonstrates that P-ECM hydrogels exhibit physical and biological attributes conducive to soft tissue reconstruction. These hydrogels establish a favorable microenvironment for cell growth and the need for investigating innovative sterilization methods.
Collapse
Affiliation(s)
- Sunil Gujjar
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Anurag Tyagi
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Saloni Sainger
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Puja Bharti
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Vaibhav Nain
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Pratibha Sood
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Prakash Jayabal
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Jagadish Chandra Sharma
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Priyanka Sharma
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Sanjay Rajput
- Shriram Institute for Industrial Research, Delhi, 110007, India
| | - Anil Kumar Pandey
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, 121012, India
| | - Amit Kumar Pandey
- Mycobacterial Pathogenesis Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| | - Prasad Abnave
- National Centre for Cell Science, Pune, Maharashtra, 411007, India
| | - Santosh Mathapati
- Biomaterials Laboratory, Translational Health Science and Technology Institute, Faridabad, Haryana, 121001, India
| |
Collapse
|
16
|
Kim S, Kim C, Lee K. Hydrogels as filler materials. HYDROGELS FOR TISSUE ENGINEERING AND REGENERATIVE MEDICINE 2024:413-432. [DOI: 10.1016/b978-0-12-823948-3.00005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
17
|
Zhai X, Tao X, Wu Y, Jin K, Tan H, Zhou T, Chen Y. Injectable and Self-Adaptive Gel Scaffold Based on Heparin Microspheres for Adipogenesis of Human Adipose-Derived Stem Cells. Biomacromolecules 2023; 24:4663-4671. [PMID: 37722066 DOI: 10.1021/acs.biomac.3c00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
An injectable and self-adaptive heparin microsphere-based cell scaffold was developed to achieve adipose regeneration. Simultaneously, the cell scaffold exhibited a dynamic architecture, self-regulated glucose levels, sustained insulin delivery, and steady viscoelastic properties for adipogenesis. The dynamic cell scaffold is cross-linked by the boronate-diol interaction among heparin-based microspheres, which have boronate and maltose groups. Because of the boronate-maltose ester bonds, the gelatinous complex would be partially dismantled and readily display glucose-sensitive performance by free glucose via competitive displacement. The dynamic cross-linking heparin microsphere scaffold can deliver the lipogenic drug insulin to enhance lipid filling, which has an impact on fat tissue enhancement. A 4-week in vitro cell culture demonstrated that the dynamic heparin microsphere-based cell scaffold, through loading with insulin, showed significantly higher efficiency in promoting ASC differentiation compared with traditional 3D culture methods. In vivo histological results further demonstrated that there was a significant increase in adipose in the proposed cell scaffold, which proved to be statistically significant compared with traditional biomaterials. Notable stain expression of the FABP4 and PPAR-γ genes was also observed in the dynamic cell scaffold containing insulin, which was more similar to natural fat.
Collapse
Affiliation(s)
- Xinyue Zhai
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Xinwei Tao
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yuqian Wu
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Kesun Jin
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Huaping Tan
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Tianle Zhou
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yong Chen
- Department of Orthopaedics, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
18
|
Turan Sorhun D, Kuşoğlu A, Öztürk E. Developing Bovine Brain-Derived Extracellular Matrix Hydrogels: a Screen of Decellularization Methods for Their Impact on Biochemical and Mechanical Properties. ACS OMEGA 2023; 8:36933-36947. [PMID: 37841171 PMCID: PMC10569007 DOI: 10.1021/acsomega.3c04064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Tissue models that recapitulate the key biochemical and physical aspects of the brain have been highly pursued in neural tissue engineering. Decellularization of native organs offers the advantage of preserving the composition of native extracellular matrix (ECM). Brain ECM has distinct features which play a major role in neural cell behavior. Cell instructive ligands and mechanical properties take part in the regulation of cellular processes in homeostasis and diseases. One of the main challenges in decellularization is maintaining mechanical integrity in reconstituted hydrogels and achieving physiologically relevant stiffness. The effect of the decellularization process on different mechanical aspects, particularly the viscoelasticity of brain-derived hydrogels, has not been addressed. In this study, we developed bovine brain-derived hydrogels for the first time. We pursued seven protocols for decellularization and screened their effect on biochemical content, hydrogel formation, and mechanical characteristics. We show that bovine brain offers an easily accessible alternative for in vitro brain tissue modeling. Our data demonstrate that the choice of decellularization method strongly alters gelation as well as the stiffness and viscoelasticity of the resulting hydrogels. Lastly, we investigated the cytocompatibility of brain ECM hydrogels and the effect of modulated mechanical properties on the growth and morphological features of neuroblastoma cells.
Collapse
Affiliation(s)
- Duygu Turan Sorhun
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Alican Kuşoğlu
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Ece Öztürk
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Department
of Medical Biology, School of Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
19
|
Nasiraee MR, Shahrivari S, Sayad S, Mahdavi H, Saraygord‐Afshari N, Bagheri Z. An agarose-alginate microfluidic device for the study of spheroid invasion, ATRA inhibits CAFs-mediated matrix remodeling. Cytotechnology 2023; 75:309-323. [PMID: 37389131 PMCID: PMC10299977 DOI: 10.1007/s10616-023-00578-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/31/2023] [Indexed: 07/01/2023] Open
Abstract
UNLABELLED Growing evidence demonstrates that cancer-associated fibroblasts (CAF) are responsible for tumor genesis, growth, metastasis, and treatment response. Therefore, targeting these cells may contribute to tumor control. It has been proposed that targeting key molecules and pathways of proliferative functions can be more effective than killing CAFs. In this regard, multicellular aggregates, like spheroids, can be used as human tumor models. Spheroids closely resemble human tumors and mimic many of their features. Microfluidic systems are ideal for cultivation and study of spheroids. These systems can be designed with different biological and synthetic matrices in order to have a more realistic simulation of the tumor microenvironment (TME). In this study, we investigated the effect of all-trans retinoic acid (ATRA) on 3D spheroid invasion of MDA-MB cells exposed to hydrogel matrix derived from CAFs. The number of invasive cells significantly decreased in CAF-ECM hydrogel treated with ATRA (p < 0.05), which indicates that ATRA could be effective for CAFs normalization. This experiment was done using an agarose-alginate microfluidic chip. As compared with common methods, such hydrogel casting is an easier method for chip fabrication and can even reduce costs. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s10616-023-00578-y.
Collapse
Affiliation(s)
- Mohammad Reza Nasiraee
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Shabnam Shahrivari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Soheila Sayad
- Department of Surgery, Firoozgar Clinical Research Development Center (FCRDC), Iran University of Medical Sciences, Tehran, Iran
| | - Hoda Mahdavi
- Department of Radiation Oncology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Saraygord‐Afshari
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Zeinab Bagheri
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University G.C, Tehran, 19839-69411 Iran
| |
Collapse
|
20
|
Zhang GK, Ren J, Li JP, Wang DX, Wang SN, Shi LY, Li CY. Injectable hydrogel made from antler mesenchyme matrix for regenerative wound healing via creating a fetal-like niche. World J Stem Cells 2023; 15:768-780. [PMID: 37545751 PMCID: PMC10401419 DOI: 10.4252/wjsc.v15.i7.768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/09/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Scar formation and loss of cutaneous appendages are the greatest challenges in cutaneous wound healing. Previous studies have indicated that antler reserve mesenchyme (RM) cells and their conditioned medium improved regenerative wound healing with partial recovery of cutaneous appendages. AIM To develop hydrogels from the antler RM matrix (HARM) and evaluate the effect on wound healing. METHODS We prepared the hydrogels from the HARM via enzymatic solubilization with pepsin. Then we investigated the therapeutic effects of HARM on a full-thickness cutaneous wound healing rat model using both local injections surrounding the wound and topical wound application. RESULTS The results showed that HARM accelerated wound healing rate and reduced scar formation. Also, HARM stimulated the regeneration of cutaneous appendages and blood vessels, and reduced collagen fiber aggregation. Further study showed that these functions might be achieved via creating a fetal-like niche at the wound site. The levels of fetal wound healing-related genes, including Collagen III and TGFβ3 treated with HARM were all increased, while the expression levels of Collagen I, TGFβ1, and Engrailed 1 were decreased in the healing. Moreover, the number of stem cells was increased in the fetal-like niche created by HARM, which may contribute to the regeneration of cutaneous appendages. CONCLUSION Overall, we successfully developed an injectable hydrogel made from antler RM matrix for the regenerative repair of full-thickness cutaneous wounds. We uncovered the molecular mechanism of the hydrogels in promoting regenerative wound healing, and thus pave the way for HARM to be developed for the clinic use.
Collapse
Affiliation(s)
- Guo-Kun Zhang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
| | - Jing Ren
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, Jilin Province, China
| | - Ji-Ping Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
| | - Dong-Xu Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
| | - Sheng-Nan Wang
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
| | - Li-Yan Shi
- China-Japan Union Hospital, Jilin University, Changchun 130033, Jilin Province, China
| | - Chun-Yi Li
- Institute of Antler Science and Product Technology, Changchun Sci-Tech University, Changchun 130600, Jilin Province, China
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, Jilin Province, China.
| |
Collapse
|
21
|
Baptista LS, Silva KR, Jobeili L, Guillot L, Sigaudo-Roussel D. Unraveling White Adipose Tissue Heterogeneity and Obesity by Adipose Stem/Stromal Cell Biology and 3D Culture Models. Cells 2023; 12:1583. [PMID: 37371053 PMCID: PMC10296800 DOI: 10.3390/cells12121583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/30/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
The immune and endocrine dysfunctions of white adipose tissue are a hallmark of metabolic disorders such as obesity and type 2 diabetes. In humans, white adipose tissue comprises distinct depots broadly distributed under the skin (hypodermis) and as internal depots (visceral). Depot-specific ASCs could account for visceral and subcutaneous adipose tissue properties, by regulating adipogenesis and immunomodulation. More importantly, visceral and subcutaneous depots account for distinct contributions to obesity and its metabolic comorbidities. Recently, distinct ASCs subpopulations were also described in subcutaneous adipose tissue. Interestingly, the superficial layer closer to the dermis shows hyperplastic and angiogenic capacities, whereas the deep layer is considered as having inflammatory properties similar to visceral. The aim of this focus review is to bring the light of recent discoveries into white adipose tissue heterogeneity together with the biology of distinct ASCs subpopulations and to explore adipose tissue 3D models revealing their advantages, disadvantages, and contributions to elucidate the role of ASCs in obesity development. Recent advances in adipose tissue organoids opened an avenue of possibilities to recreate the main cellular and molecular events of obesity leading to a deep understanding of this inflammatory disease besides contributing to drug discovery. Furthermore, 3D organ-on-a-chip will add reproducibility to these adipose tissue models contributing to their translation to the pharmaceutical industry.
Collapse
Affiliation(s)
- Leandra S. Baptista
- Numpex-bio, Campus UFRJ Duque de Caxias Prof Geraldo Cidade, Universidade Federal do Rio de Janeiro, Rio de Janeiro 25240005, Brazil
| | - Karina R. Silva
- Laboratory of Stem Cell Research, Histology and Embryology Department, Biology Institute, State University of Rio de Janeiro, Rio de Janeiro 20550900, Brazil;
- Teaching and Research Division, National Institute of Traumatology and Orthopedics, Rio de Janeiro 20940070, Brazil
| | - Lara Jobeili
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
| | - Lucile Guillot
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
- Urgo Research Innovation and Development, 21300 Chenôve, France
| | - Dominique Sigaudo-Roussel
- Laboratory of Tissue Biology and Therapeutic Engineering, University of Lyon, Claude Bernard University Lyon 1, CNRS, LBTI UMR 5305, 69367 Lyon, France; (L.J.); (L.G.); (D.S.-R.)
| |
Collapse
|
22
|
Karanfil AS, Louis F, Matsusaki M. Biofabrication of vascularized adipose tissues and their biomedical applications. MATERIALS HORIZONS 2023; 10:1539-1558. [PMID: 36789675 DOI: 10.1039/d2mh01391f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in adipose tissue engineering and cell biology have led to the development of innovative therapeutic strategies in regenerative medicine for adipose tissue reconstruction. To date, the many in vitro and in vivo models developed for vascularized adipose tissue engineering cover a wide range of research areas, including studies with cells of various origins and types, polymeric scaffolds of natural and synthetic derivation, models presented using decellularized tissues, and scaffold-free approaches. In this review, studies on adipose tissue types with different functions, characteristics and body locations have been summarized with 3D in vitro fabrication approaches. The reason for the particular focus on vascularized adipose tissue models is that current liposuction and fat transplantation methods are unsuitable for adipose tissue reconstruction as the lack of blood vessels results in inadequate nutrient and oxygen delivery, leading to necrosis in situ. In the first part of this paper, current studies and applications of white and brown adipose tissues are presented according to the polymeric materials used, focusing on the studies which could show vasculature in vitro and after in vivo implantation, and then the research on adipose tissue fabrication and applications are explained.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
23
|
Wang X, Ma Y, Lu F, Chang Q. The diversified hydrogels for biomedical applications and their imperative roles in tissue regeneration. Biomater Sci 2023; 11:2639-2660. [PMID: 36790251 DOI: 10.1039/d2bm01486f] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Repair and regeneration of tissues after injury are complex pathophysiological processes. Microbial infection, malnutrition, and an ischemic and hypoxic microenvironment in the injured area can impede the typical healing cascade. Distinguished by biomimicry of the extracellular matrix, high aqueous content, and diverse functions, hydrogels have revolutionized clinical practices in tissue regeneration owing to their outstanding hydrophilicity, biocompatibility, and biodegradability. Various hydrogels such as smart hydrogels, nanocomposite hydrogels, and acellular matrix hydrogels are widely used for applications ranging from bench-scale to an industrial scale. In this review, some emerging hydrogels in the biomedical field are briefly discussed. The protective roles of hydrogels in wound dressings and their diverse biological effects on multiple tissues such as bone, cartilage, nerve, muscle, and adipose tissue are also discussed. The vehicle functions of hydrogels for chemicals and cell payloads are detailed. Additionally, this review emphasizes the particular characteristics of hydrogel products that promote tissue repair and reconstruction such as anti-infection, inflammation regulation, and angiogenesis.
Collapse
Affiliation(s)
- Xinhui Wang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Yuan Ma
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 510515, China.
| |
Collapse
|
24
|
Ziegler ME, Sorensen AM, Banyard DA, Sayadi LR, Chnari E, Hatch MM, Tassey J, Mirzakhanyan Y, Gershon PD, Hughes CC, Evans GR, Widgerow AD. Deconstructing Allograft Adipose and Fascia Matrix: Fascia Matrix Improves Angiogenesis, Volume Retention, and Adipogenesis in a Rodent Model. Plast Reconstr Surg 2023; 151:108-117. [PMID: 36219861 PMCID: PMC10081826 DOI: 10.1097/prs.0000000000009794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Autologous fat grafting is commonly used for soft-tissue repair (approximately 90,000 cases per year in the United States), but outcomes are limited by volume loss (20% to 80%) over time. Human allograft adipose matrix (AAM) stimulates de novo adipogenesis in vivo, but retention requires optimization. The extracellular matrix derived from superficial fascia, interstitial within the adipose layer, is typically removed during AAM processing. Thus, fascia, which contains numerous important proteins, might cooperate with AAM to stimulate de novo adipogenesis, improving long-term retention compared to AAM alone. METHODS Human AAM and fascia matrix proteins (back and upper leg regions) were identified by mass spectrometry and annotated by gene ontology. A three-dimensional in vitro angiogenesis assay was performed. Finally, AAM and/or fascia (1 mL) was implanted into 6- to 8-week-old male Fischer rats. After 8 weeks, the authors assessed graft retention by gas pycnometry and angiogenesis (CD31) and adipocyte counts (hematoxylin and eosin) histologically. RESULTS Gene ontology annotation revealed an angiogenic enrichment pattern unique to the fascia, including lactadherin, collagen alpha-3(V) chain, and tenascin-C. In vitro, AAM stimulated 1.0 ± 0.17 angiogenic sprouts per bead. The addition of fascia matrix increased sprouting by 88% (2.0 ± 0.12; P < 0.001). A similar angiogenic response (CD31) was observed in vivo. Graft retention volume was 25% (0.25 ± 0.13) for AAM, significantly increasing to 60% (0.60 ± 0.14) for AAM/fascia ( P < 0.05). De novo adipogenesis was 12% (12.4 ± 7.4) for AAM, significantly increasing to 51% (51.2 ± 8.0) for AAM/fascia ( P < 0.001) by means of adipocyte quantification. CONCLUSIONS Combining fascia matrix with AAM improves angiogenesis and adipogenesis compared to AAM alone in rats. These preliminary in vitro and pilot animal studies should be further validated before definitive clinical adoption. CLINICAL RELEVANCE STATEMENT When producing an off-the-shelf adipose inducing product by adding a connective tissue fascial component (that is normally discarded) to the mix of adipose matrix, vasculogenesis is increased and, thus, adipogenesis and graft survival is improved. This is a significant advance in this line of product.
Collapse
Affiliation(s)
- Mary E. Ziegler
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| | | | - Derek A. Banyard
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| | - Lohrasb R. Sayadi
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| | | | - Michaela M. Hatch
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, UC Irvine, USA
| | - Jade Tassey
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| | - Yeva Mirzakhanyan
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, UC Irvine, USA
| | - Paul D. Gershon
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, UC Irvine, USA
| | - Christopher C.W. Hughes
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, UC Irvine, USA; Department of Biomedical Engineering, The Henry Samueli School of Engineering, UC Irvine, USA; The Edwards Lifesciences Center for Advanced Cardiovascular Technology, UC Irvine, USA
| | - Gregory R.D. Evans
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| | - Alan D. Widgerow
- Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA, USA
| |
Collapse
|
25
|
Louis F, Sowa Y, Irie S, Higuchi Y, Kitano S, Mazda O, Matsusaki M. Injectable Prevascularized Mature Adipose Tissues (iPAT) to Achieve Long-Term Survival in Soft Tissue Regeneration. Adv Healthc Mater 2022; 11:e2201440. [PMID: 36103662 DOI: 10.1002/adhm.202201440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 09/01/2022] [Indexed: 01/28/2023]
Abstract
Soft tissue regeneration remains a challenge in reconstructive surgery. So far, both autologous fat implantations and artificial implants methods used in clinical applications lead to various disadvantages and limited lifespan. To overcome these limitations and improve the graft volume maintenance, reproducing a mature adipose tissue already including vasculature structure before implantation can be the solution. Therefore, injectable prevascularized adipose tissues (iPAT) are made from physiological collagen microfibers mixed with human mature adipocytes, adipose-derived stem cells, and human umbilical vein endothelial cells, embedded in fibrin gel. Following murine subcutaneous implantation, the iPAT show a higher cell survival (84% ± 6% viability) and volume maintenance after 3 months (up to twice heavier) when compared to non-prevascularized balls and liposuctioned fat implanted controls. This higher survival can be explained by the greater amount of blood vessels found (up to 1.6-fold increase), with balanced host anastomosis (51% ± 1% of human/mouse lumens), also involving infiltration by the lymphatic and neural vasculature networks. Furthermore, with the cryopreservation possibility enabling their later reinjection, the iPAT technology has the merit to allow noninvasive soft tissue regeneration for long-term outcomes.
Collapse
Affiliation(s)
- Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, 565-0871, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan.,Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, 565-0871, Japan.,TOPPAN INC, Taito, Tokyo, 110-0016, Japan
| | - Yuriko Higuchi
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Shiro Kitano
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, 565-0871, Japan.,TOPPAN INC, Taito, Tokyo, 110-0016, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Suita, 565-0871, Japan.,Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Suita, 565-0871, Japan
| |
Collapse
|
26
|
Bobrova M, Safonova L, Efimov A, Lyundup A, Mozheiko N, Agapova O, Agapov I. Scaffolds Based on Silk Fibroin with Decellularized Rat Liver Microparticles: Investigation of the Structure, Biological Properties and Regenerative Potential for Skin Wound Healing. Pharmaceutics 2022; 14:2313. [PMID: 36365132 PMCID: PMC9693194 DOI: 10.3390/pharmaceutics14112313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 10/18/2023] Open
Abstract
The development of advanced biomaterials and constructs for accelerated recovery of damaged tissues is a key direction in regenerative medicine. Biocompatible scaffolds based on natural biopolymers are widely used for these tasks. Organ decellularization enables obtaining a cell-free extracellular matrix (ECM) with preserved composition and biological activity. The objectives of the present work were combining these two approaches for the development of a composite scaffold based on silk fibroin and ECM microparticles and assessing its structure, biological properties, and regenerative potential. ECM microparticles were obtained by grinding the decellularized matrix of Wistar rat liver in liquid nitrogen. Scaffolds in the form of films were prepared by the casting method. The sinuous and rough topography of the scaffold surface was assessed by the scanning probe nanotomography (SPNT) technique. The inclusion of ECM microparticles in the composition did not affect the elasticity and tensile strength of the scaffolds. The obtained scaffold was non-toxic to cells, maintained high levels of adhesion and proliferation of mouse 3T3 fibroblast and Hep-G2 cells, and showed high regenerative potential, which was studied in the experimental model of full-thickness rat skin wound healing. The wound healing was accelerated by 1.74 times in comparison with the control.
Collapse
Affiliation(s)
- Maria Bobrova
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| | - Liubov Safonova
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| | - Anton Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| | - Alexey Lyundup
- Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia
| | - Natalya Mozheiko
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| | - Olga Agapova
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Shchukinskaya Street, 123182 Moscow, Russia
| |
Collapse
|
27
|
Proteomic Analysis of Decellularized Extracellular Matrix: Achieving a Competent Biomaterial for Osteogenesis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6884370. [PMID: 36267842 PMCID: PMC9578822 DOI: 10.1155/2022/6884370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/25/2022]
Abstract
Decellularized ECMs have been used as biological scaffolds for tissue repair due to their tissue-specific biochemical and mechanical composition, poorly simulated by other materials. It is used as patches and powders, and it could be further processed via enzymatic digestion under acidic conditions using pepsin. However, part of the bioactivity is lost during the digestion process due to protein denaturation. Here, stepwise digestion was developed to prepare a competent biomaterial for osteogenesis from three different ECM sources. In addition, three different proteases were compared to evaluate the most effective digestion protocol for specific cellular processes. GAGs and peptide quantification showed that the stepwise method yielded a higher concentration of bioactive residues. Circular dichroism analysis also showed that the stepwise approach preserved the secondary structures better. The protein profiles of the digested ECMs were analyzed, and it was found to be highly diverse and tissue-specific. The digestion of ECM from pericardium produced peptides originated from 94 different proteins, followed by 48 proteins in ECM from tendon and 35 proteins in ECM from bone. In addition, digested products from pericardium ECM yielded increased proliferation and differentiation of bone marrow mesenchymal stem cells to mature osteoblasts.
Collapse
|
28
|
Rahmati S, Jalili A, Banitalebi Dehkordi M, Przedborski M. An Effective Method for Decellularization of Human Foreskin: Implications for Skin Regeneration in Small Wounds. CELL JOURNAL 2022; 24:506-514. [PMID: 36274203 PMCID: PMC9588162 DOI: 10.22074/cellj.2022.8005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Acellular matrices of different allogeneic or xenogeneic origins are widely used as structural scaffolds in regenerative medicine. The main goal of this research was to optimize a method for decellularization of foreskin for skin regeneration in small wounds. MATERIALS AND METHODS In this experimental study, the dermal layers of foreskin were divided into two sections and subjected to two different decellularization methods: the sodium dodecyl sulfate method (SDS-M), and our optimized foreskin decellularization method (OFD-M). A combination of non-ionic detergents and SDS were used to decellularize the foreskin in OFD-M. The histological, morphological, and biomechanical properties of both methods were compared. In addition, human umbilical cord mesenchymal stem cells (hucMSCs) were isolated, and the biocompatibility and recellularization of both scaffolds by hucMSC were subsequently determined. RESULTS We observed that OFD-M is an appropriate approach for successful removal of cellular components from the foreskin tissue, without physical disturbance to the acellular matrix. In comparison to SDS-M, this new bioscaffold possesses a fine network containing a high amount of collagen fibers and glycosaminoglycans (GAG) (P≤0.03), is biocompatible and harmless for hucMSC (viability 91.7%), and exhibits a relatively high tensile strength. CONCLUSION We found that the extracellular matrix (ECM) structural integrity, the main ECM components, and the mechanical properties of the foreskin are well maintained after applying the OFD-M decellularization technique, indicating that the resulting scaffold would be a suitable platform for culturing MSC for skin grafting in small wounds.
Collapse
Affiliation(s)
- Shima Rahmati
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences,
Sanandaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences,
Sanandaj, Iran,P.O.Box: 6618634683Cancer and Immunology Research CenterResearch Institute for Health DevelopmentKurdistan University of Medical SciencesSanandajIranP.O.Box: 8815713471Department of Molecular MedicineSchool of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
Emails:,
| | - Mehdi Banitalebi Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran,P.O.Box: 6618634683Cancer and Immunology Research CenterResearch Institute for Health DevelopmentKurdistan University of Medical SciencesSanandajIranP.O.Box: 8815713471Department of Molecular MedicineSchool of Advanced TechnologiesShahrekord University of Medical SciencesShahrekordIran
Emails:,
| | | |
Collapse
|
29
|
Wang D, Li Y, Ge H, Ghadban T, Reeh M, Güngör C. The Extracellular Matrix: A Key Accomplice of Cancer Stem Cell Migration, Metastasis Formation, and Drug Resistance in PDAC. Cancers (Basel) 2022; 14:cancers14163998. [PMID: 36010993 PMCID: PMC9406497 DOI: 10.3390/cancers14163998] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 12/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is rich in dense fibrotic stroma that are composed of extracellular matrix (ECM) proteins. A disruption of the balance between ECM synthesis and secretion and the altered expression of matrix remodeling enzymes lead to abnormal ECM dynamics in PDAC. This pathological ECM promotes cancer growth, survival, invasion, and alters the behavior of fibroblasts and immune cells leading to metastasis formation and chemotherapy resistance, which contribute to the high lethality of PDAC. Additionally, recent evidence highlights that ECM, as a major structural component of the tumor microenvironment, is a highly dynamic structure in which ECM proteins establish a physical and biochemical niche for cancer stem cells (CSCs). CSCs are characterized by self-renewal, tumor initiation, and resistance to chemotherapeutics. In this review, we will discuss the effects of the ECM on tumor biological behavior and its molecular impact on the fundamental signaling pathways in PDAC. We will also provide an overview of how the different ECM components are able to modulate CSCs properties and finally discuss the current and ongoing therapeutic strategies targeting the ECM. Given the many challenges facing current targeted therapies for PDAC, a better understanding of molecular events involving the interplay of ECM and CSC will be key in identifying more effective therapeutic strategies to eliminate CSCs and ultimately to improve survival in patients that are suffering from this deadly disease.
Collapse
|
30
|
Abstract
Biomaterials with the ability to self-heal and recover their structural integrity offer many advantages for applications in biomedicine. The past decade has witnessed the rapid emergence of a new class of self-healing biomaterials commonly termed injectable, or printable in the context of 3D printing. These self-healing injectable biomaterials, mostly hydrogels and other soft condensed matter based on reversible chemistry, are able to temporarily fluidize under shear stress and subsequently recover their original mechanical properties. Self-healing injectable hydrogels offer distinct advantages compared to traditional biomaterials. Most notably, they can be administered in a locally targeted and minimally invasive manner through a narrow syringe without the need for invasive surgery. Their moldability allows for a patient-specific intervention and shows great prospects for personalized medicine. Injected hydrogels can facilitate tissue regeneration in multiple ways owing to their viscoelastic and diffusive nature, ranging from simple mechanical support, spatiotemporally controlled delivery of cells or therapeutics, to local recruitment and modulation of host cells to promote tissue regeneration. Consequently, self-healing injectable hydrogels have been at the forefront of many cutting-edge tissue regeneration strategies. This study provides a critical review of the current state of self-healing injectable hydrogels for tissue regeneration. As key challenges toward further maturation of this exciting research field, we identify (i) the trade-off between the self-healing and injectability of hydrogels vs their physical stability, (ii) the lack of consensus on rheological characterization and quantitative benchmarks for self-healing injectable hydrogels, particularly regarding the capillary flow in syringes, and (iii) practical limitations regarding translation toward therapeutically effective formulations for regeneration of specific tissues. Hence, here we (i) review chemical and physical design strategies for self-healing injectable hydrogels, (ii) provide a practical guide for their rheological analysis, and (iii) showcase their applicability for regeneration of various tissues and 3D printing of complex tissues and organoids.
Collapse
Affiliation(s)
- Pascal Bertsch
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands
| | - Mani Diba
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - David J. Mooney
- John
A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts 02138, United States,Wyss
Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts 02115, United States
| | - Sander C. G. Leeuwenburgh
- Department
of Dentistry-Regenerative Biomaterials, Radboud Institute for Molecular
Life Sciences, Radboud University Medical
Center, 6525 EX Nijmegen, The Netherlands,
| |
Collapse
|
31
|
Kesim MG, Durucan C, Atila D, Keskin D, Tezcaner A. Decellularized adipose tissue matrix-coated and simvastatin-loaded hydroxyapatite microspheres for bone regeneration. Biotechnol Bioeng 2022; 119:2574-2589. [PMID: 35707929 DOI: 10.1002/bit.28154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 05/10/2022] [Accepted: 05/30/2022] [Indexed: 11/12/2022]
Abstract
Simvastatin (SIM)-loaded and human decellularized adipose tissue (DAT)-coated porous hydroxyapatite (HAp) microspheres were developed for the first time to investigate their potential on bone regeneration. Microspheres were loaded with SIM and then coated with DAT for modifying SIM release and improving their biological response. HAp microspheres were prepared by water-in-oil emulsion method using camphene (C10 H16 ) as porogen followed by camphene removal by freeze-drying and sintering at 1200°C for 3 h. Sintered HAp microspheres with an average particle size of ~400 µm were porous and spherical in shape. Microspheres were incubated with 1, 2.5, and 5 mg/ml SIM stock solutions for drug loading, and drug loading was determined as 7.5 ± 0.79, 20.41 ± 1.93, and 46.26 ± 0.29 µg SIM/mg microspheres, respectively. SIM loading increased with the increase of the initial SIM loading amount. Faster SIM release was observed in DAT-coated microspheres compared to bare counterparts. Higher SaoS-2 cell attachment and proliferation were observed on DAT-coated microspheres. Significantly higher alkaline phosphatase activity of SaoS-2 cells was observed on DAT-coated microspheres containing 0.01 mg/ml SIM than all other groups (p < 0.01). DAT-coated microspheres loaded with SIM at low doses hold promise for bone tissue engineering applications.
Collapse
Affiliation(s)
- Merve G Kesim
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey
| | - Caner Durucan
- Department of Metallurgical and Materials Engineering, Middle East Technical University, Ankara, Turkey.,Biomaterials and Tissue Engineering Center of Excellence, Middle East Technical University, Ankara, Turkey
| | - Deniz Atila
- Biomaterials and Tissue Engineering Center of Excellence, Middle East Technical University, Ankara, Turkey.,Department of Engineering Sciences, Middle East Technical University, Ankara, Turkey
| | - Dilek Keskin
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey.,Biomaterials and Tissue Engineering Center of Excellence, Middle East Technical University, Ankara, Turkey.,Department of Engineering Sciences, Middle East Technical University, Ankara, Turkey
| | - Ayşen Tezcaner
- Department of Biotechnology, Middle East Technical University, Ankara, Turkey.,Biomaterials and Tissue Engineering Center of Excellence, Middle East Technical University, Ankara, Turkey.,Department of Engineering Sciences, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
32
|
Neishabouri A, Soltani Khaboushan A, Daghigh F, Kajbafzadeh AM, Majidi Zolbin M. Decellularization in Tissue Engineering and Regenerative Medicine: Evaluation, Modification, and Application Methods. Front Bioeng Biotechnol 2022; 10:805299. [PMID: 35547166 PMCID: PMC9081537 DOI: 10.3389/fbioe.2022.805299] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 04/04/2022] [Indexed: 12/14/2022] Open
Abstract
Reproduction of different tissues using scaffolds and materials is a major element in regenerative medicine. The regeneration of whole organs with decellularized extracellular matrix (dECM) has remained a goal despite the use of these materials for different purposes. Recently, decellularization techniques have been widely used in producing scaffolds that are appropriate for regenerating damaged organs and may be able to overcome the shortage of donor organs. Decellularized ECM offers several advantages over synthetic compounds, including the preserved natural microenvironment features. Different decellularization methods have been developed, each of which is appropriate for removing cells from specific tissues under certain conditions. A variety of methods have been advanced for evaluating the decellularization process in terms of cell removal efficiency, tissue ultrastructure preservation, toxicity, biocompatibility, biodegradability, and mechanical resistance in order to enhance the efficacy of decellularization methods. Modification techniques improve the characteristics of decellularized scaffolds, making them available for the regeneration of damaged tissues. Moreover, modification of scaffolds makes them appropriate options for drug delivery, disease modeling, and improving stem cells growth and proliferation. However, considering different challenges in the way of decellularization methods and application of decellularized scaffolds, this field is constantly developing and progressively moving forward. This review has outlined recent decellularization and sterilization strategies, evaluation tests for efficient decellularization, materials processing, application, and challenges and future outlooks of decellularization in regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Afarin Neishabouri
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Pediatric Center of Excellence, Tehran University of Medical Science, Tehran, Iran
| | - Alireza Soltani Khaboushan
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Pediatric Center of Excellence, Tehran University of Medical Science, Tehran, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Daghigh
- Department of Physiology, Faculty of Medicine, Tabriz Medical Sciences, Islamic Azad University, Tabriz, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Pediatric Center of Excellence, Tehran University of Medical Science, Tehran, Iran
- *Correspondence: Masoumeh Majidi Zolbin, ; Abdol-Mohammad Kajbafzadeh,
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Pediatric Center of Excellence, Tehran University of Medical Science, Tehran, Iran
- *Correspondence: Masoumeh Majidi Zolbin, ; Abdol-Mohammad Kajbafzadeh,
| |
Collapse
|
33
|
Nellinger S, Mrsic I, Keller S, Heine S, Southan A, Bach M, Volz A, Chassé T, Kluger PJ. Cell‐derived and enzyme‐based decellularized extracellular matrix exhibit compositional and structural differences that are relevant for its use as a biomaterial. Biotechnol Bioeng 2022; 119:1142-1156. [DOI: 10.1002/bit.28047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/01/2022] [Accepted: 01/19/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Ivana Mrsic
- Institute of Physical and Theoretical Chemistry, University of Tuebingen Auf der Morgenstelle 18 72076 Tuebingen Germany
| | - Silke Keller
- 3R‐Center for In Vitro Models and Alternatives to Animal Testing, Eberhard Karls University Tübingen Österbergstr. 3 72074 Tübingen Germany
- Department for Microphysiological Systems Institute of Biomedical Engineering, Faculty of Medicine of the Eberhard Karls University Tübingen Österbergstr. 3 72074 Tübingen Germany
| | - Simon Heine
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology, University of Stuttgart Nobelstr. 12 70569 Stuttgart Germany
| | - Monika Bach
- Core Facility Hohenheim, University of Hohenheim Emil‐Wolff‐Str. 12 70599 Stuttgart Germany
| | - Ann‐Cathrin Volz
- Reutlingen Research Institute Alteburgstr. 150 72762 Reutlingen Germany
| | - Thomas Chassé
- Institute of Physical and Theoretical Chemistry, University of Tuebingen Auf der Morgenstelle 18 72076 Tuebingen Germany
| | - Petra J. Kluger
- School of Applied Chemistry, Reutlingen University Alteburgstr. 150 72762 Reutlingen Germany
| |
Collapse
|
34
|
Moffat D, Ye K, Jin S. Decellularization for the retention of tissue niches. J Tissue Eng 2022; 13:20417314221101151. [PMID: 35620656 PMCID: PMC9128068 DOI: 10.1177/20417314221101151] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/01/2022] [Indexed: 12/25/2022] Open
Abstract
Decellularization of natural tissues to produce extracellular matrix is a promising method for three-dimensional scaffolding and for understanding microenvironment of the tissue of interest. Due to the lack of a universal standard protocol for tissue decellularization, recent investigations seek to develop novel methods for whole or partial organ decellularization capable of supporting cell differentiation and implantation towards appropriate tissue regeneration. This review provides a comprehensive and updated perspective on the most recent advances in decellularization strategies for a variety of organs and tissues, highlighting techniques of chemical, physical, biological, enzymatic, or combinative-based methods to remove cellular contents from tissues. In addition, the review presents modernized approaches for improving standard decellularization protocols for numerous organ types.
Collapse
Affiliation(s)
- Deana Moffat
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
| | - Sha Jin
- Department of Biomedical Engineering, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, NY, USA
| |
Collapse
|
35
|
Yu L, Liu Y, Wu J, Wang S, Yu J, Wang W, Ye X. Genipin Cross-Linked Decellularized Nucleus Pulposus Hydrogel-Like Cell Delivery System Induces Differentiation of ADSCs and Retards Intervertebral Disc Degeneration. Front Bioeng Biotechnol 2021; 9:807883. [PMID: 35004657 PMCID: PMC8733700 DOI: 10.3389/fbioe.2021.807883] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/03/2021] [Indexed: 01/08/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is the pathological basis of disc degenerative diseases (DDD). Reduction in the number of cells and degeneration of the extracellular matrix (ECM) in the nucleus pulposus (NP) are characteristics of IDD. Bio-hydrogel combined with stem cell transplantation is a promising treatment. Injectable ECM hydrogels have good biological activity and in-situ gelatinization. However, its biomechanics and stability are insufficient to provide adequate mechanical support for intervertebral discs and to maintain the long-term differential stimulus for seeded stem cells. In our study, we developed genipin cross-linked decellularized nucleus pulposus hydrogel (GDH) as delivery system. We evaluated the mechanical properties, stability, biocompatibility, and differentiation induction of GDH cross-linked with different concentrations of genipin in vitro. The GDH-loaded adipose-derived mesenchymal stem cells (ADSCs) (GDHA) were injected into the rat degenerated coccygeal intervertebral disc. The effect of intervertebral disc regeneration in vivo was evaluated. The results showed that GDH with 0.02% of genipin had similar elastic modulus to human nucleus pulposus, good biocompatibility, and inducibility of expressing NP-related genes. In vivo studies showed that GDHA improved the survival of ADSCs and improved the intervertebral height, MRI index, and histological grading score. In conclusion, GDH, as an outstanding bio-hydrogel cell delivery system, has the therapeutic potential for retarding IDD.
Collapse
Affiliation(s)
- Lei Yu
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yi Liu
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jianxin Wu
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shuang Wang
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiangming Yu
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Weiheng Wang
- Department of Orthopedics, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiaojian Ye
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
36
|
Liu K, He Y, Yao Y, Zhang Y, Cai Z, Ru J, Zhang X, Jin X, Xu M, Li Y, Ma Q, Gao J, Lu F. Methoxy polyethylene glycol modification promotes adipogenesis by inducing the production of regulatory T cells in xenogeneic acellular adipose matrix. Mater Today Bio 2021; 12:100161. [PMID: 34870140 PMCID: PMC8626673 DOI: 10.1016/j.mtbio.2021.100161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/22/2022] Open
Abstract
Acellular adipose matrix (AAM) has emerged as an important biomaterial for adipose tissue regeneration. Current decellularization methods damage the bioactive components of the extracellular matrix (ECM), and the residual immunogenic antigens may induce adverse immune responses. Here, we adopted a modified decellularization method which can protect more bioactive components with less immune reaction by methoxy polyethylene glycol (mPEG). Then, we determined the adipogenic mechanisms of mPEG-modified AAM after xenogeneic transplantation. AAM transplantation caused significantly lesser adipogenesis in the wild-type group than in the immune-deficient group. The mPEG-modified AAM showed significantly lower immunogenicity and higher adipogenesis than the AAM alone after xenogeneic transplantation. Furthermore, mPEG modification increased regulatory T (Treg) cell numbers in the AAM grafts, which in turn enhanced the M2/M1 macrophage ratio by secreting IL-10, IL-13, and TGF-β1. These findings suggest that mPEG modification effectively reduces the immunogenicity of xenogeneic AAM and promotes adipogenesis in the AAM grafts. Hence, mPEG-modified AAM can serve as an ideal biomaterial for xenogeneic adipose tissue engineering.
Collapse
Affiliation(s)
- Kaiyang Liu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yunfan He
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yao Yao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yuchen Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Zihan Cai
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jiangjiang Ru
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiangdong Zhang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoxuan Jin
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Yibao Li
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Qizhuan Ma
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Jianhua Gao
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
37
|
Hackethal J, Dungel P, Teuschl AH. Frequently used strategies to isolate ECM proteins from human placenta and adipose tissue. Tissue Eng Part C Methods 2021; 27:649-660. [PMID: 34751590 DOI: 10.1089/ten.tec.2021.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The natural extracellular matrix (ECM) provides the optimal environment for cells. Many enzymatic or non-enzymatic based strategies to extract ECM proteins from tissues were published over the last years. However, every single isolation strategy reported so far is associated with specific bottlenecks. Experiment: In this study, frequently used strategies to isolate extracellular matrix (ECM) from human placenta or adipose tissue using Tris-, serum, or pepsin-based buffers were compared. The resulting ECM proteins were biochemically characterized by analysis of cellular remnants using HOECHST DNA staining, glycosaminoglycan (GAG) content by dimethylemethylene blue (DMMB), visualization of protein bands using SDS PAGE analysis combined with amino acid quantification and assessment of the pro-angiogenic profile using an angiogenesis array. RESULTS Tris-NaCl extracted ECM proteins showed a high heterogenic degree of extracted proteins, bioactive growth factors and GAGS, but no collagen-I. Active serum extracted ECM showed significant lower DNA remnants when compared to the Tris-NaCl isolation strategy. Pepsin-extracted ECM was rich in collagen-I and low amounts of remaining bioactive growth factors. This strategy was most effective to reduce DNA amounts when compared to the other isolation strategies. Pepsin-extracted ECM from both tissues easily gelled at 37°C, whereas the other extracted ECM strategies did not gel at 37°C (Tris-NaCl: liquid; serum: sponge). CONCLUSIONS All relevant characteristics (DNA residues, ECM diversity and bioactivity, shape) of the extracted ECM proteins highly depend on its isolation strategy and could still be optimized.
Collapse
Affiliation(s)
- Johannes Hackethal
- THT Biomaterials, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria;
| | - Peter Dungel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, 497572, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria;
| | - Andreas Herbert Teuschl
- University of Applied Sciences Technikum Wien, Department of Biochemical Engineering, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria;
| |
Collapse
|
38
|
Gong D, Yu F, Zhou M, Dong W, Yan D, Zhang S, Yan Y, Wang H, Tan Y, Chen Y, Feng B, Fu W, Fu Y, Lu Y. Ex Vivo and In Vivo Properties of an Injectable Hydrogel Derived From Acellular Ear Cartilage Extracellular Matrix. Front Bioeng Biotechnol 2021; 9:740635. [PMID: 34589475 PMCID: PMC8474061 DOI: 10.3389/fbioe.2021.740635] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023] Open
Abstract
Extracellular matrix (ECM) hydrogels provide advantages such as injectability, the ability to fill an irregularly shaped space, and the adequate bioactivity of native matrix. In this study, we developed decellularized cartilage ECM (dcECM) hydrogels from porcine ears innovatively via the main method of enzymatic digestion and verified good biocompatible properties of dcECM hydrogels to deliver chondrocytes and form subcutaneous cartilage in vivo. The scanning electron microscopy and turbidimetric gelation kinetics were used to characterize the material properties and gelation kinetics of the dcECM hydrogels. Then we evaluated the biocompatibility of hydrogels via the culture of chondrocytes in vitro. To further explore the dcECM hydrogels in vivo, grafts made from the mixture of dcECM hydrogels and chondrocytes were injected subcutaneously in nude mice for the gross and histological analysis. The structural and gelation kinetics of the dcECM hydrogels altered according to the variation in the ECM concentrations. The 10 mg/ml dcECM hydrogels could support the adhesion and proliferation of chondrocytes in vitro. In vivo, at 4 weeks after transplantation, cartilage-like tissues were detected in all groups with positive staining of toluidine blue, Safranin O, and collagen II, indicating the good gelation of dcECM hydrogels. While with the increasing concentration, the tissue engineering cartilages formed by 10 mg/ml dcECM hydrogel grafts were superior in weights, volumes, collagen, and glycosaminoglycan (GAG) content compared to the dcECM hydrogels of 1 mg/ml and 5 mg/ml. At 8 weeks after grafting, dcECM hydrogel grafts at 10 mg/ml showed very similar qualities to the control, collagen I grafts. After 12 weeks of in vivo culture, the histological analysis indicated that 10 mg/ml dcECM hydrogel grafts were similar to the normal cartilage from pig ears, which was the source tissue. In conclusion, dcECM hydrogel showed the promising potential as a tissue engineering biomaterial to improve the regeneration and heal injuries of ear cartilage.
Collapse
Affiliation(s)
- Danni Gong
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Fei Yu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Meng Zhou
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Wei Dong
- Shanghai Children's Medical Center, Department of Pediatric Cardiothoracic Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Siyi Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yan Yan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Huijing Wang
- Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Tan
- Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bei Feng
- Shanghai Children's Medical Center, Department of Pediatric Cardiothoracic Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Fu
- Shanghai Children's Medical Center, Department of Pediatric Cardiothoracic Surgery, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Fu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Yang Lu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
39
|
Correa S, Grosskopf AK, Lopez Hernandez H, Chan D, Yu AC, Stapleton LM, Appel EA. Translational Applications of Hydrogels. Chem Rev 2021; 121:11385-11457. [PMID: 33938724 PMCID: PMC8461619 DOI: 10.1021/acs.chemrev.0c01177] [Citation(s) in RCA: 463] [Impact Index Per Article: 115.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Indexed: 12/17/2022]
Abstract
Advances in hydrogel technology have unlocked unique and valuable capabilities that are being applied to a diverse set of translational applications. Hydrogels perform functions relevant to a range of biomedical purposes-they can deliver drugs or cells, regenerate hard and soft tissues, adhere to wet tissues, prevent bleeding, provide contrast during imaging, protect tissues or organs during radiotherapy, and improve the biocompatibility of medical implants. These capabilities make hydrogels useful for many distinct and pressing diseases and medical conditions and even for less conventional areas such as environmental engineering. In this review, we cover the major capabilities of hydrogels, with a focus on the novel benefits of injectable hydrogels, and how they relate to translational applications in medicine and the environment. We pay close attention to how the development of contemporary hydrogels requires extensive interdisciplinary collaboration to accomplish highly specific and complex biological tasks that range from cancer immunotherapy to tissue engineering to vaccination. We complement our discussion of preclinical and clinical development of hydrogels with mechanical design considerations needed for scaling injectable hydrogel technologies for clinical application. We anticipate that readers will gain a more complete picture of the expansive possibilities for hydrogels to make practical and impactful differences across numerous fields and biomedical applications.
Collapse
Affiliation(s)
- Santiago Correa
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Abigail K. Grosskopf
- Chemical
Engineering, Stanford University, Stanford, California 94305, United States
| | - Hector Lopez Hernandez
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Doreen Chan
- Chemistry, Stanford University, Stanford, California 94305, United States
| | - Anthony C. Yu
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
| | | | - Eric A. Appel
- Materials
Science & Engineering, Stanford University, Stanford, California 94305, United States
- Bioengineering, Stanford University, Stanford, California 94305, United States
- Pediatric
Endocrinology, Stanford University School
of Medicine, Stanford, California 94305, United States
- ChEM-H Institute, Stanford
University, Stanford, California 94305, United States
- Woods
Institute for the Environment, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
40
|
Nie JY, Zhu YZ, Wang JW, Hu X, Wang ZH, Wu S, Yi YY. Preparing Adipogenic Hydrogel with Neo-Mechanical Isolated Adipose-Derived Extracellular Vesicles for Adipose Tissue Engineering. Plast Reconstr Surg 2021; 148:212e-222e. [PMID: 34153018 DOI: 10.1097/prs.0000000000008186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Subcutaneous transplantation of decellularized adipose tissue was capable of recellularization during soft tissue repair. However, further improvements are required to promote angiogenesis and adipogenesis. Here, the authors proposed a neo-mechanical protocol to isolate adipose tissue-derived extracellular vesicles (ATEVs) through lipoaspirate as a mediator for both angiogenesis and adipogenesis, and prepared ATEV-rich decellularized adipose tissue hydrogel for adipose tissue engineering. METHODS Adipose liquid extract and lipid-devoid adipose tissue were extracted through homogenization and repeated freeze and thaw cycles. ATEVs were isolated from adipose liquid extract by ultracentrifugation. Decellularized adipose tissue hydrogel was prepared by optimized decellularization of lipid-devoid adipose tissue. The optimum dose of ATEVs for angiogenesis and adipogenesis was estimated by co-culturing with vascular endothelial cells and 3T3-L1 cells, then mixed with the hydrogel. ATEV-enriched hydrogel was injected subcutaneously into the back of severe combined immunodeficiency mice, and then subjected to supplementary injection of ATEVs on postoperative day 14. ATEV-free decellularized adipose tissue hydrogel was injected as control. The newly formed tissue samples were harvested at postoperative weeks 2, 4, and 8 and subjected to volume measurement, hematoxylin and eosin staining, and immunofluorescence (CD31 and perilipin) staining. RESULTS The optimum dose of ATEVs for promoting angiogenesis and adipogenesis was 50 μg/ml. The newly formed tissue mediated by ATEV-enriched hydrogel had increased volume well as improved angiogenesis and adipogenesis at postoperative week 4 and 8. CONCLUSION ATEV-enriched adipogenic hydrogel promotes enhanced angiogenesis and adipogenesis and could serve as a promising biomaterial for adipose tissue engineering.
Collapse
Affiliation(s)
- Jia-Ying Nie
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Yuan-Zheng Zhu
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Jiang-Wen Wang
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Xuan Hu
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Zhao-Hui Wang
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Shu Wu
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| | - Yang-Yan Yi
- From the Department of Plastic Surgery, The Second Affiliated Hospital of Nanchang University
| |
Collapse
|
41
|
Modular cell-assembled adipose matrix-derived bead foams as a mesenchymal stromal cell delivery platform for soft tissue regeneration. Biomaterials 2021; 275:120978. [PMID: 34182328 DOI: 10.1016/j.biomaterials.2021.120978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/19/2022]
Abstract
With the goal of establishing a new clinically-relevant bioscaffold format to enable the delivery of high densities of human adipose-derived stromal cells (ASCs) for applications in soft tissue regeneration, a novel "cell-assembly" method was developed to generate robust 3-D scaffolds comprised of fused networks of decellularized adipose tissue (DAT)-derived beads. In vitro studies confirmed that the assembly process was mediated by remodelling of the extracellular matrix by the seeded ASCs, which were well distributed throughout the scaffolds and remained highly viable after 8 days in culture. The ASC density, sulphated glycosaminoglycan content and scaffold stability were enhanced under culture conditions that included growth factor preconditioning. In vivo testing was performed to compare ASCs delivered within the cell-assembled DAT bead foams to an equivalent number of ASCs delivered on a previously-established pre-assembled DAT bead foam platform in a subcutaneous implant model in athymic nude mice. Scaffolds were fabricated with human ASCs engineered to stably co-express firefly luciferase and tdTomato to enable long-term cell tracking. Longitudinal bioluminescence imaging showed a significantly stronger signal associated with viable human ASCs at timepoints up to 7 days in the cell-assembled scaffolds, although both implant groups were found to retain similar densities of human ASCs at 28 days. Notably, the infiltration of CD31+ murine endothelial cells was enhanced in the cell-assembled implants at 28 days. Moreover, microcomputed tomography angiography revealed that there was a marked reduction in vascular permeability in the cell-assembled group, indicating that the developing vascular network was more stable in the new scaffold format. Overall, the novel cell-assembled DAT bead foams represent a promising platform to harness the pro-regenerative paracrine functionality of human ASCs and warrant further investigation as a clinically-translational approach for volume augmentation.
Collapse
|
42
|
Li W, Wang F, Barnett C, Wang B. A comparative study on fabrication techniques of gelable bone matrix derived from porcine tibia. J Biomed Mater Res B Appl Biomater 2021; 109:2131-2141. [PMID: 33964121 DOI: 10.1002/jbm.b.34860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/19/2021] [Accepted: 04/24/2021] [Indexed: 11/11/2022]
Abstract
Recently, several types of native tissues have been enzymatically digested to prepare hydrogels that have natural-mimic extracellular matrix (ECM) proteins, architecture, and biologic activities. However, the residual detergents and salts remaining in the hydrogel may cause some undesirable effects on compatibility, functionality, and bioactivity of the material. In this study, we enzymatically digested the demineralized and decellularized bone matrix (DDBM) and adopted two common methods that included dialysis against distilled water and acetone precipitation for sample desalting. Efficiency in salt removal, protein preservation, gelation ability, and in vivo biocompatibility and function were compared to the DDBM digest without a desalting treatment. After lyophilization, the dialyzed, precipitated, and non-desalted DDBM digests all exhibited cotton-like texture and were water-soluble; however, only the precipitated DDBM digest could be gelled. We also found that the method of acetone precipitation could effectively remove salt from the DDBM digest while preserving of multiple proteins from the native bone and internal porous structure. A total of 57 proteins were identified by mass spectrometry in the precipitated DDBM digest and the majority of these proteins are critical to overall protein assembly, scaffold structure and stability, and cell-activities. Additionally, the precipitated DDBM digest possessed enhanced biocompatibility and osteointegration in repairing a cranial bone defect in Sprague-Dawley (SD) rat. In conclusion, the soluble, biodegradable, and biocompatible natures of the precipitated DDBM digest allow its usage in bone tissue engineering as a protein carrier because of its resemblance to native bone-like protein composite and operative flexibility.
Collapse
Affiliation(s)
- Wuwei Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Dalian Medical University, Dalian, China
| | - Feilong Wang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Dalian Medical University, Dalian, China
| | - Cleon Barnett
- Department of Physical Sciences, Alabama State University, Montgomery, Alabama, USA
| | - Bo Wang
- Joint Department of Biomedical Engineering, Marquette University and Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
43
|
Frazier T, Williams C, Henderson M, Duplessis T, Rogers E, Wu X, Hamel K, Martin EC, Mohiuddin O, Shaik S, Devireddy R, Rowan BG, Hayes DJ, Gimble JM. Breast Cancer Reconstruction: Design Criteria for a Humanized Microphysiological System. Tissue Eng Part A 2021; 27:479-488. [PMID: 33528293 PMCID: PMC8196546 DOI: 10.1089/ten.tea.2020.0372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/21/2021] [Indexed: 11/12/2022] Open
Abstract
International regulatory agencies such as the Food and Drug Administration have mandated that the scientific community develop humanized microphysiological systems (MPS) as an in vitro alternative to animal models in the near future. While the breast cancer research community has long appreciated the importance of three-dimensional growth dynamics in their experimental models, there are remaining obstacles preventing a full conversion to humanized MPS for drug discovery and pathophysiological studies. This perspective evaluates the current status of human tissue-derived cells and scaffolds as building blocks for an "idealized" breast cancer MPS based on bioengineering design principles. It considers the utility of adipose tissue as a potential source of endothelial, lymphohematopoietic, and stromal cells for the support of breast cancer epithelial cells. The relative merits of potential MPS scaffolds derived from adipose tissue, blood components, and synthetic biomaterials is evaluated relative to the current "gold standard" material, Matrigel, a murine chondrosarcoma-derived basement membrane-enriched hydrogel. The advantages and limitations of a humanized breast cancer MPS are discussed in the context of in-process and destructive read-out assays. Impact statement Regulatory authorities have highlighted microphysiological systems as an emerging tool in breast cancer research. This has been led by calls for more predictive human models and reduced animal experimentation. This perspective describes how human-derived cells, extracellular matrices, and hydrogels will provide the building blocks to create breast cancer models that accurately reflect diversity at multiple levels, that is, patient ethnicity, pathophysiology, and metabolic status.
Collapse
Affiliation(s)
| | - Christopher Williams
- Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | | | - Tamika Duplessis
- Department of Physical Sciences, Delgado Community College, New Orleans, Louisiana, USA
| | - Emma Rogers
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Xiying Wu
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Katie Hamel
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Omair Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, Pakistan
| | - Shahensha Shaik
- Cell and Molecular Biology Core Laboratory, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | - Ram Devireddy
- Department of Mechanical Engineering, Louisiana State University, New Orleans, Louisiana, USA
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Daniel J. Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, Pennsylvania, USA
| | | |
Collapse
|
44
|
Louis F, Piantino M, Liu H, Kang DH, Sowa Y, Kitano S, Matsusaki M. Bioprinted Vascularized Mature Adipose Tissue with Collagen Microfibers for Soft Tissue Regeneration. CYBORG AND BIONIC SYSTEMS 2021; 2021:1412542. [PMID: 36285131 PMCID: PMC9494725 DOI: 10.34133/2021/1412542] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/06/2021] [Indexed: 12/02/2022] Open
Abstract
The development of soft tissue regeneration has recently gained importance due to safety concerns about artificial breast implants. Current autologous fat graft implantations can result in up to 90% of volume loss in long-term outcomes due to their limited revascularization. Adipose tissue has a highly vascularized structure which enables its proper homeostasis as well as its endocrine function. Mature adipocytes surrounded by a dense vascular network are the specific features required for efficient regeneration of the adipose tissue to perform host anastomosis after its implantation. Recently, bioprinting has been introduced as a promising solution to recreate in vitro this architecture in large-scale tissues. However, the in vitro induction of both the angiogenesis and adipogenesis differentiations from stem cells yields limited maturation states for these two pathways. To overcome these issues, we report a novel method for obtaining a fully vascularized adipose tissue reconstruction using supporting bath bioprinting. For the first time, directly isolated mature adipocytes encapsulated in a bioink containing physiological collagen microfibers (CMF) were bioprinted in a gellan gum supporting bath. These multilayered bioprinted tissues retained high viability even after 7 days of culture. Moreover, the functionality was also confirmed by the maintenance of fatty acid uptake from mature adipocytes. Therefore, this method of constructing fully functional adipose tissue regeneration holds promise for future clinical applications.
Collapse
Affiliation(s)
- Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Hao Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Dong-Hee Kang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
| | - Shiro Kitano
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
- Toppan Printing Co., Ltd., Tokyo, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
45
|
Yang J, Zhou C, Fu J, Yang Q, He T, Tan Q, Lv Q. In situ Adipogenesis in Biomaterials Without Cell Seeds: Current Status and Perspectives. Front Cell Dev Biol 2021; 9:647149. [PMID: 33763426 PMCID: PMC7982583 DOI: 10.3389/fcell.2021.647149] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/08/2021] [Indexed: 02/05/2023] Open
Abstract
For cosmetic and reconstructive purposes in the setting of small-volume adipose tissue damage due to aging, traumatic defects, oncological resections, and degenerative diseases, the current strategies for soft tissue replacement involve autologous fat grafts and tissue fillers with synthetic, bioactive, or tissue-engineered materials. However, they all have drawbacks such as volume shrinkage and foreign-body responses. Aiming to regenerate bioactive vascularized adipose tissue on biomaterial scaffolds, adipose tissue engineering (ATE) has emerged as a suitable substitute for soft tissue repair. The essential components of ATE include scaffolds as support, cells as raw materials for fat formation, and a tolerant local environment to allow regeneration to occur. The commonly loaded seeding cells are adipose-derived stem cells (ASCs), which are expected to induce stable and predictable adipose tissue formation. However, defects in stem cell enrichment, such as donor-site sacrifice, limit their wide application. As a promising alternative approach, cell-free bioactive scaffolds recruit endogenous cells for adipogenesis. In biomaterials without cell seeds, the key to sufficient adipogenesis relies on the recruitment of endogenous host cells and continuous induction of cell homing to scaffolds. Regeneration, rather than repair, is the fundamental dominance of an optimal mature product. To induce in situ adipogenesis, many researchers have focused on the mechanical and biochemical properties of scaffolds. In addition, efforts to regulate an angiogenic and adipogenic microenvironment in cell-free settings involve integrating growth factors or extracellular matrix (ECM) proteins onto bioactive scaffolds. Despite the theoretical feasibility and encouraging results in animal models, few of the reported cell-free biomaterials have been tested in humans, and failures of decellularized adipose tissues in adipogenesis have also been reported. In these cases, the most likely reason was the lack of supporting vasculature. This review summarizes the current status of biomaterials without cell seeds. Related mechanisms and influencing factors of in situ adipogenesis in cell-free biomaterials, dilemma in the development of biomaterials, and future perspectives are also addressed.
Collapse
Affiliation(s)
- Jiqiao Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Tumor Targeted and Immune Therapy, Clinical Research Center for Breast Disease, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Chen Zhou
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jingyang Fu
- West China School of Medicine/West China Hospital, Sichuan University, Chengdu, China
| | - Qianru Yang
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao He
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuwen Tan
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Lv
- Department of Breast Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
46
|
Abaci A, Guvendiren M. Designing Decellularized Extracellular Matrix-Based Bioinks for 3D Bioprinting. Adv Healthc Mater 2020; 9:e2000734. [PMID: 32691980 DOI: 10.1002/adhm.202000734] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/10/2020] [Indexed: 12/17/2022]
Abstract
3D bioprinting is an emerging technology to fabricate tissues and organs by precisely positioning cells into 3D structures using printable cell-laden formulations known as bioinks. Various bioinks are utilized in 3D bioprinting applications; however, developing the perfect bioink to fabricate constructs with biomimetic microenvironment and mechanical properties that are similar to native tissues is a challenging task. In recent years, decellularized extracellular matrix (dECM)-based bioinks have received an increasing attention in 3D bioprinting applications, since they are derived from native tissues and possess unique, complex tissue-specific biochemical properties. This review focuses on designing dECM-based bioinks for tissue and organ bioprinting, including commonly used decellularization and decellularized tissue characterization methods, bioink formulation and characterization, applications of dECM-based bioinks, and most recent advancements in dECM-based bioink design.
Collapse
Affiliation(s)
- Alperen Abaci
- Instructive Biomaterials and Additive Manufacturing Laboratory Otto H. York Chemical and Materials Engineering 138 York Center New Jersey Institute of Technology University Heights Newark NJ 07102 USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory Otto H. York Chemical and Materials Engineering 138 York Center New Jersey Institute of Technology University Heights Newark NJ 07102 USA
- Department of Biomedical Engineering New Jersey Institute of Technology University Heights Newark NJ 07102 USA
| |
Collapse
|
47
|
Pope BD, Warren CR, Dahl MO, Pizza CV, Henze DE, Sinatra NR, Gonzalez GM, Chang H, Liu Q, Glieberman AL, Ferrier JP, Cowan CA, Parker KK. Fattening chips: hypertrophy, feeding, and fasting of human white adipocytes in vitro. LAB ON A CHIP 2020; 20:4152-4165. [PMID: 33034335 PMCID: PMC7818847 DOI: 10.1039/d0lc00508h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Adipose is a distributed organ that performs vital endocrine and energy homeostatic functions. Hypertrophy of white adipocytes is a primary mode of both adaptive and maladaptive weight gain in animals and predicts metabolic syndrome independent of obesity. Due to the failure of conventional culture to recapitulate adipocyte hypertrophy, technology for production of adult-size adipocytes would enable applications such as in vitro testing of weight loss therapeutics. To model adaptive adipocyte hypertrophy in vitro, we designed and built fat-on-a-chip using fiber networks inspired by extracellular matrix in adipose tissue. Fiber networks extended the lifespan of differentiated adipocytes, enabling growth to adult sizes. By micropatterning preadipocytes in a native cytoarchitecture and by adjusting cell-to-cell spacing, rates of hypertrophy were controlled independent of culture time or differentiation efficiency. In vitro hypertrophy followed a nonlinear, nonexponential growth model similar to human development and elicited transcriptomic changes that increased overall similarity with primary tissue. Cells on the chip responded to simulated meals and starvation, which potentiated some adipocyte endocrine and metabolic functions. To test the utility of the platform for therapeutic development, transcriptional network analysis was performed, and retinoic acid receptors were identified as candidate drug targets. Regulation by retinoid signaling was suggested further by pharmacological modulation, where activation accelerated and inhibition slowed hypertrophy. Altogether, this work presents technology for mature adipocyte engineering, addresses the regulation of cell growth, and informs broader applications for synthetic adipose in pharmaceutical development, regenerative medicine, and cellular agriculture.
Collapse
Affiliation(s)
- Benjamin D Pope
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Curtis R Warren
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Madeleine O Dahl
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Christina V Pizza
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Douglas E Henze
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Nina R Sinatra
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Grant M Gonzalez
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Huibin Chang
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Qihan Liu
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Aaron L Glieberman
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - John P Ferrier
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA.
| | - Chad A Cowan
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Kevin Kit Parker
- Disease Biophysics Group, Wyss Institute for Biologically Inspired Engineering, Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Pierce Hall, Room 318, 29 Oxford Street, Cambridge, MA 02138, USA. and Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| |
Collapse
|
48
|
Pu W, Ren J, Chen Y, Shu J, Cui L, Han Y, Xi J, Pei X, Yue W, Han Y. Injectable human decellularized adipose tissue hydrogel containing stem cells enhances wound healing in mouse. Colloids Surf A Physicochem Eng Asp 2020. [DOI: 10.1016/j.colsurfa.2020.125268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Zhou C, Zhou L, Liu J, Xu L, Xu Z, Chen Z, Ge Y, Zhao F, Wu R, Wang X, Jiang N, Mao L, Jia R. Kidney extracellular matrix hydrogel enhances therapeutic potential of adipose-derived mesenchymal stem cells for renal ischemia reperfusion injury. Acta Biomater 2020; 115:250-263. [PMID: 32771597 DOI: 10.1016/j.actbio.2020.07.056] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
Stem cell-based therapy has been suggested as a promising option for the treatment of renal ischemia-reperfusion injury (IRI). However, how to efficiently deliver stem cells remains a challenge. In the present study, we firstly proposed the utilization of kidney extracellular matrix hydrogel (ECMH) as an injectable scaffold for delivering adipose-derived mesenchymal stem cells (ad-MSCs) into ischemic kidneys. A modified strategy of decellularization and gelation was introduced to prepare the ECMH, by which the bioactive ingredients were retained as much as possible. Bioluminescence living imaging and immunofluorescence revealed that ECMH could significantly elevate the retention and survival rate of transplanted ad-MSCs in damaged kidneys and reduce their escape rate to other organs, which consequently resulted to the enhanced therapeutic effect of ad-MSCs on renal IRI. Further, in vitro evidence demonstrated that ECMH could remarkably reduce the oxidative stress and apoptosis, promote the proliferation, secretion, and epithelial differentiation of ad-MSCs, as well as facilitate cell migration while acting as a sustained-release scaffold. This study establishes an effective approach to enhance the therapeutic potential of ad-MSCs for renal IRI. Our findings suggest that ECMH derived from organs or tissues would be a promising injectable scaffold for stem cell-based therapy. STATEMENT OF SIGNIFICANCE: It remains a challenge to efficiently deliver stem cells to target tissues, which may limit the clinical application of stem cell-based therapy. In this study, we developed a modified strategy of decellularization and gelation to prepare the kidney extracellular matrix hydrogel (ECMH). In vivo and in vitro evidence indicated that the kidney ECMH could improve the retention and survival rate, as well as multiple biological functions of adipose-derived mesenchymal stem cells, thereby contributing to the histological and functional recovery of injured kidneys induced by ischemia-reperfusion. Our findings highlight the use of organs or tissues derived ECMH as a promising stem cell delivery scaffold for tissue repair.
Collapse
Affiliation(s)
- Changcheng Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liuhua Zhou
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Jingyu Liu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Luwei Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Zheng Xu
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Zaozao Chen
- Institute of Biomaterials and Medical Devices, School of Biological Science and Medical Engineering, Southeast University, Dingjiaqiao 87, Nanjing 210009, China
| | - Yuzheng Ge
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Feng Zhao
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Ran Wu
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Xinning Wang
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Nan Jiang
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Liang Mao
- Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China
| | - Ruipeng Jia
- Department of Urology, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China; Center for Renal Transplantation, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing 210006, China.
| |
Collapse
|
50
|
Mohiuddin OA, Motherwell JM, Rogers E, Bratton MR, Zhang Q, Wang G, Bunnell B, Hayes DJ, Gimble JM. Characterization and Proteomic Analysis of Decellularized Adipose Tissue Hydrogels Derived from Lean and Overweight/Obese Human Donors. ACTA ACUST UNITED AC 2020; 4:e2000124. [PMID: 32914579 DOI: 10.1002/adbi.202000124] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/28/2020] [Indexed: 12/19/2022]
Abstract
While decellularized adipose tissue (DAT) has potential as an "off-the-shelf" biomaterial product for regenerative medicine, it remains to be determined if donor-source body mass index (BMI) impacts the functionality of DAT. This study set out to comparatively characterize lean versus overweight/obese-donor derived DAT hydrogel based on proteome and to analyze their respective effects on adipose stromal/stem cell (ASC) viability, and differentiation in vitro. Decellularized adipose tissue from lean (lDAT) and overweight/obese (oDAT) donors is produced and characterized. Variability in the fibril microstructures is found, with dense fibrotic fiber clusters and large pore area uniquely present in the oDAT samples. Proteomic analysis reveals that lDAT contains a greater proportion of enriched extracellular proteins and a smaller proportion of enriched intracellular proteins relative to oDAT. Biocompatibility studies show that ASCs cultured in lDAT and oDAT hydrogels remain viable. The adipogenic and osteogenic differentiation capability of ASCs seeded in lDAT and oDAT hydrogels is confirmed by an upregulation in marker gene expression and phenotypic analysis. In conclusion, this study establishes that DAT hydrogels derived from lean and overweight/obese adipose donors present similar physicochemical profiles with some distinctive features while comparably supporting the viability and adipogenic differentiation of ASCs in vitro.
Collapse
Affiliation(s)
- Omair A Mohiuddin
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Jessica M Motherwell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Emma Rogers
- Department of Biomedical Engineering, Tulane University, New Orleans, LA, 70112, USA
| | | | - Qiang Zhang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Guangdi Wang
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, 70125, USA
| | - Bruce Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, 16802, USA
| | - Jeffrey M Gimble
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- LaCell LLC and Obatala Sciences Inc., New Orleans, LA, 70148, USA
| |
Collapse
|